51
|
PIP 2: A critical regulator of vascular ion channels hiding in plain sight. Proc Natl Acad Sci U S A 2020; 117:20378-20389. [PMID: 32764146 PMCID: PMC7456132 DOI: 10.1073/pnas.2006737117] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The phosphoinositide, phosphatidylinositol 4,5-bisphosphate (PIP2), has long been established as a major contributor to intracellular signaling, primarily by virtue of its role as a substrate for phospholipase C (PLC). Signaling by Gq-protein-coupled receptors triggers PLC-mediated hydrolysis of PIP2 into inositol 1,4,5-trisphosphate and diacylglycerol, which are well known to modulate vascular ion channel activity. Often overlooked, however, is the role PIP2 itself plays in this regulation. Although numerous reports have demonstrated that PIP2 is critical for ion channel regulation, how it impacts vascular function has received scant attention. In this review, we focus on PIP2 as a regulator of ion channels in smooth muscle cells and endothelial cells-the two major classes of vascular cells. We further address the concerted effects of such regulation on vascular function and blood flow control. We close with a consideration of current knowledge regarding disruption of PIP2 regulation of vascular ion channels in disease.
Collapse
|
52
|
Bruce JIE, James AD. Targeting the Calcium Signalling Machinery in Cancer. Cancers (Basel) 2020; 12:cancers12092351. [PMID: 32825277 PMCID: PMC7565467 DOI: 10.3390/cancers12092351] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/30/2020] [Accepted: 08/08/2020] [Indexed: 12/13/2022] Open
Abstract
Cancer is caused by excessive cell proliferation and a propensity to avoid cell death, while the spread of cancer is facilitated by enhanced cellular migration, invasion, and vascularization. Cytosolic Ca2+ is central to each of these important processes, yet to date, there are no cancer drugs currently being used clinically, and very few undergoing clinical trials, that target the Ca2+ signalling machinery. The aim of this review is to highlight some of the emerging evidence that targeting key components of the Ca2+ signalling machinery represents a novel and relatively untapped therapeutic strategy for the treatment of cancer.
Collapse
Affiliation(s)
- Jason I. E. Bruce
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
- Correspondence: ; Tel.: +44-(0)-161-275-5484
| | - Andrew D. James
- Department of Biology, University of York, Heslington, York YO10 5DD, UK;
| |
Collapse
|
53
|
Abstract
Vascular smooth muscle cells (VSMCs) of small peripheral arteries contribute to blood pressure control by adapting their contractile state. These adaptations depend on the VSMC cytosolic Ca2+ concentration, regulated by complex local elementary Ca2+ signaling pathways. Ca2+ sparks represent local, transient, rapid calcium release events from a cluster of ryanodine receptors (RyRs) in the sarcoplasmic reticulum. In arterial SMCs, Ca2+ sparks activate nearby calcium-dependent potassium channels, cause membrane hyperpolarization and thus decrease the global intracellular [Ca2+] to oppose vasoconstriction. Arterial SMC Cav1.2 L-type channels regulate intracellular calcium stores content, which in turn modulates calcium efflux through RyRs. Cav3.2 T-type channels contribute to a minor extend to Ca2+ spark generation in certain types of arteries. Their localization within cell membrane caveolae is essential. We summarize present data on local elementary calcium signaling (Ca2+ sparks) in arterial SMCs with focus on RyR isoforms, large-conductance calcium-dependent potassium (BKCa) channels, and cell membrane-bound calcium channels (Cav1.2 and Cav3.2), particularly in caveolar microdomains.
Collapse
Affiliation(s)
- Gang Fan
- Charité - Universitätsmedizin Berlin, Experimental and Clinical Research Center (ECRC), a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Yingqiu Cui
- Charité - Universitätsmedizin Berlin, Experimental and Clinical Research Center (ECRC), a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Maik Gollasch
- Charité - Universitätsmedizin Berlin, Experimental and Clinical Research Center (ECRC), a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany
| | - Mario Kassmann
- Charité - Universitätsmedizin Berlin, Experimental and Clinical Research Center (ECRC), a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Germany
| |
Collapse
|
54
|
Song R, Hu XQ, Romero M, Holguin MA, Kagabo W, Xiao D, Wilson SM, Zhang L. Ryanodine receptor subtypes regulate Ca2+ sparks/spontaneous transient outward currents and myogenic tone of uterine arteries in pregnancy. Cardiovasc Res 2020; 117:792-804. [PMID: 32251501 DOI: 10.1093/cvr/cvaa089] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 02/18/2020] [Accepted: 03/31/2020] [Indexed: 12/12/2022] Open
Abstract
AIMS Our recent study demonstrated that increased Ca2+ sparks and spontaneous transient outward currents (STOCs) played an important role in uterine vascular tone and haemodynamic adaptation to pregnancy. The present study examined the role of ryanodine receptor (RyR) subtypes in regulating Ca2+ sparks/STOCs and myogenic tone in uterine arterial adaptation to pregnancy. METHODS AND RESULTS Uterine arteries isolated from non-pregnant and near-term pregnant sheep were used in the present study. Pregnancy increased the association of α and β1 subunits of large-conductance Ca2+-activated K+ (BKCa) channels and enhanced the co-localization of RyR1 and RyR2 with the β1 subunit in the uterine artery. In contrast, RyR3 was not co-localized with BKCa β1 subunit. Knockdown of RyR1 or RyR2 in uterine arteries of pregnant sheep downregulated the β1 but not α subunit of the BKCa channel and decreased the association of α and β1 subunits. Unlike RyR1 and RyR2, knockdown of RyR3 had no significant effect on either expression or association of BKCa subunits. In addition, knockdown of RyR1 or RyR2 significantly decreased Ca2+ spark frequency, suppressed STOCs frequency and amplitude, and increased pressure-dependent myogenic tone in uterine arteries of pregnant animals. RyR3 knockdown did not affect Ca2+ sparks/STOCs and myogenic tone in the uterine artery. CONCLUSION Together, the present study demonstrates a novel mechanistic paradigm of RyR subtypes in the regulation of Ca2+ sparks/STOCs and uterine vascular tone, providing new insights into the mechanisms underlying uterine vascular adaptation to pregnancy.
Collapse
Affiliation(s)
- Rui Song
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Xiang-Qun Hu
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Monica Romero
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Mark A Holguin
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Whitney Kagabo
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Daliao Xiao
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Sean M Wilson
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| | - Lubo Zhang
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92350, USA
| |
Collapse
|
55
|
Barabas P, Augustine J, Fernández JA, McGeown JG, McGahon MK, Curtis TM. Ion channels and myogenic activity in retinal arterioles. CURRENT TOPICS IN MEMBRANES 2020; 85:187-226. [PMID: 32402639 DOI: 10.1016/bs.ctm.2020.01.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Retinal pressure autoregulation is an important mechanism that protects the retina by stabilizing retinal blood flow during changes in arterial or intraocular pressure. Similar to other vascular beds, retinal pressure autoregulation is thought to be mediated largely through the myogenic response of small arteries and arterioles which constrict when transmural pressure increases or dilate when it decreases. Over recent years, we and others have investigated the signaling pathways underlying the myogenic response in retinal arterioles, with particular emphasis on the involvement of different ion channels expressed in the smooth muscle layer of these vessels. Here, we review and extend previous work on the expression and spatial distribution of the plasma membrane and sarcoplasmic reticulum ion channels present in retinal vascular smooth muscle cells (VSMCs) and discuss their contribution to pressure-induced myogenic tone in retinal arterioles. This includes new data demonstrating that several key players and modulators of the myogenic response show distinctively heterogeneous expression along the length of the retinal arteriolar network, suggesting differences in myogenic signaling between larger and smaller pre-capillary arterioles. Our immunohistochemical investigations have also highlighted the presence of actin-containing microstructures called myobridges that connect the retinal VSMCs to one another. Although further work is still needed, studies to date investigating myogenic mechanisms in the retina have contributed to a better understanding of how blood flow is regulated in this tissue. They also provide a basis to direct future research into retinal diseases where blood flow changes contribute to the pathology.
Collapse
Affiliation(s)
- Peter Barabas
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University of Belfast, Belfast, United Kingdom
| | - Josy Augustine
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University of Belfast, Belfast, United Kingdom
| | - José A Fernández
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University of Belfast, Belfast, United Kingdom
| | - J Graham McGeown
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University of Belfast, Belfast, United Kingdom
| | - Mary K McGahon
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University of Belfast, Belfast, United Kingdom
| | - Tim M Curtis
- Wellcome-Wolfson Institute for Experimental Medicine, Queen's University of Belfast, Belfast, United Kingdom.
| |
Collapse
|
56
|
Manoury B, Idres S, Leblais V, Fischmeister R. Ion channels as effectors of cyclic nucleotide pathways: Functional relevance for arterial tone regulation. Pharmacol Ther 2020; 209:107499. [PMID: 32068004 DOI: 10.1016/j.pharmthera.2020.107499] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 02/05/2020] [Indexed: 02/07/2023]
Abstract
Numerous mediators and drugs regulate blood flow or arterial pressure by acting on vascular tone, involving cyclic nucleotide intracellular pathways. These signals lead to regulation of several cellular effectors, including ion channels that tune cell membrane potential, Ca2+ influx and vascular tone. The characterization of these vasocontrictive or vasodilating mechanisms has grown in complexity due to i) the variety of ion channels that are expressed in both vascular endothelial and smooth muscle cells, ii) the heterogeneity of responses among the various vascular beds, and iii) the number of molecular mechanisms involved in cyclic nucleotide signalling in health and disease. This review synthesizes key data from literature that highlight ion channels as physiologically relevant effectors of cyclic nucleotide pathways in the vasculature, including the characterization of the molecular mechanisms involved. In smooth muscle cells, cation influx or chloride efflux through ion channels are associated with vasoconstriction, whereas K+ efflux repolarizes the cell membrane potential and mediates vasodilatation. Both categories of ion currents are under the influence of cAMP and cGMP pathways. Evidence that some ion channels are influenced by CN signalling in endothelial cells will also be presented. Emphasis will also be put on recent data touching a variety of determinants such as phosphodiesterases, EPAC and kinase anchoring, that complicate or even challenge former paradigms.
Collapse
Affiliation(s)
- Boris Manoury
- Inserm, Umr-S 1180, Université Paris-Saclay, Châtenay-Malabry, France.
| | - Sarah Idres
- Inserm, Umr-S 1180, Université Paris-Saclay, Châtenay-Malabry, France
| | - Véronique Leblais
- Inserm, Umr-S 1180, Université Paris-Saclay, Châtenay-Malabry, France
| | | |
Collapse
|
57
|
Hasan R, Leo MD, Muralidharan P, Mata-Daboin A, Yin W, Bulley S, Fernandez-Peña C, MacKay CE, Jaggar JH. SUMO1 modification of PKD2 channels regulates arterial contractility. Proc Natl Acad Sci U S A 2019; 116:27095-27104. [PMID: 31822608 PMCID: PMC6936352 DOI: 10.1073/pnas.1917264116] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
PKD2 (polycystin-2, TRPP1) channels are expressed in a wide variety of cell types and can regulate functions, including cell division and contraction. Whether posttranslational modification of PKD2 modifies channel properties is unclear. Similarly uncertain are signaling mechanisms that regulate PKD2 channels in arterial smooth muscle cells (myocytes). Here, by studying inducible, cell-specific Pkd2 knockout mice, we discovered that PKD2 channels are modified by SUMO1 (small ubiquitin-like modifier 1) protein in myocytes of resistance-size arteries. At physiological intravascular pressures, PKD2 exists in approximately equal proportions as either nonsumoylated (PKD2) or triple SUMO1-modifed (SUMO-PKD2) proteins. SUMO-PKD2 recycles, whereas unmodified PKD2 is surface-resident. Intravascular pressure activates voltage-dependent Ca2+ influx that stimulates the return of internalized SUMO-PKD2 channels to the plasma membrane. In contrast, a reduction in intravascular pressure, membrane hyperpolarization, or inhibition of Ca2+ influx leads to lysosomal degradation of internalized SUMO-PKD2 protein, which reduces surface channel abundance. Through this sumoylation-dependent mechanism, intravascular pressure regulates the surface density of SUMO-PKD2-mediated Na+ currents (INa) in myocytes to control arterial contractility. We also demonstrate that intravascular pressure activates SUMO-PKD2, not PKD2, channels, as desumoylation leads to loss of INa activation in myocytes and vasodilation. In summary, this study reveals that PKD2 channels undergo posttranslational modification by SUMO1, which enables physiological regulation of their surface abundance and pressure-mediated activation in myocytes and thus control of arterial contractility.
Collapse
Affiliation(s)
- Raquibul Hasan
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163
| | - M. Dennis Leo
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163
| | | | - Alejandro Mata-Daboin
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Wen Yin
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Simon Bulley
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Carlos Fernandez-Peña
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Charles E. MacKay
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163
| | - Jonathan H. Jaggar
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163
| |
Collapse
|
58
|
Wang FH, Peng X, Chen Y, Wang Y, Yang M, Guo MY. Se Regulates the Contractile Ability of Uterine Smooth Musclevia Selenoprotein N, Selenoprotein T, and Selenoprotein Win Mice. Biol Trace Elem Res 2019; 192:196-205. [PMID: 30756291 DOI: 10.1007/s12011-019-1647-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 01/18/2019] [Indexed: 11/27/2022]
Abstract
Selenium (Se) is an essential micronutrient that maintains normal physiological functions in humans and animals. Se plays a vital role in regulating smooth muscle contractions, and selenoprotein N (SelN), selenoprotein T (SelT), and selenoprotein W (SelW) are closely related to the release of Ca2+. The present study analyzed the effects and mechanisms of SelN, SelT, and SelW in uterine smooth muscle contractions in a mouse model fed Se. The mRNA and protein levels in the uterine smooth muscle of mice were detected by qPCR, Western blot, and immunohistochemical analysis. The results showed that Se played an indispensable role in uterine smooth muscle contractions. Increased Se concentration in food increased the release of Ca2+ to a certain extent, causing CaM expression, MLCK expression, and MLC phosphorylation, which can lead to uterine smooth muscle contractions. In contrast, Se deficiency reduced the release of Ca2+ to a certain degree, thereby reducing the contractile ability of uterine smooth muscle. In this study, genes related to SelN, SelT, and SelW expression in uterine smooth muscle cells were investigated. The results showed that the Se concentration had an effect on the expression of SelN, SelT, and SelW in uterine smooth muscle cells. Se influences the release of Ca2+ through SelN, SelT, and SelW, which changes the expression of MLCK and then affects uterine smooth muscle contractions. The three selenoproteins SelN, SelT, and SelW play a very important role in uterine smooth muscle contractions, and the absence of any of these proteins affects the contractility of the uterus.
Collapse
Affiliation(s)
- Fu-Han Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Xiao Peng
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Yu Chen
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Ying Wang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Mei Yang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Meng-Yao Guo
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China.
| |
Collapse
|
59
|
Hu XQ, Song R, Romero M, Dasgupta C, Huang X, Holguin MA, Williams V, Xiao D, Wilson SM, Zhang L. Pregnancy Increases Ca 2+ Sparks/Spontaneous Transient Outward Currents and Reduces Uterine Arterial Myogenic Tone. Hypertension 2019; 73:691-702. [PMID: 30661479 DOI: 10.1161/hypertensionaha.118.12484] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Spontaneous transient outward currents (STOCs) at physiological membrane potentials of vascular smooth muscle cells fundamentally regulate vascular myogenic tone and blood flow in an organ. We hypothesize that heightened STOCs play a key role in uterine vascular adaptation to pregnancy. Uterine arteries were isolated from nonpregnant and near-term pregnant sheep. Ca2+ sparks were measured by confocal microscopy, and STOCs were determined by electrophysiological recording in smooth muscle cells. Percentage of Ca2+ spark firing myocytes increased dramatically at the resting condition in uterine arterial smooth muscle of pregnant animals, as compared with nonpregnant animals. Pregnancy upregulated the expression of RyRs (ryanodine receptors) and significantly boosted Ca2+ spark frequency. Ex vivo treatment of uterine arteries of nonpregnant sheep with estrogen and progesterone imitated pregnancy-induced RyR upregulation. STOCs occurred at much more negative membrane potentials in uterine arterial myocytes of pregnant animals. STOCs in uterine arterial myocytes were diminished by inhibiting large-conductance Ca2+-activated K+ (BKCa) channels and RyRs, thus functionally linking Ca2+ sparks and BKCa channel activity to STOCs. Pregnancy and steroid hormone treatment significantly increased STOCs frequency and amplitude in uterine arteries. Of importance, inhibition of STOCs with RyR inhibitor ryanodine eliminated pregnancy- and steroid hormone-induced attenuation of uterine arterial myogenic tone. Thus, the present study demonstrates a novel role of Ca2+ sparks and STOCs in the regulation of uterine vascular tone and provides new insights into the mechanisms underlying uterine vascular adaptation to pregnancy.
Collapse
Affiliation(s)
- Xiang-Qun Hu
- From the Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, CA
| | - Rui Song
- From the Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, CA
| | - Monica Romero
- From the Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, CA
| | - Chiranjib Dasgupta
- From the Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, CA
| | - Xiaohui Huang
- From the Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, CA
| | - Mark A Holguin
- From the Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, CA
| | - VaShon Williams
- From the Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, CA
| | - Daliao Xiao
- From the Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, CA
| | - Sean M Wilson
- From the Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, CA
| | - Lubo Zhang
- From the Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, CA
| |
Collapse
|
60
|
Nanoscale coupling of junctophilin-2 and ryanodine receptors regulates vascular smooth muscle cell contractility. Proc Natl Acad Sci U S A 2019; 116:21874-21881. [PMID: 31591206 PMCID: PMC6815135 DOI: 10.1073/pnas.1911304116] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Junctophilin proteins maintain close contacts between the endoplasmic/sarcoplasmic reticulum (ER/SR) and the plasma membrane in many types of cells, as typified by junctophilin-2 (JPH2), which is necessary for the formation of the cardiac dyad. Here, we report that JPH2 is the most abundant junctophilin isotype in native smooth muscle cells (SMCs) isolated from cerebral arteries and that acute knockdown diminishes the area of sites of interaction between the SR and plasma membrane. Superresolution microscopy revealed nanometer-scale colocalization of JPH2 clusters with type 2 ryanodine receptor (RyR2) clusters near the cell surface. Knockdown of JPH2 had no effect on the frequency, amplitude, or kinetics of spontaneous Ca2+ sparks generated by transient release of Ca2+ from the SR through RyR2s, but it did nearly abolish Ca2+ spark-activated, large-conductance, Ca2+-activated K+ (BK) channel currents. We also found that JPH2 knockdown was associated with hypercontractility of intact cerebral arteries. We conclude that JPH2 maintains functional coupling between RyR2s and BK channels and is critically important for cerebral arterial function.
Collapse
|
61
|
Bailey CS, Moldenhauer HJ, Park SM, Keros S, Meredith AL. KCNMA1-linked channelopathy. J Gen Physiol 2019; 151:1173-1189. [PMID: 31427379 PMCID: PMC6785733 DOI: 10.1085/jgp.201912457] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 08/13/2019] [Indexed: 12/20/2022] Open
Abstract
Bailey et al. review a new neurological channelopathy associated with KCNMA1, encoding the BK voltage- and Ca2+-activated K+ channel. KCNMA1 encodes the pore-forming α subunit of the “Big K+” (BK) large conductance calcium and voltage-activated K+ channel. BK channels are widely distributed across tissues, including both excitable and nonexcitable cells. Expression levels are highest in brain and muscle, where BK channels are critical regulators of neuronal excitability and muscle contractility. A global deletion in mouse (KCNMA1−/−) is viable but exhibits pathophysiology in many organ systems. Yet despite the important roles in animal models, the consequences of dysfunctional BK channels in humans are not well characterized. Here, we summarize 16 rare KCNMA1 mutations identified in 37 patients dating back to 2005, with an array of clinically defined pathological phenotypes collectively referred to as “KCNMA1-linked channelopathy.” These mutations encompass gain-of-function (GOF) and loss-of-function (LOF) alterations in BK channel activity, as well as several variants of unknown significance (VUS). Human KCNMA1 mutations are primarily associated with neurological conditions, including seizures, movement disorders, developmental delay, and intellectual disability. Due to the recent identification of additional patients, the spectrum of symptoms associated with KCNMA1 mutations has expanded but remains primarily defined by brain and muscle dysfunction. Emerging evidence suggests the functional BK channel alterations produced by different KCNMA1 alleles may associate with semi-distinct patient symptoms, such as paroxysmal nonkinesigenic dyskinesia (PNKD) with GOF and ataxia with LOF. However, due to the de novo origins for the majority of KCNMA1 mutations identified to date and the phenotypic variability exhibited by patients, additional evidence is required to establish causality in most cases. The symptomatic picture developing from patients with KCNMA1-linked channelopathy highlights the importance of better understanding the roles BK channels play in regulating cell excitability. Establishing causality between KCNMA1-linked BK channel dysfunction and specific patient symptoms may reveal new treatment approaches with the potential to increase therapeutic efficacy over current standard regimens.
Collapse
Affiliation(s)
- Cole S Bailey
- Dept. of Physiology, University of Maryland School of Medicine, Baltimore, MD
| | - Hans J Moldenhauer
- Dept. of Physiology, University of Maryland School of Medicine, Baltimore, MD
| | - Su Mi Park
- Dept. of Physiology, University of Maryland School of Medicine, Baltimore, MD
| | - Sotirios Keros
- Department of Pediatrics, University of South Dakota Sanford School of Medicine, Sioux Falls, SD
| | - Andrea L Meredith
- Dept. of Physiology, University of Maryland School of Medicine, Baltimore, MD
| |
Collapse
|
62
|
Saeki T, Suzuki Y, Yamamura H, Takeshima H, Imaizumi Y. A junctophilin-caveolin interaction enables efficient coupling between ryanodine receptors and BK Ca channels in the Ca 2+ microdomain of vascular smooth muscle. J Biol Chem 2019; 294:13093-13105. [PMID: 31308177 PMCID: PMC6721949 DOI: 10.1074/jbc.ra119.008342] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 07/09/2019] [Indexed: 01/09/2023] Open
Abstract
Functional coupling between large-conductance Ca2+-activated K+ (BKCa) channels in the plasma membrane (PM) and ryanodine receptors (RyRs) in the sarcoplasmic reticulum (SR) is an essential mechanism for regulating mechanical force in most smooth muscle (SM) tissues. Spontaneous Ca2+ release through RyRs (Ca2+ sparks) and subsequent BKCa channel activation occur within the PM-SR junctional sites. We report here that a molecular interaction of caveolin-1 (Cav1), a caveola-forming protein, with junctophilin-2 (JP2), a bridging protein between PM and SR, positions BKCa channels near RyRs in SM cells (SMCs) and thereby contributes to the formation of a molecular complex essential for Ca2+ microdomain function. Approximately half of all Ca2+ sparks occurred within a close distance (<400 nm) from fluorescently labeled JP2 or Cav1 particles, when they were moderately expressed in primary SMCs from mouse mesenteric artery. The removal of caveolae by genetic Cav1 ablation or methyl-β-cyclodextrin treatments significantly reduced coupling efficiency between Ca2+ sparks and BKCa channel activity in SMCs, an effect also observed after JP2 knockdown in SMCs. A 20-amino acid-long region in JP2 appeared to be essential for the observed JP2-Cav1 interaction, and we also observed an interaction between JP2 and the BKCa channel. It can be concluded that the JP2-Cav1 interaction provides a structural and functional basis for the Ca2+ microdomain at PM-SR junctions and mediates cross-talk between RyRs and BKCa channels, converts local Ca2+ sparks into membrane hyperpolarization, and contributes to stabilizing resting tone in SMCs.
Collapse
Affiliation(s)
- Takanori Saeki
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan
| | - Yoshiaki Suzuki
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan
| | - Hisao Yamamura
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan
| | - Hiroshi Takeshima
- Department of Biological Chemistry, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto 606-8501, Japan
| | - Yuji Imaizumi
- Department of Molecular and Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan.
| |
Collapse
|
63
|
Drumm BT, Hwang SJ, Baker SA, Ward SM, Sanders KM. Ca 2+ signalling behaviours of intramuscular interstitial cells of Cajal in the murine colon. J Physiol 2019; 597:3587-3617. [PMID: 31124144 DOI: 10.1113/jp278036] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 05/23/2019] [Indexed: 12/13/2022] Open
Abstract
KEY POINTS Colonic intramuscular interstitial cells of Cajal (ICC-IM) exhibit spontaneous Ca2+ transients manifesting as stochastic events from multiple firing sites with propagating Ca2+ waves occasionally observed. Firing of Ca2+ transients in ICC-IM is not coordinated with adjacent ICC-IM in a field of view or even with events from other firing sites within a single cell. Ca2+ transients, through activation of Ano1 channels and generation of inward current, cause net depolarization of colonic muscles. Ca2+ transients in ICC-IM rely on Ca2+ release from the endoplasmic reticulum via IP3 receptors, spatial amplification from RyRs and ongoing refilling of ER via the sarcoplasmic/endoplasmic-reticulum-Ca2+ -ATPase. ICC-IM are sustained by voltage-independent Ca2+ influx via store-operated Ca2+ entry. Some of the properties of Ca2+ in ICC-IM in the colon are similar to the behaviour of ICC located in the deep muscular plexus region of the small intestine, suggesting there are functional similarities between these classes of ICC. ABSTRACT A component of the SIP syncytium that regulates smooth muscle excitability in the colon is the intramuscular class of interstitial cells of Cajal (ICC-IM). All classes of ICC (including ICC-IM) express Ca2+ -activated Cl- channels, encoded by Ano1, and rely upon this conductance for physiological functions. Thus, Ca2+ handling in ICC is fundamental to colonic motility. We examined Ca2+ handling mechanisms in ICC-IM of murine proximal colon expressing GCaMP6f in ICC. Several Ca2+ firing sites were detected in each cell. While individual sites displayed rhythmic Ca2+ events, the overall pattern of Ca2+ transients was stochastic. No correlation was found between discrete Ca2+ firing sites in the same cell or in adjacent cells. Ca2+ transients in some cells initiated Ca2+ waves that spread along the cell at ∼100 µm s-1 . Ca2+ transients were caused by release from intracellular stores, but depended strongly on store-operated Ca2+ entry mechanisms. ICC Ca2+ transient firing regulated the resting membrane potential of colonic tissues as a specific Ano1 antagonist hyperpolarized colonic muscles by ∼10 mV. Ca2+ transient firing was independent of membrane potential and not affected by blockade of L- or T-type Ca2+ channels. Mechanisms regulating Ca2+ transients in the proximal colon displayed both similarities to and differences from the intramuscular type of ICC in the small intestine. Similarities and differences in Ca2+ release patterns might determine how ICC respond to neurotransmission in these two regions of the gastrointestinal tract.
Collapse
Affiliation(s)
- Bernard T Drumm
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, 89557, USA
| | - Sung J Hwang
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, 89557, USA
| | - Salah A Baker
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, 89557, USA
| | - Sean M Ward
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, 89557, USA
| | - Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, NV, 89557, USA
| |
Collapse
|
64
|
Duan J, Navarro-Dorado J, Clark JH, Kinnear NP, Meinke P, Schirmer EC, Evans AM. The cell-wide web coordinates cellular processes by directing site-specific Ca 2+ flux across cytoplasmic nanocourses. Nat Commun 2019; 10:2299. [PMID: 31127110 PMCID: PMC6534574 DOI: 10.1038/s41467-019-10055-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Accepted: 04/10/2019] [Indexed: 02/01/2023] Open
Abstract
Ca2+ coordinates diverse cellular processes, yet how function-specific signals arise is enigmatic. We describe a cell-wide network of distinct cytoplasmic nanocourses with the nucleus at its centre, demarcated by sarcoplasmic reticulum (SR) junctions (≤400 nm across) that restrict Ca2+ diffusion and by nanocourse-specific Ca2+-pumps that facilitate signal segregation. Ryanodine receptor subtype 1 (RyR1) supports relaxation of arterial myocytes by unloading Ca2+ into peripheral nanocourses delimited by plasmalemma-SR junctions, fed by sarco/endoplasmic reticulum Ca2+ ATPase 2b (SERCA2b). Conversely, stimulus-specified increases in Ca2+ flux through RyR2/3 clusters selects for rapid propagation of Ca2+ signals throughout deeper extraperinuclear nanocourses and thus myocyte contraction. Nuclear envelope invaginations incorporating SERCA1 in their outer nuclear membranes demarcate further diverse networks of cytoplasmic nanocourses that receive Ca2+ signals through discrete RyR1 clusters, impacting gene expression through epigenetic marks segregated by their associated invaginations. Critically, this circuit is not hardwired and remodels for different outputs during cell proliferation. Although calcium signals are known to be critical for many cellular processes, how signaling elicits specific functions remains unclear. In visually striking work, Duan et al. reveal that networks of cytoplasmic nanocourses orchestrate cell activity by directing site-specific calcium signals.
Collapse
Affiliation(s)
- Jingxian Duan
- Centres for Discovery Brain Sciences and Cardiovascular Sciences, College of Medicine and Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Jorge Navarro-Dorado
- Centres for Discovery Brain Sciences and Cardiovascular Sciences, College of Medicine and Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Jill H Clark
- Centres for Discovery Brain Sciences and Cardiovascular Sciences, College of Medicine and Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Nicholas P Kinnear
- Centres for Discovery Brain Sciences and Cardiovascular Sciences, College of Medicine and Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh, EH8 9XD, UK
| | - Peter Meinke
- Wellcome Centre for Cell Biology, Michael Swann Building, University of Edinburgh, Edinburgh, EH9 3BF, UK
| | - Eric C Schirmer
- Wellcome Centre for Cell Biology, Michael Swann Building, University of Edinburgh, Edinburgh, EH9 3BF, UK
| | - A Mark Evans
- Centres for Discovery Brain Sciences and Cardiovascular Sciences, College of Medicine and Veterinary Medicine, Hugh Robson Building, University of Edinburgh, Edinburgh, EH8 9XD, UK.
| |
Collapse
|
65
|
Kaßmann M, Szijártó IA, García‐Prieto CF, Fan G, Schleifenbaum J, Anistan Y, Tabeling C, Shi Y, le Noble F, Witzenrath M, Huang Y, Markó L, Nelson MT, Gollasch M. Role of Ryanodine Type 2 Receptors in Elementary Ca 2+ Signaling in Arteries and Vascular Adaptive Responses. J Am Heart Assoc 2019; 8:e010090. [PMID: 31030596 PMCID: PMC6512102 DOI: 10.1161/jaha.118.010090] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 02/07/2019] [Indexed: 12/29/2022]
Abstract
Background Hypertension is the major risk factor for cardiovascular disease, the most common cause of death worldwide. Resistance arteries are capable of adapting their diameter independently in response to pressure and flow-associated shear stress. Ryanodine receptors (RyRs) are major Ca2+-release channels in the sarcoplasmic reticulum membrane of myocytes that contribute to the regulation of contractility. Vascular smooth muscle cells exhibit 3 different RyR isoforms (RyR1, RyR2, and RyR3), but the impact of individual RyR isoforms on adaptive vascular responses is largely unknown. Herein, we generated tamoxifen-inducible smooth muscle cell-specific RyR2-deficient mice and tested the hypothesis that vascular smooth muscle cell RyR2s play a specific role in elementary Ca2+ signaling and adaptive vascular responses to vascular pressure and/or flow. Methods and Results Targeted deletion of the Ryr2 gene resulted in a complete loss of sarcoplasmic reticulum-mediated Ca2+-release events and associated Ca2+-activated, large-conductance K+ channel currents in peripheral arteries, leading to increased myogenic tone and systemic blood pressure. In the absence of RyR2, the pulmonary artery pressure response to sustained hypoxia was enhanced, but flow-dependent effects, including blood flow recovery in ischemic hind limbs, were unaffected. Conclusions Our results establish that RyR2-mediated Ca2+-release events in VSCM s specifically regulate myogenic tone (systemic circulation) and arterial adaptation in response to changes in pressure (hypoxic lung model), but not flow. They further suggest that vascular smooth muscle cell-expressed RyR2 deserves scrutiny as a therapeutic target for the treatment of vascular responses in hypertension and chronic vascular diseases.
Collapse
Affiliation(s)
- Mario Kaßmann
- Experimental and Clinical Research Centera joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular MedicineCharité–Universitätsmedizin BerlinBerlinGermany
- DZHK (German Centre for Cardiovascular Research), partner site BerlinBerlinGermany
| | - István András Szijártó
- Experimental and Clinical Research Centera joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular MedicineCharité–Universitätsmedizin BerlinBerlinGermany
| | - Concha F. García‐Prieto
- Experimental and Clinical Research Centera joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular MedicineCharité–Universitätsmedizin BerlinBerlinGermany
- Department of Pharmaceutical and Health SciencesFacultad de FarmaciaUniversidad CEU San PabloMadridSpain
| | - Gang Fan
- Experimental and Clinical Research Centera joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular MedicineCharité–Universitätsmedizin BerlinBerlinGermany
| | - Johanna Schleifenbaum
- Experimental and Clinical Research Centera joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular MedicineCharité–Universitätsmedizin BerlinBerlinGermany
| | - Yoland‐Marie Anistan
- Experimental and Clinical Research Centera joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular MedicineCharité–Universitätsmedizin BerlinBerlinGermany
| | - Christoph Tabeling
- Department of Infectious Diseases and Pulmonary MedicineCharité–Universitätsmedizin BerlinBerlinGermany
| | - Yu Shi
- Medical Clinic for Hematology, Oncology and Tumor ImmunologyCharité–Universitätsmedizin BerlinBerlinGermany
| | - Ferdinand le Noble
- Department of Cell and Developmental BiologyITG (Institute of Toxicology and Genetics)Karlsruhe Institute of TechnologyKarlsruheGermany
| | - Martin Witzenrath
- Department of Infectious Diseases and Pulmonary MedicineCharité–Universitätsmedizin BerlinBerlinGermany
| | - Yu Huang
- Institute of Vascular Medicine and School of Biomedical SciencesChinese University of Hong KongChina
| | - Lajos Markó
- Medical Clinic for Hematology, Oncology and Tumor ImmunologyCharité–Universitätsmedizin BerlinBerlinGermany
| | - Mark T. Nelson
- Department of PharmacologyCollege of MedicineThe University of VermontBurlingtonVT
| | - Maik Gollasch
- Experimental and Clinical Research Centera joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular MedicineCharité–Universitätsmedizin BerlinBerlinGermany
- DZHK (German Centre for Cardiovascular Research), partner site BerlinBerlinGermany
- Medical Clinic for Nephrology and Internal Intensive CareCharité–Universitätsmedizin BerlinBerlinGermany
| |
Collapse
|
66
|
Gupta S, Manchanda R. A computational model of large conductance voltage and calcium activated potassium channels: implications for calcium dynamics and electrophysiology in detrusor smooth muscle cells. J Comput Neurosci 2019; 46:233-256. [PMID: 31025235 DOI: 10.1007/s10827-019-00713-9] [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: 09/11/2018] [Revised: 02/14/2019] [Accepted: 02/19/2019] [Indexed: 11/25/2022]
Abstract
The large conductance voltage and calcium activated potassium (BK) channels play a crucial role in regulating the excitability of detrusor smooth muscle, which lines the wall of the urinary bladder. These channels have been widely characterized in terms of their molecular structure, pharmacology and electrophysiology. They control the repolarising and hyperpolarising phases of the action potential, thereby regulating the firing frequency and contraction profiles of the smooth muscle. Several groups have reported varied profiles of BK currents and I-V curves under similar experimental conditions. However, no single computational model has been able to reconcile these apparent discrepancies. In view of the channels' physiological importance, it is imperative to understand their mechanistic underpinnings so that a realistic model can be created. This paper presents a computational model of the BK channel, based on the Hodgkin-Huxley formalism, constructed by utilising three activation processes - membrane potential, calcium inflow from voltage-gated calcium channels on the membrane and calcium released from the ryanodine receptors present on the sarcoplasmic reticulum. In our model, we attribute the discrepant profiles to the underlying cytosolic calcium received by the channel during its activation. The model enables us to make heuristic predictions regarding the nature of the sub-membrane calcium dynamics underlying the BK channel's activation. We have employed the model to reproduce various physiological characteristics of the channel and found the simulated responses to be in accordance with the experimental findings. Additionally, we have used the model to investigate the role of this channel in electrophysiological signals, such as the action potential and spontaneous transient hyperpolarisations. Furthermore, the clinical effects of BK channel openers, mallotoxin and NS19504, were simulated for the detrusor smooth muscle cells. Our findings support the proposed application of these drugs for amelioration of the condition of overactive bladder. We thus propose a physiologically realistic BK channel model which can be integrated with other biophysical mechanisms such as ion channels, pumps and exchangers to further elucidate its micro-domain interaction with the intracellular calcium environment.
Collapse
Affiliation(s)
- Suranjana Gupta
- Computational NeuroPhysiology Lab, Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai, 400076, India
| | - Rohit Manchanda
- Computational NeuroPhysiology Lab, Department of Biosciences and Bioengineering, IIT Bombay, Powai, Mumbai, 400076, India.
| |
Collapse
|
67
|
Yan H, Zhang MZ, Wong G, Liu L, Kwok YSS, Kuang SJ, Yang H, Rao F, Li X, Mai LP, Lin QX, Yang M, Zhang QH, Deng CY. Mechanisms of U46619-induced contraction in mouse intrarenal artery. Clin Exp Pharmacol Physiol 2019; 46:643-651. [PMID: 30907443 DOI: 10.1111/1440-1681.13087] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 03/15/2019] [Accepted: 03/19/2019] [Indexed: 11/27/2022]
Abstract
Thromboxane A2 (TXA2 ) has been implicated in the pathogenesis of vascular complications, but the underlying mechanism remains unclear. The contraction of renal arterial rings in mice was measured by a Multi Myograph System. The intracellular calcium concentration ([Ca2+ ]i ) in vascular smooth muscle cells (VSMCs) was obtained by using a fluo-4/AM dye and a confocal laser scanning microscopy. The results show that the U46619-induced vasoconstriction of renal artery was completely blocked by a TXA2 receptor antagonist GR32191, significantly inhibited by a selective phospholipase C (PI-PLC) inhibitor U73122 at 10 μmol/L and partially inhibited by a Phosphatidylcholine - specific phospholipase C (PC-PLC) inhibitor D609 at 50 μmol/L. Moreover, the U46619-induced vasoconstriction was inhibited by a general protein kinase C (PKC) inhibitor chelerythrine at 10 μmol/L, and a selective PKCδ inhibitor rottlerin at 10 μmol/L. In addition, the PKC-induced vasoconstriction was partially inhibited by a Rho-kinase inhibitor Y-27632 at 10 μmol/L and was further completely inhibited together with a putative IP3 receptor antagonist and store-operated Ca2+ (SOC) entry inhibitor 2-APB at 100 μmol/L. On the other hand, U46619-induced vasoconstriction was partially inhibited by L-type calcium channel (Cav1.2) inhibitor nifedipine at 1 μmol/L and 2-APB at 50 and 100 μmol/L. Last, U46619-induced vasoconstriction was partially inhibited by a cell membrane Ca2+ activated C1- channel blocker 5-Nitro-2-(3-phenylpropylamino) benzoic acid (NPPB) at 50 and 100 μmol/L. Our results suggest that the U46619-induced contraction of mouse intrarenal arteries is mediated by Cav1.2 and SOC channel, through the activation of thromboxane-prostanoid receptors and its downstream signaling pathway.
Collapse
Affiliation(s)
- Hong Yan
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangzhou, China.,Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Meng-Zhen Zhang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangzhou, China.,Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China.,Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Gordon Wong
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangzhou, China.,Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China.,Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Lin Liu
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangzhou, China.,Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China.,Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Yat Sze Shelia Kwok
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangzhou, China.,Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China.,Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Su-Juan Kuang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangzhou, China.,Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China.,Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Hui Yang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangzhou, China.,Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China.,Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Fang Rao
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangzhou, China.,Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China.,Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Xin Li
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangzhou, China.,Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China.,Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Li-Ping Mai
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangzhou, China.,Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China.,Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Qiu-Xiong Lin
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangzhou, China.,Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China.,Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Min Yang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangzhou, China.,Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China.,Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Qian-Huan Zhang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangzhou, China.,Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China.,Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China
| | - Chun-Yu Deng
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangzhou, China.,Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China.,Guangdong Provincial Key Laboratory of Clinical Pharmacology, Guangdong Academy of Medical Sciences, Guangdong Provincial People's Hospital, Guangzhou, China
| |
Collapse
|
68
|
Leo MD, Zhai X, Yin W, Jaggar JH. Impaired Trafficking of β1 Subunits Inhibits BK Channels in Cerebral Arteries of Hypertensive Rats. Hypertension 2019; 72:765-775. [PMID: 30012867 DOI: 10.1161/hypertensionaha.118.11147] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Hypertension is a risk factor for cerebrovascular diseases, including stroke and dementia. During hypertension, arteries become constricted and are less responsive to vasodilators, including nitric oxide (NO). The regulation of arterial contractility by smooth muscle cell (myocyte) large-conductance calcium (Ca2+)-activated potassium (BK) channels is altered during hypertension, although mechanisms involved are unclear. We tested the hypothesis that dysfunctional trafficking of pore-forming BK channel (BKα) and auxiliary β1 subunits contributes to changes in cerebral artery contractility of stroke-prone spontaneously hypertensive rats (SP-SHRs). Our data indicate that the amounts of total and surface BKα and β1 proteins are similar in unstimulated arteries of age-matched SP-SHRs and normotensive Wistar-Kyoto rats. In contrast, stimulated surface-trafficking of β1 subunits by NO or membrane depolarization is inhibited in SP-SHR myocytes. PKCα (protein kinase C α) and PKCβII total protein and activity were both higher in SP-SHR than in Wistar-Kyoto rat arteries. NO or depolarization robustly activated Rab11, a small trafficking GTPase, in Wistar-Kyoto rat arteries but weakly activated Rab11 in SP-SHRs. Bisindolylmaleimide, a PKC inhibitor, and overexpression of a PKC phosphorylation-deficient Rab11A mutant (Rab11A S177A) restored stimulated β1 subunit surface-trafficking in SP-SHR myocytes. BK channel activation by NO was inhibited in SP-SHR myocytes and restored by Rab11A S177A expression. Vasodilation to NO and lithocholate, a BKα/β1 channel activator, was inhibited in pressurized SP-SHR arteries and reestablished by bisindolylmaleimide. In summary, data indicate that spontaneously active PKC inhibits Rab11A-mediated β1 subunit trafficking in arterial myocytes of SP-SHRs, leading to dysfunctional NO-induced BK channel activation and vasodilation.
Collapse
Affiliation(s)
- M Dennis Leo
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis
| | - Xue Zhai
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis
| | - Wen Yin
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis
| | - Jonathan H Jaggar
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis
| |
Collapse
|
69
|
|
70
|
Shen CH, Wang ST, Wang SC, Lin SM, Lin LC, Dai YC, Liu YW. Ketamine‑induced bladder dysfunction is associated with extracellular matrix accumulation and impairment of calcium signaling in a mouse model. Mol Med Rep 2019; 19:2716-2728. [PMID: 30720140 PMCID: PMC6423593 DOI: 10.3892/mmr.2019.9907] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 01/07/2019] [Indexed: 02/06/2023] Open
Abstract
Due to the rising abuse of ketamine usage in recent years, ketamine-induced urinary tract syndrome has received increasing attention. The present study aimed to investigate the molecular mechanism underlying ketamine-associated cystitis in a mouse model. Female C57BL/6 mice were randomly divided into two groups: One group was treated with ketamine (100 mg/kg/day of ketamine for 20 weeks), whereas, the control group was treated with saline solution. In each group, micturition frequency and urine volume were examined to assess urinary voiding functions. Mouse bladders were extracted and samples were examined for pathological and morphological alterations using hematoxylin and eosin staining, Masson's trichrome staining and scanning electron microscopy. A cDNA microarray was conducted to investigate the differentially expressed genes following treatment with ketamine. The results suggested that bladder hyperactivity increased in the mice treated with ketamine. Furthermore, treatment with ketamine resulted in a smooth apical epithelial surface, subepithelial vascular congestion and lymphoplasmacytic aggregation. Microarray analysis identified a number of genes involved in extracellular matrix accumulation, which is associated with connective tissue fibrosis progression, and in calcium signaling regulation, that was associated with urinary bladder smooth muscle contraction. Collectively, the present results suggested that these differentially expressed genes may serve critical roles in ketamine-induced alterations of micturition patterns and urothelial pathogenesis. Furthermore, the present findings may provide a theoretical basis for the development of effective therapies to treat ketamine-induced urinary tract syndrome.
Collapse
Affiliation(s)
- Cheng-Huang Shen
- Department of Urology, Chiayi Christian Hospital, Chiayi 600, Taiwan, R.O.C
| | - Shou-Tsung Wang
- Department of Food Science, National Chiayi University, Chiayi 600, Taiwan, R.O.C
| | - Shou-Chieh Wang
- Department of Food Science, National Chiayi University, Chiayi 600, Taiwan, R.O.C
| | - Shu-Mei Lin
- Department of Food Science, National Chiayi University, Chiayi 600, Taiwan, R.O.C
| | - Lei-Chen Lin
- Department of Forestry and Natural Resources, National Chiayi University, Chiayi 600, Taiwan, R.O.C
| | - Yuan-Chang Dai
- Department of Pathology, Chiayi Christian Hospital, Chiayi 600, Taiwan, R.O.C
| | - Yi-Wen Liu
- Department of Microbiology, Immunology and Biopharmaceuticals, National Chiayi University, Chiayi 600, Taiwan, R.O.C
| |
Collapse
|
71
|
Drumm BT, Hennig GW, Baker SA, Sanders KM. Applications of Spatio-temporal Mapping and Particle Analysis Techniques to Quantify Intracellular Ca2+ Signaling In Situ. J Vis Exp 2019. [PMID: 30663707 DOI: 10.3791/58989] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Ca2+ imaging of isolated cells or specific types of cells within intact tissues often reveals complex patterns of Ca2+ signaling. This activity requires careful and in-depth analyses and quantification to capture as much information about the underlying events as possible. Spatial, temporal and intensity parameters intrinsic to Ca2+ signals such as frequency, duration, propagation, velocity and amplitude may provide some biological information required for intracellular signalling. High-resolution Ca2+ imaging typically results in the acquisition of large data files that are time consuming to process in terms of translating the imaging information into quantifiable data, and this process can be susceptible to human error and bias. Analysis of Ca2+ signals from cells in situ typically relies on simple intensity measurements from arbitrarily selected regions of interest (ROI) within a field of view (FOV). This approach ignores much of the important signaling information contained in the FOV. Thus, in order to maximize recovery of information from such high-resolution recordings obtained with Ca2+dyes or optogenetic Ca2+ imaging, appropriate spatial and temporal analysis of the Ca2+ signals is required. The protocols outlined in this paper will describe how a high volume of data can be obtained from Ca2+ imaging recordings to facilitate more complete analysis and quantification of Ca2+ signals recorded from cells using a combination of spatiotemporal map (STM)-based analysis and particle-based analysis. The protocols also describe how different patterns of Ca2+ signaling observed in different cell populations in situ can be analyzed appropriately. For illustration, the method will examine Ca2+ signaling in a specialized population of cells in the small intestine, interstitial cells of Cajal (ICC), using GECIs.
Collapse
Affiliation(s)
- Bernard T Drumm
- Department of Physiology and Cell Biology, University of Nevada Reno School of Medicine;
| | - Grant W Hennig
- Department of Pharmacology, The Robert Larner, M.D. College of Medicine, University of Vermont
| | - Salah A Baker
- Department of Physiology and Cell Biology, University of Nevada Reno School of Medicine
| | - Kenton M Sanders
- Department of Physiology and Cell Biology, University of Nevada Reno School of Medicine
| |
Collapse
|
72
|
Thornbury KD, Hollywood MA, Sergeant GP. Ion Channels and Intracellular Calcium Signalling in Corpus Cavernosum. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1124:171-194. [PMID: 31183827 DOI: 10.1007/978-981-13-5895-1_7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The corpus cavernosum smooth muscle is important for both erection of the penis and for maintaining penile flaccidity. Most of the time, the smooth muscle cells are in a contracted state, which limits filling of the corpus sinuses with blood. Occasionally, however, they relax in a co-ordinated manner, allowing filling to occur. This results in an erection. When contractions of the corpus cavernosum are measured, it can be deduced that the muscle cells work together in a syncytium, for not only do they spontaneously contract in a co-ordinated manner, but they also synchronously relax. It is challenging to understand how they achieve this.In this review we will attempt to explain the activity of the corpus cavernosum, firstly by summarising current knowledge regarding the role of ion channels and how they influence tone, and secondly by presenting data on the intracellular Ca2+ signals that interact with the ion channels. We propose that spontaneous Ca2+ waves act as a primary event, driving transient depolarisation by activating Ca2+-activated Cl- channels. Depolarisation then facilitates Ca2+ influx via L-type voltage-dependent Ca2+ channels. We propose that the spontaneous Ca2+ oscillations depend on Ca2+ release from both ryanodine- and inositol trisphosphate (IP3)-sensitive stores and that modulation by signalling molecules is achieved mainly by interactions with the IP3-sensitive mechanism. This pacemaker mechanism is inhibited by nitric oxide (acting through cyclic GMP) and enhanced by noradrenaline. By understanding these mechanisms better, it might be possible to design new treatments for erectile dysfunction.
Collapse
Affiliation(s)
- Keith D Thornbury
- Smooth Muscle Research Centre, Regional Development Centre, Dundalk Institute of Technology, Dundalk, Co. Louth, Ireland.
| | - Mark A Hollywood
- Smooth Muscle Research Centre, Regional Development Centre, Dundalk Institute of Technology, Dundalk, Co. Louth, Ireland
| | - Gerard P Sergeant
- Smooth Muscle Research Centre, Regional Development Centre, Dundalk Institute of Technology, Dundalk, Co. Louth, Ireland
| |
Collapse
|
73
|
Li X, Feng X, Lu L, He A, Liu B, Zhang Y, Shi R, Liu Y, Chen X, Sun M, Xu Z. Prenatal hypoxia plus postnatal high-fat diet exacerbated vascular dysfunction via up-regulated vascular Cav1.2 channels in offspring rats. J Cell Mol Med 2018; 23:1183-1196. [PMID: 30556291 PMCID: PMC6349350 DOI: 10.1111/jcmm.14020] [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] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 10/10/2018] [Accepted: 10/20/2018] [Indexed: 12/13/2022] Open
Abstract
Background This study aimed to examine whether and how postnatal high‐fat diet had additional impact on promoting vascular dysfunction in the offspring exposed to prenatal hypoxia. Methods and Results Pregnant Sprague‐Dawley rats were randomly assigned to hypoxia (10.5% oxygen) or normoxia (21% O2) groups from gestation days 5‐21. A subset of male offspring was placed on a high‐fat diet (HF, 45% fat) from 4‐16 weeks of age. Prenatal hypoxia induced a decrease in birth weight. In offspring‐fed HF diet, prenatal hypoxia was associated with increased fasting plasma triglyceride, total cholesterol, free fatty acids, and low‐density lipoprotein‐cholesterol. Compared with the other three groups, prenatal hypoxic offspring with high‐fat diet showed a significant increase in blood pressure, phenylephrine‐mediated vasoconstrictions, L‐type voltage‐gated Ca2+ (Cav1.2) channel currents, and elevated mRNA and protein expression of Cav1.2 α1 subunit in mesenteric arteries or myocytes. The large‐conductance Ca2+‐activated K+ (BK) channels currents and the BK channel units (β1, not α‐subunits) were significantly increased in mesenteric arteries or myocytes in HF offspring independent of prenatal hypoxia factor. Conclusion The results demonstrated that prenatal hypoxia followed by postnatal HF caused vascular dysfunction through ion channel remodelling in myocytes.
Collapse
Affiliation(s)
- Xiang Li
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Xueqin Feng
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Likui Lu
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Axin He
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Bailin Liu
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Yingying Zhang
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Ruixiu Shi
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Yanping Liu
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Xueyi Chen
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Miao Sun
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Zhice Xu
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China.,Center for Perinatal Biology, Loma Linda University, Loma Linda, California
| |
Collapse
|
74
|
Bulley S, Fernández-Peña C, Hasan R, Leo MD, Muralidharan P, Mackay CE, Evanson KW, Moreira-Junior L, Mata-Daboin A, Burris SK, Wang Q, Kuruvilla KP, Jaggar JH. Arterial smooth muscle cell PKD2 (TRPP1) channels regulate systemic blood pressure. eLife 2018; 7:42628. [PMID: 30511640 PMCID: PMC6281320 DOI: 10.7554/elife.42628] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 11/22/2018] [Indexed: 01/12/2023] Open
Abstract
Systemic blood pressure is determined, in part, by arterial smooth muscle cells (myocytes). Several Transient Receptor Potential (TRP) channels are proposed to be expressed in arterial myocytes, but it is unclear if these proteins control physiological blood pressure and contribute to hypertension in vivo. We generated the first inducible, smooth muscle-specific knockout mice for a TRP channel, namely for PKD2 (TRPP1), to investigate arterial myocyte and blood pressure regulation by this protein. Using this model, we show that intravascular pressure and α1-adrenoceptors activate PKD2 channels in arterial myocytes of different systemic organs. PKD2 channel activation in arterial myocytes leads to an inward Na+ current, membrane depolarization and vasoconstriction. Inducible, smooth muscle cell-specific PKD2 knockout lowers both physiological blood pressure and hypertension and prevents pathological arterial remodeling during hypertension. Thus, arterial myocyte PKD2 controls systemic blood pressure and targeting this TRP channel reduces high blood pressure.
Collapse
Affiliation(s)
- Simon Bulley
- Department of Physiology, University of Tennessee Health Science Center, Memphis, United States
| | - Carlos Fernández-Peña
- Department of Physiology, University of Tennessee Health Science Center, Memphis, United States
| | - Raquibul Hasan
- Department of Physiology, University of Tennessee Health Science Center, Memphis, United States
| | - M Dennis Leo
- Department of Physiology, University of Tennessee Health Science Center, Memphis, United States
| | - Padmapriya Muralidharan
- Department of Physiology, University of Tennessee Health Science Center, Memphis, United States
| | - Charles E Mackay
- Department of Physiology, University of Tennessee Health Science Center, Memphis, United States
| | - Kirk W Evanson
- Department of Physiology, University of Tennessee Health Science Center, Memphis, United States
| | - Luiz Moreira-Junior
- Department of Physiology, University of Tennessee Health Science Center, Memphis, United States
| | - Alejandro Mata-Daboin
- Department of Physiology, University of Tennessee Health Science Center, Memphis, United States
| | - Sarah K Burris
- Department of Physiology, University of Tennessee Health Science Center, Memphis, United States
| | - Qian Wang
- Department of Physiology, University of Tennessee Health Science Center, Memphis, United States
| | - Korah P Kuruvilla
- Department of Physiology, University of Tennessee Health Science Center, Memphis, United States
| | - Jonathan H Jaggar
- Department of Physiology, University of Tennessee Health Science Center, Memphis, United States
| |
Collapse
|
75
|
Ryanodine receptor cluster size sets the tone in cerebral smooth muscle. Proc Natl Acad Sci U S A 2018; 115:10195-10197. [PMID: 30257942 DOI: 10.1073/pnas.1814207115] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
|
76
|
Fan G, Kaßmann M, Hashad AM, Welsh DG, Gollasch M. Differential targeting and signalling of voltage-gated T-type Ca v 3.2 and L-type Ca v 1.2 channels to ryanodine receptors in mesenteric arteries. J Physiol 2018; 596:4863-4877. [PMID: 30146760 PMCID: PMC6187032 DOI: 10.1113/jp276923] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 08/24/2018] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS In arterial smooth muscle, Ca2+ sparks are elementary Ca2+ -release events generated by ryanodine receptors (RyRs) to cause vasodilatation by opening maxi Ca2+ -sensitive K+ (BKCa ) channels. This study elucidated the contribution of T-type Cav 3.2 channels in caveolae and their functional interaction with L-type Cav 1.2 channels to trigger Ca2+ sparks in vascular smooth muscle cells (VSMCs). Our data demonstrate that L-type Cav 1.2 channels provide the predominant Ca2+ pathway for the generation of Ca2+ sparks in murine arterial VSMCs. T-type Cav 3.2 channels represent an additional source for generation of VSMC Ca2+ sparks. They are located in pit structures of caveolae to provide locally restricted, tight coupling between T-type Cav 3.2 channels and RyRs to ignite Ca2+ sparks. ABSTRACT Recent data suggest that T-type Cav 3.2 channels in arterial vascular smooth muscle cells (VSMCs) and pits structure of caveolae could contribute to elementary Ca2+ signalling (Ca2+ sparks) via ryanodine receptors (RyRs) to cause vasodilatation. While plausible, their precise involvement in igniting Ca2+ sparks remains largely unexplored. The goal of this study was to elucidate the contribution of caveolar Cav 3.2 channels and their functional interaction with Cav 1.2 channels to trigger Ca2+ sparks in VSMCs from mesenteric, tibial and cerebral arteries. We used tamoxifen-inducible smooth muscle-specific Cav 1.2-/- (SMAKO) mice and laser scanning confocal microscopy to assess Ca2+ spark generation in VSMCs. Ni2+ , Cd2+ and methyl-β-cyclodextrin were used to inhibit Cav 3.2 channels, Cav 1.2 channels and caveolae, respectively. Ni2+ (50 μmol L-1 ) and methyl-β-cyclodextrin (10 mmol L-1 ) decreased Ca2+ spark frequency by ∼20-30% in mesenteric VSMCs in a non-additive manner, but failed to inhibit Ca2+ sparks in tibial and cerebral artery VSMCs. Cd2+ (200 μmol L-1 ) suppressed Ca2+ sparks in mesenteric arteries by ∼70-80%. A similar suppression of Ca2+ sparks was seen in mesenteric artery VSMCs of SMAKO mice. The remaining Ca2+ sparks were fully abolished by Ni2+ or methyl-β-cyclodextrin. Our data demonstrate that Ca2+ influx through CaV 1.2 channels is the primary means of triggering Ca2+ sparks in murine arterial VSMCs. CaV 3.2 channels, localized to caveolae and tightly coupled to RyR, provide an additional Ca2+ source for Ca2+ spark generation in mesenteric, but not tibial and cerebral, arteries.
Collapse
Affiliation(s)
- Gang Fan
- Charité – Universitätsmedizin BerlinExperimental and Clinical Research Center (ECRC)Campus BuchBerlinGermany
| | - Mario Kaßmann
- Charité – Universitätsmedizin BerlinExperimental and Clinical Research Center (ECRC)Campus BuchBerlinGermany
- DZHK (German Centre for Cardiovascular Research)BerlinGermany
| | - Ahmed M. Hashad
- Department of Physiology and PharmacologyHotchkiss Brain and Libin Cardiovascular InstitutesUniversity of CalgaryAlbertaCanada
| | - Donald G. Welsh
- Department of Physiology and PharmacologyWestern UniversityLondonONCanada
| | - Maik Gollasch
- Charité – Universitätsmedizin BerlinExperimental and Clinical Research Center (ECRC)Campus BuchBerlinGermany
- DZHK (German Centre for Cardiovascular Research)BerlinGermany
- Charité – Universitätsmedizin BerlinMedical Clinic for Nephrology and Internal Intensive CareCampus VirchowBerlinGermany
| |
Collapse
|
77
|
Nanoscale remodeling of ryanodine receptor cluster size underlies cerebral microvascular dysfunction in Duchenne muscular dystrophy. Proc Natl Acad Sci U S A 2018; 115:E9745-E9752. [PMID: 30181262 PMCID: PMC6187127 DOI: 10.1073/pnas.1804593115] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a hereditary neuromuscular disease that results from mutations in the gene encoding dystrophin. The effects of the disease on cardiac and skeletal muscle have been intensely investigated, but much less is known about how DMD impacts vascular smooth muscle cells (SMCs). Using superresolution nanoscopy, we demonstrate that clusters of ryanodine receptors (RyR2s) on the sarcoplasmic reticulum (SR) of cerebral artery SMCs from the mdx mouse model of DMD are larger compared with controls. Increased RyR2 cluster size is associated with augmented SR Ca2+ release and Ca2+-activated K+ channel activity, resulting in impaired vasoconstriction of cerebral microvessels. Our findings demonstrate that remodeling of RyR2 clusters at the molecular level results in cerebral microvascular dysfunction during DMD. Duchenne muscular dystrophy (DMD) results from mutations in the gene encoding dystrophin which lead to impaired function of skeletal and cardiac muscle, but little is known about the effects of the disease on vascular smooth muscle cells (SMCs). Here we used the mdx mouse model to study the effects of mutant dystrophin on the regulation of cerebral artery and arteriole SMC contractility, focusing on an important Ca2+-signaling pathway composed of type 2 ryanodine receptors (RyR2s) on the sarcoplasmic reticulum (SR) and large-conductance Ca2+-activated K+ (BK) channels on the plasma membrane. Nanoscale superresolution image analysis revealed that RyR2 and BKα were organized into discrete clusters, and that the mean size of RyR2 clusters that colocalized with BKα was larger in SMCs from mdx mice (∼62 RyR2 monomers) than in controls (∼40 RyR2 monomers). We further found that the frequency and signal mass of spontaneous, transient Ca2+-release events through SR RyR2s (“Ca2+ sparks”) were greater in SMCs from mdx mice. Patch-clamp electrophysiological recordings indicated a corresponding increase in Ca2+-dependent BK channel activity. Using pressure myography, we found that cerebral pial arteries and parenchymal arterioles from mdx mice failed to develop appreciable spontaneous myogenic tone. Inhibition of RyRs with tetracaine and blocking of BK channels with paxilline restored myogenic tone to control levels, demonstrating that enhanced RyR and BK channel activity is responsible for the diminished pressure-induced constriction of arteries and arterioles from mdx mice. We conclude that increased size of RyR2 protein clusters in SMCs from mdx mice increases Ca2+ spark and BK channel activity, resulting in cerebral microvascular dysfunction.
Collapse
|
78
|
Agrawal A, Suryakumar G, Rathor R. Role of defective Ca 2+ signaling in skeletal muscle weakness: Pharmacological implications. J Cell Commun Signal 2018; 12:645-659. [PMID: 29982883 DOI: 10.1007/s12079-018-0477-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 06/27/2018] [Indexed: 01/19/2023] Open
Abstract
The misbehaving attitude of Ca2+ signaling pathways could be the probable reason in many muscular disorders such as myopathies, systemic disorders like hypoxia, sepsis, cachexia, sarcopenia, heart failure, and dystrophy. The present review throws light upon the calcium flux regulating signaling channels like ryanodine receptor complex (RyR1), SERCA (Sarco-endoplasmic Reticulum Calcium ATPase), DHPR (Dihydropyridine Receptor) or Cav1.1 and Na+/Ca2+ exchange pump in detail and how remodelling of these channels contribute towards disturbed calcium homeostasis. Understanding these pathways will further provide an insight for establishing new therapeutic approaches for the prevention and treatment of muscle atrophy under stress conditions, targeting calcium ion channels and associated regulatory proteins.
Collapse
Affiliation(s)
- Akanksha Agrawal
- DRDO, Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi, 110054, India
| | - Geetha Suryakumar
- DRDO, Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi, 110054, India
| | - Richa Rathor
- DRDO, Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi, 110054, India.
| |
Collapse
|
79
|
Evanson KW, Goldsmith JA, Ghosh P, Delp MD. The G protein-coupled estrogen receptor agonist, G-1, attenuates BK channel activation in cerebral arterial smooth muscle cells. Pharmacol Res Perspect 2018; 6:e00409. [PMID: 29938113 PMCID: PMC6011940 DOI: 10.1002/prp2.409] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Accepted: 05/22/2018] [Indexed: 11/07/2022] Open
Abstract
The G protein-coupled estrogen receptor (GPER) is a significant modulator of arterial contractility and blood flow. The GPER-specific activator, G-1, has been widely used to characterize GPER function in a variety of tissue types. Large conductance, calcium (Ca2+)-activated K+ (BK) channels are sensitive to 17β-estradiol (17β-E2) and estrogenic compounds (e.g., tamoxifen, ICI 182 780) that target estrogen receptors. The purpose of this study was to investigate the effects of G-1 on BK channel activation and function in cerebral arterial myocytes. Inside-out and perforated patch clamp were utilized to assess the effects of G-1 (50 nmol·L-1-5 μmol·L-1) on BK channel activation and currents in cerebral arterial myocytes. Pressurized artery myography was used to investigate the effects of G-1 on vasodilatory response and BK channel function of cerebral resistance size arteries. G-1 reduced BK channel activation in cerebral arterial myocytes through elevations in BK channel mean close times. Depressed BK channel activation following G-1 application resulted in attenuated physiological BK currents (transient BK currents). G-1 elicited vasodilation, but reduced BK channel function, in pressurized, endothelium-denuded cerebral arteries. These data suggest that G-1 directly suppresses BK channel activation and currents in cerebral arterial myocytes, BK channels being critically important in the regulation of myocyte membrane potential and arterial contractility. Thus, GPER-mediated vasodilation using G-1 to activate the receptor may underestimate the physiological function and relevance of GPER in the cardiovascular system.
Collapse
Affiliation(s)
- Kirk W. Evanson
- Department of Nutrition, Food, and Exercise SciencesFlorida State UniversityTallahasseeFlorida
| | - Jacob A. Goldsmith
- Department of Nutrition, Food, and Exercise SciencesFlorida State UniversityTallahasseeFlorida
| | - Payal Ghosh
- Department of Nutrition, Food, and Exercise SciencesFlorida State UniversityTallahasseeFlorida
| | - Michael D. Delp
- Department of Nutrition, Food, and Exercise SciencesFlorida State UniversityTallahasseeFlorida
| |
Collapse
|
80
|
Dopico AM, Bukiya AN, Jaggar JH. Calcium- and voltage-gated BK channels in vascular smooth muscle. Pflugers Arch 2018; 470:1271-1289. [PMID: 29748711 DOI: 10.1007/s00424-018-2151-y] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 04/27/2018] [Indexed: 02/04/2023]
Abstract
Ion channels in vascular smooth muscle regulate myogenic tone and vessel contractility. In particular, activation of calcium- and voltage-gated potassium channels of large conductance (BK channels) results in outward current that shifts the membrane potential toward more negative values, triggering a negative feed-back loop on depolarization-induced calcium influx and SM contraction. In this short review, we first present the molecular basis of vascular smooth muscle BK channels and the role of subunit composition and trafficking in the regulation of myogenic tone and vascular contractility. BK channel modulation by endogenous signaling molecules, and paracrine and endocrine mediators follows. Lastly, we describe the functional changes in smooth muscle BK channels that contribute to, or are triggered by, common physiological conditions and pathologies, including obesity, diabetes, and systemic hypertension.
Collapse
Affiliation(s)
- Alex M Dopico
- Department of Pharmacology, College of Medicine, The University of Tennessee Health Science Center, 71 South Manassas St., Memphis, TN, 38163, USA.
| | - Anna N Bukiya
- Department of Pharmacology, College of Medicine, The University of Tennessee Health Science Center, 71 South Manassas St., Memphis, TN, 38163, USA
| | - Jonathan H Jaggar
- Department of Physiology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
81
|
Drumm BT, Rembetski BE, Cobine CA, Baker SA, Sergeant GP, Hollywood MA, Thornbury KD, Sanders KM. Ca 2+ signalling in mouse urethral smooth muscle in situ: role of Ca 2+ stores and Ca 2+ influx mechanisms. J Physiol 2018; 596:1433-1466. [PMID: 29383731 PMCID: PMC5899989 DOI: 10.1113/jp275719] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 01/17/2018] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Contraction of urethral smooth muscle cells (USMCs) contributes to urinary continence. Ca2+ signalling in USMCs was investigated in intact urethral muscles using a genetically encoded Ca2+ sensor, GCaMP3, expressed selectively in USMCs. USMCs were spontaneously active in situ, firing intracellular Ca2+ waves that were asynchronous at different sites within cells and between adjacent cells. Spontaneous Ca2+ waves in USMCs were myogenic but enhanced by adrenergic or purinergic agonists and decreased by nitric oxide. Ca2+ waves arose from inositol trisphosphate type 1 receptors and ryanodine receptors, and Ca2+ influx by store-operated calcium entry was required to maintain Ca2+ release events. Ca2+ release and development of Ca2+ waves appear to be the primary source of Ca2+ for excitation-contraction coupling in the mouse urethra, and no evidence was found that voltage-dependent Ca2+ entry via L-type or T-type channels was required for responses to α adrenergic responses. ABSTRACT Urethral smooth muscle cells (USMCs) generate myogenic tone and contribute to urinary continence. Currently, little is known about Ca2+ signalling in USMCs in situ, and therefore little is known about the source(s) of Ca2+ required for excitation-contraction coupling. We characterized Ca2+ signalling in USMCs within intact urethral muscles using a genetically encoded Ca2+ sensor, GCaMP3, expressed selectively in USMCs. USMCs fired spontaneous intracellular Ca2+ waves that did not propagate cell-to-cell across muscle bundles. Ca2+ waves increased dramatically in response to the α1 adrenoceptor agonist phenylephrine (10 μm) and to ATP (10 μm). Ca2+ waves were inhibited by the nitric oxide donor DEA NONOate (10 μm). Ca2+ influx and release from sarcoplasmic reticulum stores contributed to Ca2+ waves, as Ca2+ free bathing solution and blocking the sarcoplasmic Ca2+ -ATPase abolished activity. Intracellular Ca2+ release involved cooperation between ryanadine receptors and inositol trisphosphate receptors, as tetracaine and ryanodine (100 μm) and xestospongin C (1 μm) reduced Ca2+ waves. Ca2+ waves were insensitive to L-type Ca2+ channel modulators nifedipine (1 μm), nicardipine (1 μm), isradipine (1 μm) and FPL 64176 (1 μm), and were unaffected by the T-type Ca2+ channel antagonists NNC-550396 (1 μm) and TTA-A2 (1 μm). Ca2+ waves were reduced by the store operated Ca2+ entry blocker SKF 96365 (10 μm) and by an Orai antagonist, GSK-7975A (1 μm). The latter also reduced urethral contractions induced by phenylephrine, suggesting that Orai can function effectively as a receptor-operated channel. In conclusion, Ca2+ waves in mouse USMCs are a source of Ca2+ for excitation-contraction coupling in urethral muscles.
Collapse
Affiliation(s)
- Bernard T. Drumm
- Department of Physiology and Cell BiologyUniversity of Nevada, Reno School of MedicineRenoNV89557USA
| | - Benjamin E. Rembetski
- Department of Physiology and Cell BiologyUniversity of Nevada, Reno School of MedicineRenoNV89557USA
| | - Caroline A. Cobine
- Department of Physiology and Cell BiologyUniversity of Nevada, Reno School of MedicineRenoNV89557USA
| | - Salah A. Baker
- Department of Physiology and Cell BiologyUniversity of Nevada, Reno School of MedicineRenoNV89557USA
| | - Gerard P. Sergeant
- Smooth Muscle Research CentreDundalk Institute of TechnologyCo. LouthDundalkRepublic of Ireland
| | - Mark A. Hollywood
- Smooth Muscle Research CentreDundalk Institute of TechnologyCo. LouthDundalkRepublic of Ireland
| | - Keith D. Thornbury
- Smooth Muscle Research CentreDundalk Institute of TechnologyCo. LouthDundalkRepublic of Ireland
| | - Kenton M. Sanders
- Department of Physiology and Cell BiologyUniversity of Nevada, Reno School of MedicineRenoNV89557USA
| |
Collapse
|
82
|
Liu B, Shi R, Li X, Liu Y, Feng X, Chen X, Fan X, Zhang Y, Zhang W, Tang J, Zhou X, Li N, Lu X, Xu Z. Downregulation of L-Type Voltage-Gated Ca 2+, Voltage-Gated K +, and Large-Conductance Ca 2+-Activated K + Channels in Vascular Myocytes From Salt-Loading Offspring Rats Exposed to Prenatal Hypoxia. J Am Heart Assoc 2018; 7:JAHA.117.008148. [PMID: 29545262 PMCID: PMC5907567 DOI: 10.1161/jaha.117.008148] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Background Prenatal hypoxia is suggested to be associated with increased risks of hypertension in offspring. This study tested whether prenatal hypoxia resulted in salt‐sensitive offspring and its related mechanisms of vascular ion channel remodeling. Methods and Results Pregnant rats were housed in a normoxic (21% O2) or hypoxic (10.5% O2) chamber from gestation days 5 to 21. A subset of male offspring received a high‐salt diet (8% NaCl) from 4 to 12 weeks after birth. Blood pressure was significantly increased only in the salt‐loading offspring exposed to prenatal hypoxia, not in the offspring that received regular diets and in control offspring provided with high‐salt diets. In mesenteric artery myocytes from the salt‐loading offspring with prenatal hypoxia, depolarized resting membrane potential was associated with decreased density of L‐type voltage‐gated Ca2+ (Cav1.2) and voltage‐gated K+ channel currents and decreased calcium sensitive to the large‐conductance Ca2+‐activated K+ channels. Protein expression of the L‐type voltage‐gated Ca2+ α1C subunit, large‐conductance calcium‐activated K+ channel (β1, not α subunits), and voltage‐gated K+ channel (KV2.1, not KV1.5 subunits) was also decreased in the arteries of salt‐loading offspring with prenatal hypoxia. Conclusions The results demonstrated that chronic prenatal hypoxia may program salt‐sensitive hypertension in male offspring, providing new information of ion channel remodeling in hypertensive myocytes. This information paves the way for early prevention and treatments of salt‐induced hypertension related to developmental problems in fetal origins.
Collapse
Affiliation(s)
- Bailin Liu
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Ruixiu Shi
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Xiang Li
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Yanping Liu
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Xueqin Feng
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Xueyi Chen
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Xiaorong Fan
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Yingying Zhang
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Wenna Zhang
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Jiaqi Tang
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Xiuwen Zhou
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Na Li
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Xiyuan Lu
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China
| | - Zhice Xu
- Institute for Fetology, First Hospital of Soochow University, Suzhou, China .,Center for Perinatal Biology, Loma Linda University, Loma Linda, CA
| |
Collapse
|
83
|
Lorca RA, Wakle‐Prabagaran M, Freeman WE, Pillai MK, England SK. The large-conductance voltage- and Ca 2+ -activated K + channel and its γ1-subunit modulate mouse uterine artery function during pregnancy. J Physiol 2018; 596:1019-1033. [PMID: 29319186 PMCID: PMC5851882 DOI: 10.1113/jp274524] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 01/05/2018] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS The uterine artery (UA) markedly vasodilates during pregnancy to direct blood flow to the developing fetus. Inadequate UA vasodilatation leads to intrauterine growth restriction and fetal death. The large-conductance voltage- and Ca2+ -activated K+ (BKCa ) channel promotes UA vasodilatation during pregnancy. We report that BKCa channel activation increases the UA diameter at late pregnancy stages in mice. Additionally, a BKCa channel auxiliary subunit, γ1, participates in this process by increasing channel activation and inducing UA vasodilatation at late pregnancy stages. Our results highlight the importance of the BKCa channel and its γ1-subunit for UA functional changes during pregnancy. ABSTRACT Insufficient vasodilatation of the uterine artery (UA) during pregnancy leads to poor utero-placental perfusion, contributing to intrauterine growth restriction and fetal loss. Activity of the large-conductance Ca2+ -activated K+ (BKCa ) channel increases in the UA during pregnancy, and its inhibition reduces uterine blood flow, highlighting a role of this channel in UA adaptation to pregnancy. The auxiliary γ1-subunit increases BKCa activation in vascular smooth muscle, but its role in pregnancy-associated UA remodelling is unknown. We explored whether the BKCa and its γ1-subunit contribute to UA remodelling during pregnancy. Doppler imaging revealed that, compared to UAs from wild-type (WT) mice, UAs from BKCa knockout (BKCa-/- ) mice had lower resistance at pregnancy day 14 (P14) but not at P18. Lumen diameters were twofold larger in pressurized UAs from P18 WT mice than in those from non-pregnant mice, but this difference was not seen in UAs from BKCa-/- mice. UAs from pregnant WT mice constricted 20-50% in response to the BKCa blocker iberiotoxin (IbTX), whereas UAs from non-pregnant WT mice only constricted 15%. Patch-clamp analysis of WT UA smooth muscle cells confirmed that BKCa activity increased over pregnancy, showing three distinct voltage sensitivities. The γ1-subunit transcript increased 7- to 10-fold during pregnancy. Furthermore, γ1-subunit knockdown reduced IbTX sensitivity in UAs from pregnant mice, whereas γ1-subunit overexpression increased IbTX sensitivity in UAs from non-pregnant mice. Finally, at P18, γ1-knockout (γ1-/- ) mice had smaller UA diameters than WT mice, and IbTX-mediated vasoconstriction was prevented in UAs from γ1-/- mice. Our results suggest that the γ1-subunit increases BKCa activation in UAs during pregnancy.
Collapse
Affiliation(s)
- Ramón A. Lorca
- Center for Reproductive Health SciencesDepartment of Obstetrics and GynecologyWashington University in St Louis School of MedicineSt LouisMO63110USA
| | - Monali Wakle‐Prabagaran
- Center for Reproductive Health SciencesDepartment of Obstetrics and GynecologyWashington University in St Louis School of MedicineSt LouisMO63110USA
| | - William E. Freeman
- Center for Reproductive Health SciencesDepartment of Obstetrics and GynecologyWashington University in St Louis School of MedicineSt LouisMO63110USA
| | - Meghan K. Pillai
- Center for Reproductive Health SciencesDepartment of Obstetrics and GynecologyWashington University in St Louis School of MedicineSt LouisMO63110USA
| | - Sarah K. England
- Center for Reproductive Health SciencesDepartment of Obstetrics and GynecologyWashington University in St Louis School of MedicineSt LouisMO63110USA
| |
Collapse
|
84
|
Activation of human smooth muscle BK channels by hydrochlorothiazide requires cell integrity and the presence of BK β 1 subunit. Acta Pharmacol Sin 2018; 39:371-381. [PMID: 29188803 DOI: 10.1038/aps.2017.133] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 08/28/2017] [Indexed: 12/13/2022] Open
Abstract
Thiazide-like diuretics are the most commonly used drugs to treat arterial hypertension, with their efficacy being linked to their chronic vasodilatory effect. Previous studies suggest that activation of the large conductance voltage- and Ca2+-dependent K+ (BK) channel (Slo 1, MaxiK channel) is responsible for the thiazide-induced vasodilatory effect. But the direct electrophysiological evidence supporting this claim is lacking. BK channels can be associated with one small accessory β-subunit (β1-β4) that confers specific biophysical and pharmacological characteristics to the current phenotype. The β1-subunit is primarily expressed in smooth muscle cells (SMCs). In this study we investigated the effect of hydrochlorothiazide (HCTZ) on BK channel activity in native SMCs from human umbilical artery (HUASMCs) and HEK293T cells expressing the BK channel (with and without the β1-subunit). Bath application of HCTZ (10 μmol/L) significantly augmented the BK current in HUASMCs when recorded using the whole-cell configurations, but it did not affect the unitary conductance and open probability of the BK channel in HUASMCs evaluated in the inside-out configuration, suggesting an indirect mechanism requiring cell integrity. In HEK293T cells expressing BK channels, HCTZ-augmented BK channel activity was only observed when the β1-subunit was co-expressed, being concentration-dependent with an EC50 of 28.4 μmol/L, whereas membrane potential did not influence the concentration relationship. Moreover, HCTZ did not affect the BK channel current in HEK293T cells evaluated in the inside-out configuration, but significantly increases the open probability in the cell-attached configuration. Our data demonstrate that a β1-subunit-dependent mechanism that requires SMC integrity leads to HCTZ-induced BK channel activation.
Collapse
|
85
|
Zhang XD, He CX, Cheng J, Wen J, Li PY, Wang N, Li G, Zeng XR, Cao JM, Yang Y. Sodium Tanshinone II-A Sulfonate (DS-201) Induces Vasorelaxation of Rat Mesenteric Arteries via Inhibition of L-Type Ca 2+ Channel. Front Pharmacol 2018; 9:62. [PMID: 29456510 PMCID: PMC5801295 DOI: 10.3389/fphar.2018.00062] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Accepted: 01/17/2018] [Indexed: 11/13/2022] Open
Abstract
Background: We previously have proved that sodium tanshinone II-A sulfonate (DS-201), a derivative of traditional Chinese medicinal herb Danshen (Salvia miltiorrhiza), is an opener and vasodilator of BKCa channel in the vascular smooth muscle cells (VSMCs). Vascular tension is closely associated with Ca2+ dynamics and activation of BKCa channel may not be the sole mechanism for the relaxation of the vascular tension by DS-201. Therefore, we hypothesized that the vasorelaxing effect of DS-20 may be also related to Ca2+ channel and cytoplasmic Ca2+ level in the VSMCs. Methods: Arterial tension was measured by Danish Myo Technology (DMT) myograph system in the mesentery vessels of rats, intracellular Ca2+ level by fluorescence imaging system in the VSMCs of rats, and L-type Ca2+ current by patch clamp technique in Ca2+ channels transfected human embryonic kidney 293 (HEK-293) cells. Results: DS-201 relaxed the endothelium-denuded artery rings pre-constricted with PE or high K+ and the vasorelaxation was reversible. Blockade of K+ channel did not totally block the effect of DS-201 on vasorelaxation. DS-201 suppressed [Ca2+]i transient induced by high K+ in a concentration-dependent manner in the VSMCs, including the amplitude of Ca2+ transient, the time for Ca2+ transient reaching to the [Ca2+]i peak and the time to remove Ca2+ from the cytoplasm. DS-201 inhibited L-type Ca2+ channel with an EC50 of 59.5 μM and at about 40% efficacy of inhibition. However, DS-201did not significantly affect the kinetics of Ca2+ channel. The effect of DS-201 on L-type Ca2+ channel was rate-independent. Conclusion: The effect of DS-201 on vasorelaxation was not only via activating BKCa channel, but also blocking Ca2+ channel and inhibiting Ca2+ influx in the VSMCs of rats. The results favor the use of DS-201 and Danshen in the treatment of cardiovascular diseases clinically.
Collapse
Affiliation(s)
- Xiao-Dong Zhang
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Chun-Xia He
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Jun Cheng
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Jing Wen
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Peng-Yun Li
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Na Wang
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Guang Li
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Xiao-Rong Zeng
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Ji-Min Cao
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| | - Yan Yang
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, China
| |
Collapse
|
86
|
Leo MD, Jaggar JH. Trafficking of BK channel subunits controls arterial contractility. Oncotarget 2017; 8:106149-106150. [PMID: 29290926 PMCID: PMC5739711 DOI: 10.18632/oncotarget.22280] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 10/30/2017] [Indexed: 11/25/2022] Open
Affiliation(s)
- M Dennis Leo
- Jonathan H. Jaggar: Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Jonathan H Jaggar
- Jonathan H. Jaggar: Department of Physiology, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
87
|
Kyle BD, Mishra RC, Braun AP. The augmentation of BK channel activity by nitric oxide signaling in rat cerebral arteries involves co-localized regulatory elements. J Cereb Blood Flow Metab 2017; 37:3759-3773. [PMID: 28155571 PMCID: PMC5718322 DOI: 10.1177/0271678x17691291] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Large conductance, Ca2+-activated K+ (BK) channels control cerebrovascular tone; however, the regulatory processes influencing these channels remain poorly understood. Here, we investigate the cellular mechanisms underlying the enhancement of BK current in rat cerebral arteries by nitric oxide (NO) signaling. In isolated cerebral myocytes, BK current magnitude was reversibly increased by sodium nitroprusside (SNP, 100 μM) and sensitive to the BK channel inhibitor, penitrem-A (100 nM). Fostriecin (30 nM), a protein phosphatase type 2A (PP2A) inhibitor, significantly prolonged the SNP-induced augmentation of BK current and a similar effect was produced by sildenafil (30 nM), a phosphodiesterase 5 (PDE5) inhibitor. Using proximity ligation assay (PLA)-based co-immunostaining, BK channels were observed to co-localize with PP2A, PDE5, and cGMP-dependent protein kinase (cGKI) (spatial restriction < 40 nm); cGKI co-localization increased following SNP exposure. SNP (10 μM) reversibly inhibited myogenic tone in cannulated cerebral arteries, which was augmented by either fostriecin or sildenafil and inhibited by penitrem-A. Collectively, these data suggest that (1) cGKI, PDE5, and PP2A are compartmentalized with cerebrovascular BK channels and determine the extent of BK current augmentation by NO/cGMP signaling, and (2) the dynamic regulation of BK activity by co-localized signaling enzymes modulates NO-evoked dilation of cerebral resistance arteries.
Collapse
Affiliation(s)
- Barry D Kyle
- Department of Physiology and Pharmacology and Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Ramesh C Mishra
- Department of Physiology and Pharmacology and Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Andrew P Braun
- Department of Physiology and Pharmacology and Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
88
|
Shen CP, Romero M, Brunelle A, Wolfe C, Dobyns A, Francis M, Taylor MS, Puglisi JL, Longo LD, Zhang L, Wilson CG, Wilson SM. Long-term high-altitude hypoxia influences pulmonary arterial L-type calcium channel-mediated Ca 2+ signals and contraction in fetal and adult sheep. Am J Physiol Regul Integr Comp Physiol 2017; 314:R433-R446. [PMID: 29167165 DOI: 10.1152/ajpregu.00154.2017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Long-term hypoxia (LTH) has a profound effect on pulmonary arterial vasoconstriction in the fetus and adult. Dysregulation in Ca2+ signaling is important during the development of LTH-induced pulmonary hypertension. In the present study, we tested the hypothesis that L-type Ca2+ channels (CaL), which are voltage dependent and found in smooth, skeletal, and cardiac muscle, are important in the adaptation of pulmonary arterial contractions in postnatal maturation and in response to LTH. Pulmonary arteries were isolated from fetal or adult sheep maintained at low or high altitude (3,801 m) for >100 days. The effects were measured using an L-type Ca2+ channel opener FPL 64176 (FPL) in the presence or absence of an inhibitor, Nifedipine (NIF) on arterial contractions, intracellular Ca2+ oscillations, and ryanodine receptor-driven Ca2+ sparks. FPL induced pulmonary arterial contractions in all groups were sensitive to NIF. However, when compared with 125 mM K+, FPL contractions were greater in fetuses than in adults. FPL reduced Ca2+ oscillations in myocytes of adult but not fetal arteries, independently of altitude. The FPL effects on Ca2+ oscillations were reversed by NIF in myocytes of hypoxic but not normoxic adults. FPL failed to enhance Ca2+ spark frequency and had little impact on spatiotemporal firing characteristics. These data suggest that CaL-dependent contractions are largely uncoupled from intracellular Ca2+ oscillations and the development of Ca2+ sparks. This raises questions regarding the coupling of pulmonary arterial contractility to membrane depolarization, attendant CaL facilitation, and the related associations with the activation of Ca2+ oscillations and Ca2+ sparks.
Collapse
Affiliation(s)
- Christine P Shen
- Lawrence D. Longo, MD Center for Perinatal Biology, Loma Linda University School of Medicine , Loma Linda, California
| | - Monica Romero
- Advanced Imaging and Microscopy Core, Loma Linda University School of Medicine , Loma Linda, California
| | - Alexander Brunelle
- Lawrence D. Longo, MD Center for Perinatal Biology, Loma Linda University School of Medicine , Loma Linda, California
| | - Craig Wolfe
- Lawrence D. Longo, MD Center for Perinatal Biology, Loma Linda University School of Medicine , Loma Linda, California
| | - Abigail Dobyns
- Lawrence D. Longo, MD Center for Perinatal Biology, Loma Linda University School of Medicine , Loma Linda, California
| | - Michael Francis
- Department of Physiology, University of South Alabama College of Medicine , Mobile, Alabama
| | - Mark S Taylor
- Department of Physiology, University of South Alabama College of Medicine , Mobile, Alabama
| | - Jose L Puglisi
- Department of Biostatistics, California Northstate University School of Medicine , Elk Grove, California
| | - Lawrence D Longo
- Lawrence D. Longo, MD Center for Perinatal Biology, Loma Linda University School of Medicine , Loma Linda, California
| | - Lubo Zhang
- Lawrence D. Longo, MD Center for Perinatal Biology, Loma Linda University School of Medicine , Loma Linda, California
| | - Christopher G Wilson
- Lawrence D. Longo, MD Center for Perinatal Biology, Loma Linda University School of Medicine , Loma Linda, California
| | - Sean M Wilson
- Lawrence D. Longo, MD Center for Perinatal Biology, Loma Linda University School of Medicine , Loma Linda, California
| |
Collapse
|
89
|
Chloroform Extract of Artemisia annua L. Relaxes Mouse Airway Smooth Muscle. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 2017:9870414. [PMID: 29259649 PMCID: PMC5702405 DOI: 10.1155/2017/9870414] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 09/06/2017] [Accepted: 10/10/2017] [Indexed: 11/18/2022]
Abstract
Artemisia annua L. belongs to the Asteraceae family, which is indigenous to China. It has valuable pharmacological properties, such as antimalarial, anti-inflammatory, and anticancer properties. However, whether it possesses antiasthma properties is unknown. In the current study, chloroform extract of Artemisia annua L. (CEAA) was prepared, and we found that CEAA completely eliminated acetylcholine (ACh) or high K+-elicited (80 mM) contractions of mouse tracheal rings (TRs). Patch-clamp technique and ion channel blockers were employed to explore the underlying mechanisms of the relaxant effect of CEAA. In whole-cell current recording, CEAA almost fully abolished voltage-dependent Ca2+ channel (VDCC) currents and markedly enhanced large conductance Ca2+-activated K+ (BK) channel currents on airway smooth muscle cells (ASMCs). In single channel current recording, CEAA increased the opening probability but had no effect on the single channel conductance of BK channels. However, under paxilline-preincubated (a selective BK channel blocker) conditions, CEAA only slightly increased BK channel currents. These results indicate that CEAA may contain active components with potent antiasthma activity. The abolished VDCCs by CEAA may mainly contribute to the underlying mechanism through which it acts as an effective antiasthmatic compound, but the enhanced BK currents might play a less important role in the antiasthmatic effects.
Collapse
|
90
|
Zhao QY, Peng YB, Luo XJ, Luo X, Xu H, Wei MY, Jiang QJ, Li WE, Ma LQ, Xu JC, Liu XC, Zang DA, She YS, Zhu H, Shen J, Zhao P, Xue L, Yu MF, Chen W, Zhang P, Fu X, Chen J, Nie X, Shen C, Chen S, Chen S, Chen J, Hu S, Zou C, Qin G, Fang Y, Ding J, Ji G, Zheng YM, Song T, Wang YX, Liu QH. Distinct Effects of Ca 2+ Sparks on Cerebral Artery and Airway Smooth Muscle Cell Tone in Mice and Humans. Int J Biol Sci 2017; 13:1242-1253. [PMID: 29104491 PMCID: PMC5666523 DOI: 10.7150/ijbs.21475] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 08/10/2017] [Indexed: 11/21/2022] Open
Abstract
The effects of Ca2+ sparks on cerebral artery smooth muscle cells (CASMCs) and airway smooth muscle cells (ASMCs) tone, as well as the underlying mechanisms, are not clear. In this investigation, we elucidated the underlying mechanisms of the distinct effects of Ca2+ sparks on cerebral artery smooth muscle cells (CASMCs) and airway smooth muscle cells (ASMCs) tone. In CASMCs, owing to the functional loss of Ca2+-activated Cl- (Clca) channels, Ca2+ sparks activated large-conductance Ca2+-activated K+ channels (BKs), resulting in a decreases in tone against a spontaneous depolarization-caused high tone in the resting state. In ASMCs, Ca2+ sparks induced relaxation through BKs and contraction via Clca channels. However, the integrated result was contraction because Ca2+ sparks activated BKs prior to Clca channels and Clca channels-induced depolarization was larger than BKs-caused hyperpolarization. However, the effects of Ca2+ sparks on both cell types were determined by L-type voltage-dependent Ca2+ channels (LVDCCs). In addition, compared with ASMCs, CASMCs had great and higher amplitude Ca2+ sparks, a higher density of BKs, and higher Ca2+ and voltage sensitivity of BKs. These differences enhanced the ability of Ca2+ sparks to decrease CASMC and to increase ASMC tone. The higher Ca2+ and voltage sensitivity of BKs in CASMCs than ASMCs were determined by the β1 subunits. Moreover, Ca2+ sparks showed the similar effects on human CASMC and ASMC tone. In conclusions, Ca2+ sparks decrease CASMC tone and increase ASMC tone, mediated by BKs and Clca channels, respectively, and finally determined by LVDCCs.
Collapse
Affiliation(s)
- Qing-Yang Zhao
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Yong-Bo Peng
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Xiao-Jing Luo
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Xi Luo
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Hao Xu
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Ming-Yu Wei
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Qiu-Ju Jiang
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Wen-Er Li
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Li-Qun Ma
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Jin-Chao Xu
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Xiao-Cao Liu
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Dun-An Zang
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Yu-San She
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - He Zhu
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Jinhua Shen
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Ping Zhao
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Lu Xue
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Meng-Fei Yu
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Weiwei Chen
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| | - Ping Zhang
- Department of Cerebral Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430032, Hubei, China
| | - Xiangning Fu
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430032, Hubei, China
| | - Jingyu Chen
- Wuxi &Jiangsu Key Laboratory of Organ Transplantation, Department of Cardiothoracic Surgery, Lung Transplant Group, Wuxi People's Hospital, Nanjing Medical University, Jiangsu, China
| | - Xiaowei Nie
- Wuxi &Jiangsu Key Laboratory of Organ Transplantation, Department of Cardiothoracic Surgery, Lung Transplant Group, Wuxi People's Hospital, Nanjing Medical University, Jiangsu, China
| | - Chenyou Shen
- Wuxi &Jiangsu Key Laboratory of Organ Transplantation, Department of Cardiothoracic Surgery, Lung Transplant Group, Wuxi People's Hospital, Nanjing Medical University, Jiangsu, China
| | - Shu Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430032, Hubei, China
| | - Shanshan Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430032, Hubei, China
| | - Jingcao Chen
- Department of Cerebral Surgery, Zhongnan Hospital, Wuhan University Medical College, Wuhan, 430071, Hubei, China
| | - Sheng Hu
- Department of Medical Oncology, Hubei Cancer Hospital, Wuhan, 430079, Hubei, China
| | - Chunbin Zou
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Gangjian Qin
- Department of Biomedical Engineering, School of Medicine & School of Engineering, University of Alabama Birmingham, AL, 35294, USA
| | - Ying Fang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jiuping Ding
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Guangju Ji
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yun-Min Zheng
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, 12208, USA
| | - Tengyao Song
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, 12208, USA
| | - Yong-Xiao Wang
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, 12208, USA
| | - Qing-Hua Liu
- Institute for Medical Biology and Hubei Provincial Key Laboratory for Protection and Application of Special Plants in Wuling Area of China, College of Life Sciences, South-Central University for Nationalities, Wuhan 430074, China
| |
Collapse
|
91
|
Pritchard HAT, Gonzales AL, Pires PW, Drumm BT, Ko EA, Sanders KM, Hennig GW, Earley S. Microtubule structures underlying the sarcoplasmic reticulum support peripheral coupling sites to regulate smooth muscle contractility. Sci Signal 2017; 10:eaan2694. [PMID: 28928237 PMCID: PMC6328376 DOI: 10.1126/scisignal.aan2694] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Junctional membrane complexes facilitate excitation-contraction coupling in skeletal and cardiac muscle cells by forming subcellular invaginations that maintain close (≤20 nm) proximity of ryanodine receptors (RyRs) on the sarcoplasmic reticulum (SR) with voltage-dependent Ca2+ channels in the plasma membrane. In fully differentiated smooth muscle cells, junctional membrane complexes occur as distributed sites of peripheral coupling. We investigated the role of the cytoskeleton in maintaining peripheral coupling and associated Ca2+ signaling networks within native smooth muscle cells of mouse and rat cerebral arteries. Using live-cell confocal and superresolution microscopy, we found that the tight interactions between the SR and the plasma membrane in these cells relied on arching microtubule structures present at the periphery of smooth muscle cells and were independent of the actin cytoskeleton. Loss of peripheral coupling associated with microtubule depolymerization altered the spatiotemporal properties of localized Ca2+ sparks generated by the release of Ca2+ through type 2 RyRs (RyR2s) on the SR and decreased the number of sites of colocalization between RyR2s and large-conductance Ca2+-activated K+ (BK) channels. The reduced BK channel activity associated with the loss of SR-plasma membrane interactions was accompanied by increased pressure-induced constriction of cerebral resistance arteries. We conclude that microtubule structures maintain peripheral coupling in contractile smooth muscle cells, which is crucial for the regulation of contractility and cerebral vascular tone.
Collapse
Affiliation(s)
- Harry A T Pritchard
- Department of Pharmacology, Center for Cardiovascular Research, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
| | - Albert L Gonzales
- Department of Pharmacology, University of Vermont, Burlington, VT 05405, USA
| | - Paulo W Pires
- Department of Pharmacology, Center for Cardiovascular Research, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
| | - Bernard T Drumm
- Department of Physiology and Cell Biology, Center for Cardiovascular Research, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
| | - Eun A Ko
- Department of Physiology and Cell Biology, Center for Cardiovascular Research, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
| | - Kenton M Sanders
- Department of Physiology and Cell Biology, Center for Cardiovascular Research, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA
| | - Grant W Hennig
- Department of Pharmacology, University of Vermont, Burlington, VT 05405, USA
| | - Scott Earley
- Department of Pharmacology, Center for Cardiovascular Research, University of Nevada, Reno School of Medicine, Reno, NV 89557, USA.
| |
Collapse
|
92
|
Humphries ESA, Kamishima T, Quayle JM, Dart C. Calcium/calmodulin-dependent kinase 2 mediates Epac-induced spontaneous transient outward currents in rat vascular smooth muscle. J Physiol 2017; 595:6147-6164. [PMID: 28731505 PMCID: PMC5599484 DOI: 10.1113/jp274754] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 07/18/2017] [Indexed: 01/02/2023] Open
Abstract
KEY POINTS The Ca2+ and redox-sensing enzyme Ca2+ /calmodulin-dependent kinase 2 (CaMKII) is a crucial and well-established signalling molecule in the heart and brain. In vascular smooth muscle, which controls blood flow by contracting and relaxing in response to complex Ca2+ signals and oxidative stress, surprisingly little is known about the role of CaMKII. The vasodilator-induced second messenger cAMP can relax vascular smooth muscle via its effector, exchange protein directly activated by cAMP (Epac), by activating spontaneous transient outward currents (STOCs) that hyperpolarize the cell membrane and reduce voltage-dependent Ca2+ influx. How Epac activates STOCs is unknown. In the present study, we map the pathway by which Epac increases STOC activity in contractile vascular smooth muscle and show that a critical step is the activation of CaMKII. To our knowledge, this is the first report of CaMKII activation triggering cellular activity known to induce vasorelaxation. ABSTRACT Activation of the major cAMP effector, exchange protein directly activated by cAMP (Epac), induces vascular smooth muscle relaxation by increasing the activity of ryanodine (RyR)-sensitive release channels on the peripheral sarcoplasmic reticulum. Resultant Ca2+ sparks activate plasma membrane Ca2+ -activated K+ (BKCa ) channels, evoking spontaneous transient outward currents (STOCs) that hyperpolarize the cell and reduce voltage-dependent Ca2+ entry. In the present study, we investigate the mechanism by which Epac increases STOC activity. We show that the selective Epac activator 8-(4-chloro-phenylthio)-2'-O-methyladenosine-3', 5-cyclic monophosphate-AM (8-pCPT-AM) induces autophosphorylation (activation) of calcium/calmodulin-dependent kinase 2 (CaMKII) and also that inhibition of CaMKII abolishes 8-pCPT-AM-induced increases in STOC activity. Epac-induced CaMKII activation is probably initiated by inositol 1,4,5-trisphosphate (IP3 )-mobilized Ca2+ : 8-pCPT-AM fails to induce CaMKII activation following intracellular Ca2+ store depletion and inhibition of IP3 receptors blocks both 8-pCPT-AM-mediated CaMKII phosphorylation and STOC activity. 8-pCPT-AM does not directly activate BKCa channels, but STOCs cannot be generated by 8-pCPT-AM in the presence of ryanodine. Furthermore, exposure to 8-pCPT-AM significantly slows the initial rate of [Ca2+ ]i rise induced by the RyR activator caffeine without significantly affecting the caffeine-induced Ca2+ transient amplitude, a measure of Ca2+ store content. We conclude that Epac-mediated STOC activity (i) occurs via activation of CaMKII and (ii) is driven by changes in the underlying behaviour of RyR channels. To our knowledge, this is the first report of CaMKII initiating cellular activity linked to vasorelaxation and suggests novel roles for this Ca2+ and redox-sensing enzyme in the regulation of vascular tone and blood flow.
Collapse
MESH Headings
- Action Potentials
- Animals
- Calcium/metabolism
- Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics
- Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism
- Cells, Cultured
- Guanine Nucleotide Exchange Factors/metabolism
- Large-Conductance Calcium-Activated Potassium Channels/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/physiology
- Myocytes, Smooth Muscle/metabolism
- Rats
- Rats, Wistar
- Vasodilation
Collapse
Affiliation(s)
| | | | - John M. Quayle
- Translational MedicineUniversity of LiverpoolLiverpoolUK
| | | |
Collapse
|
93
|
Dopico AM, Bukiya AN. Regulation of Ca 2+-Sensitive K + Channels by Cholesterol and Bile Acids via Distinct Channel Subunits and Sites. CURRENT TOPICS IN MEMBRANES 2017; 80:53-93. [PMID: 28863822 DOI: 10.1016/bs.ctm.2017.07.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Cholesterol (CLR) conversion into bile acids (BAs) in the liver constitutes the major pathway for CLR elimination from the body. Moreover, these steroids regulate each other's metabolism. While the roles of CLR and BAs in regulating metabolism and tissue function are well known, research of the last two decades revealed the existence of specific protein receptors for CLR or BAs in tissues with minor contribution to lipid metabolism, raising the possibility that these lipids serve as signaling molecules throughout the body. Among other lipids, CLR and BAs regulate ionic current mediated by the activity of voltage- and Ca2+-gated, K+ channels of large conductance (BK channels) and, thus, modulate cell physiology and participate in tissue pathophysiology. Initial work attributed modification of BK channel function by CLR or BAs to the capability of these steroids to directly interact with bilayer lipids and thus alter the physicochemical properties of the bilayer with eventual modification of BK channel function. Based on our own work and that of others, we now review evidence that supports direct interactions between CLR or BA and specific BK protein subunits, and the consequence of such interactions on channel activity and organ function, with a particular emphasis on arterial smooth muscle. For each steroid type, we will also briefly discuss several mechanisms that may underlie modification of channel steady-state activity. Finally, we will present novel computational data that provide a chemical basis for differential recognition of CLR vs lithocholic acid by distinct BK channel subunits and recognition sites.
Collapse
Affiliation(s)
- Alex M Dopico
- College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, United States.
| | - Anna N Bukiya
- College of Medicine, The University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
94
|
Getz MC, Nirody JA, Rangamani P. Stability analysis in spatial modeling of cell signaling. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2017; 10. [PMID: 28787545 DOI: 10.1002/wsbm.1395] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 06/06/2017] [Accepted: 06/11/2017] [Indexed: 11/08/2022]
Abstract
Advances in high-resolution microscopy and other techniques have emphasized the spatio-temporal nature of information transfer through signal transduction pathways. The compartmentalization of signaling molecules and the existence of microdomains are now widely acknowledged as key features in biochemical signaling. To complement experimental observations of spatio-temporal dynamics, mathematical modeling has emerged as a powerful tool. Using modeling, one can not only recapitulate experimentally observed dynamics of signaling molecules, but also gain an understanding of the underlying mechanisms in order to generate experimentally testable predictions. Reaction-diffusion systems are commonly used to this end; however, the analysis of coupled nonlinear systems of partial differential equations, generated by considering large reaction networks is often challenging. Here, we aim to provide an introductory tutorial for the application of reaction-diffusion models to the spatio-temporal dynamics of signaling pathways. In particular, we outline the steps for stability analysis of such models, with a focus on biochemical signal transduction. WIREs Syst Biol Med 2018, 10:e1395. doi: 10.1002/wsbm.1395 This article is categorized under: Biological Mechanisms > Cell Signaling Analytical and Computational Methods > Dynamical Methods Models of Systems Properties and Processes > Mechanistic Models.
Collapse
Affiliation(s)
- Michael C Getz
- Chemical Engineering Graduate Program, University of California San Diego, La Jolla, CA, USA
| | - Jasmine A Nirody
- Biophysics Graduate Program, University of California, Berkeley, CA, USA
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
95
|
Imaging sympathetic neurogenic Ca 2+ signaling in blood vessels. Auton Neurosci 2017; 207:59-66. [PMID: 28781164 DOI: 10.1016/j.autneu.2017.07.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 06/20/2017] [Accepted: 07/27/2017] [Indexed: 12/30/2022]
Abstract
We review the information that has been provided by optical imaging experiments directed at understanding the role and effects of sympathetic nerve activity (SNA) in the functioning of blood vessels. Earlier studies utilized electric field stimulation of nerve terminals (EFS) in isolated arteries and vascular tissues (ex vivo) to elicit SNA, but more recently, imaging studies have been conducted in vivo, enabling the study of SNA in truly physiological conditions. Ex vivo: In vascular smooth muscle cells (VSMC) of isolated arteries, the three sympathetic neurotransmitters, norepinephrine (NE), ATP and neuropeptide Y (NPY), elicit or modulate distinct patterns of Ca2+ signaling, as revealed by confocal imaging of exogenous fluorescent Ca2+ indicators. Purinergic junctional Ca2+ transients (jCaTs) arise from Ca2+ influx during excitatory junction potentials (eJPs), and are associated with the initial neurogenic contraction. Adrenergic Ca2+ waves and oscillations cause contraction while SNA-induced endothelial Ca2+ 'pulsars' cause relaxation. In vivo: optical biosensor mice, which express genetically encoded Ca2+ indicators (GECI's) specifically in smooth muscle, combined with non-invasive imaging techniques has enabled imaging SNA-induced Ca2+ signaling and arterial diameter in vivo. SNA induces Ca2+ oscillations in intact arteries. [Ca2+] of arterial smooth muscle cells increased in hypertension, in association with increased SNA. High resolution imaging has revealed local sympathetic, neurogenic Ca2+ signaling within smooth muscle and endothelial cells of the vasculature. The ongoing development of in vivo imaging together with an expanding availability of different biosensor animals promises to enable the further assessment of SNA and its effects in the vasculature of living animals.
Collapse
|
96
|
Zhai X, Leo MD, Jaggar JH. Endothelin-1 Stimulates Vasoconstriction Through Rab11A Serine 177 Phosphorylation. Circ Res 2017; 121:650-661. [PMID: 28696251 DOI: 10.1161/circresaha.117.311102] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 07/06/2017] [Accepted: 07/07/2017] [Indexed: 01/18/2023]
Abstract
RATIONALE Large-conductance calcium-activated potassium channels (BK) are composed of pore-forming BKα and auxiliary β1 subunits in arterial smooth muscle cells (myocytes). Vasoconstrictors, including endothelin-1 (ET-1), inhibit myocyte BK channels, leading to contraction, but mechanisms involved are unclear. Recent evidence indicates that BKα is primarily plasma membrane localized, whereas the cellular location of β1 can be rapidly altered by Rab11A-positive recycling endosomes. Whether vasoconstrictors regulate the multisubunit composition of surface BK channels to stimulate contraction is unclear. OBJECTIVE Test the hypothesis that ET-1 inhibits BK channels by altering BKα and β1 surface trafficking in myocytes, identify mechanisms involved, and determine functional significance in myocytes of small cerebral arteries. METHODS AND RESULTS ET-1, through activation of PKC (protein kinase C), reduced surface β1 abundance and the proximity of β1 to surface BKα in myocytes. In contrast, ET-1 did not alter surface BKα, total β1, or total BKα proteins. ET-1 stimulated Rab11A phosphorylation, which reduced Rab11A activity. Rab11A serine 177 was identified as a high-probability PKC phosphorylation site. Expression of a phosphorylation-incapable Rab11A construct (Rab11A S177A) blocked the ET-1-induced Rab11A phosphorylation, reduction in Rab11A activity, and decrease in surface β1 protein. ET-1 inhibited single BK channels and transient BK currents in myocytes and stimulated vasoconstriction via a PKC-dependent mechanism that required Rab11A S177. In contrast, NO-induced Rab11A activation, surface trafficking of β1 subunits, BK channel and transient BK current activation, and vasodilation did not involve Rab11A S177. CONCLUSIONS ET-1 stimulates PKC-mediated phosphorylation of Rab11A at serine 177, which inhibits Rab11A and Rab11A-dependent surface trafficking of β1 subunits. The decrease in surface β1 subunits leads to a reduction in BK channel calcium-sensitivity, inhibition of transient BK currents, and vasoconstriction. We describe a unique mechanism by which a vasoconstrictor inhibits BK channels and identify Rab11A serine 177 as a modulator of arterial contractility.
Collapse
Affiliation(s)
- Xue Zhai
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis
| | - M Dennis Leo
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis
| | - Jonathan H Jaggar
- From the Department of Physiology, University of Tennessee Health Science Center, Memphis.
| |
Collapse
|
97
|
The Slo(w) path to identifying the mitochondrial channels responsible for ischemic protection. Biochem J 2017; 474:2067-2094. [PMID: 28600454 DOI: 10.1042/bcj20160623] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 02/10/2017] [Accepted: 02/13/2017] [Indexed: 12/19/2022]
Abstract
Mitochondria play an important role in tissue ischemia and reperfusion (IR) injury, with energetic failure and the opening of the mitochondrial permeability transition pore being the major causes of IR-induced cell death. Thus, mitochondria are an appropriate focus for strategies to protect against IR injury. Two widely studied paradigms of IR protection, particularly in the field of cardiac IR, are ischemic preconditioning (IPC) and volatile anesthetic preconditioning (APC). While the molecular mechanisms recruited by these protective paradigms are not fully elucidated, a commonality is the involvement of mitochondrial K+ channel opening. In the case of IPC, research has focused on a mitochondrial ATP-sensitive K+ channel (mitoKATP), but, despite recent progress, the molecular identity of this channel remains a subject of contention. In the case of APC, early research suggested the existence of a mitochondrial large-conductance K+ (BK, big conductance of potassium) channel encoded by the Kcnma1 gene, although more recent work has shown that the channel that underlies APC is in fact encoded by Kcnt2 In this review, we discuss both the pharmacologic and genetic evidence for the existence and identity of mitochondrial K+ channels, and the role of these channels both in IR protection and in regulating normal mitochondrial function.
Collapse
|
98
|
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
|
99
|
Leo MD, Zhai X, Muralidharan P, Kuruvilla KP, Bulley S, Boop FA, Jaggar JH. Membrane depolarization activates BK channels through ROCK-mediated β1 subunit surface trafficking to limit vasoconstriction. Sci Signal 2017; 10:10/478/eaah5417. [PMID: 28487419 DOI: 10.1126/scisignal.aah5417] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Membrane depolarization of smooth muscle cells (myocytes) in the small arteries that regulate regional organ blood flow leads to vasoconstriction. Membrane depolarization also activates large-conductance calcium (Ca2+)-activated potassium (BK) channels, which limits Ca2+ channel activity that promotes vasoconstriction, thus leading to vasodilation. We showed that in human and rat arterial myocytes, membrane depolarization rapidly increased the cell surface abundance of auxiliary BK β1 subunits but not that of the pore-forming BKα channels. Membrane depolarization stimulated voltage-dependent Ca2+ channels, leading to Ca2+ influx and the activation of Rho kinase (ROCK) 1 and 2. ROCK1/2-mediated activation of Rab11A promoted the delivery of β1 subunits to the plasma membrane by Rab11A-positive recycling endosomes. These additional β1 subunits associated with BKα channels already at the plasma membrane, leading to an increase in apparent Ca2+ sensitivity and activation of the channels in pressurized arterial myocytes and vasodilation. Thus, membrane depolarization activates BK channels through stimulation of ROCK- and Rab11A-dependent trafficking of β1 subunits to the surface of arterial myocytes.
Collapse
Affiliation(s)
- M Dennis Leo
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Xue Zhai
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Padmapriya Muralidharan
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Korah P Kuruvilla
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Simon Bulley
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Frederick A Boop
- Department of Neurosurgery, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jonathan H Jaggar
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| |
Collapse
|
100
|
Tykocki NR, Boerman EM, Jackson WF. Smooth Muscle Ion Channels and Regulation of Vascular Tone in Resistance Arteries and Arterioles. Compr Physiol 2017; 7:485-581. [PMID: 28333380 DOI: 10.1002/cphy.c160011] [Citation(s) in RCA: 233] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Vascular tone of resistance arteries and arterioles determines peripheral vascular resistance, contributing to the regulation of blood pressure and blood flow to, and within the body's tissues and organs. Ion channels in the plasma membrane and endoplasmic reticulum of vascular smooth muscle cells (SMCs) in these blood vessels importantly contribute to the regulation of intracellular Ca2+ concentration, the primary determinant of SMC contractile activity and vascular tone. Ion channels provide the main source of activator Ca2+ that determines vascular tone, and strongly contribute to setting and regulating membrane potential, which, in turn, regulates the open-state-probability of voltage gated Ca2+ channels (VGCCs), the primary source of Ca2+ in resistance artery and arteriolar SMCs. Ion channel function is also modulated by vasoconstrictors and vasodilators, contributing to all aspects of the regulation of vascular tone. This review will focus on the physiology of VGCCs, voltage-gated K+ (KV) channels, large-conductance Ca2+-activated K+ (BKCa) channels, strong-inward-rectifier K+ (KIR) channels, ATP-sensitive K+ (KATP) channels, ryanodine receptors (RyRs), inositol 1,4,5-trisphosphate receptors (IP3Rs), and a variety of transient receptor potential (TRP) channels that contribute to pressure-induced myogenic tone in resistance arteries and arterioles, the modulation of the function of these ion channels by vasoconstrictors and vasodilators, their role in the functional regulation of tissue blood flow and their dysfunction in diseases such as hypertension, obesity, and diabetes. © 2017 American Physiological Society. Compr Physiol 7:485-581, 2017.
Collapse
Affiliation(s)
- Nathan R Tykocki
- Department of Pharmacology, University of Vermont, Burlington, Vermont, USA
| | - Erika M Boerman
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, USA
| | - William F Jackson
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|