1
|
Hashitani H, Takeya M, van Helden DF. Commonality and heterogeneity of pacemaker mechanisms in the male reproductive organs. J Physiol 2024. [PMID: 38607187 DOI: 10.1113/jp284756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 03/26/2024] [Indexed: 04/13/2024] Open
Abstract
During emission, the first phase of ejaculation, smooth muscle in organs of the male reproductive tract (MRT) vigorously contract upon sympathetic nerve excitation to expel semen consisting of sperm and seminal plasma. During inter-ejaculation phases, the epididymis, seminal vesicles and prostate undergo spontaneous phasic contractions (SPCs), this transporting and maintaining the quality of sperm and seminal plasma. Recent studies have revealed platelet-derived growth factor receptor α-expressing (PDGFRα+) subepithelial interstitial cells in seminal vesicles subserve the role of pacemaker cells that electrically drive SPCs in this organ. PDGFRα+ smooth muscle cells in the epididymis also appear to function as pacemaker cells implicating PDGFRα as a potential signature molecule in MRT pacemaking. The dominant mechanism driving pacemaking in these organs is the cytosolic Ca2+ oscillator. This operates through entrainment of the release-refill cycle of Ca2+ stores, the released Ca2+ ions opening Ca2+-activated chloride channels, including in some cases ANO1 (TMEM16A), with the resultant pacemaker potential activating L-type voltage-dependent Ca2+ channels in the smooth muscle causing contraction (viz. SPCs). A second pacemaker mechanism, namely the membrane oscillator also has a role in specific cases. Further investigations into the commonality and heterogeneity of MRT pacemakers will open an avenue for understanding the pathogenesis of male infertility associated with deterioration of seminal plasma.
Collapse
Affiliation(s)
- Hikaru Hashitani
- Department of Cell Physiology, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Mitsue Takeya
- Division of Integrated Autonomic Function, Department of Physiology, Kurume University School of Medicine, Kurume, Japan
| | - Dirk F van Helden
- School of Biomedical Sciences and Pharmacy, Faculty of Health, Medicine and Wellbeing & Hunter Medical Research Institute, The University of Newcastle, Callaghan, New South Wales, Australia
| |
Collapse
|
2
|
Sanders KM, Drumm BT, Cobine CA, Baker SA. Ca 2+ dynamics in interstitial cells: foundational mechanisms for the motor patterns in the gastrointestinal tract. Physiol Rev 2024; 104:329-398. [PMID: 37561138 PMCID: PMC11281822 DOI: 10.1152/physrev.00036.2022] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 06/29/2023] [Accepted: 08/06/2023] [Indexed: 08/11/2023] Open
Abstract
The gastrointestinal (GI) tract displays multiple motor patterns that move nutrients and wastes through the body. Smooth muscle cells (SMCs) provide the forces necessary for GI motility, but interstitial cells, electrically coupled to SMCs, tune SMC excitability, transduce inputs from enteric motor neurons, and generate pacemaker activity that underlies major motor patterns, such as peristalsis and segmentation. The interstitial cells regulating SMCs are interstitial cells of Cajal (ICC) and PDGF receptor (PDGFR)α+ cells. Together these cells form the SIP syncytium. ICC and PDGFRα+ cells express signature Ca2+-dependent conductances: ICC express Ca2+-activated Cl- channels, encoded by Ano1, that generate inward current, and PDGFRα+ cells express Ca2+-activated K+ channels, encoded by Kcnn3, that generate outward current. The open probabilities of interstitial cell conductances are controlled by Ca2+ release from the endoplasmic reticulum. The resulting Ca2+ transients occur spontaneously in a stochastic manner. Ca2+ transients in ICC induce spontaneous transient inward currents and spontaneous transient depolarizations (STDs). Neurotransmission increases or decreases Ca2+ transients, and the resulting depolarizing or hyperpolarizing responses conduct to other cells in the SIP syncytium. In pacemaker ICC, STDs activate voltage-dependent Ca2+ influx, which initiates a cluster of Ca2+ transients and sustains activation of ANO1 channels and depolarization during slow waves. Regulation of GI motility has traditionally been described as neurogenic and myogenic. Recent advances in understanding Ca2+ handling mechanisms in interstitial cells and how these mechanisms influence motor patterns of the GI tract suggest that the term "myogenic" should be replaced by the term "SIPgenic," as this review discusses.
Collapse
Affiliation(s)
- Kenton M Sanders
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada-Reno, Reno, Nevada, United States
| | - Bernard T Drumm
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Ireland
| | - Caroline A Cobine
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Ireland
| | - Salah A Baker
- Department of Physiology and Cell Biology, School of Medicine, University of Nevada-Reno, Reno, Nevada, United States
| |
Collapse
|
3
|
Zawieja SD, Pea GA, Broyhill SE, Patro A, Bromert KH, Li M, Norton CE, Castorena-Gonzalez JA, Hancock EJ, Bertram CD, Davis MJ. IP3R1 underlies diastolic ANO1 activation and pressure-dependent chronotropy in lymphatic collecting vessels. J Gen Physiol 2023; 155:e202313358. [PMID: 37851027 PMCID: PMC10585095 DOI: 10.1085/jgp.202313358] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 08/11/2023] [Accepted: 09/22/2023] [Indexed: 10/19/2023] Open
Abstract
Pressure-dependent chronotropy of murine lymphatic collecting vessels relies on the activation of the Ca2+-activated chloride channel encoded by Anoctamin 1 (Ano1) in lymphatic muscle cells. Genetic ablation or pharmacological inhibition of ANO1 results in a significant reduction in basal contraction frequency and essentially complete loss of pressure-dependent frequency modulation by decreasing the rate of the diastolic depolarization phase of the ionic pacemaker in lymphatic muscle cells (LMCs). Oscillating Ca2+ release from sarcoendoplasmic reticulum Ca2+ channels has been hypothesized to drive ANO1 activity during diastole, but the source of Ca2+ for ANO1 activation in smooth muscle remains unclear. Here, we investigated the role of the inositol triphosphate receptor 1 (Itpr1; Ip3r1) in this process using pressure myography, Ca2+ imaging, and membrane potential recordings in LMCs of ex vivo pressurized inguinal-axillary lymphatic vessels from control or Myh11CreERT2;Ip3r1fl/fl (Ip3r1ismKO) mice. Ip3r1ismKO vessels had significant reductions in contraction frequency and tone but an increased contraction amplitude. Membrane potential recordings from LMCs of Ip3r1ismKO vessels revealed a depressed diastolic depolarization rate and an elongation of the plateau phase of the action potential (AP). Ca2+ imaging of LMCs using the genetically encoded Ca2+ sensor GCaMP6f demonstrated an elongation of the Ca2+ flash associated with an AP-driven contraction. Critically, diastolic subcellular Ca2+ transients were absent in LMCs of Ip3r1ismKO mice, demonstrating the necessity of IP3R1 activity in controlling ANO1-mediated diastolic depolarization. These findings indicate a critical role for IP3R1 in lymphatic vessel pressure-dependent chronotropy and contractile regulation.
Collapse
Affiliation(s)
- Scott D. Zawieja
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| | - Grace A. Pea
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| | - Sarah E. Broyhill
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| | - Advaya Patro
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| | - Karen H. Bromert
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| | - Min Li
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| | - Charles E. Norton
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| | | | - Edward J. Hancock
- School of Mathematics and Statistics, University of Sydney, Sydney, Australia
| | | | - Michael J. Davis
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, USA
| |
Collapse
|
4
|
Grainger N, Shonnard CC, Quiggle SK, Fox EB, Presley H, Daugherty R, Shonnard MC, Drumm BT, Sanders KM. Propagation of Pacemaker Activity and Peristaltic Contractions in the Mouse Renal Pelvis Rely on Ca 2+-activated Cl - Channels and T-Type Ca 2+ Channels. FUNCTION (OXFORD, ENGLAND) 2022; 3:zqac041. [PMID: 36325511 PMCID: PMC9614935 DOI: 10.1093/function/zqac041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 08/15/2022] [Accepted: 08/16/2022] [Indexed: 01/07/2023]
Abstract
The process of urine removal from the kidney occurs via the renal pelvis (RP). The RP demarcates the beginning of the upper urinary tract and is endowed with smooth muscle cells. Along the RP, organized contraction of smooth muscle cells generates the force required to move urine boluses toward the ureters and bladder. This process is mediated by specialized pacemaker cells that are highly expressed in the proximal RP that generate spontaneous rhythmic electrical activity to drive smooth muscle depolarization. The mechanisms by which peristaltic contractions propagate from the proximal to distal RP are not fully understood. In this study, we utilized a transgenic mouse that expresses the genetically encoded Ca2+ indicator, GCaMP3, under a myosin heavy chain promotor to visualize spreading peristaltic contractions in high spatial detail. Using this approach, we discovered variable effects of L-type Ca2+ channel antagonists on contraction parameters. Inhibition of T-type Ca2+ channels reduced the frequency and propagation distance of contractions. Similarly, antagonizing Ca2+-activated Cl- channels or altering the transmembrane Cl- gradient decreased contractile frequency and significantly inhibited peristaltic propagation. These data suggest that voltage-gated Ca2+ channels are important determinants of contraction initiation and maintain the fidelity of peristalsis as the spreading contraction moves further toward the ureter. Recruitment of Ca2+-activated Cl- channels, likely Anoctamin-1, and T-type Ca2+ channels are required for efficiently conducting the depolarizing current throughout the length of the RP. These mechanisms are necessary for the efficient removal of urine from the kidney.
Collapse
Affiliation(s)
| | - Cameron C Shonnard
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, 1664 North Virginia Street, Reno, NV, 89557, USA
| | - Sage K Quiggle
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, 1664 North Virginia Street, Reno, NV, 89557, USA
| | - Emily B Fox
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, 1664 North Virginia Street, Reno, NV, 89557, USA
| | - Hannah Presley
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, 1664 North Virginia Street, Reno, NV, 89557, USA
| | - Robbie Daugherty
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, 1664 North Virginia Street, Reno, NV, 89557, USA
| | - Matthew C Shonnard
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, 1664 North Virginia Street, Reno, NV, 89557, USA
| | - Bernard T Drumm
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, 1664 North Virginia Street, Reno, NV, 89557, USA,Department of Life and Health Science, Dundalk Institute of Technology, Dublin Road, Dundalk, Co. Louth, A91 K584, Ireland
| | - Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, 1664 North Virginia Street, Reno, NV, 89557, USA
| |
Collapse
|
5
|
Foong D, Liyanage L, Zhou J, Zarrouk A, Ho V, O'Connor MD. Single-cell RNA sequencing predicts motility networks in purified human gastric interstitial cells of Cajal. Neurogastroenterol Motil 2022; 34:e14303. [PMID: 34913225 DOI: 10.1111/nmo.14303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 10/25/2021] [Accepted: 11/17/2021] [Indexed: 01/19/2023]
Abstract
BACKGROUND Gastrointestinal (GI) motility disorders affect millions of people worldwide, yet they remain poorly treated in part due to insufficient knowledge of the molecular networks controlling GI motility. Interstitial cells of Cajal (ICC) are critical GI pacemaker cells, and abnormalities in ICC are implicated in GI motility disorders. Two cell surface proteins, KIT and ANO1, are used for identifying ICC. However, difficulties accessing human tissue and the low frequency of ICC in GI tissues have meant human ICC are insufficiently characterized. Here, a range of characterization assays including single-cell RNA sequencing (scRNA-seq) was performed using KIT+ CD45- CD11B- primary human gastric ICC to better understand networks controlling human ICC biology. METHODS Excess sleeve gastrectomy tissues were dissected; ICC were analyzed by immunofluorescence, fluorescence-activated cell sorting (FACSorting), real-time PCR, mass spectrometry, and scRNA-seq. KEY RESULTS Immunofluorescence identified ANO1+ /KIT+ cells throughout the gastric muscle. Compared to the FACSorted negative cells, PCR showed the KIT+ CD45- CD11B- ICC were enriched 28-fold in ANO1 expression (p < 0.01). scRNA-seq analysis of the KIT- CD45+ CD11B+ and KIT+ CD45- CD11B- ICC revealed separate clusters of immune cells and ICC (respectively); cells in the ICC cluster expressed critical GI motility genes (eg, CAV1 and PRKG1). The scRNA-seq data for these two cell clusters predicted protein interaction networks consistent with immune cell and ICC biology, respectively. CONCLUSIONS & INFERENCES The single-cell transcriptome of purified KIT+ CD45- CD11B- human gastric ICC presented here provides new molecular insights and hypotheses into evolving models of GI motility. This knowledge will provide an improved framework to investigate targeted therapies for GI motility disorders.
Collapse
Affiliation(s)
- Daphne Foong
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Liwan Liyanage
- School of Computing, Data and Mathematical Sciences, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Jerry Zhou
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Ali Zarrouk
- Campbelltown Private Hospital, Campbelltown, New South Wales, Australia
| | - Vincent Ho
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia.,Campbelltown Private Hospital, Campbelltown, New South Wales, Australia
| | - Michael D O'Connor
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| |
Collapse
|
6
|
Straface M, Koussai MA, Makwana R, Crawley E, Palmer A, Cai W, Gharibans A, Adebibe M, Loy J, O’Grady G, Andrews PLR, Sanger GJ. A multi-parameter approach to measurement of spontaneous myogenic contractions in human stomach: Utilization to assess potential modulators of myogenic contractions. Pharmacol Res 2022; 180:106247. [DOI: 10.1016/j.phrs.2022.106247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/22/2022] [Accepted: 05/03/2022] [Indexed: 10/18/2022]
|
7
|
Sanders KM, Baker SA, Drumm BT, Kurahashi M. Ca 2+ Signaling Is the Basis for Pacemaker Activity and Neurotransduction in Interstitial Cells of the GI Tract. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1383:229-241. [PMID: 36587162 DOI: 10.1007/978-3-031-05843-1_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Years ago gastrointestinal motility was thought to be due to interactions between enteric nerves and smooth muscle cells (SMCs) in the tunica muscularis. Thus, regulatory mechanisms controlling motility were either myogenic or neurogenic. Now we know that populations of interstitial cells, c-Kit+ (interstitial cells of Cajal or ICC), and PDGFRα+ cells (formerly "fibroblast-like" cells) are electrically coupled to SMCs, forming the SIP syncytium. Pacemaker and neurotransduction functions are provided by interstitial cells through Ca2+ release from the endoplasmic reticulum (ER) and activation of Ca2+-activated ion channels in the plasma membrane (PM). ICC express Ca2+-activated Cl- channels encoded by Ano1. When activated, Ano1 channels produce inward current and, therefore, depolarizing or excitatory effects in the SIP syncytium. PDGFRα+ cells express Ca2+-activated K+ channels encoded by Kcnn3. These channels generate outward current when activated and hyperpolarizing or membrane-stabilizing effects in the SIP syncytium. Inputs from enteric and sympathetic neurons regulate Ca2+ transients in ICC and PDGFRα+ cells, and currents activated in these cells conduct to SMCs and regulate contractile behaviors. ICC also serve as pacemakers, generating slow waves that are the electrophysiological basis for gastric peristalsis and intestinal segmentation. Pacemaker types of ICC express voltage-dependent Ca2+ conductances that organize Ca2+ transients, and therefore Ano1 channel openings, into clusters that define the amplitude and duration of slow waves. Ca2+ handling mechanisms are at the heart of interstitial cell function, yet little is known about what happens to Ca2+ dynamics in these cells in GI motility disorders.
Collapse
Affiliation(s)
- Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, NV, USA.
| | - Salah A Baker
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, NV, USA
| | - Bernard T Drumm
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, NV, USA
| | - Masaaki Kurahashi
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, NV, USA
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Iowa, Iowa, Iowa City, USA
| |
Collapse
|
8
|
Zhao W, Zhang L, Ermilov LG, Colmenares Aguilar MG, Linden DR, Eisenman ST, Romero MF, Farrugia G, Sha L, Gibbons SJ. Bicarbonate ion transport by the electrogenic Na + /HCO 3- cotransporter, NBCe1, is required for normal electrical slow-wave activity in mouse small intestine. Neurogastroenterol Motil 2021; 33:e14149. [PMID: 33837991 PMCID: PMC8485339 DOI: 10.1111/nmo.14149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 02/26/2021] [Accepted: 03/21/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Normal gastrointestinal motility depends on electrical slow-wave activity generated by interstitial cells of Cajal (ICC) in the tunica muscularis of the gastrointestinal tract. A requirement for HCO3- in extracellular solutions used to record slow waves indicates a role for HCO3- transport in ICC pacemaking. The Slc4a4 gene transcript encoding the electrogenic Na+ /HCO3- cotransporter, NBCe1, is enriched in mouse small intestinal myenteric region ICC (ICC-MY) that generate slow waves. This study aimed to determine how extracellular HCO3- concentrations affect electrical activity in mouse small intestine and to determine the contribution of NBCe1 activity to these effects. METHODS Immunohistochemistry and sharp electrode electrical recordings were used. KEY RESULTS The NBCe1 immunoreactivity was localized to ICC-MY of the tunica muscularis. In sharp electrode electrical recordings, removal of HCO3- from extracellular solutions caused significant, reversible, depolarization of the smooth muscle and a reduction in slow-wave amplitude and frequency. In 100 mM HCO3- , the muscle hyperpolarized and slow wave amplitude and frequency increased. The effects of replacing extracellular Na+ with Li+ , an ion that does not support NBCe1 activity, were similar to, but larger than, the effects of removing HCO3- . There were no additional changes to electrical activity when HCO3- was removed from Li+ containing solutions. The Na+ /HCO3- cotransport inhibitor, S-0859 (30µM) significantly reduced the effect of removing HCO3- on electrical activity. CONCLUSIONS & INFERENCES These studies demonstrate a major role for Na+ /HCO3- cotransport by NBCe1 in electrical activity of mouse small intestine and indicated that regulation of intracellular acid:base homeostasis contributes to generation of normal pacemaker activity in the gastrointestinal tract.
Collapse
Affiliation(s)
- Wenchang Zhao
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Rochester, Minnesota, USA.,Physiology and Biomedical Engineering, Rochester, Minnesota, USA.,Neuroendocrine Pharmacology, China Medical University, Shenyang, Liaoning Province, P. R. China
| | - Liwen Zhang
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Rochester, Minnesota, USA.,Physiology and Biomedical Engineering, Rochester, Minnesota, USA.,Neuroendocrine Pharmacology, China Medical University, Shenyang, Liaoning Province, P. R. China
| | - Leonid G. Ermilov
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Rochester, Minnesota, USA.,Physiology and Biomedical Engineering, Rochester, Minnesota, USA
| | - Maria Gabriela Colmenares Aguilar
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Rochester, Minnesota, USA.,Physiology and Biomedical Engineering, Rochester, Minnesota, USA
| | - David R. Linden
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Rochester, Minnesota, USA.,Physiology and Biomedical Engineering, Rochester, Minnesota, USA
| | - Seth T. Eisenman
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Rochester, Minnesota, USA.,Physiology and Biomedical Engineering, Rochester, Minnesota, USA
| | - Michael F. Romero
- Physiology and Biomedical Engineering, Rochester, Minnesota, USA.,Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Gianrico Farrugia
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Rochester, Minnesota, USA.,Physiology and Biomedical Engineering, Rochester, Minnesota, USA
| | - Lei Sha
- Neuroendocrine Pharmacology, China Medical University, Shenyang, Liaoning Province, P. R. China.,Corresponding Authors: Simon J Gibbons, Ph.D., Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, 55905, USA. . Telephone: +1 507 284 9652, Lei Sha, M.D., China Medical University, 77 Pu He Road, Shenbei New District, Shenyang, Liaoning Province, P. R. China, 110122, , . Telephone: +86 18900911003
| | - Simon J. Gibbons
- Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Rochester, Minnesota, USA.,Physiology and Biomedical Engineering, Rochester, Minnesota, USA.,Corresponding Authors: Simon J Gibbons, Ph.D., Enteric Neuroscience Program, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, 55905, USA. . Telephone: +1 507 284 9652, Lei Sha, M.D., China Medical University, 77 Pu He Road, Shenbei New District, Shenyang, Liaoning Province, P. R. China, 110122, , . Telephone: +86 18900911003
| |
Collapse
|
9
|
Fu BB, Zhao JN, Wu SD, Fan Y. Cholesterol gallstones: Focusing on the role of interstitial Cajal-like cells. World J Clin Cases 2021; 9:3498-3505. [PMID: 34046450 PMCID: PMC8130069 DOI: 10.12998/wjcc.v9.i15.3498] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 01/08/2021] [Accepted: 03/17/2021] [Indexed: 02/06/2023] Open
Abstract
Cholesterol gallstone (CG) is a common, frequent biliary system disease in China, with a complex and multifactorial etiology. Declined gallbladder motility reportedly contributes to CG pathogenesis. Furthermore, interstitial Cajal-like cells (ICLCs) are reportedly present in human and guinea pig gallbladder tissue. ICLCs potentially contribute to the regulation of gallbladder motility, and aberrant conditions involving the loss of ICLCs and/or a reduction in its pacing potential and reactivity to cholecystokinin may promote CG pathogenesis. This review discusses the association between ICLCs and CG pathogenesis and provides a basis for further studies on the functions of ICLCs and the etiologies of CG.
Collapse
Affiliation(s)
- Bei-Bei Fu
- Department of Health Management, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Jian-Nan Zhao
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Shuo-Dong Wu
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Ying Fan
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| |
Collapse
|
10
|
Liu JYH, Du P, Lu Z, Kung JSC, Huang IB, Hui JCM, Ng HSH, Ngan MP, Cui D, Jiang B, Chan SW, Rudd JA. Involvement of TRPV1 and TRPA1 in the modulation of pacemaker potentials in the mouse ileum. Cell Calcium 2021; 97:102417. [PMID: 33962108 DOI: 10.1016/j.ceca.2021.102417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 04/23/2021] [Accepted: 04/26/2021] [Indexed: 11/21/2022]
Abstract
BACKGROUND The roles of transient receptor potential cation channel, subfamily V, member 1 (TRPV1) and subfamily A, member 1 (TRPA1) in mechanisms of gastrointestinal motility are complex. This study aimed to clarify the effects of several TRPV1 and TRPA1 ligands on the electrical potentials generated by pacemaker cells in the mouse-isolated ileum. METHOD The pacemaker potentials of ileal segments of mice were recorded extracellularly using a 60-channel microelectrode array. The dominant frequencies, average waveform periods and propagation velocities were quantified. The effects of TRPV1 and TRPA1 agonist and antagonist were compared with the baseline recordings. RESULTS The electrophysiological recordings showed that capsaicin (30 μM to 3 mM), resiniferatoxin (300 μM), capsazepine (100-300 μM), allyl isothiocyanate (300 μM), isovelleral (300 μM), icilin (300 μM), A-967,079 (10 μM), AP18 (20 μM) and HC-030,031 (50 μM) significantly reduced the pacemaker frequency and increased the waveform period relative to the baseline. Conversely, ruthenium red (300 μM) significantly increased the pacemaker frequency and reduced the waveform period. Capsaicin (3 mM) and AP18 (20 μM) also significantly reduced the propagation velocity. However, all tested antagonists failed to inhibit the effects of agonists. AMG9810 (300 μM), but not A-967,079 (300 μM), significantly inhibited the increases in pacemaker frequency caused by increased temperatures. CONCLUSION Our findings suggest that TRPV1 and TRPA1 play a minor role in regulating pacemaker potentials and that at non-specific actions at other TRP and ion channels most likely contributed to the overall effects on the electrophysiological recordings that we observed.
Collapse
Affiliation(s)
- Julia Y H Liu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China.
| | - Peng Du
- Auckland Bioengineering Institute, University of Auckland, New Zealand
| | - Zengbing Lu
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Jeng S C Kung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Ianto B Huang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Jessica C M Hui
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Heidi S H Ng
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - M P Ngan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Dexuan Cui
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| | - Bin Jiang
- School of Health Sciences, Caritas Institute of Higher Education, Tseung Kwan O, Hong Kong SAR, China
| | - S W Chan
- School of Health Sciences, Caritas Institute of Higher Education, Tseung Kwan O, Hong Kong SAR, China
| | - John A Rudd
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong SAR, China
| |
Collapse
|
11
|
Malik M, Roh M, England SK. Uterine contractions in rodent models and humans. Acta Physiol (Oxf) 2021; 231:e13607. [PMID: 33337577 PMCID: PMC8047897 DOI: 10.1111/apha.13607] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 12/07/2020] [Accepted: 12/11/2020] [Indexed: 12/18/2022]
Abstract
Aberrant uterine contractions can lead to preterm birth and other labour complications and are a significant cause of maternal morbidity and mortality. To investigate the mechanisms underlying dysfunctional uterine contractions, researchers have used experimentally tractable small animal models. However, biological differences between humans and rodents change how researchers select their animal model and interpret their results. Here, we provide a general review of studies of uterine excitation and contractions in mice, rats, guinea pigs, and humans, in an effort to introduce new researchers to the field and help in the design and interpretation of experiments in rodent models.
Collapse
Affiliation(s)
- Manasi Malik
- Center for Reproductive Health SciencesDepartment of Obstetrics and GynecologyWashington University School of MedicineSt. LouisMOUSA
| | - Michelle Roh
- Center for Reproductive Health SciencesDepartment of Obstetrics and GynecologyWashington University School of MedicineSt. LouisMOUSA
| | - Sarah K. England
- Center for Reproductive Health SciencesDepartment of Obstetrics and GynecologyWashington University School of MedicineSt. LouisMOUSA
| |
Collapse
|
12
|
Lee JY, Zheng H, Sanders KM, Koh SD. Low-voltage-activated inward current in murine antral smooth muscle cells is an artifact. Am J Physiol Cell Physiol 2021; 320:C966-C973. [PMID: 33788632 DOI: 10.1152/ajpcell.00031.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Two types of voltage-dependent inward currents were evoked by depolarization in murine antral smooth muscle cells (SMCs) bathed in Ca2+-containing physiological solution: high-voltage-activated (HVA) and low-voltage-activated (LVA) inward currents. We examined whether the LVA current was due to: 1) T-type Ca2+ channels, 2) Ca2+-activated Cl-channels, 3) nonselective cation channels (NSCC), or 4) voltage-dependent K+ channels. Replacement of external Ca2+ (2 mM) with equimolar Ba2+ increased the amplitude of the HVA current but blocked the LVA current. Nicardipine blocked the HVA current, and in the presence of nicardipine, T-type Ca2+ blockers failed to block LVA current. A Cl- channel antagonist had little effect on LVA current. Cation-free external solution completely abolished both HVA and LVA currents. Addition of Ca2+ to the solution restored only HVA currents. Addition of K+ (5 mM) to otherwise cation-free solution induced LVA current that reversed at -20 mV. These data suggest that LVA current is not due to T-type Ca2+ channels, Ca2+-activated Cl- channels, or NSCC. A-type K+ (KA) currents and delayed rectifying K+ (KDR) currents can be resolved in antral SMCs dialyzed with a solution containing 140 mM K+. When cells were exposed to high K+ external solution and dialyzed with Cs+-rich solution in the presence of nicardipine, LVA current was evoked and reversed at positive potentials. LVA currents were blocked by K+ channel blockers, 4-aminopyridine, and tetraethylammonium. In conclusion, LVA inward currents can be generated by K+ influx via KA channels in murine antral SMCs when cells were dialyzed with Cs+-rich solution.
Collapse
Affiliation(s)
- Ji Yeon Lee
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Haifeng Zheng
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Sang Don Koh
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| |
Collapse
|
13
|
Baker SA, Leigh WA, Del Valle G, De Yturriaga IF, Ward SM, Cobine CA, Drumm BT, Sanders KM. Ca 2+ signaling driving pacemaker activity in submucosal interstitial cells of Cajal in the murine colon. eLife 2021; 10:64099. [PMID: 33399536 PMCID: PMC7806270 DOI: 10.7554/elife.64099] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 01/04/2021] [Indexed: 02/06/2023] Open
Abstract
Interstitial cells of Cajal (ICC) generate pacemaker activity responsible for phasic contractions in colonic segmentation and peristalsis. ICC along the submucosal border (ICC-SM) contribute to mixing and more complex patterns of colonic motility. We show the complex patterns of Ca2+ signaling in ICC-SM and the relationship between ICC-SM Ca2+ transients and activation of smooth muscle cells (SMCs) using optogenetic tools. ICC-SM displayed rhythmic firing of Ca2+transients ~ 15 cpm and paced adjacent SMCs. The majority of spontaneous activity occurred in regular Ca2+ transients clusters (CTCs) that propagated through the network. CTCs were organized and dependent upon Ca2+ entry through voltage-dependent Ca2+ conductances, L- and T-type Ca2+ channels. Removal of Ca2+ from the external solution abolished CTCs. Ca2+ release mechanisms reduced the duration and amplitude of Ca2+ transients but did not block CTCs. These data reveal how colonic pacemaker ICC-SM exhibit complex Ca2+-firing patterns and drive smooth muscle activity and overall colonic contractions.
Collapse
Affiliation(s)
- Salah A Baker
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, United States
| | - Wesley A Leigh
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, United States
| | - Guillermo Del Valle
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, United States
| | - Inigo F De Yturriaga
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, United States
| | - Sean M Ward
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, United States
| | - Caroline A Cobine
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, United States
| | - Bernard T Drumm
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, United States
| | - Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada, Reno, School of Medicine, Reno, United States
| |
Collapse
|
14
|
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
|
15
|
Zheng H, Drumm BT, Zhu MH, Xie Y, O'Driscoll KE, Baker SA, Perrino BA, Koh SD, Sanders KM. Na +/Ca 2 + Exchange and Pacemaker Activity of Interstitial Cells of Cajal. Front Physiol 2020; 11:230. [PMID: 32256387 PMCID: PMC7093646 DOI: 10.3389/fphys.2020.00230] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 02/27/2020] [Indexed: 01/30/2023] Open
Abstract
Interstitial cells of Cajal (ICC) are pacemaker cells that generate electrical slow waves in gastrointestinal (GI) smooth muscles. Slow waves organize basic motor patterns, such as peristalsis and segmentation in the GI tract. Slow waves depend upon activation of Ca2+-activated Cl– channels (CaCC) encoded by Ano1. Slow waves consist of an upstroke depolarization and a sustained plateau potential that is the main factor leading to excitation-contraction coupling. The plateau phase can last for seconds in some regions of the GI tract. How elevated Ca2+ is maintained throughout the duration of slow waves, which is necessary for sustained activation of CaCC, is unknown. Modeling has suggested a role for Na+/Ca2+ exchanger (NCX) in regulating CaCC currents in ICC, so we tested this idea on murine intestinal ICC. ICC of small and large intestine express NCX isoforms. NCX3 is closely associated with ANO1 in ICC, as shown by immunoprecipitation and proximity ligation assays (PLA). KB-R7943, an inhibitor of NCX, increased CaCC current in ICC, suggesting that NCX, acting in Ca2+ exit mode, helps to regulate basal [Ca2+]i in these cells. Shifting NCX into Ca2+ entry mode by replacing extracellular Na+ with Li+ increased spontaneous transient inward currents (STICs), due to activation of CaCC. Stepping ICC from −80 to −40 mV activated slow wave currents that were reduced in amplitude and duration by NCX inhibitors, KB-R7943 and SN-6, and enhanced by increasing the NCX driving force. SN-6 reduced the duration of clustered Ca2+ transients that underlie the activation of CaCC and the plateau phase of slow waves. Our results suggest that NCX participates in slow waves as modeling has predicted. Dynamic changes in membrane potential and ionic gradients during slow waves appear to flip the directionality of NCX, facilitating removal of Ca2+ during the inter-slow wave interval and providing Ca2+ for sustained activation of ANO1 during the slow wave plateau phase.
Collapse
Affiliation(s)
- Haifeng Zheng
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, United States
| | - Bernard T Drumm
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, United States
| | - Mei Hong Zhu
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, United States
| | - Yeming Xie
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, United States
| | - Kate E O'Driscoll
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, United States
| | - Salah A Baker
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, United States
| | - Brian A Perrino
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, United States
| | - Sang Don Koh
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, United States
| | - Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, United States
| |
Collapse
|
16
|
Role of Pericytes in the Initiation and Propagation of Spontaneous Activity in the Microvasculature. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1124:329-356. [PMID: 31183834 DOI: 10.1007/978-981-13-5895-1_14] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The microvasculature is composed of arterioles, capillaries and venules. Spontaneous arteriolar constrictions reduce effective vascular resistance to enhance tissue perfusion, while spontaneous venular constrictions facilitate the drainage of tissue metabolites by pumping blood. In the venules of visceral organs, mural cells, i.e. smooth muscle cells (SMCs) or pericytes, periodically generate spontaneous phasic constrictions, Ca2+ transients and transient depolarisations. These events arise from spontaneous Ca2+ release from the sarco-endoplasmic reticulum (SR/ER) and the subsequent opening of Ca2+-activated chloride channels (CaCCs). CaCC-dependent depolarisation further activates L-type voltage-dependent Ca2+ channels (LVDCCs) that play a critical role in maintaining the synchrony amongst mural cells. Mural cells in arterioles or capillaries are also capable of developing spontaneous activity. Non-contractile capillary pericytes generate spontaneous Ca2+ transients primarily relying on SR/ER Ca2+ release. Synchrony amongst capillary pericytes depends on gap junction-mediated spread of depolarisations resulting from the opening of either CaCCs or T-type VDCCs (TVDCCs) in a microvascular bed-dependent manner. The propagation of capillary Ca2+ transients into arterioles requires the opening of either L- or TVDCCs again depending on the microvascular bed. Since the blockade of gap junctions or CaCCs prevents spontaneous Ca2+ transients in arterioles and venules but not capillaries, capillary pericytes appear to play a primary role in generating spontaneous activity of the microvasculature unit. Pericytes in capillaries where the interchange of substances between tissues and the circulation takes place may provide the fundamental drive for upstream arterioles and downstream venules so that the microvasculature network functions as an integrated unit.
Collapse
|
17
|
Use of a microelectrode array to record extracellular pacemaker potentials from the gastrointestinal tracts of the ICR mouse and house musk shrew (Suncus murinus). Cell Calcium 2019; 80:175-188. [DOI: 10.1016/j.ceca.2019.05.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/23/2019] [Accepted: 05/08/2019] [Indexed: 12/17/2022]
|
18
|
Hwang SJ, Pardo DM, Zheng H, Bayguinov Y, Blair PJ, Fortune‐Grant R, Cook RS, Hennig GW, Shonnard MC, Grainger N, Peri LE, Verma SD, Rock J, Sanders KM, Ward SM. Differential sensitivity of gastric and small intestinal muscles to inducible knockdown of anoctamin 1 and the effects on gastrointestinal motility. J Physiol 2019; 597:2337-2360. [PMID: 30843201 PMCID: PMC6487927 DOI: 10.1113/jp277335] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 02/22/2019] [Indexed: 12/13/2022] Open
Abstract
KEY POINTS Electrical pacemaking in gastrointestinal muscles is generated by specialized interstitial cells of Cajal that produce the patterns of contractions required for peristalsis and segmentation in the gut. The calcium-activated chloride conductance anoctamin-1 (Ano1) has been shown to be responsible for the generation of pacemaker activity in GI muscles, but this conclusion is established from studies of juvenile animals in which effects of reduced Ano1 on gastric emptying and motor patterns could not be evaluated. Knocking down Ano1 expression using Cre/LoxP technology caused dramatic changes in in gastric motor activity, with disrupted slow waves, abnormal phasic contractions and delayed gastric emptying; modest changes were noted in the small intestine. Comparison of the effects of Ano1 antagonists on muscles from juvenile and adult small intestinal muscles suggests that conductances in addition to Ano1 may develop with age and contribute to pacemaker activity. ABSTRACT Interstitial cells of Cajal (ICC) generate slow waves and transduce neurotransmitter signals in the gastrointestinal (GI) tract, facilitating normal motility patterns. ICC express a Ca2+ -activated Cl- conductance (CaCC), and constitutive knockout of the channel protein anoctamin-1 leads to loss of slow waves in gastric and intestinal muscles. These knockout experiments were performed on juvenile mice. However, additional experiments demonstrated significant differences in the sensitivity of gastric and intestinal muscles to antagonists of anoctamin-1 channels. Furthermore, the significance of anoctamin-1 and the electrical and mechanical behaviours facilitated by this conductance have not been evaluated on the motor behaviours of adult animals. Cre/loxP technology was used to generate cell-specific knockdowns of anoctamin-1 in ICC (KitCreERT2/+ ;Ano1tm2jrr/+ ) in GI muscles. The recombination efficiency of KitCreERT was evaluated with an eGFP reporter, molecular techniques and immunohistochemistry. Electrical and contractile experiments were used to examine the consequences of anoctamin-1 knockdown on pacemaker activity, mechanical responses, gastric motility patterns, gastric emptying and GI transit. Reduced anoctamin-1 caused loss of gastric, but not intestinal slow waves. Irregular spike complexes developed in gastric muscles, leading to uncoordinated antral contractions, delayed gastric emptying and increased total GI transit time. Slow waves in intestinal muscles of juvenile mice were more sensitive to anoctamin-1 antagonists than slow waves in adult muscles. The low susceptibility to anoctamin-1 knockdown and weak efficacy of anoctamin-1 antagonists in inhibiting slow waves in adult small intestinal muscles suggest that a conductance in addition to anoctamin-1 may develop in small intestinal ICC with ageing and contribute to pacemaker activity.
Collapse
Affiliation(s)
- Sung Jin Hwang
- Department of Physiology & Cell BiologyUniversity of NevadaReno School of MedicineRenoNV89557USA
| | - David M. Pardo
- Department of AnatomyUniversity of CaliforniaSan FranciscoSan FranciscoCA94143USA
| | - Haifeng Zheng
- Department of Physiology & Cell BiologyUniversity of NevadaReno School of MedicineRenoNV89557USA
| | - Yulia Bayguinov
- Department of Physiology & Cell BiologyUniversity of NevadaReno School of MedicineRenoNV89557USA
| | - Peter J. Blair
- Department of Physiology & Cell BiologyUniversity of NevadaReno School of MedicineRenoNV89557USA
| | - Rachael Fortune‐Grant
- Faculty of BiologyMedicine and HealthSchool of Biological SciencesUniversity of ManchesterUK
| | - Robert S. Cook
- School of Molecular and Cellular BiologyUniversity of LeedsLeedsLS2 9JTUK
| | - Grant W. Hennig
- Department of PharmacologyThe University of VermontUVM College of MedicineBurlingtonVT05405USA
| | - Matthew C. Shonnard
- Department of Physiology & Cell BiologyUniversity of NevadaReno School of MedicineRenoNV89557USA
| | - Nathan Grainger
- Department of Physiology & Cell BiologyUniversity of NevadaReno School of MedicineRenoNV89557USA
| | - Lauren E. Peri
- Department of Physiology & Cell BiologyUniversity of NevadaReno School of MedicineRenoNV89557USA
| | - Sonali Deep Verma
- Department of AnatomyUniversity of CaliforniaSan FranciscoSan FranciscoCA94143USA
| | - Jason Rock
- Centre for Regenerative MedicineBoston University School of MedicineBostonMA02118USA
| | - Kenton M. Sanders
- Department of Physiology & Cell BiologyUniversity of NevadaReno School of MedicineRenoNV89557USA
| | - Sean M. Ward
- Department of Physiology & Cell BiologyUniversity of NevadaReno School of MedicineRenoNV89557USA
| |
Collapse
|
19
|
|
20
|
Sanders KM. Spontaneous Electrical Activity and Rhythmicity in Gastrointestinal Smooth Muscles. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1124:3-46. [PMID: 31183821 PMCID: PMC7035145 DOI: 10.1007/978-981-13-5895-1_1] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The gastrointestinal (GI) tract has multifold tasks of ingesting, processing, and assimilating nutrients and disposing of wastes at appropriate times. These tasks are facilitated by several stereotypical motor patterns that build upon the intrinsic rhythmicity of the smooth muscles that generate phasic contractions in many regions of the gut. Phasic contractions result from a cyclical depolarization/repolarization cycle, known as electrical slow waves, which result from intrinsic pacemaker activity. Interstitial cells of Cajal (ICC) are electrically coupled to smooth muscle cells (SMCs) and generate and propagate pacemaker activity and slow waves. The mechanism of slow waves is dependent upon specialized conductances expressed by pacemaker ICC. The primary conductances responsible for slow waves in mice are Ano1, Ca2+-activated Cl- channels (CaCCs), and CaV3.2, T-type, voltage-dependent Ca2+ channels. Release of Ca2+ from intracellular stores in ICC appears to be the initiator of pacemaker depolarizations, activation of T-type current provides voltage-dependent Ca2+ entry into ICC, as slow waves propagate through ICC networks, and Ca2+-induced Ca2+ release and activation of Ano1 in ICC amplifies slow wave depolarizations. Slow waves conduct to coupled SMCs, and depolarization elicited by these events enhances the open-probability of L-type voltage-dependent Ca2+ channels, promotes Ca2+ entry, and initiates contraction. Phasic contractions timed by the occurrence of slow waves provide the basis for motility patterns such as gastric peristalsis and segmentation. This chapter discusses the properties of ICC and proposed mechanism of electrical rhythmicity in GI muscles.
Collapse
Affiliation(s)
- Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, USA.
| |
Collapse
|
21
|
Abstract
Veins exhibit spontaneous contractile activity, a phenomenon generally termed vasomotion. This is mediated by spontaneous rhythmical contractions of mural cells (i.e. smooth muscle cells (SMCs) or pericytes) in the wall of the vessel. Vasomotion occurs through interconnected oscillators within and between mural cells, entraining their cycles. Pharmacological studies indicate that a key oscillator underlying vasomotion is the rhythmical calcium ion (Ca2+) release-refill cycle of Ca2+ stores. This occurs through opening of inositol 1,4,5-trisphosphate receptor (IP3R)- and/or ryanodine receptor (RyR)-operated Ca2+ release channels in the sarcoplasmic/endoplasmic (SR/ER) reticulum and refilling by the SR/ER reticulum Ca2+ATPase (SERCA). Released Ca2+ from stores near the plasma membrane diffuse through the cytosol to open Ca2+-activated chloride (Cl-) channels, this generating inward current through an efflux of Cl-. The resultant depolarisation leads to the opening of voltage-dependent Ca2+ channels and possibly increased production of IP3, which through Ca2+-induced Ca2+ release (CICR) of IP3Rs and/or RyRs and IP3R-mediated Ca2+ release provide a means by which store oscillators entrain their activity. Intercellular entrainment normally involves current flow through gap junctions that interconnect mural cells and in many cases this is aided by additional connectivity through the endothelium. Once entrainment has occurred the substantial Ca2+ entry that results from the near-synchronous depolarisations leads to rhythmical contractions of the mural cells, this often leading to vessel constriction. The basis for venous/venular vasomotion has yet to be fully delineated but could improve both venous drainage and capillary/venular absorption of blood plasma-associated fluids.
Collapse
|
22
|
Zheng H, Drumm BT, Earley S, Sung TS, Koh SD, Sanders KM. SOCE mediated by STIM and Orai is essential for pacemaker activity in the interstitial cells of Cajal in the gastrointestinal tract. Sci Signal 2018; 11:eaaq0918. [PMID: 29895614 PMCID: PMC6310020 DOI: 10.1126/scisignal.aaq0918] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Electrical pacemaker activity generates phasic contractions and motility patterns such as segmentation and peristalsis in the gastrointestinal tract. Pacemaker currents are generated in interstitial cells of Cajal (ICC), which release Ca2+ from intracellular stores that stimulates Ca2+-activated Cl- channels (CaCCs) in the plasma membrane. Thus, Ca2+ stores must be maintained to sustain pacemaker activity. Store-operated Ca2+ entry (SOCE) facilitates the refilling of Ca2+ stores by a mechanism dependent upon interactions between STIM and Orai proteins. We investigated the role of SOCE in ICC pacemaker activity. Reintroduction of extracellular Ca2+ in store-depleted ICC resulted in CaCC activation. Blocking CaCCs revealed an inwardly rectifying current with properties of a Ca2+ release-activated current (ICRAC). An inhibitory peptide that interfered with the STIM-Orai interaction blocked ICRAC in HEK 293 cells expressing STIM1 and Orai1 and blocked spontaneous transient inward currents (STICs) and slow wave currents in ICC. STICs, which are fundamental pacemaker events in ICC, were blocked by an Orai antagonist. Imaging of Ca2+ transients linked to pacemaker activity in ICC in intact muscles showed that the Orai antagonist blocked Ca2+ transients in ICC. These data suggest that Ca2+ recovery through STIM-Orai interactions is necessary to maintain ICC pacemaker activity.
Collapse
Affiliation(s)
- Haifeng Zheng
- 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
| | - Scott Earley
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
| | - Tae Sik Sung
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
| | - Sang Don Koh
- 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.
| |
Collapse
|
23
|
Drumm BT, Sung TS, Zheng H, Baker SA, Koh SD, Sanders KM. The effects of mitochondrial inhibitors on Ca 2+ signalling and electrical conductances required for pacemaking in interstitial cells of Cajal in the mouse small intestine. Cell Calcium 2018; 72:1-17. [PMID: 29748128 DOI: 10.1016/j.ceca.2018.01.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 01/26/2018] [Accepted: 01/27/2018] [Indexed: 01/16/2023]
Abstract
Interstitial cells of Cajal (ICC-MY) are pacemakers that generate and propagate electrical slow waves in gastrointestinal (GI) muscles. Slow waves appear to be generated by the release of Ca2+ from intracellular stores and activation of Ca2+-activated Cl- channels (Ano1). Conduction of slow waves to smooth muscle cells coordinates rhythmic contractions. Mitochondrial Ca2+ handling is currently thought to be critical for ICC pacemaking. Protonophores, inhibitors of the electron transport chain (FCCP, CCCP or antimycin) or mitochondrial Na+/Ca2+ exchange blockers inhibited slow waves in several GI muscles. Here we utilized Ca2+ imaging of ICC in small intestinal muscles in situ to determine the effects of mitochondrial drugs on Ca2+ transients in ICC. Muscles were obtained from mice expressing a genetically encoded Ca2+ indicator (GCaMP3) in ICC. FCCP, CCCP, antimycin, a uniporter blocker, Ru360, and a mitochondrial Na+/Ca2+ exchange inhibitor, CGP-37157 inhibited Ca2+ transients in ICC-MY. Effects were not due to depletion of ATP, as oligomycin did not affect Ca2+ transients. Patch-clamp experiments were performed to test the effects of the mitochondrial drugs on key pacemaker conductances, Ano1 and T-type Ca2+ (CaV3.2), in HEK293 cells. Antimycin blocked Ano1 and reduced CaV3.2 currents. CCCP blocked CaV3.2 current but did not affect Ano1 current. Ano1 and Cav3.2 currents were inhibited by CGP-37157. Inhibitory effects of mitochondrial drugs on slow waves and Ca2+ signalling in ICC can be explained by direct antagonism of key pacemaker conductances in ICC that generate and propagate slow waves. A direct obligatory role for mitochondria in pacemaker activity is therefore questionable.
Collapse
Affiliation(s)
- Bernard T Drumm
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA
| | - Tae S Sung
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV 89557, USA
| | - Haifeng Zheng
- 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
| | - Sang D Koh
- 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
|
24
|
Park CG, Wu MJ, Hong C, Jo JY, Jiao HY, Park H, Jun JY, Choi S. Regulation of Intracellular Calcium by Endoplasmic Reticulum Proteins in Small Intestinal Interstitial Cells of Cajal. J Neurogastroenterol Motil 2018; 24:128-137. [PMID: 28774158 PMCID: PMC5753911 DOI: 10.5056/jnm16212] [Citation(s) in RCA: 7] [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: 12/13/2016] [Revised: 03/13/2017] [Accepted: 04/07/2017] [Indexed: 12/23/2022] Open
Abstract
Background/Aims We investigated the role of representative endoplasmic reticulum proteins, stromal interaction molecule 1 (STIM1), and store-operated calcium entry-associated regulatory factor (SARAF) in pacemaker activity in cultured interstitial cells of Cajal (ICCs) isolated from mouse small intestine. Methods The whole-cell patch clamp technique applied for intracellular calcium ions ([Ca2+]i) analysis with STIM1 or SARAF overexpressed cultured ICCs from mouse small intestine. Results In the current-clamping mode, cultured ICCs displayed spontaneous pacemaker potentials. External carbachol exposure produced tonic membrane depolarization in the current-clamp mode, which recovered within a few seconds into normal pacemaker potentials. In STIM1-overexpressing cultured ICCs pacemaker potential frequency was increased, and in SARAF-overexpressing ICCs pacemaker potential frequency was strongly inhibited. The application of gadolinium (a non-selective cation channel inhibitor) or a Ca2+-free solution to understand Orai channel involvement abolished the generation of pacemaker potentials. When recording intracellular Ca2+ concentration with Fluo 3-AM, STIM1-overexpressing ICCs showed an increased number of spontaneous intracellular Ca2+ oscillations. However, SARAF-overexpressing ICCs showed fewer spontaneous intracellular Ca2+ oscillations. Conclusion Endoplasmic reticulum proteins modulated the frequency of pacemaker activity in ICCs, and levels of STIM1 and SARAF may determine slow wave patterns in the gastrointestinal tract.
Collapse
Affiliation(s)
- Chan Guk Park
- Department of Internal Medicine, College of Medicine, Chosun University, Gwangju, Korea
| | - Mei Jin Wu
- Department of Medicine, Graduate School, Chosun University, Gwangju, Korea
| | - Chansik Hong
- Department of Medicine, Graduate School, Chosun University, Gwangju, Korea
| | - Ju Yeon Jo
- Department of Internal Medicine, College of Medicine, Chosun University, Gwangju, Korea
| | - Han Yi Jiao
- Department of Medicine, Graduate School, Chosun University, Gwangju, Korea
| | - Hyun Park
- Department of Physiology, College of Medicine, Chosun University, Gwangju, Korea
| | - Jae Yeoul Jun
- Department of Medicine, Graduate School, Chosun University, Gwangju, Korea
| | - Seok Choi
- Department of Medicine, Graduate School, Chosun University, Gwangju, Korea
| |
Collapse
|
25
|
Sedwick C. Many parts make a whole: Calcium transients sum for slow waves. J Gen Physiol 2017; 149:681. [PMID: 28619745 PMCID: PMC5496511 DOI: 10.1085/jgp.201711831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
New JGP study shows how calcium events drive long intestinal slow wave plateaus.
Collapse
|
26
|
Drumm BT, Hennig GW, Battersby MJ, Cunningham EK, Sung TS, Ward SM, Sanders KM, Baker SA. Clustering of Ca 2+ transients in interstitial cells of Cajal defines slow wave duration. J Gen Physiol 2017; 149:703-725. [PMID: 28592421 PMCID: PMC5496507 DOI: 10.1085/jgp.201711771] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 05/02/2017] [Indexed: 12/13/2022] Open
Abstract
Electrical slow waves in the small intestine are generated by pacemaker cells called interstitial cells of Cajal. Drumm et al. record clusters of Ca2+ transients in these cells that are entrained by voltage-dependent Ca2+ entry and which define the duration of the electrical slow waves. Interstitial cells of Cajal (ICC) in the myenteric plexus region (ICC-MY) of the small intestine are pacemakers that generate rhythmic depolarizations known as slow waves. Slow waves depend on activation of Ca2+-activated Cl− channels (ANO1) in ICC, propagate actively within networks of ICC-MY, and conduct to smooth muscle cells where they generate action potentials and phasic contractions. Thus, mechanisms of Ca2+ regulation in ICC are fundamental to the motor patterns of the bowel. Here, we characterize the nature of Ca2+ transients in ICC-MY within intact muscles, using mice expressing a genetically encoded Ca2+ sensor, GCaMP3, in ICC. Ca2+ transients in ICC-MY display a complex firing pattern caused by localized Ca2+ release events arising from multiple sites in cell somata and processes. Ca2+ transients are clustered within the time course of slow waves but fire asynchronously during these clusters. The durations of Ca2+ transient clusters (CTCs) correspond to slow wave durations (plateau phase). Simultaneous imaging and intracellular electrical recordings revealed that the upstroke depolarization of slow waves precedes clusters of Ca2+ transients. Summation of CTCs results in relatively uniform Ca2+ responses from one slow wave to another. These Ca2+ transients are caused by Ca2+ release from intracellular stores and depend on ryanodine receptors as well as amplification from IP3 receptors. Reduced extracellular Ca2+ concentrations and T-type Ca2+ channel blockers decreased the number of firing sites and firing probability of Ca2+ transients. In summary, the fundamental electrical events of small intestinal muscles generated by ICC-MY depend on asynchronous firing of Ca2+ transients from multiple intracellular release sites. These events are organized into clusters by Ca2+ influx through T-type Ca2+ channels to sustain activation of ANO1 channels and generate the plateau phase of slow waves.
Collapse
Affiliation(s)
- Bernard T Drumm
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV
| | - Grant W Hennig
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV
| | - Matthew J Battersby
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV
| | - Erin K Cunningham
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV
| | - Tae Sik Sung
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV
| | - Sean M Ward
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV
| | - Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV
| | - Salah A Baker
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV
| |
Collapse
|
27
|
Sánchez M, Suárez L, Andrés MT, Flórez BH, Bordallo J, Riestra S, Cantabrana B. Modulatory effect of intestinal polyamines and trace amines on the spontaneous phasic contractions of the isolated ileum and colon rings of mice. Food Nutr Res 2017; 61:1321948. [PMID: 28659731 PMCID: PMC5475348 DOI: 10.1080/16546628.2017.1321948] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 03/20/2017] [Indexed: 11/08/2022] Open
Abstract
Background: Gastrointestinal motility modulatory factors include substances of the intestinal content, such as polyamines and trace amines (TAs), the focus of this study. Methods: The amines of food, intestinal content and from faecal bacteria of Swiss mice were determined by HPLC and functionally characterised in isolated distal ileum and medial colon rings. Results: Mouse food and intestinal content contain polyamines (spermidine>putrescine>spermine) and TAs (isoamylamine>cadaverine). Intestinal bacteria mainly produce putrescine and cadaverine. The amines inhibited the spontaneous motility of the ileum (0.1-3 mM) and colon rings (0.01-3 mM, with lower IC50), with: spermine~isoamylamine~spermidine. Spermine inhibition was tetrodotoxin (TTX)-insensitive, while isoamylamine was TTX-sensitive, suggesting neural control. Mainly in the ileum, isoamylamine (3 mM) elicited acute effects modified by TTX, atropine and propranolol, and suppressed by spermine (3 mM), not being localized at the smooth muscle level. The amines assayed (3 mM), except putrescine and cadaverine in the ileum and isoamylamine in the colon, antagonised acetylcholine (ACh, 0.1 mM)-elicited phasic contractions. Isoamylamine and spermine in colon relaxed KCl (100 mM)-elicited tonic contractions, suggesting an effect on smooth muscle, but did not justify the suppression of motility caused by spermine and isoamylamine. Conclusions: Polyamines and TAs of the intestinal content might act on chemosensors and modulate intestinal peristalsis.
Collapse
Affiliation(s)
- Manuel Sánchez
- Farmacología, Departamento de Medicina, Universidad de Oviedo, Oviedo, Spain.,Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Oviedo, Spain
| | - Lorena Suárez
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Oviedo, Spain
| | - María Teresa Andrés
- Laboratorio de Microbiología Oral, Escuela de Estomatología, Universidad de Oviedo, Oviedo, Spain
| | - Blanca Henar Flórez
- Farmacología, Departamento de Medicina, Universidad de Oviedo, Oviedo, Spain
| | - Javier Bordallo
- Farmacología, Departamento de Medicina, Universidad de Oviedo, Oviedo, Spain.,Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Oviedo, Spain
| | - Sabino Riestra
- Servicio de Aparato Digestivo, Unidad de Enfermedad Inflamatoria Intestinal, Hospital Universitario Central de Asturias (HUCA), Oviedo, Spain
| | - Begoña Cantabrana
- Farmacología, Departamento de Medicina, Universidad de Oviedo, Oviedo, Spain.,Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Oviedo, Spain
| |
Collapse
|
28
|
Sanders KM, Kito Y, Hwang SJ, Ward SM. Regulation of Gastrointestinal Smooth Muscle Function by Interstitial Cells. Physiology (Bethesda) 2017; 31:316-26. [PMID: 27488743 DOI: 10.1152/physiol.00006.2016] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Interstitial cells of mesenchymal origin form gap junctions with smooth muscle cells in visceral smooth muscles and provide important regulatory functions. In gastrointestinal (GI) muscles, there are two distinct classes of interstitial cells, c-Kit(+) interstitial cells of Cajal and PDGFRα(+) cells, that regulate motility patterns. Loss of these cells may contribute to symptoms in GI motility disorders.
Collapse
Affiliation(s)
- Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada Reno School of Medicine, Reno, Nevada; and
| | - Yoshihiko Kito
- Department of Pharmacology, Faculty of Medicine, Saga University, Nabeshima, Japan
| | - Sung Jin Hwang
- Department of Physiology and Cell Biology, University of Nevada Reno School of Medicine, Reno, Nevada; and
| | - Sean M Ward
- Department of Physiology and Cell Biology, University of Nevada Reno School of Medicine, Reno, Nevada; and
| |
Collapse
|
29
|
Cobine CA, Hannah EE, Zhu MH, Lyle HE, Rock JR, Sanders KM, Ward SM, Keef KD. ANO1 in intramuscular interstitial cells of Cajal plays a key role in the generation of slow waves and tone in the internal anal sphincter. J Physiol 2017; 595:2021-2041. [PMID: 28054347 DOI: 10.1113/jp273618] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 12/16/2016] [Indexed: 12/13/2022] Open
Abstract
KEY POINTS The internal anal sphincter develops tone important for maintaining high anal pressure and continence. Controversy exists regarding the mechanisms underlying tone development. We examined the hypothesis that tone depends upon electrical slow waves (SWs) initiated in intramuscular interstitial cells of Cajal (ICC-IM) by activation of Ca2+ -activated Cl- channels (ANO1, encoded by Ano1) and voltage-dependent L-type Ca2+ channels (CavL , encoded by Cacna1c). Measurement of membrane potential and contraction indicated that ANO1 and CavL have a central role in SW generation, phasic contractions and tone, independent of stretch. ANO1 expression was examined in wildtype and Ano1/+egfp mice with immunohistochemical techniques. Ano1 and Cacna1c expression levels were examined by quantitative PCR in fluorescence-activated cell sorting. ICC-IM were the predominant cell type expressing ANO1 and the most likely candidate for SW generation. SWs in ICC-IM are proposed to conduct to smooth muscle where Ca2+ entry via CavL results in phasic activity that sums to produce tone. ABSTRACT The mechanism underlying tone generation in the internal anal sphincter (IAS) is controversial. We examined the hypothesis that tone depends upon generation of electrical slow waves (SWs) initiated in intramuscular interstitial cells of Cajal (ICC-IM) by activation of Ca2+ -activated Cl- channels (encoded by Ano1) and voltage-dependent L-type Ca2+ channels (encoded by Cacna1c). Phasic contractions and tone in the IAS were nearly abolished by ANO1 and CavL antagonists. ANO1 antagonists also abolished SWs as well as transient depolarizations that persisted after addition of CavL antagonists. Tone development in the IAS did not require stretch of muscles, and the sensitivity of contraction to ANO1 antagonists was the same in stretched versus un-stretched muscles. ANO1 expression was examined in wildtype and Ano1/+egfp mice with immunohistochemical techniques. Dual labelling revealed that ANO1 expression could be resolved in ICC but not smooth muscle cells (SMCs) in the IAS and rectum. Ano1, Cacna1c and Kit gene expression were the same in extracts of IAS and rectum muscles. In IAS cells isolated with fluorescence-activated cell sorting, Ano1 expression was 26.5-fold greater in ICC than in SMCs while Cacna1c expression was only 2-fold greater in SMCs than in ICC. These data support a central role for ANO1 and CavL in the generation of SWs and tone in the IAS. ICC-IM are the probable cellular candidate for ANO1 currents and SW generation. We propose that ANO1 and CavL collaborate to generate SWs in ICC-IM followed by conduction to adjacent SMCs where phasic calcium entry through CavL sums to produce tone.
Collapse
Affiliation(s)
- C A Cobine
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, 89557, USA
| | - E E Hannah
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, 89557, USA
| | - M H Zhu
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, 89557, USA
| | - H E Lyle
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, 89557, USA
| | - J R Rock
- Department of Anatomy, UCSF School of Medicine, San Francisco, CA, 94143, USA
| | - K M Sanders
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, 89557, USA
| | - S M Ward
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, 89557, USA
| | - K D Keef
- Department of Physiology and Cell Biology, University of Nevada, Reno School of Medicine, Reno, NV, 89557, USA
| |
Collapse
|
30
|
Guo T, Li J, Li J, Kong D, Bi C, He Z, Tang D, Jin X, Jin L. Association between hyperpolarization-activated channel in interstitial cells of Cajal and gastrointestinal dysmotility induced by malignant ascites. Oncol Lett 2017; 13:1601-1608. [PMID: 28454297 PMCID: PMC5403200 DOI: 10.3892/ol.2017.5652] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 11/03/2016] [Indexed: 12/20/2022] Open
Abstract
Advanced malignant ascites is accompanied by gastrointestinal dysmotility, and patients often feel abdominal pain, abdominal distention, nausea and constipation. Gastrointestinal dysmotility is not only painful for the patients, but it reduces the absorption of nutrients and affects the physical recovery of patients with malignant ascites. It is reported that changes in interstitial cells of Cajal (ICCs) are responsible for the gastrointestinal dysmotility induced by malignant ascites, but the mechanism is not completely understood. The present study observed a significantly decreased expression of ion channels, including hyperpolarization-activated cyclic nucleotide-gated potassium channel 2 (HCN2) and cyclic adenosine monophosphate, in the condition of malignant ascites. Using electrophysiology, it was identified that malignant ascites led to lower amplitude and slower frequency signals in cells of the small intestine. In addition, when ICCs were cultured with malignant ascites in vitro, the expression of HCN2 of ICCs was significantly reduced, and the data of flow cytometry revealed that the Ca2+ concentration of ICCs was also decreased. The results of electron microscopy analysis demonstrated the nuclei of ICCs were pyknotic, and the processes of ICCs were reduced in malignant ascites. The present study suggests the small intestinal dysmotility caused by malignant ascites may be associated with changes in HCN2 of ICCs, which offers a potential therapeutic target for gastrointestinal dysmotility in advanced malignant ascites.
Collapse
Affiliation(s)
- Tieyun Guo
- Department of Histology and Embryology, Basic Medical Science College, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Jiade Li
- Department of Pathology, Basic Medical Science College, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Jing Li
- Department of Pathology, Basic Medical Science College, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Dan Kong
- Department of Gynecology, The Third Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Chunli Bi
- Department of Pathology, Basic Medical Science College, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Zheng He
- Department of Pathology, Basic Medical Science College, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Dai Tang
- Department of Pathology, Basic Medical Science College, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Xiaoming Jin
- Department of Pathology, Basic Medical Science College, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Lianhong Jin
- Department of Histology and Embryology, Basic Medical Science College, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| |
Collapse
|
31
|
Zhu MH, Sung TS, Kurahashi M, O'Kane LE, O'Driscoll K, Koh SD, Sanders KM. Na+-K+-Cl- cotransporter (NKCC) maintains the chloride gradient to sustain pacemaker activity in interstitial cells of Cajal. Am J Physiol Gastrointest Liver Physiol 2016; 311:G1037-G1046. [PMID: 27742704 PMCID: PMC5206290 DOI: 10.1152/ajpgi.00277.2016] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 10/11/2016] [Indexed: 01/31/2023]
Abstract
Interstitial cells of Cajal (ICC) generate electrical slow waves by coordinated openings of ANO1 channels, a Ca2+-activated Cl- (CaCC) conductance. Efflux of Cl- during slow waves must be significant, as there is high current density during slow-wave currents and slow waves are of sufficient magnitude to depolarize the syncytium of smooth muscle cells and PDGFRα+ cells to which they are electrically coupled. We investigated how the driving force for Cl- current is maintained in ICC. We found robust expression of Slc12a2 (which encodes an Na+-K+-Cl- cotransporter, NKCC1) and immunohistochemical confirmation that NKCC1 is expressed in ICC. With the use of the gramicidin permeabilized-patch technique, which is reported to not disturb [Cl-]i, the reversal potential for spontaneous transient inward currents (ESTICs) was -10.5 mV. This value corresponds to the peak of slow waves when they are recorded directly from ICC in situ. Inhibition of NKCC1 with bumetanide shifted ESTICs to more negative potentials within a few minutes and reduced pacemaker activity. Bumetanide had no direct effects on ANO1 or CaV3.2 channels expressed in HEK293 cells or L-type Ca2+ currents. Reducing extracellular Cl- to 10 mM shifted ESTICs to positive potentials as predicted by the Nernst equation. The relatively rapid shift in ESTICs when NKCC1 was blocked suggests that significant changes in the transmembrane Cl- gradient occur during the slow-wave cycle, possibly within microdomains formed between endoplasmic reticulum and the plasma membrane in ICC. Recovery of Cl- via NKCC1 might have additional consequences on shaping the waveforms of slow waves via Na+ entry into microdomains.
Collapse
Affiliation(s)
- Mei Hong Zhu
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Tae Sik Sung
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Masaaki Kurahashi
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Lauren E. O'Kane
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Kate O'Driscoll
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Sang Don Koh
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Kenton M. Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| |
Collapse
|
32
|
Abstract
Functions of the gastrointestinal tract include motility, digestion and absorption of nutrients. These functions are mediated by several specialized cell types including smooth muscle cells, neurons, interstitial cells and epithelial cells. In gastrointestinal diseases, some of the cells become degenerated or fail to accomplish their normal functions. Surgical resection of the diseased segments of the gastrointestinal tract is considered the gold-standard treatment in many cases, but patients might have surgical complications and quality of life can remain low. Tissue engineering and regenerative medicine aim to restore, repair, or regenerate the function of the tissues. Gastrointestinal tissue engineering is a challenging process given the specific phenotype and alignment of each cell type that colonizes the tract - these properties are critical for proper functionality. In this Review, we summarize advances in the field of gastrointestinal tissue engineering and regenerative medicine. Although the findings are promising, additional studies and optimizations are needed for translational purposes.
Collapse
Affiliation(s)
- Khalil N Bitar
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, 391 Technology Way NE, Winston Salem, North Carolina 27101, USA.,Department of Molecular Medicine and Translational Sciences, Wake Forest School of Medicine, 1 Medical Center Blvd, Winston Salem, North Carolina 27157, USA.,Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, 391 Technology Way NE, Winston Salem, North Carolina 27101, USA
| | - Elie Zakhem
- Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, 391 Technology Way NE, Winston Salem, North Carolina 27101, USA.,Department of Molecular Medicine and Translational Sciences, Wake Forest School of Medicine, 1 Medical Center Blvd, Winston Salem, North Carolina 27157, USA
| |
Collapse
|
33
|
Taylor CE. A novel treatment for "morning sickness": Nausea of pregnancy could be induced by excess sulfite which molybdenum can help alleviate. Med Hypotheses 2016; 95:31-33. [PMID: 27692161 DOI: 10.1016/j.mehy.2016.08.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 08/13/2016] [Indexed: 12/28/2022]
Abstract
Nausea and vomiting of pregnancy (NVP) remains difficult to treat. Last century, thalidomide was used to alleviate NVP, but it caused teratogenesis by interfering with angiogenesis. The gasotransmitters hydrogen sulfide (H2S) and nitric oxide are mutually dependent on each other for their angiogenesis-related functions. Pregnancy-related requirements for increased endogenous H2S could create a temporary excess of sulfite, an H2S catabolite, which is toxic and can induce nausea. Sulfite oxidase, a molybdenum-containing enzyme, catalyzes oxidation of sulfite to sulfate, which can then be excreted or reused by the body. Supplementation with molybdenum should facilitate enhanced sulfite oxidase activity, thus lowering gestationally-elevated sulfite levels in the gastrointestinal tract and easing NVP.
Collapse
|
34
|
Kaji N, Horiguchi K, Iino S, Nakayama S, Ohwada T, Otani Y, Firman, Murata T, Sanders KM, Ozaki H, Hori M. Nitric oxide-induced oxidative stress impairs pacemaker function of murine interstitial cells of Cajal during inflammation. Pharmacol Res 2016; 111:838-848. [PMID: 27468647 DOI: 10.1016/j.phrs.2016.07.030] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 06/24/2016] [Accepted: 07/22/2016] [Indexed: 12/22/2022]
Abstract
The pacemaker function of interstitial cells of Cajal (ICC) is impaired during intestinal inflammation. The aim of this study is to clarify the pathophysiological mechanisms of ICC dysfunction during inflammatory condition by using intestinal cell clusters. Cell clusters were prepared from smooth muscle layer of murine jejunum and treated with interferon-gamma and lipopolysaccharide (IFN-γ+LPS) for 24h to induce inflammation. Pacemaker function of ICC was monitored by measuring cytosolic Ca(2+) oscillation in the presence of nifedipine. Treatment with IFN-γ+LPS impaired the pacemaker activity of ICC with increasing mRNA level of interleukin-1 beta, tumor necrosis factor-alpha and interleukin-6 in cell clusters; however, treatment with these cytokines individually had little effect on pacemaker activity of ICC. Treatment with IFN-γ+LPS also induced the expression of inducible nitric oxide synthase (iNOS) in smooth muscle cells and resident macrophages, but not in ICC. Pretreatment with NOS inhibitor, L-NAME or iNOS inhibitor, 1400W ameliorated IFN-γ+LPS-induced pacemaker dysfunction of ICC. Pretreatment with guanylate cyclase inhibitor, ODQ did not, but antioxidant, apocynin, to suppress NO-induced oxidative stress, significantly suppressed the impairment of ICC function induced by IFN-γ+LPS. Treatment with IFN-γ+LPS also decreased c-Kit-positive ICC, which was prevented by pretreatment with L-NAME. However, apoptotic ICC were not detected in IFN-γ+LPS-treated clusters, suggesting IFN-γ+LPS stimulation just changed the phenotype of ICC but not induced cell death. Moreover, ultrastructure of ICC was not disturbed by IFN-γ+LPS. In conclusion, ICC dysfunction during inflammation is induced by NO-induced oxidative stress rather than NO/cGMP signaling. NO-induced oxidative stress might be the main factor to induce phenotypic changes of ICC.
Collapse
Affiliation(s)
- Noriyuki Kaji
- Department of Veterinary Pharmacology, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Kazuhide Horiguchi
- Division of Anatomy and Neuroscience, Department of Morphological and Physiological Sciences, University of Fukui Faculty of Medical Sciences, 23-3 Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui 910-1193, Japan
| | - Satoshi Iino
- Division of Anatomy and Neuroscience, Department of Morphological and Physiological Sciences, University of Fukui Faculty of Medical Sciences, 23-3 Matsuokashimoaizuki, Eiheiji-cho, Yoshida-gun, Fukui 910-1193, Japan
| | - Shinsuke Nakayama
- Department of Cell Physiology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Tomohiko Ohwada
- Laboratory of Organic and Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Yuko Otani
- Laboratory of Organic and Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Firman
- Laboratory of Organic and Medicinal Chemistry, Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Takahisa Murata
- Department of Animal Radiology, Graduate School of Agriculture and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, 1664 North Virginia Street, Reno, NV 89557-0357, USA
| | - Hiroshi Ozaki
- Department of Veterinary Pharmacology, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Masatoshi Hori
- Department of Veterinary Pharmacology, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan.
| |
Collapse
|
35
|
Schwerdtfeger LA, Ryan EP, Tobet SA. An organotypic slice model for ex vivo study of neural, immune, and microbial interactions of mouse intestine. Am J Physiol Gastrointest Liver Physiol 2016; 310:G240-8. [PMID: 26680736 PMCID: PMC4754739 DOI: 10.1152/ajpgi.00299.2015] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 12/07/2015] [Indexed: 01/31/2023]
Abstract
Organotypic tissue slices provide seminatural, three-dimensional microenvironments for use in ex vivo study of specific organs and have advanced investigative capabilities compared with isolated cell cultures. Several characteristics of the gastrointestinal tract have made in vitro models for studying the intestine challenging, such as maintaining the intricate structure of microvilli, the intrinsic enteric nervous system, Peyer's patches, the microbiome, and the active contraction of gut muscles. In the present study, an organotypic intestinal slice model was developed that allows for functional investigation across regions of the intestine. Intestinal tissue slices were maintained ex vivo for several days in a physiologically relevant environment that preserved normal enterocyte structure, intact and proliferating crypt cells, submucosal organization, and muscle wall composure. Cell death was measured by a membrane-impermeable DNA binding indicator, ethidium homodimer, and less than 5% of cells were labeled in all regions of the villi and crypt epithelia at 24 h ex vivo. This tissue slice model demonstrated intact myenteric and submucosal neuronal plexuses and functional interstitial cells of Cajal to the extent that nonstimulated, segmental contractions occurred for up to 48 h ex vivo. To detect changes in physiological responses, slices were also assessed for segmental contractions in the presence and absence of antibiotic treatment, which resulted in slices with lesser or greater amounts of commensal bacteria, respectively. Segmental contractions were significantly greater in slices without antibiotics and increased native microbiota. This model renders mechanisms of neuroimmune-microbiome interactions in a complex gut environment available to direct observation and controlled perturbation.
Collapse
Affiliation(s)
- Luke A. Schwerdtfeger
- 1Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado;
| | - Elizabeth P. Ryan
- 3Department of Environmental & Radiological Health Sciences, Colorado State University, Fort Collins, Colorado
| | - Stuart A. Tobet
- 1Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado; ,2School of Biomedical Engineering, Colorado State University, Fort Collins, Colorado; and
| |
Collapse
|
36
|
Hashitani H, Lang RJ. Spontaneous activity in the microvasculature of visceral organs: role of pericytes and voltage-dependent Ca(2+) channels. J Physiol 2016; 594:555-65. [PMID: 26607499 DOI: 10.1113/jp271438] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 10/31/2015] [Indexed: 12/21/2022] Open
Abstract
The microvasculature plays a primary role in the interchange of substances between tissues and the circulation. In visceral organs that undergo considerable distension upon filling, the microvasculature appears to display intrinsic contractile properties to maintain their flow. Submucosal venules in the bladder or gastrointestinal tract generate rhythmic spontaneous phasic constrictions and associated Ca(2+) transients. These events are initiated within either venular pericytes or smooth muscle cells (SMCs) arising from spontaneous Ca(2+) release from the sarcoplasmic reticulum (SR) and the opening of Ca(2+) -activated chloride channels (CaCCs) that trigger Ca(2+) influx through L-type voltage-dependent Ca(2+) channels (VDCCs). L-type VDCCs also play a critical role in maintaining synchrony within the contractile mural cells. In the stomach myenteric layer, spontaneous Ca(2+) transients originating in capillary pericytes appear to spread to their neighbouring arteriolar SMCs. Capillary Ca(2+) transients primarily rely on SR Ca(2+) release, but also require Ca(2+) influx through T-type VDCCs for their synchrony. The opening of T-type VDCCs also contribute to the propagation of Ca(2+) transients into SMCs. In visceral microvasculature, pericytes act as either spontaneously active contractile machinery of the venules or as pacemaker cells generating synchronous Ca(2+) transients that drive spontaneous contractions in upstream arterioles. Thus pericytes play different roles in different vascular beds in a manner that may well depend on the selective expression of T-type and L-type Ca(2+) channels.
Collapse
Affiliation(s)
- Hikaru Hashitani
- Department of Cell Physiology, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Richard J Lang
- Department of Physiology, School of Biomedical Sciences, Monash University, Clayton, Victoria, 3800, Australia
| |
Collapse
|
37
|
Cipriani G, Gibbons SJ, Arumugam SS, Malysz J, Sha L, Szurszewski JH, Linden DR, Evangelista S, Faussone-Pellegrini MS, Vannucchi MG, Farrugia G. Changes in nitrergic and tachykininergic pathways in rat proximal colon in response to chronic treatment with otilonium bromide. Neurogastroenterol Motil 2015; 27:997-1009. [PMID: 25930994 PMCID: PMC4478139 DOI: 10.1111/nmo.12576] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 03/30/2015] [Indexed: 12/20/2022]
Abstract
BACKGROUND Otilonium bromide (OB) is used as a spasmolytic drug in the treatment of the functional bowel disorder irritable bowel syndrome. Although its acute effects on colonic relaxation are well-characterized, little is known about the effects of chronic administration of OB on enteric neurons, neuromuscular transmission, and interstitial cells of Cajal (ICC), key regulators of the gut function. METHODS Adult Sprague-Dawley rats were treated with OB in drinking water at a dose of 2 mg/kg for 30 days. The colons of OB-treated and age-matched control rats were studied by confocal immunohistochemistry to detect immunoreactivity (IR) in myenteric plexus neurons for nitrergic and tachykininergic markers, and also by microelectrode electrophysiology. KEY RESULTS Using immunohistochemistry, chronic OB administration did not change total neuron number, assessed by anti-Hu IR, but resulted in a significant increase in NK1 receptor positive neurons, a decrease in neuronal nitric oxide synthase expressing neurons, and a reduction in volume of substance P in nerve fibers in the myenteric plexus. Chronic OB administration potentiated inhibitory and excitatory junction potentials evoked by repetitive electrical field stimulation. The various types of colonic ICC, detected by Kit IR, were not altered nor were slow waves or smooth muscle membrane potential. CONCLUSIONS & INFERENCES Chronic treatment with OB caused significant changes in the nitrergic and tachykinergic components of the myenteric plexus and in both inhibitory and excitatory neurotransmission in the rat colon.
Collapse
Affiliation(s)
- Gianluca Cipriani
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Mayo Clinic Rochester, Rochester, MN, USA
| | - Simon J. Gibbons
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Mayo Clinic Rochester, Rochester, MN, USA
| | - Saravanaperumal Siva Arumugam
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Mayo Clinic Rochester, Rochester, MN, USA
| | - John Malysz
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Mayo Clinic Rochester, Rochester, MN, USA
| | - Lei Sha
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Mayo Clinic Rochester, Rochester, MN, USA
| | - Joseph H. Szurszewski
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Mayo Clinic Rochester, Rochester, MN, USA
| | - David R. Linden
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Mayo Clinic Rochester, Rochester, MN, USA
| | | | | | - Maria Giuliana Vannucchi
- Dept. Experimental and Clinical Medicine, Histology and Embryology Research Unit, Florence, Italy
| | - Gianrico Farrugia
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology, Mayo Clinic Rochester, Rochester, MN, USA
| |
Collapse
|
38
|
Hashitani H, Mitsui R, Masaki S, Van Helden DF. Pacemaker role of pericytes in generating synchronized spontaneous Ca2+ transients in the myenteric microvasculature of the guinea-pig gastric antrum. Cell Calcium 2015; 58:442-56. [PMID: 26153078 DOI: 10.1016/j.ceca.2015.06.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 06/08/2015] [Accepted: 06/28/2015] [Indexed: 11/25/2022]
Abstract
Properties of spontaneous Ca(2+) transients in the myenteric microvasculature of the guinea-pig stomach were investigated. Specifically, we explored the spatio-temporal origin of Ca(2+) transients and the role of voltage-dependent Ca(2+) channels (VDCCs) in their intercellular synchrony using fluorescence Ca(2+) imaging and immunohistochemistry. The microvasculature generated spontaneous Ca(2+) transients that were independent of both Ca(2+) transients in interstitial cells of Cajal (ICC) and neural activity. Spontaneous Ca(2+) transients were highly synchronous along the length of microvasculature, and appeared to be initiated in pericytes and spread to arteriolar smooth muscle cells (SMCs). In most cases, the generation or synchrony of Ca(2+) transients was not affected by blockers of L-type VDCCs. In nifedipine-treated preparations, synchronous spontaneous Ca(2+) transients were readily blocked by Ni(2+), mibefradil or ML216, blockers for T-type VDCCs. These blockers also suppressed the known T-type VDCC dependent component of ICC Ca(2+) transients or slow waves. Spontaneous Ca(2+) transients were also suppressed by caffeine, tetracaine or cyclopiazonic acid (CPA). After the blockade of both L- and T-type VDCCs, asynchronous Ca(2+) transients were generated in pericytes on precapillary arterioles and/or capillaries but not in arteriolar SMCs, and were abolished by CPA or nominally Ca(2+) free solution. Together these data indicate that pericytes in the myenteric microvasculature may act as the origin of synchronous spontaneous Ca(2+) transients. Pericyte Ca(2+) transients arise from Ca(2+) release from the sarco-endoplasmic reticulum and the opening of T-type Ca(2+) VDCCs is required for their synchrony and propagation to arteriolar SMCs.
Collapse
Affiliation(s)
- Hikaru Hashitani
- Department of Cell Physiology, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan.
| | - Retsu Mitsui
- Department of Cell Physiology, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Shota Masaki
- Department of Cell Physiology, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Dirk F Van Helden
- School of Biomedical Sciences and Pharmacy, University of Newcastle, NSW, Australia
| |
Collapse
|
39
|
Worth AA, Forrest AS, Peri LE, Ward SM, Hennig GW, Sanders KM. Regulation of gastric electrical and mechanical activity by cholinesterases in mice. J Neurogastroenterol Motil 2015; 21:200-16. [PMID: 25843073 PMCID: PMC4398240 DOI: 10.5056/jnm14120] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 11/15/2014] [Accepted: 11/18/2014] [Indexed: 12/03/2022] Open
Abstract
Background/Aims Gastric peristalsis begins in the orad corpus and propagates to the pylorus. Directionality of peristalsis depends upon orderly generation and propagation of electrical slow waves and a frequency gradient between proximal and distal pacemakers. We sought to understand how chronotropic agonists affect coupling between corpus and antrum. Methods Electrophysiological and imaging techniques were used to investigate regulation of gastric slow wave frequency by muscarinic agonists in mice. We also investigated the expression and role of cholinesterases in regulating slow wave frequency and motor patterns in the stomach. Results Both acetycholinesterase (Ache) and butyrylcholine esterase (Bche) are expressed in gastric muscles and AChE is localized to varicose processes of motor neurons. Inhibition of AChE in the absence of stimulation increased slow wave frequency in corpus and throughout muscle strips containing corpus and antrum. CCh caused depolarization and increased slow wave frequency. Stimulation of cholinergic neurons increased slow wave frequency but did not cause depolarization. Neostigmine (1 μM) increased slow wave frequency, but uncoupling between corpus and antrum was not detected. Motility mapping of contractile activity in gastric muscles showed similar effects of enteric nerve stimulation on the frequency and propagation of slow waves, but neostigmine (> 1 μM) caused aberrant contractile frequency and propagation and ectopic pacemaking. Conclusions Our data show that slow wave uncoupling is difficult to assess with electrical recording from a single or double sites and suggest that efficient metabolism of ACh released from motor neurons is an extremely important regulator of slow wave frequency and propagation and gastric motility patterns.
Collapse
Affiliation(s)
- Amy A Worth
- Faculty of Life Sciences, University of Manchester, Manchester, United Kingdom
| | | | - Lauren E Peri
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Sean M Ward
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Grant W Hennig
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, USA
| |
Collapse
|
40
|
Sung TS, Kim HU, Kim JH, Lu H, Sanders KM, Koh SD. Protease-activated receptors modulate excitability of murine colonic smooth muscles by differential effects on interstitial cells. J Physiol 2015; 593:1169-81. [PMID: 25641660 PMCID: PMC4358678 DOI: 10.1113/jphysiol.2014.285148] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 12/22/2014] [Indexed: 12/12/2022] Open
Abstract
Protease-activated receptors (PARs) are G protein-coupled receptors activated by proteolytic cleavage at their amino termini by serine proteases. PAR activation contributes to the inflammatory response in the gastrointestinal (GI) tract and alters GI motility, but little is known about the specific cells within the tunica muscularis that express PARs and the mechanisms leading to contractile responses. Using real time PCR, we found PARs to be expressed in smooth muscle cells (SMCs), interstitial cells of Cajal (ICC) and platelet-derived growth factor receptor α positive (PDGFRα(+)) cells. The latter cell-type showed dominant expression of F2r (encodes PAR1) and F2rl1 (encodes PAR2). Contractile and intracellular electrical activities were measured to characterize the integrated responses to PAR activation in whole muscles. Cells were isolated and ICC and PDGFRα(+) cells were identified by constitutive expression of fluorescent reporters. Thrombin (PAR1 agonist) and trypsin (PAR2 agonist) caused biphasic responses in colonic muscles: transient hyperpolarization and relaxation followed by repolarization and excitation. The inhibitory phase was blocked by apamin, revealing a distinct excitatory component. Patch clamp studies showed that the inhibitory response was mediated by activation of small conductance calcium-activated K(+) channels in PDGFRα(+) cells, and the excitatory response was mediated by activation of a Cl(-) conductance in ICC. SMCs contributed little to PAR responses in colonic muscles. In summary, PARs regulate the excitability of colonic muscles; different conductances are activated in each cell type of the SMC-ICC-PDGFRα(+) cell (SIP) syncytium. Motor responses to PAR agonists are integrated responses of the SIP syncytium.
Collapse
MESH Headings
- Action Potentials
- Animals
- Cells, Cultured
- Chloride Channels/antagonists & inhibitors
- Chloride Channels/metabolism
- Colon/cytology
- Colon/metabolism
- Interstitial Cells of Cajal/metabolism
- Interstitial Cells of Cajal/physiology
- Mice
- Mice, Inbred C57BL
- Muscle Contraction
- Muscle, Smooth/metabolism
- Muscle, Smooth/physiology
- Potassium Channels, Calcium-Activated/metabolism
- Potassium Channels, Inwardly Rectifying/metabolism
- Receptor, PAR-1/agonists
- Receptor, PAR-1/genetics
- Receptor, PAR-1/metabolism
- Receptor, PAR-2/agonists
- Receptor, PAR-2/genetics
- Receptor, PAR-2/metabolism
Collapse
Affiliation(s)
- Tae Sik Sung
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, RenoNV 89557, USA
| | - Heung Up Kim
- Department of Internal Medicine, School of Medicine, Jeju National UniversityJeju, Korea
| | - Jeong Hwan Kim
- Department of Internal Medicine, School of Medicine, Konkuk University Medical CentreSeoul, Korea
| | - Hongli Lu
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, RenoNV 89557, USA
| | - Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, RenoNV 89557, USA
| | - Sang Don Koh
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, RenoNV 89557, USA
| |
Collapse
|
41
|
Kito Y, Mitsui R, Ward SM, Sanders KM. Characterization of slow waves generated by myenteric interstitial cells of Cajal of the rabbit small intestine. Am J Physiol Gastrointest Liver Physiol 2015; 308:G378-88. [PMID: 25540230 PMCID: PMC4346752 DOI: 10.1152/ajpgi.00308.2014] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Slow waves (slow wavesICC) were recorded from myenteric interstitial cells of Cajal (ICC-MY) in situ in the rabbit small intestine, and their properties were compared with those of mouse small intestine. Rabbit slow wavesICC consisted of an upstroke depolarization followed by a distinct plateau component. Ni(2+) and nominally Ca(2+)-free solutions reduced the rate-of-rise and amplitude of the upstroke depolarization. Replacement of Ca(2+) with Sr(2+) enhanced the upstroke component but decreased the plateau component of rabbit slow wavesICC. In contrast, replacing Ca(2+) with Sr(2+) decreased both components of mouse slow wavesICC. The plateau component of rabbit slow wavesICC was inhibited in low-extracellular-Cl(-)-concentration (low-[Cl(-)]o) solutions and by 4,4'-diisothiocyanostilbene-2,2'-disulfonic acid (DIDS), an inhibitor of Cl(-) channels, cyclopiazonic acid (CPA), an inhibitor of internal Ca(2+) pumps, or bumetanide, an inhibitor of Na(+)-K(+)-2Cl(-) cotransporter (NKCC1). Bumetanide also inhibited the plateau component of mouse slow wavesICC. NKCC1-like immunoreactivity was observed mainly in ICC-MY in the rabbit small intestine. Membrane depolarization with a high-K(+) solution reduced the upstroke component of rabbit slow wavesICC. In cells depolarized with elevated external K(+), DIDS, CPA, and bumetanide blocked slow wavesICC. These results suggest that the upstroke component of rabbit slow wavesICC is partially mediated by voltage-dependent Ca(2+) influx, whereas the plateau component is dependent on Ca(2+)-activated Cl(-) efflux. NKCC1 is likely to be responsible for Cl(-) accumulation in ICC-MY. The results also suggest that the mechanism of the upstroke component differs in rabbit and mouse slow wavesICC in the small intestine.
Collapse
Affiliation(s)
- Yoshihiko Kito
- Department of Pharmacology, Faculty of Medicine, Saga University, Nabeshima, Saga, Japan; Department of Cell Physiology, Nagoya City University Medical School, Mizuho-ku, Nagoya, Japan; and
| | - Retsu Mitsui
- 2Department of Cell Physiology, Nagoya City University Medical School, Mizuho-ku, Nagoya, Japan; and
| | - Sean M. Ward
- 3Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Kenton M. Sanders
- 3Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| |
Collapse
|
42
|
Zhu MH, Sung TS, O'Driscoll K, Koh SD, Sanders KM. Intracellular Ca(2+) release from endoplasmic reticulum regulates slow wave currents and pacemaker activity of interstitial cells of Cajal. Am J Physiol Cell Physiol 2015; 308:C608-20. [PMID: 25631870 DOI: 10.1152/ajpcell.00360.2014] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 01/16/2015] [Indexed: 02/02/2023]
Abstract
Interstitial cells of Cajal (ICC) provide pacemaker activity in gastrointestinal muscles that underlies segmental and peristaltic contractions. ICC generate electrical slow waves that are due to large-amplitude inward currents resulting from anoctamin 1 (ANO1) channels, which are Ca(2+)-activated Cl(-) channels. We investigated the hypothesis that the Ca(2+) responsible for the stochastic activation of ANO1 channels during spontaneous transient inward currents (STICs) and synchronized activation of ANO1 channels during slow wave currents comes from intracellular Ca(2+) stores. ICC, obtained from the small intestine of Kit(+/copGFP) mice, were studied under voltage and current clamp to determine the effects of blocking Ca(2+) uptake into stores and release of Ca(2+) via inositol 1,4,5-trisphosphate (IP3)-dependent and ryanodine-sensitive channels. Cyclocpiazonic acid, thapsigargin, 2-APB, and xestospongin C inhibited STICs and slow wave currents. Ryanodine and tetracaine also inhibited STICs and slow wave currents. Store-active compounds had no direct effects on ANO1 channels expressed in human embryonic kidney-293 cells. Under current clamp, store-active drugs caused significant depolarization of ICC and reduced spontaneous transient depolarizations (STDs). After block of ryanodine receptors with ryanodine and tetracaine, repolarization did not restore STDs. ANO1 expressed in ICC has limited access to cytoplasmic Ca(2+) concentration, suggesting that pacemaker activity depends on Ca(2+) dynamics in restricted microdomains. Our data from studies of isolated ICC differ somewhat from studies on intact muscles and suggest that release of Ca(2+) from both IP3 and ryanodine receptors is important in generating pacemaker activity in ICC.
Collapse
Affiliation(s)
- Mei Hong Zhu
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Tae Sik Sung
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Kate O'Driscoll
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Sang Don Koh
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| | - Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada
| |
Collapse
|
43
|
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
|
44
|
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
|
45
|
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
|