1
|
Werner CM, Willing LB, Goudsward HJ, McBride AR, Stella SL, Holmes GM. Plasticity of colonic enteric nervous system following spinal cord injury in male and female rats. Neurogastroenterol Motil 2023; 35:e14646. [PMID: 37480186 PMCID: PMC11298951 DOI: 10.1111/nmo.14646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/30/2023] [Accepted: 06/27/2023] [Indexed: 07/23/2023]
Abstract
BACKGROUND Neurogenic bowel is a dysmotility disorder following spinal cord injury (SCI) that negatively impacts quality of life, social integration, and physical health. Colonic transit is directly modulated by the enteric nervous system. Interstitial Cells of Cajal (ICC) distributed throughout the small intestine and colon serve as specialized pacemaker cells, generating rhythmic electrical slow waves within intestinal smooth muscle, or serve as an interface between smooth muscle cells and enteric motor neurons of the myenteric plexus. Interstitial Cells of Cajal loss has been reported for other preclinical models of dysmotility, and our previous experimental SCI study provided evidence of reduced excitatory and inhibitory enteric neuronal count and smooth muscle neural control. METHODS Immunohistochemistry for the ICC-specific marker c-Kit was utilized to examine neuromuscular remodeling of the distal colon in male and female rats with experimental SCI. KEY RESULTS Myenteric plexus ICC (ICC-MP) exhibited increased cell counts 3 days following SCI in male rats, but did not significantly increase in females until 3 weeks after SCI. On average, ICC-MP total primary arborization length increased significantly in male rats at 3-day, 3-week, and 6-week time points, whereas in females, this increase occurred most frequently at 6 weeks post-SCI. Conversely, circular muscle ICC (ICC-CM) did not demonstrate post-SCI changes. CONCLUSIONS AND INFERENCES These data demonstrate resiliency of the ICC-MP in neurogenic bowel following SCI, unlike seen in other related disease states. This plasticity underscores the need to further understand neuromuscular changes driving colonic dysmotility after SCI in order to advance therapeutic targets for neurogenic bowel treatment.
Collapse
Affiliation(s)
- Claire M Werner
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine, Hershey, Pennsylvania, USA
| | - Lisa B Willing
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine, Hershey, Pennsylvania, USA
| | - Hannah J Goudsward
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine, Hershey, Pennsylvania, USA
| | - Amanda R McBride
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine, Hershey, Pennsylvania, USA
| | - Salvatore L Stella
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine, Hershey, Pennsylvania, USA
| | - Gregory M Holmes
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine, Hershey, Pennsylvania, USA
| |
Collapse
|
2
|
Kim DH, Choi JY, Kim SM, Son SM, Choi SY, Koo B, Rah CS, Nam JH, Ju MJ, Lee JS, You RY, Hong SH, Lee J, Bae JW, Kim CH, Choi W, Kim HS, Xu WX, Lee SJ, Kim YC, Yun HY. Vasomotion in human arteries and their regulations based on ion channel regulations: 10 years study. J Cell Physiol 2023; 238:2076-2089. [PMID: 37672477 DOI: 10.1002/jcp.31067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 05/21/2023] [Accepted: 05/30/2023] [Indexed: 09/08/2023]
Abstract
Vasomotion is the oscillation of vascular tone which gives rise to flow motion of blood into an organ. As is well known, spontaneous contractile organs such as heart, GI, and genitourinary tract produce rhythmic contraction. It imposes or removes pressure on their vessels alternatively for exchange of many substances. It was first described over 150 years ago, however the physiological mechanism and pathophysiological implications are not well understood. This study aimed to elucidate underlying mechanisms and physiological function of vasomotion in human arteries. Conventional contractile force measurement, immunohistochemistry, and Western blot analysis were employed to study human left gastric artery (HLGA) and uterine arteries (HUA). RESULTS: Circular muscle of HLGA and/or HUA produced sustained tonic contraction by high K+ (50 mM) which was blocked by 2 µM nifedipine. Stepwise stretch and high K+ produced nerve-independent spontaneous contraction (vasomotion) (around 45% of tested tissues). Vasomotion was also produced by application of BayK 8644, 5-HT, prostagrandins, oxytocin. It was blocked by nifedipine (2 µM) and blockers of intracellular Ca2+ stores. Inhibitors of Ca2+ -activated Cl- channels (DIDS and/or niflumic acid) and ATP-sensitive K+ (KATP ) channels inhibited vasomotion reversibly. Metabolic inhibition by sodium cyanide (NaCN) and several neuropeptides also regulated vasomotion in KATP channel-sensitive and -insensitive manner. Finally, we identified TMEM16A Ca2+ -activated Cl- channels and subunits of KATP channels (Kir 6.1/6.2 and sulfonylurea receptor 2B [SUR2B]), and c-Kit positivity by Western blot analysis. We conclude that vasomotion is sensitive to TMEM16A Ca2+ -activated Cl- channels and metabolic changes in human gastric and uterine arteries. Vasomotion might play an important role in the regulation of microcirculation dynamics even in pacemaker-related autonomic contractile organs in humans.
Collapse
Affiliation(s)
- Dae Hoon Kim
- Department of Surgery, College of Medicine, Chungbuk National University Hospital (CBNUH), Chungbuk National University (CBNU), Cheongju, Chungbuk, Korea
| | - Jin Young Choi
- Department of OBGY, College of Medicine, CBNU, College of Medicine, CBNU, (CBNUH), Cheongju, Korea
| | - Su Mi Kim
- Department of OBGY, College of Medicine, CBNU, College of Medicine, CBNU, (CBNUH), Cheongju, Korea
| | - Seung-Myoung Son
- Department of Pathology, College of Medicine, CBNU, Cheongju, Korea
| | - Song-Yi Choi
- Department of Pathology, College of Medicine, Chungnam National University, Daejeon, Korea
| | - Beommo Koo
- College of Medicine, CBNU, Cheongju, Korea
| | - Cheong-Sil Rah
- Department of Surgery, Uijeongbu Eulji Medical Center, Eulji University, Uijeongbu-si, Gyeonggi-do, Korea
| | | | | | - Jong Sung Lee
- Department of Family Medicine, Korea University College of Medicine, Seoul, Korea
| | - Ra Young You
- Department of Physiology, College of Medicine, CBNU, Cheongju, Korea
| | - Seung Hwa Hong
- Department of OBGY, College of Medicine, CBNU, College of Medicine, CBNU, (CBNUH), Cheongju, Korea
| | - Junyoung Lee
- Department of Internal Medicine, College of Medicine, CBNU & CBNUH, Cheongju, Korea
| | - Jang-Whan Bae
- Department of Internal Medicine, College of Medicine, CBNU & CBNUH, Cheongju, Korea
| | - Chan Hyung Kim
- Department of Pharmacology, College of Medicine, CBNU, Cheongju, Korea
| | - Woong Choi
- Department of Pharmacology, College of Medicine, CBNU, Cheongju, Korea
| | - Hun Sik Kim
- Department of Pharmacology, College of Medicine, CBNU, Cheongju, Korea
| | - Wen-Xie Xu
- Department of Physiology, College of Medcine, Shanghai Jiaotong University, Shanghai, People's Republic of China
| | - Sang Jin Lee
- Department of Physiology, College of Medicine, CBNU, Cheongju, Korea
| | - Young Chul Kim
- Department of Physiology, College of Medicine, CBNU, Cheongju, Korea
| | - Hyo-Yung Yun
- Department of Surgery, College of Medicine, Chungbuk National University Hospital (CBNUH), Chungbuk National University (CBNU), Cheongju, Chungbuk, Korea
| |
Collapse
|
3
|
Choi S, Seo H, Lee K, Shin DH, Wu MJ, Wu W, Huang X, Zhang J, Hong C, Jun JY. Hyperpolarization-activated cyclic nucleotide-gated channels working as pacemaker channels in colonic interstitial cells of Cajal. J Cell Mol Med 2021; 26:364-374. [PMID: 34845842 PMCID: PMC8743669 DOI: 10.1111/jcmm.17087] [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] [Received: 07/08/2021] [Revised: 10/28/2021] [Accepted: 11/17/2021] [Indexed: 11/29/2022] Open
Abstract
Hyperpolarization‐activated cyclic nucleotide‐gated (HCN) channels function as pacemaker channels in spontaneously active cells. We studied the existence of HCN channels and their functional roles in the interstitial cells of Cajal (ICC) from the mouse colon using electrophysiological, immunohistochemical and molecular techniques. HCN1 and HCN3 channels were detected in anoctamin‐1 (Ca2+‐activated Cl− channel; ANO1)‐positive cells within the muscular and myenteric layers in colonic tissues. The mRNA transcripts of HCN1 and HCN3 channels were expressed in ANO1‐positive ICC. In the deletion of HCN1 and HCN3 channels in colonic ICC, the pacemaking potential frequency was reduced. Basal cellular adenylate cyclase activity was decreased by adenylate cyclase inhibitor in colonic ICC, whereas cAMP‐specific phosphodiesterase inhibitors increased it. 8‐Bromo‐cyclic AMP and rolipram increased spontaneous intracellular Ca2+ oscillations. In addition, Ca2+‐dependent adenylate cyclase 1 (AC1) mRNA was detected in colonic ICC. Sulprostone, a PGE2‐EP3 agonist, increased the pacemaking potential frequency, maximum rate of rise of resting membrane in pacemaker potentials and basal cellular adenylate cyclase activity in colonic ICC. These results indicate that HCN channels exist in colonic ICC and participate in generating pacemaking potentials. Thus, HCN channels may be therapeutic targets in disturbed colonic motility disorders.
Collapse
Affiliation(s)
- Seok Choi
- Department of Physiology, College of Medicine, Chosun University, Gwangju, Korea
| | - Hyunhyo Seo
- Department of Anatomy, Brain Science & Engineering Institute, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Kyungmin Lee
- Department of Anatomy, Brain Science & Engineering Institute, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Dong Hoon Shin
- Department of Physiology, College of Medicine, Chosun University, Gwangju, Korea
| | - Mei Jin Wu
- Department of Physiology, College of Medicine, Chosun University, Gwangju, Korea
| | - Wenhao Wu
- Department of Physiology, College of Medicine, Chosun University, Gwangju, Korea
| | - Xingyou Huang
- Department of Physiology, College of Medicine, Chosun University, Gwangju, Korea
| | - Jingwei Zhang
- Department of Physiology, College of Medicine, Chosun University, Gwangju, Korea
| | - Chansik Hong
- Department of Physiology, College of Medicine, Chosun University, Gwangju, Korea
| | - Jae Yeoul Jun
- Department of Physiology, College of Medicine, Chosun University, Gwangju, Korea
| |
Collapse
|
4
|
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
|
5
|
Hannigan KI, Bossey AP, Foulkes HJL, Drumm BT, Baker SA, Ward SM, Sanders KM, Keef KD, Cobine CA. A novel intramuscular Interstitial Cell of Cajal is a candidate for generating pacemaker activity in the mouse internal anal sphincter. Sci Rep 2020; 10:10378. [PMID: 32587396 PMCID: PMC7316801 DOI: 10.1038/s41598-020-67142-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 06/03/2020] [Indexed: 12/14/2022] Open
Abstract
The internal anal sphincter (IAS) generates phasic contractions and tone. Slow waves (SWs) produced by interstitial cells of Cajal (ICC) underlie phasic contractions in other gastrointestinal regions. SWs are also present in the IAS where only intramuscular ICC (ICC-IM) are found, however the evidence linking ICC-IM to SWs is limited. This study examined the possible relationship between ICC-IM and SWs by recording Ca2+ transients in mice expressing a genetically-encoded Ca2+-indicator in ICC (Kit-Cre-GCaMP6f). A role for L-type Ca2+ channels (CavL) and anoctamin 1 (ANO1) was tested since each is essential for SW and tone generation. Two distinct ICC-IM populations were identified. Type I cells (36% of total) displayed localised asynchronous Ca2+ transients not dependent on CavL or ANO1; properties typical of ICC-IM mediating neural responses in other gastrointestinal regions. A second novel sub-type, i.e., Type II cells (64% of total) generated rhythmic, global Ca2+ transients at the SW frequency that were synchronised with neighbouring Type II cells and were abolished following blockade of either CavL or ANO1. Thus, the spatiotemporal characteristics of Type II cells and their dependence upon CavL and ANO1 all suggest that these cells are viable candidates for the generation of SWs and tone in the IAS.
Collapse
Affiliation(s)
- Karen I Hannigan
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, 89557, USA
| | - Aaron P Bossey
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, 89557, USA
| | - Holly J L Foulkes
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, 89557, USA
| | - Bernard T Drumm
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, 89557, USA
| | - Salah A Baker
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, 89557, USA
| | - Sean M Ward
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, 89557, USA
| | - Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, 89557, USA
| | - Kathleen D Keef
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, 89557, USA
| | - Caroline A Cobine
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, 89557, USA.
| |
Collapse
|
6
|
Drumm BT, Hwang SJ, Baker SA, Ward SM, Sanders KM. Ca 2+ signalling behaviours of intramuscular interstitial cells of Cajal in the murine colon. J Physiol 2019; 597:3587-3617. [PMID: 31124144 DOI: 10.1113/jp278036] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 05/23/2019] [Indexed: 12/13/2022] Open
Abstract
KEY POINTS Colonic intramuscular interstitial cells of Cajal (ICC-IM) exhibit spontaneous Ca2+ transients manifesting as stochastic events from multiple firing sites with propagating Ca2+ waves occasionally observed. Firing of Ca2+ transients in ICC-IM is not coordinated with adjacent ICC-IM in a field of view or even with events from other firing sites within a single cell. Ca2+ transients, through activation of Ano1 channels and generation of inward current, cause net depolarization of colonic muscles. Ca2+ transients in ICC-IM rely on Ca2+ release from the endoplasmic reticulum via IP3 receptors, spatial amplification from RyRs and ongoing refilling of ER via the sarcoplasmic/endoplasmic-reticulum-Ca2+ -ATPase. ICC-IM are sustained by voltage-independent Ca2+ influx via store-operated Ca2+ entry. Some of the properties of Ca2+ in ICC-IM in the colon are similar to the behaviour of ICC located in the deep muscular plexus region of the small intestine, suggesting there are functional similarities between these classes of ICC. ABSTRACT A component of the SIP syncytium that regulates smooth muscle excitability in the colon is the intramuscular class of interstitial cells of Cajal (ICC-IM). All classes of ICC (including ICC-IM) express Ca2+ -activated Cl- channels, encoded by Ano1, and rely upon this conductance for physiological functions. Thus, Ca2+ handling in ICC is fundamental to colonic motility. We examined Ca2+ handling mechanisms in ICC-IM of murine proximal colon expressing GCaMP6f in ICC. Several Ca2+ firing sites were detected in each cell. While individual sites displayed rhythmic Ca2+ events, the overall pattern of Ca2+ transients was stochastic. No correlation was found between discrete Ca2+ firing sites in the same cell or in adjacent cells. Ca2+ transients in some cells initiated Ca2+ waves that spread along the cell at ∼100 µm s-1 . Ca2+ transients were caused by release from intracellular stores, but depended strongly on store-operated Ca2+ entry mechanisms. ICC Ca2+ transient firing regulated the resting membrane potential of colonic tissues as a specific Ano1 antagonist hyperpolarized colonic muscles by ∼10 mV. Ca2+ transient firing was independent of membrane potential and not affected by blockade of L- or T-type Ca2+ channels. Mechanisms regulating Ca2+ transients in the proximal colon displayed both similarities to and differences from the intramuscular type of ICC in the small intestine. Similarities and differences in Ca2+ release patterns might determine how ICC respond to neurotransmission in these two regions of the gastrointestinal tract.
Collapse
Affiliation(s)
- Bernard T Drumm
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, 89557, USA
| | - Sung J Hwang
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, 89557, USA
| | - Salah A Baker
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, 89557, USA
| | - Sean M Ward
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, 89557, USA
| | - Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, 89557, USA
| |
Collapse
|
7
|
Ryoo SB, Oh HK, Moon SH, Choe EK, Yu SA, Park SH, Park KJ. Electrophysiological and Mechanical Characteristics in Human Ileal Motility: Recordings of Slow Waves Conductions and Contractions, In vitro. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2015; 19:533-42. [PMID: 26557020 PMCID: PMC4637356 DOI: 10.4196/kjpp.2015.19.6.533] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 08/14/2015] [Accepted: 08/14/2015] [Indexed: 12/13/2022]
Abstract
Little human tissue data are available for slow waves and migrating motor complexes, which are the main components of small bowel motility. We investigated the electrophysiological and mechanical characteristics of human ileal motility, in vitro. Ileum was obtained from patients undergoing bowel resection. Electrophysiological microelectrode recordings for membrane potential changes and mechanical tension recordings for contraction from smooth muscle strips and ileal segments were performed. Drugs affecting the enteric nervous system were applied to measure the changes in activity. Slow waves were detected with a frequency of 9~10/min. There were no cross-sectional differences in resting membrane potential (RMP), amplitude or frequency between outer and inner circular muscle (CM), suggesting that electrical activities could be effectively transmitted from outer to inner CM. The presence of the interstitial cell of Cajal (ICC) at the linia septa was verified by immunohistochemistry. Contractions of strips and segments occurred at a frequency of 3~4/min and 1~2/min, respectively. The frequency, amplitude and area under the curve were similar between CM and LM. In segments, contractions of CM were associated with LM, but propagation varied with antegrade and retrograde directions. Atropine, NW-oxide-L-arginine, and sodium nitroprusside exhibited different effects on RMP and contractions. There were no cross-sectional differences with regard to the characteristics of slow waves in CM. The frequency of contractions in smooth muscle strips and ileal segments was lower than slow waves. The directions of propagation were diverse, indicating both mixing and transport functions of the ileum.
Collapse
Affiliation(s)
- Seung-Bum Ryoo
- Department of Surgery, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Heung-Kwon Oh
- Department of Surgery, Seoul National University College of Medicine, Seoul 03080, Korea. ; Department of Surgery, Seoul National University Bundang Hospital, Seongnam 13620, Korea
| | - Sang Hui Moon
- Department of Surgery, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Eun Kyung Choe
- Department of Surgery, Seoul National University College of Medicine, Seoul 03080, Korea. ; Healthcare Research Institute, Seoul National University Hospital Healthcare System Gangnam Center, Seoul 06236, Korea
| | - Sung A Yu
- Department of Surgery, Seoul National University College of Medicine, Seoul 03080, Korea. ; Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Sung-Hye Park
- Department of Pathology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Kyu Joo Park
- Department of Surgery, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
8
|
Radulovic M, Anand P, Korsten MA, Gong B. Targeting Ion Channels: An Important Therapeutic Implication in Gastrointestinal Dysmotility in Patients With Spinal Cord Injury. J Neurogastroenterol Motil 2015; 21:494-502. [PMID: 26424038 PMCID: PMC4622131 DOI: 10.5056/jnm15061] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2015] [Revised: 06/10/2015] [Accepted: 07/06/2015] [Indexed: 01/04/2023] Open
Abstract
Gastrointestinal (GI) dysmotility is a severe, and common complication in patients with spinal cord injury (SCI). Current therapeutic methods using acetylcholine analogs or laxative agents have unwanted side effects, besides often fail to have desired effect. Various ion channels such as ATP-sensitive potassium (KATP) channel, calcium ions (Ca2+)-activated potassium ions (K+) channels, voltage-sensitive Ca2+ channels and chloride ion (Cl−) channels are abundantly expressed in GI tissues, and play an important role in regulating GI motility. The release of neurotransmitters from the enteric nerve terminal, innervating GI interstitial cells of Cajal (ICC), and smooth muscle cells (SMC), causes inactivation of K+ and Cl− channels, increasing Ca2+ influx into cytoplasm, resulting in membrane depolarization and smooth muscle contraction. Thus, agents directly regulating ion channels activity either in ICC or in SMC may affect GI peristalsis and would be potential therapeutic target for the treatment of GI dysmotility with SCI.
Collapse
Affiliation(s)
- Miroslav Radulovic
- Department of Medicine Icahn School of Medicine at Mount Sinai, New York, NY, USA.,National Center of Excellence for the Medical Consequences of SCI, James J Peters Veteran Affairs Medical Center, Bronx, NY, USA
| | - Preeti Anand
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mark A Korsten
- Department of Medicine Icahn School of Medicine at Mount Sinai, New York, NY, USA.,National Center of Excellence for the Medical Consequences of SCI, James J Peters Veteran Affairs Medical Center, Bronx, NY, USA
| | - Bing Gong
- Department of Medicine Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
9
|
Parsons SP, Huizinga JD. Effects of gap junction inhibition on contraction waves in the murine small intestine in relation to coupled oscillator theory. Am J Physiol Gastrointest Liver Physiol 2015; 308:G287-97. [PMID: 25501550 PMCID: PMC4329477 DOI: 10.1152/ajpgi.00338.2014] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Waves of contraction in the small intestine correlate with slow waves generated by the myenteric network of interstitial cells of Cajal. Coupled oscillator theory has been used to explain steplike gradients in the frequency (frequency plateaux) of contraction waves along the length of the small intestine. Inhibition of gap junction coupling between oscillators should lead to predictable effects on these plateaux and the wave dislocation (wave drop) phenomena associated with their boundaries. It is these predictions that we wished to test. We used a novel multicamera diameter-mapping system to measure contraction along 25- to 30-cm lengths of murine small intestine. There were typically two to three plateaux per length of intestine. Dislocations could be limited to the wavefronts immediately about the terminated wave, giving the appearance of a three-pronged fork, i.e., a fork dislocation; additionally, localized decreases in velocity developed across a number of wavefronts, ending with the terminated wave, which could appear as a fork, i.e., slip dislocations. The gap junction inhibitor carbenoxolone increased the number of plateaux and dislocations and decreased contraction wave velocity. In some cases, the usual frequency gradient was reversed, with a plateau at a higher frequency than its proximal neighbor; thus fork dislocations were inverted, and the direction of propagation was reversed. Heptanol had no effect on the frequency or velocity of contractions but did reduce their amplitude. To understand intestinal motor patterns, the pacemaker network of the interstitial cells of Cajal is best evaluated as a system of coupled oscillators.
Collapse
Affiliation(s)
- Sean P. Parsons
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Jan D. Huizinga
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
10
|
Jin X, Shah S, Du X, Zhang H, Gamper N. Activation of Ca(2+) -activated Cl(-) channel ANO1 by localized Ca(2+) signals. J Physiol 2014; 594:19-30. [PMID: 25398532 PMCID: PMC4704509 DOI: 10.1113/jphysiol.2014.275107] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 11/08/2014] [Indexed: 12/13/2022] Open
Abstract
Ca2+‐activated chloride channels (CaCCs) regulate numerous physiological processes including epithelial transport, smooth muscle contraction and sensory processing. Anoctamin‐1 (ANO1, TMEM16A) is a principal CaCC subunit in many cell types, yet our understanding of the mechanisms of ANO1 activation and regulation are only beginning to emerge. Ca2+ sensitivity of ANO1 is rather low and at negative membrane potentials the channel requires several micromoles of intracellular Ca2+ for activation. However, global Ca2+ levels in cells rarely reach such levels and, therefore, there must be mechanisms that focus intracellular Ca2+ transients towards the ANO1 channels. Recent findings indeed indicate that ANO1 channels often co‐localize with sources of intracellular Ca2+ signals. Interestingly, it appears that in many cell types ANO1 is particularly tightly coupled to the Ca2+ release sites of the intracellular Ca2+ stores. Such preferential coupling may represent a general mechanism of ANO1 activation in native tissues.
Collapse
Affiliation(s)
- Xin Jin
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Sihab Shah
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Xiaona Du
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Hailin Zhang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Nikita Gamper
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK.,Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
11
|
Sanders KM, Ward SM, Koh SD. Interstitial cells: regulators of smooth muscle function. Physiol Rev 2014; 94:859-907. [PMID: 24987007 DOI: 10.1152/physrev.00037.2013] [Citation(s) in RCA: 333] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Smooth muscles are complex tissues containing a variety of cells in addition to muscle cells. Interstitial cells of mesenchymal origin interact with and form electrical connectivity with smooth muscle cells in many organs, and these cells provide important regulatory functions. For example, in the gastrointestinal tract, interstitial cells of Cajal (ICC) and PDGFRα(+) cells have been described, in detail, and represent distinct classes of cells with unique ultrastructure, molecular phenotypes, and functions. Smooth muscle cells are electrically coupled to ICC and PDGFRα(+) cells, forming an integrated unit called the SIP syncytium. SIP cells express a variety of receptors and ion channels, and conductance changes in any type of SIP cell affect the excitability and responses of the syncytium. SIP cells are known to provide pacemaker activity, propagation pathways for slow waves, transduction of inputs from motor neurons, and mechanosensitivity. Loss of interstitial cells has been associated with motor disorders of the gut. Interstitial cells are also found in a variety of other smooth muscles; however, in most cases, the physiological and pathophysiological roles for these cells have not been clearly defined. This review describes structural, functional, and molecular features of interstitial cells and discusses their contributions in determining the behaviors of smooth muscle tissues.
Collapse
Affiliation(s)
- Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Sean M Ward
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Sang Don Koh
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| |
Collapse
|
12
|
Blair PJ, Rhee PL, Sanders KM, Ward SM. The significance of interstitial cells in neurogastroenterology. J Neurogastroenterol Motil 2014; 20:294-317. [PMID: 24948131 PMCID: PMC4102150 DOI: 10.5056/jnm14060] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 06/06/2014] [Accepted: 06/07/2014] [Indexed: 12/21/2022] Open
Abstract
Smooth muscle layers of the gastrointestinal tract consist of a heterogeneous population of cells that include enteric neurons, several classes of interstitial cells of mesenchymal origin, a variety of immune cells and smooth muscle cells (SMCs). Over the last number of years the complexity of the interactions between these cell types has begun to emerge. For example, interstitial cells, consisting of both interstitial cells of Cajal (ICC) and platelet-derived growth factor receptor alpha-positive (PDGFRα(+)) cells generate pacemaker activity throughout the gastrointestinal (GI) tract and also transduce enteric motor nerve signals and mechanosensitivity to adjacent SMCs. ICC and PDGFRα(+) cells are electrically coupled to SMCs possibly via gap junctions forming a multicellular functional syncytium termed the SIP syncytium. Cells that make up the SIP syncytium are highly specialized containing unique receptors, ion channels and intracellular signaling pathways that regulate the excitability of GI muscles. The unique role of these cells in coordinating GI motility is evident by the altered motility patterns in animal models where interstitial cell networks are disrupted. Although considerable advances have been made in recent years on our understanding of the roles of these cells within the SIP syncytium, the full physiological functions of these cells and the consequences of their disruption in GI muscles have not been clearly defined. This review gives a synopsis of the history of interstitial cell discovery and highlights recent advances in structural, molecular expression and functional roles of these cells in the GI tract.
Collapse
Affiliation(s)
- Peter J Blair
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, USA; and
| | - Poong-Lyul Rhee
- Division of Gastroenterology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, USA; and
| | - Sean M Ward
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, USA; and
| |
Collapse
|
13
|
Blair PJ, Rhee PL, Sanders KM, Ward SM. The significance of interstitial cells in neurogastroenterology. J Neurogastroenterol Motil 2014. [PMID: 24948131 DOI: 10.5056/jnm140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Smooth muscle layers of the gastrointestinal tract consist of a heterogeneous population of cells that include enteric neurons, several classes of interstitial cells of mesenchymal origin, a variety of immune cells and smooth muscle cells (SMCs). Over the last number of years the complexity of the interactions between these cell types has begun to emerge. For example, interstitial cells, consisting of both interstitial cells of Cajal (ICC) and platelet-derived growth factor receptor alpha-positive (PDGFRα(+)) cells generate pacemaker activity throughout the gastrointestinal (GI) tract and also transduce enteric motor nerve signals and mechanosensitivity to adjacent SMCs. ICC and PDGFRα(+) cells are electrically coupled to SMCs possibly via gap junctions forming a multicellular functional syncytium termed the SIP syncytium. Cells that make up the SIP syncytium are highly specialized containing unique receptors, ion channels and intracellular signaling pathways that regulate the excitability of GI muscles. The unique role of these cells in coordinating GI motility is evident by the altered motility patterns in animal models where interstitial cell networks are disrupted. Although considerable advances have been made in recent years on our understanding of the roles of these cells within the SIP syncytium, the full physiological functions of these cells and the consequences of their disruption in GI muscles have not been clearly defined. This review gives a synopsis of the history of interstitial cell discovery and highlights recent advances in structural, molecular expression and functional roles of these cells in the GI tract.
Collapse
Affiliation(s)
- Peter J Blair
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, USA
| | - Poong-Lyul Rhee
- Division of Gastroenterology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, USA
| | - Sean M Ward
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, USA
| |
Collapse
|
14
|
Lees-Green R, Gibbons SJ, Farrugia G, Sneyd J, Cheng LK. Computational modeling of anoctamin 1 calcium-activated chloride channels as pacemaker channels in interstitial cells of Cajal. Am J Physiol Gastrointest Liver Physiol 2014; 306:G711-27. [PMID: 24481603 PMCID: PMC3989704 DOI: 10.1152/ajpgi.00449.2013] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Interstitial cells of Cajal (ICC) act as pacemaker cells in the gastrointestinal tract by generating electrical slow waves to regulate rhythmic smooth muscle contractions. Intrinsic Ca(2+) oscillations in ICC appear to produce the slow waves by activating pacemaker currents, currently thought to be carried by the Ca(2+)-activated Cl(-) channel anoctamin 1 (Ano1). In this article we present a novel model of small intestinal ICC pacemaker activity that incorporates store-operated Ca(2+) entry and a new model of Ano1 current. A series of simulations were carried out with the ICC model to investigate current controversies about the reversal potential of the Ano1 Cl(-) current in ICC and to predict the characteristics of the other ion channels that are necessary to generate slow waves. The model results show that Ano1 is a plausible pacemaker channel when coupled to a store-operated Ca(2+) channel but suggest that small cyclical depolarizations may still occur in ICC in Ano1 knockout mice. The results predict that voltage-dependent Ca(2+) current is likely to be negligible during the slow wave plateau phase. The model shows that the Cl(-) equilibrium potential is an important modulator of slow wave morphology, highlighting the need for a better understanding of Cl(-) dynamics in ICC.
Collapse
Affiliation(s)
- Rachel Lees-Green
- 1Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand;
| | - Simon J. Gibbons
- 2Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota; and
| | - Gianrico Farrugia
- 2Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota; and
| | - James Sneyd
- 3Department of Mathematics, University of Auckland, New Zealand; and
| | - Leo K. Cheng
- 1Auckland Bioengineering Institute, University of Auckland, Auckland, New Zealand; ,4Department of Surgery, Vanderbilt University, Nashville, Tennessee
| |
Collapse
|
15
|
Zheng H, Park KS, Koh SD, Sanders KM. Expression and function of a T-type Ca2+ conductance in interstitial cells of Cajal of the murine small intestine. Am J Physiol Cell Physiol 2014; 306:C705-13. [PMID: 24477235 DOI: 10.1152/ajpcell.00390.2013] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Interstitial cells of Cajal (ICC) generate slow waves in gastrointestinal (GI) muscles. Previous studies have suggested that slow wave generation and propagation depends on a voltage-dependent Ca(2+) entry mechanism with the signature of a T-type Ca(2+) conductance. We studied voltage-dependent inward currents in isolated ICC. ICC displayed two phases of inward current upon depolarization: a low voltage-activated inward current and a high voltage-activated current. The latter was of smaller current density and blocked by nicardipine. Ni(2+) (30 μM) or mibefradil (1 μM) blocked the low voltage-activated current. Replacement of extracellular Ca(2+) with Ba(2+) did not affect the current, suggesting that either charge carrier was equally permeable. Half-activation and half-inactivation occurred at -36 and -59 mV, respectively. Temperature sensitivity of the Ca(2+) current was also characterized. Increasing temperature (20-30°C) augmented peak current from -7 to -19 pA and decreased the activation time from 20.6 to 7.5 ms [temperature coefficient (Q10) = 3.0]. Molecular studies showed expression of Cacna1g (Cav3.1) and Cacna1h (Cav3.2) in ICC. The temperature dependence of slow waves in intact jejunal muscles of wild-type and Cacna1h(-/-) mice was tested. Reducing temperature decreased the upstroke velocity significantly. Upstroke velocity was also reduced in muscles of Cacna1h(-/-) mice, and Ni(2+) or reduced temperature had little effect on these muscles. Our data show that a T-type conductance is expressed and functional in ICC. With previous studies our data suggest that T-type current is required for entrainment of pacemaker activity within ICC and for active propagation of slow waves in ICC networks.
Collapse
Affiliation(s)
- Haifeng Zheng
- Department of Physiology and Cell Biology, University of Nevada, School of Medicine, Reno, Nevada; and
| | | | | | | |
Collapse
|
16
|
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: 50] [Impact Index Per Article: 3.8] [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
|
17
|
Lowie BJ, Wang XY, White EJ, Huizinga JD. On the origin of rhythmic calcium transients in the ICC-MP of the mouse small intestine. Am J Physiol Gastrointest Liver Physiol 2011; 301:G835-45. [PMID: 21836058 DOI: 10.1152/ajpgi.00077.2011] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Interstitial cells of Cajal associated with the myenteric plexus (ICC-MP) are pacemaker cells of the small intestine, producing the characteristic omnipresent electrical slow waves, which orchestrate peristaltic motor activity and are associated with rhythmic intracellular calcium oscillations. Our objective was to elucidate the origins of the calcium transients. We hypothesized that calcium oscillations in the ICC-MP are primarily regulated by the sarcoplasmic reticulum (SR) calcium release system. With the use of calcium imaging, study of the effect of T-type calcium channel blocker mibefradil revealed that T-type channels did not play a major role in generating the calcium transients. 2-Aminoethoxydiphenyl borate, an inositol 1,4,5 trisphosphate receptor (IP(3)R) inhibitor, and U73122, a phospholipase C inhibitor, both drastically decreased the frequency of calcium oscillations, suggesting a major role of IP(3) and IP(3)-induced calcium release from the SR. Immunohistochemistry proved the expression of IP(3)R type I (IP(3)R-I), but not type II (IP(3)R-II) and type III (IP(3)R-III) in ICC-MP, indicating the involvement of the IP(3)R-I subtype in calcium release from the SR. Cyclopiazonic acid, a SR/endoplasmic reticulum calcium ATPase pump inhibitor, strongly reduced or abolished calcium oscillations. The Na-Ca exchanger (NCX) in reverse mode is likely involved in refilling the SR because the NCX inhibitor KB-R7943 markedly reduced the frequency of calcium oscillations. Immunohistochemistry revealed 100% colocalization of NCX and c-Kit in ICC-MP. Testing a mitochondrial NCX inhibitor, we were unable to show an essential role for mitochondria in regulating calcium oscillations in the ICC-MP. In summary, ongoing IP(3) synthesis and IP(3)-induced calcium release from the SR, via the IP(3)R-I, are the major drivers of the calcium transients associated with ICC pacemaker activity. This suggests that a biochemical clock intrinsic to ICC determines the pacemaker frequency, which is likely directly linked to kinetics of the IP(3)-activated SR calcium channel and IP(3) metabolism.
Collapse
Affiliation(s)
- Bobbi-Jo Lowie
- Farncombe Family Digestive Health Research Institute, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | | | | | | |
Collapse
|
18
|
Serotonin augments gut pacemaker activity via 5-HT3 receptors. PLoS One 2011; 6:e24928. [PMID: 21949791 PMCID: PMC3174222 DOI: 10.1371/journal.pone.0024928] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Accepted: 08/19/2011] [Indexed: 12/21/2022] Open
Abstract
Serotonin (5-hydroxytryptamine: 5-HT) affects numerous functions in the gut, such as secretion, muscle contraction, and enteric nervous activity, and therefore to clarify details of 5-HT's actions leads to good therapeutic strategies for gut functional disorders. The role of interstitial cells of Cajal (ICC), as pacemaker cells, has been recognised relatively recently. We thus investigated 5-HT actions on ICC pacemaker activity. Muscle preparations with myenteric plexus were isolated from the murine ileum. Spatio-temporal measurements of intracellular Ca(2+) and electric activities in ICC were performed by employing fluorescent Ca(2+) imaging and microelectrode array (MEA) systems, respectively. Dihydropyridine (DHP) Ca(2+) antagonists and tetrodotoxin (TTX) were applied to suppress smooth muscle and nerve activities, respectively. 5-HT significantly enhanced spontaneous Ca(2+) oscillations that are considered to underlie electric pacemaker activity in ICC. LY-278584, a 5-HT(3) receptor antagonist suppressed spontaneous Ca(2+) activity in ICC, while 2-methylserotonin (2-Me-5-HT), a 5-HT(3) receptor agonist, restored it. GR113808, a selective antagonist for 5-HT(4), and O-methyl-5-HT (O-Me-5-HT), a non-selective 5-HT receptor agonist lacking affinity for 5-HT(3) receptors, had little effect on ICC Ca(2+) activity. In MEA measurements of ICC electric activity, 5-HT and 2-Me-5-HT caused excitatory effects. RT-PCR and immunostaining confirmed expression of 5-HT(3) receptors in ICC. The results indicate that 5-HT augments ICC pacemaker activity via 5-HT(3) receptors. ICC appear to be a promising target for treatment of functional motility disorders of the gut, for example, irritable bowel syndrome.
Collapse
|
19
|
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: 36] [Impact Index Per Article: 2.6] [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
|
20
|
Means SA, Sneyd J. Spatio-temporal calcium dynamics in pacemaking units of the interstitial cells of Cajal. J Theor Biol 2010; 267:137-52. [PMID: 20705074 DOI: 10.1016/j.jtbi.2010.08.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2010] [Revised: 08/05/2010] [Accepted: 08/05/2010] [Indexed: 01/17/2023]
Abstract
The interstitial cells of Cajal (ICC) are responsible for producing pacemaking signals that stimulate rhythmic contractions in the gastro-intestinal system. The pacemaking signals are generated by membrane depolarizations, which are in turn linked to the integrated transport of calcium between the endoplasmic reticulum (ER), through inositol-trisphosphate receptor (IP(3)R) release, and mitochondria, through the uniporter. A non-specific cation channel (NSCC) is associated with the membrane depolarizations, and is inhibited by intracellular calcium. One theory proposes that the integrated calcium transport occurs within specific regions of the ICC called "pacemaker units," and results in localized calcium concentration reductions within these units, which in turn activate the NSCC and depolarize the membrane. We have constructed a model of the spatio-temporal calcium dynamics within an ICC pacemaker unit to determine under what conditions the local calcium concentrations may reduce below baseline. We obtain reductions of calcium concentrations below baseline but only under certain conditions. Without strong and persistent stimulation of the IP(3)R, reductions of calcium below baseline occur only with a non-physiological, time-dependent uniporter. Alternatively, sufficient IP(3)R release leads to reductions of calcium below baseline, due to depletion of the ER calcium store over the time scale of seconds, although these reductions require strong mitochondrial and ER calcium uptake.
Collapse
Affiliation(s)
- Shawn A Means
- Department of Mathematics, University of Auckland, Private Bag 92019, Auckland, New Zealand.
| | | |
Collapse
|
21
|
Fintl C, Hudson NPH. The interstitial cells of Cajal of the equine gastrointestinal tract: what we know so far. Equine Vet J 2010; 42:372-7. [PMID: 20525058 DOI: 10.1111/j.2042-3306.2010.00073.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: 12/19/2022]
Abstract
Gastrointestinal motility disorders are a serious problem in both veterinary and human medicine and may represent a dysfunction of the neural, muscular or pacemaker components (interstitial cells of Cajal) of bowel control. The interstitial cells of Cajal are considered to be the pacemakers and mediators of certain forms of neurotransmission in the gastrointestinal tract. These cells have been implicated, either primarily or secondarily, in the pathogenesis of gastrointestinal disease processes in which there is a prominent element of disturbance to intestinal motility. In the horse, their involvement has been implicated in large intestinal obstructive colic and grass sickness (equine dysautonomia). This review highlights the properties of the interstitial cells of Cajal and the role these cells play in orchestrating gastrointestinal motility patterns. In addition, it examines their role in intestinal motility disorders and summarises our current understanding of their importance in the equine gastrointestinal tract.
Collapse
Affiliation(s)
- C Fintl
- Norwegian School of Veterinary Science, Department of Companion Animal Clinical Sciences, PO Box 8146 Dep., 0033 Oslo, Norway
| | | |
Collapse
|
22
|
Li S, Chen JDZ. Cellular effects of gastric electrical stimulation on antral smooth muscle cells in rats. Am J Physiol Regul Integr Comp Physiol 2010; 298:R1580-7. [DOI: 10.1152/ajpregu.00024.2010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The cellular effects of gastric electrical stimulation (GES), which has recently been introduced as a potential therapy for the treatment of gastroparesis and obesity, were investigated in rat antrum smooth muscle cells (SMCs). Effects on cell membrane potentials of single electrical current pulses (pulse width from 0.1 ms to 200 ms) and 2-s pulse train stimuli with different pulse widths (0.1–4 ms), different frequencies (20–200 Hz), and different intensities were studied: 1) the stimulus amplitude had an exponential relationship to the pulse width from 2 ms to 200 ms, along with a rapidly rising strength-duration curve at pulse widths less than 5 ms, and a relatively flat curve at pulse widths greater than 50 ms; 2) when the pulse frequency was at 80 Hz or above, pulse train electrical stimulation, with a pulse width of 2 ms or above but not ≤1 ms, was able to depolarize cell membrane potentials to above −30 mV and/or generate action potentials. Electrical stimulation with a single long pulse and a width of 50 ms or greater is effective in depolarizing cell membrane potentials of SMCs with low amplitude. Pulse train electrical stimulation with a pulse width of ≤1 ms fails to generate action potentials in SMCs, whereas pulse train electrical stimulation with a pulse width of 2–4 ms and a sufficiently high pulse frequency is able to generate action potentials. These cellular findings may be useful in optimizing stimulation parameters of GES.
Collapse
Affiliation(s)
- Shiying Li
- Veterans Research and Education Foundation, Veterans Affairs Medical Center, Oklahoma City, Oklahoma; and
| | - Jiande D. Z. Chen
- Veterans Research and Education Foundation, Veterans Affairs Medical Center, Oklahoma City, Oklahoma; and
- Division of Gastroenterology, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
23
|
Takaki M, Suzuki H, Nakayama S. Recent advances in studies of spontaneous activity in smooth muscle: ubiquitous pacemaker cells. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2010; 102:129-35. [PMID: 20553741 DOI: 10.1016/j.pbiomolbio.2010.05.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2010] [Accepted: 05/19/2010] [Indexed: 02/08/2023]
Abstract
The general and specific properties of pacemaker cells, including Kit-negative cells, that are distributed in gastrointestinal, urethral and uterine smooth muscle tissues, are discussed herein. In intestinal tissues, interstitial cells of Cajal (ICC) are heterogeneous in both their forms and roles. ICC distributed in the myenteric layer (ICC-MY) act as primary pacemaker cells for intestinal mechanical and electrical activity. ICC distributed in muscle bundles play a role as mediators of signals from autonomic nerves to smooth muscle cells. A group of ICC also appears to act as a stretch sensor. Intracellular Ca2+ dynamics play a crucial role in ICC-MY pacemaking; intracellular Ca2+ ([Ca2+](i)) oscillations periodically activate plasmalemmal Ca2+-activated ion channels, such as Ca2+-activated Cl(-) channels and/or non-selective cation channels, although the relative contributions of these channels are not defined. With respect to gut motility, both the ICC network and enteric nervous system, including excitatory and inhibitory enteric neurons, play an essential role in producing highly coordinated peristalsis.
Collapse
Affiliation(s)
- Miyako Takaki
- Department of Physiology II, Nara Medical University, 840 Shijo-cho, Kashihara 634-8521, Japan.
| | | | | |
Collapse
|
24
|
Strege PR, Sha L, Beyder A, Bernard CE, Perez-Reyes E, Evangelista S, Gibbons SJ, Szurszewski JH, Farrugia G. T-type Ca(2+) channel modulation by otilonium bromide. Am J Physiol Gastrointest Liver Physiol 2010; 298:G706-13. [PMID: 20203058 PMCID: PMC2867415 DOI: 10.1152/ajpgi.00437.2009] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Antispasmodics are used clinically to treat a variety of gastrointestinal disorders by inhibition of smooth muscle contraction. The main pathway for smooth muscle Ca(2+) entry is through L-type channels; however, there is increasing evidence that T-type Ca(2+) channels also play a role in regulating contractility. Otilonium bromide, an antispasmodic, has previously been shown to inhibit L-type Ca(2+) channels and colonic contractile activity. The objective of this study was to determine whether otilonium bromide also inhibits T-type Ca(2+) channels. Whole cell currents were recorded by patch-clamp technique from HEK293 cells transfected with cDNAs encoding the T-type Ca(2+) channels, Ca(V)3.1 (alpha1G), Ca(V)3.2 (alpha1H), or Ca(V)3.3 (alpha1I) alpha subunits. Extracellular solution was exchanged with otilonium bromide (10(-8) to 10(-5) M). Otilonium bromide reversibly blocked all T-type Ca(2+) channels with a significantly greater affinity for Ca(V)3.3 than Ca(V)3.1 or Ca(V)3.2. Additionally, the drug slowed inactivation in Ca(V)3.1 and Ca(V)3.3. Inhibition of T-type Ca(2+) channels may contribute to inhibition of contractility by otilonium bromide. This may represent a new mechanism of action for antispasmodics and may contribute to the observed increased clinical effectiveness of antispasmodics compared with selective L-type Ca(2+) channel blockers.
Collapse
Affiliation(s)
- Peter R. Strege
- 1Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota;
| | - Lei Sha
- 1Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota;
| | - Arthur Beyder
- 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;
| | - Edward Perez-Reyes
- 2Department of Pharmacology, University of Virginia, Charlottesville, Virginia; and
| | | | - Simon J. Gibbons
- 1Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota;
| | - Joseph H. Szurszewski
- 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
|
25
|
Abstract
Interstitial cells of Cajal (ICC) are the pacemakers of the gut, initiating slow-wave activity. Several ion channels have been identified that contribute to the depolarization phase of the slow wave. Our aim was to contribute to knowledge about the identity and role of ICC potassium channels in pacemaking. Here we describe a transient outward potassium current in cell-attached patches of ICC. This current was activated almost instantaneously at potentials positive of the resting membrane potential and inactivated as a single exponential or biexponential with time constants that varied widely from patch to patch. Averaged traces gave a biexponential inactivation with time constants of approximately 40 and approximately 500 ms, with no clear voltage dependence. Analysis of single-channel openings and closings indicated a channel conductance of 5 pS and permeability sequence of K(+) (111) > Na(+) (1) > N-methyl-d-glucamine(+) (0.11). The current was completely blocked by 20 microM clotrimazole but was unaffected by 20 microM ketoconazole, 10 microM E4031, or 20 microM clofilium; 5 mM 4-aminopyridine slowed the activation of the current. The transient outward current may be important in moderating the upstroke of the pacemaker potential.
Collapse
Affiliation(s)
- Sean P Parsons
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | | |
Collapse
|
26
|
Park SY, Je HD, Shim JH, Sohn UD. Characteristics of spontaneous contraction in the circular smooth muscles of cat ileum. Arch Pharm Res 2010; 33:159-65. [DOI: 10.1007/s12272-010-2238-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2009] [Revised: 10/27/2009] [Accepted: 11/16/2009] [Indexed: 12/01/2022]
|
27
|
Wang ZY, Xu WX. Advances in research on pacemaking function of gastrointestinal smooth muscle cells. Shijie Huaren Xiaohua Zazhi 2010; 18:319-323. [DOI: 10.11569/wcjd.v18.i4.319] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Gastrointestinal smooth muscle has spontaneous contractile activity, which is very important to digestion and absorption function. However, the pacemaking mechanism in gastrointestinal smooth muscle is still not clear. In this article, we review the recent advances in research on the mechanisms underlying gastrointestinal pacemaker activity. We summarize the classification, function and pacemaking mechanisms of pacemaker cells, and the relationship between pacemaker cells and gastrointestinal motility dysfunction. As abnormal pacemaking activity is often associated with gastrointestinal motility dysfunction, it is of great clinical significance to clarify the pacemaking mechanisms in the gastrointestinal tract.
Collapse
|
28
|
Biophysically based mathematical modeling of interstitial cells of Cajal slow wave activity generated from a discrete unitary potential basis. Biophys J 2009; 96:4834-52. [PMID: 19527643 DOI: 10.1016/j.bpj.2009.03.058] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2008] [Revised: 02/26/2009] [Accepted: 03/26/2009] [Indexed: 11/23/2022] Open
Abstract
Spontaneously rhythmic pacemaker activity produced by interstitial cells of Cajal (ICC) is the result of the entrainment of unitary potential depolarizations generated at intracellular sites termed pacemaker units. In this study, we present a mathematical modeling framework that quantitatively represents the transmembrane ion flows and intracellular Ca2+ dynamics from a single ICC operating over the physiological membrane potential range. The mathematical model presented here extends our recently developed biophysically based pacemaker unit modeling framework by including mechanisms necessary for coordinating unitary potential events, such as a T-Type Ca2+ current, Vm-dependent K+ currents, and global Ca2+ diffusion. Model simulations produce spontaneously rhythmic slow wave depolarizations with an amplitude of 65 mV at a frequency of 17.4 cpm. Our model predicts that activity at the spatial scale of the pacemaker unit is fundamental for ICC slow wave generation, and Ca2+ influx from activation of the T-Type Ca2+ current is required for unitary potential entrainment. These results suggest that intracellular Ca2+ levels, particularly in the region local to the mitochondria and endoplasmic reticulum, significantly influence pacing frequency and synchronization of pacemaker unit discharge. Moreover, numerical investigations show that our ICC model is capable of qualitatively replicating a wide range of experimental observations.
Collapse
|
29
|
Zhu MH, Kim TW, Ro S, Yan W, Ward SM, Koh SD, Sanders KM. A Ca(2+)-activated Cl(-) conductance in interstitial cells of Cajal linked to slow wave currents and pacemaker activity. J Physiol 2009; 587:4905-18. [PMID: 19703958 DOI: 10.1113/jphysiol.2009.176206] [Citation(s) in RCA: 219] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Interstitial cells of Cajal (ICC) are unique cells that generate electrical pacemaker activity in gastrointestinal (GI) muscles. Many previous studies have attempted to characterize the conductances responsible for pacemaker current and slow waves in the GI tract, but the precise mechanism of electrical rhythmicity is still debated. We used a new transgenic mouse with a bright green fluorescent protein (copGFP) constitutively expressed in ICC to facilitate study of these cells in mixed cell dispersions. We found that ICC express a specialized 'slow wave' current. Reversal of tail current analysis showed this current was due to a Cl(-) selective conductance. ICC express ANO1, a Ca(2+)-activated Cl(-) channel. Slow wave currents are not voltage dependent, but a secondary voltage-dependent process underlies activation of these currents. Removal of extracellular Ca(2+), replacement of Ca(2+) with Ba(2+), or extracellular Ni(2+) (30 microm) blocked the slow wave current. Single Ca(2+)-activated Cl() channels with a unitary conductance of 7.8 pS were resolved in excised patches of ICC. These are similar in conductance to ANO1 channels (8 pS) expressed in HEK293 cells. Slow wave current was blocked in a concentration-dependent manner by niflumic acid (IC(50) = 4.8 microm). Slow wave currents are associated with transient depolarizations of ICC in current clamp, and these events were blocked by niflumic acid. These findings demonstrate a role for a Ca(2+)-activated Cl(-) conductance in slow wave current in ICC and are consistent with the idea that ANO1 participates in pacemaker activity.
Collapse
Affiliation(s)
- Mei Hong Zhu
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Zhu MH, Kim TW, Ro S, Yan W, Ward SM, Koh SD, Sanders KM. A Ca(2+)-activated Cl(-) conductance in interstitial cells of Cajal linked to slow wave currents and pacemaker activity. J Physiol 2009. [PMID: 19703958 DOI: 10.1113/jphysiol.2009.176206.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Interstitial cells of Cajal (ICC) are unique cells that generate electrical pacemaker activity in gastrointestinal (GI) muscles. Many previous studies have attempted to characterize the conductances responsible for pacemaker current and slow waves in the GI tract, but the precise mechanism of electrical rhythmicity is still debated. We used a new transgenic mouse with a bright green fluorescent protein (copGFP) constitutively expressed in ICC to facilitate study of these cells in mixed cell dispersions. We found that ICC express a specialized 'slow wave' current. Reversal of tail current analysis showed this current was due to a Cl(-) selective conductance. ICC express ANO1, a Ca(2+)-activated Cl(-) channel. Slow wave currents are not voltage dependent, but a secondary voltage-dependent process underlies activation of these currents. Removal of extracellular Ca(2+), replacement of Ca(2+) with Ba(2+), or extracellular Ni(2+) (30 microm) blocked the slow wave current. Single Ca(2+)-activated Cl() channels with a unitary conductance of 7.8 pS were resolved in excised patches of ICC. These are similar in conductance to ANO1 channels (8 pS) expressed in HEK293 cells. Slow wave current was blocked in a concentration-dependent manner by niflumic acid (IC(50) = 4.8 microm). Slow wave currents are associated with transient depolarizations of ICC in current clamp, and these events were blocked by niflumic acid. These findings demonstrate a role for a Ca(2+)-activated Cl(-) conductance in slow wave current in ICC and are consistent with the idea that ANO1 participates in pacemaker activity.
Collapse
Affiliation(s)
- Mei Hong Zhu
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Jin NG, Koh SD, Sanders KM. Caffeine inhibits nonselective cationic currents in interstitial cells of Cajal from the murine jejunum. Am J Physiol Cell Physiol 2009; 297:C971-8. [PMID: 19625609 DOI: 10.1152/ajpcell.00155.2009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Interstitial cells of Cajal (ICC) discharge unitary potentials in gastrointestinal muscles that constitute the basis for pacemaker activity. Caffeine has been used to block unitary potentials, but the ionic conductance responsible for unitary potentials is controversial. We investigated currents in cultured ICC from murine jejunum that may underlie unitary potentials and studied the effects of caffeine. Networks of ICC generated slow wave events under current clamp, and these events were blocked by caffeine in a concentration-dependent manner. Single ICC generated spontaneous transient inward currents (STICs) under voltage clamp at -60 mV and noisy voltage fluctuations in current clamp. STICs were unaffected when the equilibrium potential for Cl- (ECl) was set to -60 mV (excluding Cl- currents) and reversed at 0 mV, demonstrating that a nonselective cationic conductance, and not a Cl- conductance, is responsible for STICs in ICC. Caffeine inhibited STICs in a concentration-dependent manner. Reduced intracellular Ca2+ and calmidazolium (CMZ; 1 microM) activated persistent inward, nonselective cation currents in ICC. Currents activated by CMZ and by dialysis of cells with 10 mM BAPTA were also inhibited by caffeine. Excised inside-out patches contained channels that exhibited spontaneous openings, and resulting currents reversed at 0 mV. Channel openings were increased by reducing Ca2+ concentration from 10(-6) M to 10(-8) M. CMZ (1 microM) also increased openings of nonselective cation channels. Spontaneous currents and channels activated by CMZ were inhibited by caffeine (5 mM). The findings demonstrate that the Ca2+-inhibited nonselective cation channels that generate STICs in ICC are blocked directly by caffeine. STICs are responsible for unitary potentials in intact muscles, and the block of these events by caffeine is consistent with the idea that a nonselective cation conductance underlies unitary potentials in ICC.
Collapse
Affiliation(s)
- Nan Ge Jin
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada 89557, USA
| | | | | |
Collapse
|
32
|
Kim YC, Suzuki H, Xu WX, Choi W, Kim SH, Lee SJ. Ca2+-activated K+ current in freshly isolated c-Kit positive cells in guinea-pig stomach. J Korean Med Sci 2009; 24:384-91. [PMID: 19543421 PMCID: PMC2698181 DOI: 10.3346/jkms.2009.24.3.384] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2007] [Accepted: 01/29/2008] [Indexed: 11/22/2022] Open
Abstract
This study was designed to isolate Ca2+-activated K+ current (I(KCa)) and elucidate its physiological significance in freshly isolated interstitial cells of Cajal (ICCs) of guinea-pig stomach. Single ICC was freshly isolated by enzymatically dissociating from myenteric border of gastric antrum free of circular muscles, and conventional whole-cell voltage clamp technique including immunohistochemical techniques were employed to characterize the cells: In myenteric border of gastric antrum, ICC-MY (ICCs from myenteric border) were detected by immunohistochemical reactivity, and single ICC-MY which has many branches was immunohistochemically c-Kit positive. Under K+-rich and 0.1 mM ethylene glycol-bis (2-aminoethyl ether)-N,N,N',N'-tetraacetic acid pipette solution, ICC produced spontaneous inward current (-256 +/- 92.2 pA). When step-depolarizing pulse from -80 to +80 mV was applied at holding potential (V(h)) of -80 mV, voltage-dependent outward currents were recorded with superimposed spontaneous transient outward currents (STOCs). Both STOCs and outward currents were reversibly affected by tetraethylammonium chloride (TEA) and iberiotoxin (IbTX); 2 mM TEA and 200 nM IbTX completely abolished STOCs and significantly inhibited outward K+ current over the whole potential range tested for current/voltage (I/V) relationship. In addition, TEA delayed repolarization phase of spontaneous inward current. The present results indicate the presence of I(KCa) in a single ICC, and it might be involved in regulation of repolarizing phase of spontaneous inward current in guinea-pig stomach.
Collapse
Affiliation(s)
- Young Chul Kim
- Department of Physiology, Chungbuk National University, College of Medicine, Cheongju, Korea.
| | | | | | | | | | | |
Collapse
|
33
|
Choi S, Choi JJ, Jun JY, Koh JW, Kim SH, Kim DH, Pyo MY, Choi S, Son JP, Lee I, Son M, Jin M. Induction of pacemaker currents by DA-9701, a prokinetic agent, in interstitial cells of Cajal from murine small intestine. Mol Cells 2009; 27:307-12. [PMID: 19326077 DOI: 10.1007/s10059-009-0039-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2008] [Revised: 12/09/2008] [Accepted: 12/26/2008] [Indexed: 12/16/2022] Open
Abstract
The interstitial cells of Cajal (ICC) are pacemaking cells required for gastrointestinal motility. The possibility of whether DA-9701, a novel prokinetic agent formulated with Pharbitis Semen and Corydalis Tuber, modulates pacemaker activities in the ICC was tested using the whole cell patch clamp technique. DA-9701 produced membrane depolarization and increased tonic inward pacemaker currents in the voltage-clamp mode. The application of flufenamic acid, a non-selective cation channel blocker, but not niflumic acid, abolished the generation of pacemaker currents induced by DA-9701. Pretreatment with a Ca2+-free solution and thapsigargin, a Ca2+-ATPase inhibitor in the endoplasmic reticulum, abolished the generation of pacemaker currents. In addition, the tonic inward currents were inhibited by U-73122, an active phospholipase C inhibitor, but not by GDP-beta-S, which permanently binds G-binding proteins. Furthermore, the protein kinase C inhibitors, chelerythrine and calphostin C, did not block the DA-9701-induced pacemaker currents. These results suggest that DA-9701 might affect gastrointestinal motility by the modulation of pacemaker activity in the ICC, and the activation is associated with the non-selective cationic channels via external Ca2+ influx, phospholipase C activation, and Ca2+ release from internal storage in a G protein-independent and protein kinase C-independent manner.
Collapse
Affiliation(s)
- Seok Choi
- Department of Physiology, College of Medicine, Chosun University, Gwangju 501-759, Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Kim YC, Suzuki H, Xu WX, Hashitani H, Choi W, Yun HY, Park SM, Youn SJ, Lee SJ, Lee SJ. Voltage-dependent Ca Current Identified in Freshly Isolated Interstitial Cells of Cajal (ICC) of Guinea-pig Stomach. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2008; 12:323-30. [PMID: 19967074 DOI: 10.4196/kjpp.2008.12.6.323] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The properties of voltage dependent Ca(2+) current (VDCC) were investigated in interstitial cells of Cajal (ICC) distributed in the myenteric layer (ICC-MY) of guinea-pig antrum. In tissue, ICC-MY showed c-Kit positive reactions and produced driving potentials with the amplitude and frequency of about 62 mV and 2 times min(-1), respectively, in the presence of 1 microM nifedipine. Single ICC-MY isolated by enzyme treatment also showed c-Kit immunohistochemical reactivity. These cells were also identified by generation of spontaneous inward current under K(+) -rich pipette solution. The voltage clamp experiments revealed the amplitude of - 329 pA inward current at irregular frequency. With Cs(+)-rich pipette solution at V(h)=-80 mV, ICC-MY produced voltage-dependent inward currents (VDIC), and nifedipine (1 microM) blocked VDIC. Therefore, we successfully isolated c-Kit positive single ICC from guinea-pig stomach, and found that ICC-MY potently produced dihydropiridine sensitive L-type voltage-dependent Ca(2+) currents (VDCC(L)).
Collapse
Affiliation(s)
- Young Chul Kim
- Department of Physiology, Chungbuk National University, College of Medicine, Cheongju 361-763, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Gibbons SJ, Strege PR, Lei S, Roeder JL, Mazzone A, Ou Y, Rich A, Farrugia G. The alpha1H Ca2+ channel subunit is expressed in mouse jejunal interstitial cells of Cajal and myocytes. J Cell Mol Med 2008; 13:4422-31. [PMID: 19413888 PMCID: PMC2855776 DOI: 10.1111/j.1582-4934.2008.00623.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
T-type Ca2+ currents have been detected in cells from the external muscular layers of gastrointestinal smooth muscles and appear to contribute to the generation of pacemaker potentials in interstitial cells of Cajal from those tissues. However, the Ca2+ channel α subunit responsible for these currents has not been determined. We established that the α subunit of the α1H Ca2+ channel is expressed in single myocytes and interstitial cells of Cajal using reverse transcription and polymerase chain reaction from whole tissue, laser capture microdissected tissue and single cells isolated from the mouse jejunum. Whole-cell voltage clamp recordings demonstrated that a nifedipine and Cd2+ resistant, mibefradil-sensitive current is present in myocytes dissociated from the jejunum. Electrical recordings from the circular muscle layer demonstrated that mibefradil reduced the frequency and initial rate of rise of the electrical slow wave. Gene targeted knockout of both alleles of the cacna1h gene, which encodes the α1H Ca2+ channel subunit, resulted in embryonic lethality because of death of the homozygous knockouts prior to E13.5 days in utero. We conclude that a channel with the pharmacological and molecular characteristics of the α1H Ca2+ channel subunit is expressed in interstitial cells of Cajal and myocytes from the mouse jejunum, and that ionic conductances through the α1H Ca2+ channel contribute to the upstroke of the pacemaker potential. Furthermore, the survival of mice that do not express the α1H Ca2+ channel protein is dependent on the genetic background and targeting approach used to generate the knockout mice.
Collapse
Affiliation(s)
- Simon J Gibbons
- Enteric Neuroscience Program, Mayo Clinic College of Medicine, Rochester, MN, USA.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Smooth muscle cell (SMC) contraction is controlled by the Ca2+ and Rho kinase signalling pathways. While the SMC Rho kinase system seems to be reasonably constant, there is enormous variation with regard to the mechanisms responsible for generating Ca2+ signals. One way of dealing with this diversity is to consider how this system has been adapted to control different SMC functions. Phasic SMCs (vas deferens, uterus and bladder) rely on membrane depolarization to drive Ca2+ influx across the plasma membrane. This depolarization can be induced by neurotransmitters or through the operation of a membrane oscillator. Many tonic SMCs (vascular, airway and corpus cavernosum) are driven by a cytosolic Ca2+ oscillator that generates periodic pulses of Ca2+. A similar oscillator is present in pacemaker cells such as the interstitial cells of Cajal (ICCs) and atypical SMCs that control other tonic SMCs (gastrointestinal, urethra, ureter). The changes in membrane potential induced by these cytosolic oscillators does not drive contraction directly but it functions to couple together individual oscillators to provide the synchronization that is a characteristic feature of many tonic SMCs.
Collapse
|
37
|
Corrias A, Buist ML. Quantitative cellular description of gastric slow wave activity. Am J Physiol Gastrointest Liver Physiol 2008; 294:G989-95. [PMID: 18276830 DOI: 10.1152/ajpgi.00528.2007] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Interstitial cells of Cajal (ICC) are responsible for the spontaneous and omnipresent electrical activity in the stomach. A quantitative description of the intracellular processes whose coordinated activity is believed to generate electrical slow waves has been developed and is presented here. In line with recent experimental evidence, the model describes how the interplay between the mitochondria and the endoplasmic reticulum in cycling intracellular Ca(2+) provides the primary regulatory signal for the initiation of the slow wave. The major ion channels that have been identified as influencing slow wave activity have been modeled according to data obtained from isolated ICC. The model has been validated by comparing the simulated profile of the slow waves with experimental recordings and shows good correspondence in terms of frequency, amplitude, and shape in both control and pharmacologically altered conditions.
Collapse
Affiliation(s)
- Alberto Corrias
- Division of Bioengineering, National Univ. of Singapore, 9 Engineering Dr. 1, Singapore 117576
| | | |
Collapse
|
38
|
A biophysically based mathematical model of unitary potential activity in interstitial cells of Cajal. Biophys J 2008; 95:88-104. [PMID: 18339738 DOI: 10.1529/biophysj.107.122507] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Unitary potential (UP) depolarizations are the basic intracellular events responsible for pacemaker activity in interstitial cells of Cajal (ICCs), and are generated at intracellular sites termed "pacemaker units". In this study, we present a mathematical model of the transmembrane ion flows and intracellular Ca(2+) dynamics from a single ICC pacemaker unit acting at near-resting membrane potential. This model quantitatively formalizes the framework of a novel ICC pacemaking mechanism that has recently been proposed. Model simulations produce spontaneously rhythmic UP depolarizations with an amplitude of approximately 3 mV at a frequency of 0.05 Hz. The model predicts that the main inward currents, carried by a Ca(2+)-inhibited nonselective cation conductance, are activated by depletion of sub-plasma-membrane [Ca(2+)] caused by sarcoendoplasmic reticulum calcium ATPase Ca(2+) sequestration. Furthermore, pacemaker activity predicted by our model persists under simulated voltage clamp and is independent of [IP(3)] oscillations. The model presented here provides a basis to quantitatively analyze UP depolarizations and the biophysical mechanisms underlying their production.
Collapse
|
39
|
Zhu Y, Huizinga JD. Nitric oxide decreases the excitability of interstitial cells of Cajal through activation of the BK channel. J Cell Mol Med 2008; 12:1718-27. [PMID: 18194464 PMCID: PMC3918088 DOI: 10.1111/j.1582-4934.2008.00217.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Nitrergic nerves are structurally and functionally associated with ICC. To further understand mechanisms of communication, the hypothesis was investigated that NO might affect large conductance K channels. To that end, we searched for IbTX-sensitive currents in ICC obtained through explant cultures from the mouse small intestine and studied effects of the NOS inhibitor omega N-nitro-L-arginine (LNNA) and the NO donor sodium nitroprusside (SNP). IbTX-sensitive currents acquired in the whole-cell configuration through nystatin perforated patches exhibited high noise levels but relatively low amplitude, whereas currents obtained in the conventional whole-cell configuration exhibited less noise and higher amplitudes; depolarization from -80 to + 40 mV evoked 357 +/- 159 pA current in the nystatin perforated patch configuration and 1075 +/- 597 pA using the conventional whole-cell configuration. Immunohistochemistry showed that ICC associated with ganglia and Auerbach's plexus nerve fibers were immunoreactive to BK antibodies. The IbTX-sensitive currents were increased by SNP and inhibited by LNNA. BK blockers suppressed spontaneous transit outward currents in ICC. After block of BK currents, or before these currents became prominent, calcium currents were activated by depolarization in the same cells. Their peak amplitude occurred at -25 mV and the currents were increased with increasing extracellular calcium and inhibited by cobalt. The hypothesis is warranted that nitrergic innervation inhibits ICC excitability in part through activation of BK channels. In addition, NO is an intracellular regulator of ICC excitability.
Collapse
Affiliation(s)
- Yaohui Zhu
- McMaster University, Intestinal Disease Research Programme, Department of Medicine, Hamilton, Ontario, Canada
| | | |
Collapse
|
40
|
Nakayama S, Kajioka S, Goto K, Takaki M, Liu HN. Calcium-associated mechanisms in gut pacemaker activity. J Cell Mol Med 2008; 11:958-68. [PMID: 17979877 PMCID: PMC4401267 DOI: 10.1111/j.1582-4934.2007.00107.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
A considerable body of evidence has revealed that interstitial cells of Cajal (ICC), identified with c-Kit-immunoreactivity, act as gut pacemaker cells, with spontaneous Ca2+ activity in ICC as the probable primary mechanism. Namely, intracellular (cytosolic) Ca2+ oscillations in ICC periodically activate plasmalemmal Ca2+-dependent ion channels and thereby generate pacemaker potentials. This review will, thus, focus on Ca2+-associated mechanisms in ICC in the gastrointestinal (GI) tract, including auxiliary organs.
Collapse
Affiliation(s)
- Shinsuke Nakayama
- Department of Cell Physiology, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | | | | | | | | |
Collapse
|
41
|
Abstract
C-Kit positive interstitial cells of Cajal (ICC) play an important role in the regulation of the smooth muscle motility, acting as internal pacemakers to provide the slow wave activity within various luminal organs. Recently c-Kit-(CD117)-positive interstitial cells (IC) have been shown in the genitourinary tract, but systematic studies on the distribution and density of IC within the urinary tract are still lacking. Therefore the aim of the present study was to analyze systematically the localization and distribution of the c-Kit receptor in the urinary tract of the pig using immunohistochemical and molecular methods. Tissue samples were harvested from the porcine urinary tract including renal calices and pelvis, ureteropelvic junction, proximal, middle and distal ureter, ureteral orifice, fundus, and corpus of the bladder and the internal urethral orifice. Small and large intestine specimen served as controls. Immunohistochemistry (APAAP, IF) was applied on serial frozen sections using four monoclonal and polyclonal antibodies recognizing CD117. Whole mounts of the porcine upper urinary tract were prepared and investigated using conventional and confocal fluorescence microscopy followed by three-dimensional reconstruction. UV-laser microdissection and RT-PCR were applied to confirm the immunohistochemical results. CD117-immunoreactivity labeled bipolar IC and round-shaped mast cells (MC) throughout the adventitia, tunica muscularis and submucosa within the whole porcine urinary tract. While MC were found continuously in all investigated segments, a gradient of bipolar IC was evident. The whole mount preparations gave a detailed cytomorphology of IC within the various layers of the porcine urinary tract. Whole mount preparations revealed closed apposition of bi- and tripolar c-Kit positive IC parallel to the smooth muscle bundles and to veins of the tunica muscularis and adventitia. In the urothelium single CD117-positive interepithelial cells were found. The highest density of CD117-positive cells was found at the ureteropelvic junction, however the differences in between the segments were minimal. Microdissection and RT-PCR confirmed the results uncovered by immunohistochemistry. The ubiquitous distribution of IC and their close relationship to smooth muscle provides strong evidence that IC could contribute to the intrinsic pacemaker activity within the porcine (upper and lower) urinary tract. The role of the interepithelial CD117-positive cells as mechanosensors or as a precursor cell in the regeneration of the urothelium, is conceivable.
Collapse
|
42
|
Takeda Y, Koh SD, Sanders KM, Ward SM. Differential expression of ionic conductances in interstitial cells of Cajal in the murine gastric antrum. J Physiol 2007; 586:859-73. [PMID: 18033817 DOI: 10.1113/jphysiol.2007.140293] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Two distinct populations of interstitial cells of Cajal (ICC) exist within the tunica muscularis of the gastric antrum, and these cells serve different physiological functions. One population of ICC generates and actively propagates electrical slow waves, and the other population of ICC is innervated by excitatory and inhibitory motor neurons and mediates enteric motor neurotransmission. In spite of the key role of ICC in gastric excitability, little is known about the ionic conductances that underlie the functional diversity of these cells. In the present study we isolated ICC from the murine gastric antrum and investigated the Ca(2+)-dependent ionic conductances expressed by these cells using the patch clamp technique. Conductances in ICC were compared with those expressed in smooth muscle cells. The cells studied were identified by RT-PCR using cell-specific primers that included Myh11 (smooth muscle cells), Kit (ICC) and Uchl1 (enteric neurons) following electrophysiolgical recordings. Distinct ionic conductances were observed in Kit-positive cells. One group of ICC expressed a basal non-selective cation conductance (NSCC) that was inhibited by an increase in [Ca(2+)](i) in a calmodulin (CaM)-dependent manner. A second population of ICC generated spontaneous transient inward currents (STICs) and expressed a basal noisy NSCC that was facilitated by an increase in [Ca(2+)](i) in a CaM-dependent manner. The [Ca(2+)](i)-facilitated NSCC in ICC was blocked by the Cl(-) channel antagonists 4,4'-diisothiocyanatostilbene-2,2'-disulphonic acid (DIDS), anthracene-9-carboxylate (9-AC) and niflumic acid. These data suggest that distinct NSCC are expressed in subpopulations of ICC and these conductances may underlie the functional differences of these cells within the gastric antrum.
Collapse
Affiliation(s)
- Yukari Takeda
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA
| | | | | | | |
Collapse
|
43
|
Chen H, Ordög T, Chen J, Young DL, Bardsley MR, Redelman D, Ward SM, Sanders KM. Differential gene expression in functional classes of interstitial cells of Cajal in murine small intestine. Physiol Genomics 2007; 31:492-509. [PMID: 17895395 DOI: 10.1152/physiolgenomics.00113.2007] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Interstitial cells of Cajal (ICC) have important functions in regulation of motor activity in the gastrointestinal tract. In murine small intestine, ICC are gathered in the regions of the myenteric plexus (ICC-MY) and the deep muscular plexus (ICC-DMP). These two classes of ICC have different physiological functions. ICC-MY are pacemaker cells and generate the slow-wave electrical rhythmicity of gastrointestinal organs. ICC-DMP form synaptic connections with the varicose nerve terminals of enteric motor neurons and are involved in reception and transduction of motor neurotransmission. Gene expression underlying specific functions of ICC classes is incompletely understood. In the present study, we used recently developed highly selective techniques to isolate the two functional ICC classes from enzymatically dispersed intestinal muscles by fluorescence-activated cell sorting. The transcriptomes of ICC-MY and ICC-DMP were investigated using oligonucleotide microarray analysis. Differential expression of functional groups of genes defined by standard gene ontology terms was also studied. There were substantial numbers of genes expressed more abundantly in ICC than in the tunica muscularis, and we also detected marked phenotypic differences between ICC-MY and ICC-DMP. Notably, genes related to cell junction, process guidance, and vesicle trafficking were upregulated in ICC. Consistent with their specific functions, metabolic and Ca(2+) transport genes were relatively upregulated in ICC-MY, whereas genes for signaling proteins involved in transduction of neurotransmitter functions were relatively upregulated in ICC-DMP. Our results may lead to the identification of novel biomarkers for ICC and provide directions for further studies designed to understand ICC function in health and disease.
Collapse
Affiliation(s)
- Hui Chen
- Department of Physiology and Cell Biology, University of Nevada, School of Medicine, Reno, NV 89557, USA
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Bayguinov O, Ward SM, Kenyon JL, Sanders KM. Voltage-gated Ca2+ currents are necessary for slow-wave propagation in the canine gastric antrum. Am J Physiol Cell Physiol 2007; 293:C1645-59. [PMID: 17855773 DOI: 10.1152/ajpcell.00165.2007] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Electrical slow waves determine the timing and force of peristaltic contractions in the stomach. Slow waves originate from a dominant pacemaker in the orad corpus and propagate actively around and down the stomach to the pylorus. The mechanism of slow-wave propagation is controversial. We tested whether Ca(2+) entry via a voltage-dependent, dihydropyridine-resistant Ca(2+) conductance is necessary for active propagation in canine gastric antral muscles. Muscle strips cut parallel to the circular muscle were studied with intracellular electrophysiological techniques using a partitioned-chamber apparatus. Slow-wave upstroke velocity and plateau amplitude decreased from the greater to the lesser curvature, and this corresponded to a decrease in the density of interstitial cells of Cajal in the lesser curvature. Slow-wave propagation velocity between electrodes impaling cells in two regions of muscle and slow-wave upstroke and plateau were measured in response to experimental conditions that reduce the driving force for Ca(2+) entry or block voltage-dependent Ca(2+) currents. Nicardipine (0.1-1 microM) did not affect slow-wave upstroke or propagation velocities. Upstroke velocity, amplitude, and propagation velocity were reduced in a concentration-dependent manner by Ni(2+) (1-100 microM), mibefradil (10-30 microM), and reduced extracellular Ca(2+) (0.5-1.5 mM). Depolarization (by 10-15 mM K(+)) or hyperpolarization (10 microM pinacidil) also reduced upstroke and propagation velocities. The higher concentrations (or lowest Ca(2+)) of these drugs and ionic conditions tested blocked slow-wave propagation. Treatment with cyclopiazonic acid to empty Ca(2+) stores did not affect propagation. These experiments show that voltage-dependent Ca(2+) entry is obligatory for the upstroke phase of slow waves and active propagation.
Collapse
Affiliation(s)
- Orline Bayguinov
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada 89557, USA
| | | | | | | |
Collapse
|
45
|
Strege PR, Mazzone A, Kraichely RE, Sha L, Holm AN, Ou Y, Lim I, Gibbons SJ, Sarr MG, Farrugia G. Species dependent expression of intestinal smooth muscle mechanosensitive sodium channels. Neurogastroenterol Motil 2007; 19:135-43. [PMID: 17244168 DOI: 10.1111/j.1365-2982.2006.00844.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A mechanosensitive Na(+) current carried by Na(v)1.5 is present in human intestinal circular smooth muscle and contributes to regulation of intestinal motor function. Expression of this channel in different species is unknown. Our aim was to determine if Na(+) currents and message for the alpha subunit of the Na(+) channel (SCN5A) are found in circular smooth muscle cells of human, dog, pig, mouse and guinea pig jejunum. Currents were recorded using patch clamp techniques. Message for SCN5A was investigated using laser capture microdissection and reverse transcription polymerase chain reaction (RT-PCR). Na(+) currents were identified consistently in human and dog smooth muscle cells; however, Na(+) current was not found in pig (0/20) or guinea pig smooth muscle cells (0/21) and found only one mouse cell (1/21). SCN5A mRNA was found in circular muscle of human, dog, and mouse, but not in pig or guinea pig, and not in mouse longitudinal or mucosal layers. In summary, SCN5A message is expressed in, and Na(+) current recorded from, circular muscle layer of human and dog but not from pig and guinea pig. These data show that there are species differences in expression of the SCN5A-encoded Na(v)1.5 channel, suggesting species-specific differences in the electrophysiological response to mechanical and depolarizing stimuli.
Collapse
Affiliation(s)
- P R Strege
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Sui GP, Wu C, Severs N, Newgreen D, Fry CH. The association between t-type Ca2+current and outward current in isolated human detrusor cells from stable and overactive bladders. BJU Int 2007; 99:436-41. [PMID: 17026591 DOI: 10.1111/j.1464-410x.2006.06568.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To determine if bladder overactivity in humans is associated with an altered activity of Ca(2+) channels in detrusor smooth muscle and the consequent activation of other ion channels. MATERIAL AND METHODS Samples of bladder were obtained from patients with urodynamically stable bladders, or with idiopathic detrusor overactivity. Isolated cells were patch-clamped with pipettes containing a Cs(+)-based filling solution to isolate inward currents, or a K(+)-filling solution to measure outward current. Components of inward current were separated according to their sensitivity to NiCl(2) (< or =100 microm) and nifedipine. RESULTS Ni(2+)-sensitive (T-type) and nifedipine-sensitive (L-type) current was recorded in all cells. The voltage- and time-dependent properties were similar in cells from both patient groups. However, the current density of the L-type current was less, and that of the T-type current was greater, in myocytes from overactive bladders. In cells from overactive bladders, the mean K(+) current over the range - 80 to - 50 mV was also higher than in control cells. This current was sensitive to the large-conductance channel modulator iberiotoxin and to NiCl(2) (100 microm) CONCLUSIONS Detrusor myocytes from overactive human bladders have a higher T-type Ca(2+) current density; we propose that this increases transient outward currents, and so might contribute to higher levels of spontaneous activity.
Collapse
Affiliation(s)
- Gui-Ping Sui
- The Institute of Urology, University College London, UK
| | | | | | | | | |
Collapse
|
47
|
Nikitina E, Zhang ZD, Kawashima A, Jahromi BS, Bouryi VA, Takahashi M, Xie A, Macdonald RL. Voltage-dependent calcium channels of dog basilar artery. J Physiol 2006; 580:523-41. [PMID: 17185332 PMCID: PMC2075556 DOI: 10.1113/jphysiol.2006.126128] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Electrophysiological and molecular characteristics of voltage-dependent calcium (Ca(2+)) channels were studied using whole-cell patch clamp, polymerase chain reaction and Western blotting in smooth muscle cells freshly isolated from dog basilar artery. Inward currents evoked by depolarizing steps from a holding potential of -50 or -90 mV in 10 mm barium consisted of low- (LVA) and high-voltage activated (HVA) components. LVA current comprised more than half of total current in 24 (12%) of 203 cells and less than 10% of total current in 52 (26%) cells. The remaining cells (127 cells, 62%) had LVA currents between one tenth and one half of total current. LVA current was rapidly inactivating, slowly deactivating, inhibited by high doses of nimodipine and mibefradil (> 0.3 microM), not affected by omega-agatoxin GVIA (gamma100 nM), omega-conotoxin IVA (1 microM) or SNX-482 (200 nM) and probably carried by T-type Ca(2+) channels based on the presence of messenger ribonucleic acid (mRNA) and protein for Ca(v3.1) and Ca(v3.3) alpha(1) subunits of these channels. LVA currents exhibited window current with a maximum of 13% of the LVA current at -37.4 mV. HVA current was slowly inactivating and rapidly deactivating. It was inhibited by nimodipine (IC(50) = 0.018 microM), mibefradil (IC(50) = 0.39 microM) and omega-conotoxin IV (1 microM). Smooth muscle cells also contained mRNA and protein for L- (Ca(v1.2) and Ca(v1.3)), N- (Ca(v2.2)) and T-type (Ca(v3.1) and Ca(v3.3)) alpha(1) Ca(2+) channel subunits. Confocal microscopy showed Ca(v1.2) and Ca(v1.3) (L-type), Ca(v2.2) (N-type) and Ca(v3.1) and Ca(v3.3) (T-type) protein in smooth muscle cells. Relaxation of intact arteries under isometric tension in vitro to nimodipine (1 microM) and mibefradil (1 microM) but not to omega-agatoxin GVIA (100 nM), omega-conotoxin IVA (1 microM) or SNX-482 (1 microM) confirmed the functional significance of L- and T-type voltage-dependent Ca(2+) channel subtypes but not N-type. These results show that dog basilar artery smooth muscle cells express functional voltage-dependent Ca(2+) channels of multiple types.
Collapse
Affiliation(s)
- Elena Nikitina
- Department of Surgery, University of Chicago Medical Center, IL, USA
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Hotta A, Okada N, Suzuki H. Mibefradil-sensitive component involved in the plateau potential in submucosal interstitial cells of the murine proximal colon. Biochem Biophys Res Commun 2006; 353:170-6. [PMID: 17174936 DOI: 10.1016/j.bbrc.2006.12.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2006] [Accepted: 12/01/2006] [Indexed: 11/15/2022]
Abstract
Submucosal interstitial cells of Cajal (ICC(SM)) produce plateau potentials comprised of initial fast and subsequent plateau components. The possible involvement of voltage-dependent Ca(2+) channels in plateau potentials was examined in ICC(SM) of the murine proximal colon. Increases in external K(+) concentration ([K(+)](o)) changed the rise rate of the initial component in a biphasic way, an increase in 10.6 or 15.3mM [K(+)](o) and a decrease in 20.0mM [K(+)](o). The rise rate of plateau potentials was significantly reduced by the application of 3 microM mibefradil or 100 microM Ni(2+) but not by 0.3 microM nifedipine. The inhibitory effect of mibefradil on the rise rate of plateau potentials was concentration-dependent with an IC(50) value of 1.0 microM. In conclusion, the initial phase of plateau potentials is partly due to the activation of T-type Ca(2+) channel in ICC(SM) from the murine proximal colon.
Collapse
Affiliation(s)
- Aya Hotta
- Department of Regulatory Cell Physiology, Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan.
| | | | | |
Collapse
|
49
|
Abstract
There has been considerable speculation about the function of interstitial cells of Cajal (ICC) since their discovery more than 100 years ago. It has been difficult to study these cells under native conditions, but great insights about the function of ICC have come from studies of genetic models with loss-of function mutations in the Kit signalling pathway. First it was discovered that signalling via Kit (a receptor tyrosine kinase) was vital for the development and maintenance of the ICC phenotype in gastrointestinal (GI) muscles. In compound heterozygotes (W/W(V) and Sl/Sl(d) animals), where there are partial loss-of-function mutations in Kit receptors or Kit ligand (stem cell factor), ICC failed to develop in various regions of the GI tract, but no major changes in the smooth muscle layers or enteric nervous system occurred in the absence of these cells. Animals with these mutations provided an unprecedented opportunity to understand the role of ICC in GI motor function, and it is now clear from these studies that ICC serve as: (i) pacemaker cells, generating the spontaneous electrical rhythms of the gut known as slow waves; (ii) a propagation pathway for slow waves so that large areas of the musculature can be entrained to a dominant pacemaker frequency; (iii) mediators of excitatory cholinergic and inhibitory nitrergic neural inputs from the enteric nervous system, and (iv) stretch receptors that modulate membrane potential and electrical slow wave frequency. This review describes the use of genetic models to understand the important physiological role of ICC in the GI tract.
Collapse
Affiliation(s)
- Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA.
| | | |
Collapse
|
50
|
Chen H, Redelman D, Ro S, Ward SM, Ordög T, Sanders KM. Selective labeling and isolation of functional classes of interstitial cells of Cajal of human and murine small intestine. Am J Physiol Cell Physiol 2006; 292:C497-507. [PMID: 16943245 DOI: 10.1152/ajpcell.00147.2006] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Specific functions of interstitial cells of Cajal (ICC) have been linked to distinct classes that differ by morphology and distribution. In the small intestine, slow wave-generating ICC are located in the myenteric region (ICC-MY), whereas ICC that mediate neuromuscular neurotransmission occur either throughout the circular muscle layer (intramuscular ICC, ICC-IM) or in association with the deep muscular plexus (ICC-DMP). Selective isolation of ICC to characterize specific properties has been difficult. Recently, neurokinin-1 receptors have been detected in murine ICC-DMP and neurons but not in ICC-MY. Here we identified and isolated ICC-DMP/IM by receptor-mediated internalization of fluorescent substance P and Kit immunofluorescence. Specificity of labeling was verified by confocal microscopy. Mouse and human ICC-DMP/IM were detected in suspension by fluorescent microscopy and harvested for RT-PCR with micropipettes. The isolated cells expressed Kit but not markers for neurons, smooth muscle, or antigen-presenting cells. ICC-DMP expressed neurokinin-1 receptor, M(2) and M(3) muscarinic receptors, P2Y(1) and P2Y(4) purinergic receptors, VIP receptor 2, soluble guanylate cyclase-1 subunits, and protein kinase G. L- or T-type Ca(2+) channels were not detected in these cells. ICC-MY and ICC-DMP were simultaneously detected and enumerated by flow cytometry and sorted to purity by fluorescence-activated cell sorting. In summary, functional classes of ICC have distinct molecular identities that can be used to selectively identify and harvest these cells with, for example, receptor-mediated uptake of substance P and Kit immunofluorescence. ICC-DMP express neurotransmitter receptors and signaling intermediate molecules that are consistent with their role in neuromuscular neurotransmission.
Collapse
MESH Headings
- Animals
- Cell Separation
- Cells/classification
- Flow Cytometry
- Fluorescent Antibody Technique
- Humans
- Intestine, Small/cytology
- Intestine, Small/metabolism
- Intestine, Small/physiology
- Mice/anatomy & histology
- Mice, Inbred BALB C
- Microscopy, Confocal
- Microscopy, Fluorescence
- Muscle, Smooth/cytology
- Muscle, Smooth/metabolism
- Muscle, Smooth/physiology
- Proto-Oncogene Proteins c-kit/metabolism
- Receptors, Neurokinin-1/metabolism
- Receptors, Neurotransmitter/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Substance P/metabolism
Collapse
Affiliation(s)
- Hui Chen
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada-Reno, Reno, NV 89557, USA
| | | | | | | | | | | |
Collapse
|