1
|
Pyanova A, Serebryakov VN, Gagov H, Mladenov M, Schubert R. BK Channels in Tail Artery Vascular Smooth Muscle Cells of Normotensive (WKY) and Hypertensive (SHR) Rats Possess Similar Calcium Sensitivity But Different Responses to the Vasodilator Iloprost. Int J Mol Sci 2024; 25:7140. [PMID: 39000253 PMCID: PMC11241265 DOI: 10.3390/ijms25137140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/25/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
It has been reported that, in the spontaneously hypertensive rat (SHR) model of hypertension, different components of the G-protein/adenylate cyclase (AC)/Calcium-activated potassium channel of high conductance (BK) channel signaling pathway are altered differently. In the upstream part of the pathway (G-protein/AC), a comparatively low efficacy has been established, whereas downstream BK currents seem to be increased. Thus, the overall performance of this signaling pathway in SHR is elusive. For a better understanding, we focused on one aspect, the direct targeting of the BK channel by the G-protein/AC pathway and tested the hypothesis that the comparatively low AC pathway efficacy in SHR results in a reduced agonist-induced stimulation of BK currents. This hypothesis was investigated using freshly isolated smooth muscle cells from WKY and SHR rat tail artery and the patch-clamp technique. It was observed that: (1) single BK channels have similar current-voltage relationships, voltage-dependence and calcium sensitivity; (2) BK currents in cells with a strong buffering of the BK channel activator calcium have similar current-voltage relationships; (3) the iloprost-induced concentration-dependent increase of the BK current is larger in WKY compared to SHR; (4) the effects of activators of the PKA pathway, the catalytic subunit of PKA and the potent and selective cAMP-analogue Sp-5,6-DCl-cBIMPS on BK currents are similar. Thus, our data suggest that the lower iloprost-induced stimulation of the BK current in freshly isolated rat tail artery smooth muscle cells from SHR compared with WKY is due to the lower efficacy of upstream elements of the G-Protein/AC/BK channel pathway.
Collapse
MESH Headings
- Animals
- Rats, Inbred SHR
- Large-Conductance Calcium-Activated Potassium Channels/metabolism
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/cytology
- Rats
- Calcium/metabolism
- Iloprost/pharmacology
- Rats, Inbred WKY
- Hypertension/metabolism
- Hypertension/drug therapy
- Vasodilator Agents/pharmacology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/drug effects
- Male
- Arteries/drug effects
- Arteries/metabolism
- Tail/blood supply
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Anastasia Pyanova
- Physiology, Institute of Theoretical Medicine, Faculty of Medicine, University of Augsburg, 86159 Augsburg, Germany;
| | | | - Hristo Gagov
- Department of Animal and Human Physiology, Faculty of Biology, Sofia University “St. Kliment Ohridski”, 1164 Sofia, Bulgaria;
| | - Mitko Mladenov
- Institute of Biology, Faculty of Natural Sciences and Mathematics, University of Ss. Cyril and Methodius, 1000 Skopje, North Macedonia;
- Department of Fundamental and Applied Physiology, Russian States Medical University, 117997 Moscow, Russia
| | - Rudolf Schubert
- Physiology, Institute of Theoretical Medicine, Faculty of Medicine, University of Augsburg, 86159 Augsburg, Germany;
| |
Collapse
|
2
|
Naser N, Lonj CK, Rikard-Bell M, Sandow SL, Murphy TV. Advanced glycated end-products inhibit dilation through constitutive endothelial RAGE and Nox1/4 in rat isolated skeletal muscle arteries. Microcirculation 2024; 31:e12837. [PMID: 37985248 DOI: 10.1111/micc.12837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/17/2023] [Accepted: 11/07/2023] [Indexed: 11/22/2023]
Abstract
OBJECTIVE This study investigated the actions of advanced glycated end-products (AGE), their receptors (RAGE), and NAD(P)H oxidase (Nox) subtypes 1, 2, and 4 on mechanisms of endothelium-dependent dilation of the rat cremaster muscle artery (CMA). METHODS Immunofluorescence studies were used to examine expression of RAGE in rat arteries. ROS accumulation was measured using luminescence and fluorescence assays. Functional studies were performed using pressure myography. RESULTS High levels of RAGE expression were shown in the endothelial cells of the CMA, compared with low endothelial expression in middle cerebral and mesenteric arteries and the aorta. Exogenous AGE (in vitro glycated bovine serum albumin) stimulated H2O2 accumulation in CMA, which was prevented by the RAGE antagonist FPS-ZM1, the NAD(P)H oxidase (Nox) inhibitor apocynin and inhibited by the Nox1/4 inhibitor setanaxib, but not the Nox2 inhibitor GSK2795039. In functional studies, AGE inhibited vasodilation of CMA stimulated by acetylcholine, sodium nitroprusside, and the BKCa activator NS1619, but not adenosine-induced dilation. FPS-ZM1, apocynin, and setanaxib prevented the inhibitory effects of AGE on responses to acetylcholine and NS-1619. CONCLUSION These observations suggest RAGE are constitutively expressed in the endothelium of the rat CMA and may be activated by AGE to stimulate Nox1/4 and ROS formation with resulting inhibition of NO and BKCa-mediated endothelium-dependent dilation.
Collapse
Affiliation(s)
- Nadim Naser
- Physiology, School of Biomedical Sciences, UNSW Sydney, Sydney, Australia
| | - Chenchel K Lonj
- Physiology, School of Biomedical Sciences, UNSW Sydney, Sydney, Australia
| | - Matthew Rikard-Bell
- Physiology, School of Biomedical Sciences, UNSW Sydney, Sydney, Australia
- Townsville University Hospital, Townsville, Queensland, Australia
| | - Shaun L Sandow
- Biomedical Science, University of the Sunshine Coast, Maroochydore, Australia
- Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Timothy V Murphy
- Physiology, School of Biomedical Sciences, UNSW Sydney, Sydney, Australia
| |
Collapse
|
3
|
Yang Y, Nourian Z, Li M, Sun Z, Zhang L, Davis MJ, Meininger GA, Wu J, Braun AP, Hill MA. Modification of Fibronectin by Non-Enzymatic Glycation Impairs K + Channel Function in Rat Cerebral Artery Smooth Muscle Cells. Front Physiol 2022; 13:871968. [PMID: 35832482 PMCID: PMC9272009 DOI: 10.3389/fphys.2022.871968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Fibronectin (FN) enhances K+ channel activity by integrin-mediated mechanisms. As vascular smooth muscle (VSM) K+ channels mediate vasodilation, we hypothesized that modification of fibronectin, via advanced non-enzymatic glycation, would alter signaling of this extracellular matrix protein through these channels. Bovine FN (1 mg/ml) was glycated (gFN) for 5 days using methylglyoxal (50 mM), and albumin was similarly glycated as a non-matrix protein control. VSM cells were isolated from rat cerebral arteries for measurement of macroscopic K+ channel activity using whole cell patch clamp methodology. Pharmacological inhibitors, iberiotoxin (0.1 μM) and 4-aminopyridine (0.1 mM), were used to identify contributions of large-conductance, Ca2+-activated, K+ channels and voltage-gated K+ channels, respectively. Compared with baseline, native FN enhanced whole cell K+ current in a concentration-dependent manner, whereas gFN inhibited basal current. Furthermore, native albumin did not enhance basal K+ current, but the glycated form (gAlb) caused inhibition. gFN was shown to impair both the Kv and BKCa components of total macroscopic K+ current. Anti-integrin α5 and β1 antibodies attenuated the effects of both FN and gFN on macroscopic K+ current at +70 mV. Consistent with an action on BKCa activity, FN increased, whereas gFN decreased the frequency of spontaneous transient outward current (STOCs). In contrast, gAlb inhibited whole cell K+ current predominantly through Kv, showing little effect on STOCs. A function-blocking, anti-RAGE antibody partially reversed the inhibitory effects of gFN, suggesting involvement of this receptor. Further, gFN caused production of reactive oxygen species (ROS) by isolated VSMCs as revealed by the fluorescent indicator, DHE. Evoked ROS production was attenuated by the RAGE blocking antibody. Collectively, these studies identify ion channel-related mechanisms (integrin and ROS-mediated) by which protein glycation may modify VSMC function.
Collapse
Affiliation(s)
- Yan Yang
- Dalton Cardiovascular Research Center, Columbia, MO, United States
| | - Zahra Nourian
- Dalton Cardiovascular Research Center, Columbia, MO, United States
| | - Min Li
- Dalton Cardiovascular Research Center, Columbia, MO, United States
| | - Zhe Sun
- Dalton Cardiovascular Research Center, Columbia, MO, United States
| | | | - Michael J. Davis
- Dalton Cardiovascular Research Center, Columbia, MO, United States
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, United States
| | - Gerald A. Meininger
- Dalton Cardiovascular Research Center, Columbia, MO, United States
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, United States
| | - Jianbo Wu
- Southwest Medical University, Luzhou, China
| | - Andrew P. Braun
- Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Michael A. Hill
- Dalton Cardiovascular Research Center, Columbia, MO, United States
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, United States
| |
Collapse
|
4
|
Jackson WF. Calcium-Dependent Ion Channels and the Regulation of Arteriolar Myogenic Tone. Front Physiol 2021; 12:770450. [PMID: 34819877 PMCID: PMC8607693 DOI: 10.3389/fphys.2021.770450] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 10/11/2021] [Indexed: 11/25/2022] Open
Abstract
Arterioles in the peripheral microcirculation regulate blood flow to and within tissues and organs, control capillary blood pressure and microvascular fluid exchange, govern peripheral vascular resistance, and contribute to the regulation of blood pressure. These important microvessels display pressure-dependent myogenic tone, the steady state level of contractile activity of vascular smooth muscle cells (VSMCs) that sets resting arteriolar internal diameter such that arterioles can both dilate and constrict to meet the blood flow and pressure needs of the tissues and organs that they perfuse. This perspective will focus on the Ca2+-dependent ion channels in the plasma and endoplasmic reticulum membranes of arteriolar VSMCs and endothelial cells (ECs) that regulate arteriolar tone. In VSMCs, Ca2+-dependent negative feedback regulation of myogenic tone is mediated by Ca2+-activated K+ (BKCa) channels and also Ca2+-dependent inactivation of voltage-gated Ca2+ channels (VGCC). Transient receptor potential subfamily M, member 4 channels (TRPM4); Ca2+-activated Cl− channels (CaCCs; TMEM16A/ANO1), Ca2+-dependent inhibition of voltage-gated K+ (KV) and ATP-sensitive K+ (KATP) channels; and Ca2+-induced-Ca2+ release through inositol 1,4,5-trisphosphate receptors (IP3Rs) participate in Ca2+-dependent positive-feedback regulation of myogenic tone. Calcium release from VSMC ryanodine receptors (RyRs) provide negative-feedback through Ca2+-spark-mediated control of BKCa channel activity, or positive-feedback regulation in cooperation with IP3Rs or CaCCs. In some arterioles, VSMC RyRs are silent. In ECs, transient receptor potential vanilloid subfamily, member 4 (TRPV4) channels produce Ca2+ sparklets that activate IP3Rs and intermediate and small conductance Ca2+ activated K+ (IKCa and sKCa) channels causing membrane hyperpolarization that is conducted to overlying VSMCs producing endothelium-dependent hyperpolarization and vasodilation. Endothelial IP3Rs produce Ca2+ pulsars, Ca2+ wavelets, Ca2+ waves and increased global Ca2+ levels activating EC sKCa and IKCa channels and causing Ca2+-dependent production of endothelial vasodilator autacoids such as NO, prostaglandin I2 and epoxides of arachidonic acid that mediate negative-feedback regulation of myogenic tone. Thus, Ca2+-dependent ion channels importantly contribute to many aspects of the regulation of myogenic tone in arterioles in the microcirculation.
Collapse
Affiliation(s)
- William F Jackson
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
5
|
Liao LM, Zhou L, Wang CR, Hu JY, Lu YJ, Huang S. Opposing responses of the rat pulmonary artery and vein to phenylephrine and other agents in vitro. BMC Pulm Med 2021; 21:189. [PMID: 34090386 PMCID: PMC8180060 DOI: 10.1186/s12890-021-01558-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 05/27/2021] [Indexed: 11/10/2022] Open
Abstract
Background Different from current cognition, our study demonstrated that adrenergic receptors agonist phenylephrine significantly relaxed isolated pulmonary artery but constricted pulmonary veins. Through comparing differences in the effects of commonly used vasoactive drugs on pulmonary artery and veins, the study aimed to improve efficiency and accuracy of isolated pulmonary vascular experiments, and to provide experimental basis for clinical drug use. Methods The contractile responses of pulmonary arteries and veins from twelve-week-old Male Sprague-Dawley rats to phenylephrine, arginine vasopressin (AVP), U46619, endothelin-1, and potassium chloride (KCl) were recorded, as well as the relaxation in response to phenylephrine, AVP, acetylcholine. To further explore the mechanism, some vessels was also pre-incubated with adrenergic receptors antagonists propranolol, prazosin and nitric oxide synthesis inhibitor N[gamma]-nitro-L-arginine methyl ester (L-NAME) before addition of the experimental drugs. Results Phenylephrine constricted pulmonary veins directly, but constricted pulmonary artery only after incubation with propranolol or/and L-NAME. The pulmonary artery exhibited significant relaxation to AVP with or without L-NAME incubation. AVP more clearly constricted the veins after incubation with L-NAME. Changes in vascular tension also varied from pulmonary artery to veins for KCl stimulation. Different from phenomena presented in veins, acetylcholine did not relax pulmonary artery preconstricted by KCl, U46619, and endothelin-1. Conclusions According to the results, phenylephrine, KCl, AVP, and acetylcholine could be used to distinguish pulmonary arteries and pulmonary veins in vitro. This also suggested that the pulmonary arteries and pulmonary veins have great differences in physiology and drug reactivity.
Collapse
Affiliation(s)
- Li-Mei Liao
- Department of Anaesthesia, Obstetrics and Gynecology Hospital, Fudan University, 128 Shenyang road, Shanghai, 200090, China.,Department of Anesthesiology and Perioperative Medicine, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, 9# Jinsui Road, Guangzhou, 510623, China
| | - Li Zhou
- Department of Physiology and Pathophysiology, School of Basic Medicine Science, Fudan University, 130 Dongan Road, Shanghai, 200032, China.
| | - Chen-Ran Wang
- Department of Anaesthesia, Obstetrics and Gynecology Hospital, Fudan University, 128 Shenyang road, Shanghai, 200090, China
| | - Jian-Ying Hu
- Department of Anaesthesia, Obstetrics and Gynecology Hospital, Fudan University, 128 Shenyang road, Shanghai, 200090, China
| | - Yao-Jun Lu
- Department of Anaesthesia, Obstetrics and Gynecology Hospital, Fudan University, 128 Shenyang road, Shanghai, 200090, China
| | - Shaoqiang Huang
- Department of Anaesthesia, Obstetrics and Gynecology Hospital, Fudan University, 128 Shenyang road, Shanghai, 200090, China.
| |
Collapse
|
6
|
Mondéjar-Parreño G, Cogolludo A, Perez-Vizcaino F. Potassium (K +) channels in the pulmonary vasculature: Implications in pulmonary hypertension Physiological, pathophysiological and pharmacological regulation. Pharmacol Ther 2021; 225:107835. [PMID: 33744261 DOI: 10.1016/j.pharmthera.2021.107835] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/26/2021] [Accepted: 03/02/2021] [Indexed: 02/06/2023]
Abstract
The large K+ channel functional diversity in the pulmonary vasculature results from the multitude of genes expressed encoding K+ channels, alternative RNA splicing, the post-transcriptional modifications, the presence of homomeric or heteromeric assemblies of the pore-forming α-subunits and the existence of accessory β-subunits modulating the functional properties of the channel. K+ channels can also be regulated at multiple levels by different factors controlling channel activity, trafficking, recycling and degradation. The activity of these channels is the primary determinant of membrane potential (Em) in pulmonary artery smooth muscle cells (PASMC), providing an essential regulatory mechanism to dilate or contract pulmonary arteries (PA). K+ channels are also expressed in pulmonary artery endothelial cells (PAEC) where they control resting Em, Ca2+ entry and the production of different vasoactive factors. The activity of K+ channels is also important in regulating the population and phenotype of PASMC in the pulmonary vasculature, since they are involved in cell apoptosis, survival and proliferation. Notably, K+ channels play a major role in the development of pulmonary hypertension (PH). Impaired K+ channel activity in PH results from: 1) loss of function mutations, 2) downregulation of its expression, which involves transcription factors and microRNAs, or 3) decreased channel current as a result of increased vasoactive factors (e.g., hypoxia, 5-HT, endothelin-1 or thromboxane), exposure to drugs with channel-blocking properties, or by a reduction in factors that positively regulate K+ channel activity (e.g., NO and prostacyclin). Restoring K+ channel expression, its intracellular trafficking and the channel activity is an attractive therapeutic strategy in PH.
Collapse
Affiliation(s)
- Gema Mondéjar-Parreño
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain; Ciber Enfermedades Respiratorias (CIBERES), Spain
| | - Angel Cogolludo
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain; Ciber Enfermedades Respiratorias (CIBERES), Spain
| | - Francisco Perez-Vizcaino
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Spain; Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain; Ciber Enfermedades Respiratorias (CIBERES), Spain.
| |
Collapse
|
7
|
Saddala MS, Lennikov A, Mukwaya A, Yang Y, Hill MA, Lagali N, Huang H. Discovery of novel L-type voltage-gated calcium channel blockers and application for the prevention of inflammation and angiogenesis. J Neuroinflammation 2020; 17:132. [PMID: 32334630 PMCID: PMC7183139 DOI: 10.1186/s12974-020-01801-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 04/02/2020] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The ways in which microglia activate and promote neovascularization (NV) are not fully understood. Recent in vivo evidence supports the theory that calcium is required for the transition of microglia from a surveillance state to an active one. The objectives of this study were to discover novel L-type voltage-gated channel (L-VGCC) blockers and investigate their application for the prevention of inflammation and angiogenesis. METHODS Pharmacophore-based computational modeling methods were used to screen for novel calcium channel blockers (CCBs) from the ZINC compound library. The effects of CCBs on calcium blockade, microglial pro-inflammatory activation, and cell toxicity were validated in BV-2 microglial cell and freshly isolated smooth muscle cell (SMC) cultures. Laser-induced choroidal neovascularization (NV) and the suture-induced inflammatory corneal NV models of angiogenesis were used for in vivo validation of the novel CCBs. CX3CR1gfp/+ mice were used to examine the infiltration of GFP-labeled microglial cells. RESULTS We identified three compounds from the ZINC database (Zinc20267861, Zinc18204217, and Zinc33254827) as new blockers of L-type voltage-gated calcium channels (L-VGCC) using a structure-based pharmacophore approach. The effects of the three CCBs on Ca2+ influx into cells were verified in BV-2 microglial cells using Fura-2 fluorescent dye and in freshly isolated SMCs using the voltage-patch clamp. All three CCBs reduced microglial cell migration, activation stimulated by lipopolysaccharide (LPS), and reduced the expression of the inflammatory markers NF-κB (phospho-IκBα) and cyclooxygenase-2 (COX-2) as well as reactive oxygen species. Of the three compounds, we further examined the in vivo activity of Zinc20267861. Topical treatment with Zinc20267861 in a rat model of suture-induced inflammatory cornea neovascularization demonstrated efficacy of the compound in reducing monocyte infiltration and overall corneal NV response. Subconjunctival administration of the compound in the choroidal NV mouse model effectively prevented CNV and microglial infiltration. CONCLUSIONS Our findings suggest that the novel CCBs identified here are effective anti-inflammatory agents that can be further evaluated for treating NV disorders and can be potentially applied in the treatment of ocular inflammatory and pathological angiogenetic disorders.
Collapse
Affiliation(s)
- Madhu Sudhana Saddala
- Department of Ophthalmology, School of Medicine, University of Missouri-Columbia, 1 Hospital Drive, MA102C, Columbia, MO, 65212, USA
| | - Anton Lennikov
- Department of Ophthalmology, School of Medicine, University of Missouri-Columbia, 1 Hospital Drive, MA102C, Columbia, MO, 65212, USA
| | - Anthony Mukwaya
- Department of Ophthalmology, Institute for Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | - Yan Yang
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, MO, USA
| | - Michael A Hill
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, MO, USA
| | - Neil Lagali
- Department of Ophthalmology, Institute for Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, Linköping, Sweden
| | - Hu Huang
- Department of Ophthalmology, School of Medicine, University of Missouri-Columbia, 1 Hospital Drive, MA102C, Columbia, MO, 65212, USA.
| |
Collapse
|
8
|
Ion channels and the regulation of myogenic tone in peripheral arterioles. CURRENT TOPICS IN MEMBRANES 2020; 85:19-58. [DOI: 10.1016/bs.ctm.2020.01.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
9
|
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
|
10
|
Dora KA. Endothelial-smooth muscle cell interactions in the regulation of vascular tone in skeletal muscle. Microcirculation 2018; 23:626-630. [PMID: 27653241 DOI: 10.1111/micc.12322] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 09/19/2016] [Indexed: 11/28/2022]
Abstract
The SMCs of skeletal muscle arterioles are intricately sensitive to changes in membrane potential. Upon increasing luminal pressure, the SMCs depolarize, thereby opening VDCCs, which leads to contraction. Mechanisms that oppose this myogenic tone can involve voltage-dependent and independent dilator pathways, and can be endothelium-dependent or independent. Of particular interest are the pathways leading to hyperpolarization of SMCs, as these can potentially evoke both local and conducted dilation. This review focuses on three agonists that cause local and conducted dilation in skeletal muscle: ACh, ATP, and KCl. The mechanisms for the release of these agonists during motor nerve stimulation and/or hypoxia, and their actions to open either Ca2+ -activated K+ channels (KCa ) or inwardly rectifying K+ channels (KIR ) are described. By causing local and conducted dilation, each agonist has the ability to improve skeletal muscle blood flow during exercise and ischemia.
Collapse
Affiliation(s)
- Kim A Dora
- Department of Pharmacology, University of Oxford, Oxford, UK
| |
Collapse
|
11
|
Dick GM, Tune JD. Dynamic Regulation of the Subunit Composition of BK Channels in Smooth Muscle. Circ Res 2017; 121:594-595. [PMID: 28860314 DOI: 10.1161/circresaha.117.311723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Gregory M Dick
- From the California Medical Innovations Institute, San Diego (G.M.D.); and Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis (J.D.T.)
| | - Johnathan D Tune
- From the California Medical Innovations Institute, San Diego (G.M.D.); and Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis (J.D.T.).
| |
Collapse
|
12
|
Suh EY, Yin MZ, Lin H, Zhang YH, Yoo HY, Kim SJ. Maxi-K channel (BK Ca) activity veils the myogenic tone of mesenteric artery in rats. Physiol Rep 2017; 5:5/14/e13330. [PMID: 28720714 PMCID: PMC5532478 DOI: 10.14814/phy2.13330] [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: 01/25/2017] [Revised: 05/30/2017] [Accepted: 05/31/2017] [Indexed: 01/27/2023] Open
Abstract
Arterioles and small arteries change their tone in response to transmural pressure changes, called myogenic tone (MT). In comparison to the branches of cerebral arteries (CA) showing prominent MT, the third branches of mesenteric arteries (MA) with similar diameters show weaker MT. Here, we aimed to analyze the electrophysiological differences responsible for the weaker MT in MA (MTMA) than MT in CA (MTCA). We measured ionic current using patch clamp in isolated MA smooth muscle cells (MASMCs) and CA smooth muscle cells (CASMCs) of rats. MT was analyzed using video analysis of pressurized small arteries. Quantitative‐PCR (q‐PCR) and immunofluorescence confocal microscopy were used to compare the mRNA and protein expression level of big‐conductance Ca2+‐activated K+ channel (BKCa) subunits (Slo1α and Sloβ1). Whole‐cell patch clamp study revealed higher density of voltage‐operated Ca2+ channel current (ICaV) in the MASMCs than in CASMCs. Although voltage‐gated K+ channel current (IKv) was also higher in MASMCs, treatment with Kv inhibitor (4‐aminopyridine) did not affect MTMA. Interestingly, BKCa current density and the frequency of spontaneous transient outward currents (STOCs) were consistently higher in MASMCs than in CASMCs. Inside‐out patch clamp showed that the Ca2+‐sensitivity of BKCa is higher in MASMCs than in CASMCs. Iberiotoxin, a selective BKCa inhibitor, augmented MTMA by a larger extent than MTCA. Although q‐PCR analysis did not reveal a significant difference of mRNAs for Slo1α and Sloβ1, immunofluorescence image suggested higher expression of Slo1α in MASMCs than in CASMCs. Despite the large ICaV density, the high activities of BKCa including the more frequent STOCs in MASMCs veils the potentially strong MTMA.
Collapse
Affiliation(s)
- Eun Yeong Suh
- Departments of Physiology and of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Ming Zhe Yin
- Departments of Physiology and of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Haiyue Lin
- Departments of Physiology and of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Yin Hua Zhang
- Departments of Physiology and of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| | - Hae Young Yoo
- Chung-Ang University Red Cross College of Nursing, Seoul, Korea
| | - Sung Joon Kim
- Departments of Physiology and of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
13
|
Garland CJ, Bagher P, Powell C, Ye X, Lemmey HAL, Borysova L, Dora KA. Voltage-dependent Ca 2+ entry into smooth muscle during contraction promotes endothelium-mediated feedback vasodilation in arterioles. Sci Signal 2017; 10:10/486/eaal3806. [PMID: 28676489 DOI: 10.1126/scisignal.aal3806] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Vascular smooth muscle contraction is suppressed by feedback dilation mediated by the endothelium. In skeletal muscle arterioles, this feedback can be activated by Ca2+ signals passing from smooth muscle through gap junctions to endothelial cells, which protrude through holes in the internal elastic lamina to make contact with vascular smooth muscle cells. Although hypothetically either Ca2+ or inositol trisphosphate (IP3) may provide the intercellular signal, it is generally thought that IP3 diffusion is responsible. We provide evidence that Ca2+ entry through L-type voltage-dependent Ca2+ channels (VDCCs) in vascular smooth muscle can pass to the endothelium through positions aligned with holes in the internal elastic lamina in amounts sufficient to activate endothelial cell Ca2+ signaling. In endothelial cells in which IP3 receptors (IP3Rs) were blocked, VDCC-driven Ca2+ events were transient and localized to the endothelium that protrudes through the internal elastic lamina to contact vascular smooth muscle cells. In endothelial cells in which IP3Rs were not blocked, VDCC-driven Ca2+ events in endothelial cells were amplified to form propagating waves. These waves activated voltage-insensitive, intermediate-conductance, Ca2+-activated K+ (IKCa) channels, thereby providing feedback that effectively suppressed vasoconstriction and enabled cycles of constriction and dilation called vasomotion. Thus, agonists that stimulate vascular smooth muscle depolarization provide Ca2+ to endothelial cells to activate a feedback circuit that protects tissue blood flow.
Collapse
Affiliation(s)
- Christopher J Garland
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Pooneh Bagher
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Chloe Powell
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Xi Ye
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Hamish A L Lemmey
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Lyudmyla Borysova
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Kim A Dora
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK.
| |
Collapse
|
14
|
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: 228] [Impact Index Per Article: 28.5] [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
|
15
|
Hong K, Lee S, Li R, Yang Y, Tanner MA, Wu J, Hill MA. Adiponectin Receptor Agonist, AdipoRon, Causes Vasorelaxation Predominantly Via a Direct Smooth Muscle Action. Microcirculation 2016; 23:207-20. [PMID: 26728950 DOI: 10.1111/micc.12266] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 12/31/2015] [Indexed: 12/24/2022]
Abstract
OBJECTIVE AdipoRon, an adiponectin receptor agonist, was recently proposed for treating insulin resistance and hyperglycemia. As adiponectin is vasoprotective via NO-mediated signaling, it was hypothesized that adipoRon similarly exerts potentially beneficial vasodilator effects. We therefore examined if adipoRon induces vasorelaxation and via what contributing mechanisms. METHODS Vascular function was assessed in skeletal muscle arteries from rats and cerebral/coronary arteries from mice using pressure and wire myography. RESULTS Using qPCR, mRNA for adiponectin receptors was demonstrated in skeletal muscle, cerebral and coronary arteries. AdipoRon-caused vasorelaxation was not abolished by compound C (10 μM; AMPK inhibitor). Inhibition of endothelium-dependent relaxation with combinations of l-NAME/indomethacin/apamin/TRAM-34 only slightly reduced adipoRon-mediated vasorelaxation in cerebral and coronary arteries. EC-denuded cremaster arteries showed similar relaxant responses to adipoRon as in intact vessels, suggesting adipoRon directly impacts VSMCs. K(+) currents measured in VSMCs isolated from mouse basilar and LAD arteries were not altered by adiopRon. In cremaster arteries, adipoRon induced vasorelaxation without a marked decrease in VSMC [Ca(2+)]i . Adiponectin, itself, caused vasodilation in intact cremaster arteries while failing to cause significant dilation in EC-denuded arteries, consistent with endothelium dependency of adiponectin. CONCLUSIONS AdipoRon exerts vasodilation by mechanisms distinct to adiponectin. The dominant mechanism for adipoRon-induced vasorelaxation occurs independently of endothelium-dependent relaxing factors, AMPK activation, K(+) efflux-mediated hyperpolarization and reductions in cytosolic [Ca(2+)]i .
Collapse
Affiliation(s)
- Kwangseok Hong
- Department of Medical Pharmacology & Physiology, University of Missouri-Columbia, Columbia, Missouri, USA.,Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri, USA
| | - Sewon Lee
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri, USA.,Division of Sport Science & Sport Science Institute, Incheon National University, Incheon, South Korea
| | - Rong Li
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri, USA.,Drug Discovery Research Center, Luzhou Medical College, Luzhou, China
| | - Yan Yang
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri, USA
| | - Miles A Tanner
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri, USA
| | - Jianbo Wu
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri, USA.,Drug Discovery Research Center, Luzhou Medical College, Luzhou, China
| | - Michael A Hill
- Department of Medical Pharmacology & Physiology, University of Missouri-Columbia, Columbia, Missouri, USA.,Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri, USA
| |
Collapse
|
16
|
Hong K, Zhao G, Hong Z, Sun Z, Yang Y, Clifford PS, Davis MJ, Meininger GA, Hill MA. Mechanical activation of angiotensin II type 1 receptors causes actin remodelling and myogenic responsiveness in skeletal muscle arterioles. J Physiol 2016; 594:7027-7047. [PMID: 27531064 PMCID: PMC5134373 DOI: 10.1113/jp272834] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 08/09/2016] [Indexed: 12/15/2022] Open
Abstract
KEY POINTS Candesartan, an inverse agonist of the type 1 angiotensin II receptor (AT1 R), causes a concentration-dependent inhibition of pressure-dependent myogenic tone consistent with previous reports of mechanosensitivity of this G protein-coupled receptor. Mechanoactivation of the AT1 R occurs independently of local angiotensin II production and the type 2 angiotensin receptor. Mechanoactivation of the AT1 R stimulates actin polymerization by a protein kinase C-dependent mechanism, but independently of a change in intracellular Ca2+ . Using atomic force microscopy, changes in single vascular smooth muscle cell cortical actin are observed to remodel following mechanoactivation of the AT1 R. ABSTRACT The Gq/11 protein-coupled angiotensin II type 1 receptor (AT1 R) has been shown to be activated by mechanical stimuli. In the vascular system, evidence supports the AT1 R being a mechanosensor that contributes to arteriolar myogenic constriction. The aim of this study was to determine if AT1 R mechanoactivation affects myogenic constriction in skeletal muscle arterioles and to determine underlying cellular mechanisms. Using pressure myography to study rat isolated first-order cremaster muscle arterioles the AT1 R inhibitor candesartan (10-7 -10-5 m) showed partial but concentration-dependent inhibition of myogenic reactivity. Inhibition was demonstrated by a rightward shift in the pressure-diameter relationship over the intraluminal pressure range, 30-110 mmHg. Pressure-induced changes in global vascular smooth muscle intracellular Ca2+ (using Fura-2) were similar in the absence or presence of candesartan, indicating that AT1 R-mediated myogenic constriction relies on Ca2+ -independent downstream signalling. The diacylglycerol analogue 1-oleoyl-2-acetyl-sn-glycerol (OAG) reversed the inhibitory effect of candesartan, while this rescue effect was prevented by the protein kinase C (PKC) inhibitor GF 109203X. Both candesartan and PKC inhibition caused increased G-actin levels, as determined by Western blotting of vessel lysates, supporting involvement of cytoskeletal remodelling. At the single vascular smooth muscle cell level, atomic force microscopy showed that cell swelling (stretch) with hypotonic buffer also caused thickening of cortical actin fibres and this was blocked by candesartan. Collectively, the present studies support growing evidence for novel modes of activation of the AT1 R in arterioles and suggest that mechanically activated AT1 R generates diacylglycerol, which in turn activates PKC which induces the actin cytoskeleton reorganization that is required for pressure-induced vasoconstriction.
Collapse
Affiliation(s)
- Kwangseok Hong
- Dalton Cardiovascular Research CentreUniversity of MissouriColumbiaMO65211USA
- Department of Medical Pharmacology and PhysiologyUniversity of MissouriColumbiaMO65211USA
- Robert M. Berne Cardiovascular Research Centre and Department of Molecular Physiology and Biological PhysicsUniversity of VirginiaCharlottesvilleVA22908USA
| | - Guiling Zhao
- College of Applied Health SciencesUniversity of Illinois at ChicagoChicagoIL60612USA
| | - Zhongkui Hong
- Dalton Cardiovascular Research CentreUniversity of MissouriColumbiaMO65211USA
- Department of Biomedical EngineeringUniversity of South DakotaSioux FallsSD57107USA
| | - Zhe Sun
- Dalton Cardiovascular Research CentreUniversity of MissouriColumbiaMO65211USA
- Department of Medical Pharmacology and PhysiologyUniversity of MissouriColumbiaMO65211USA
| | - Yan Yang
- Dalton Cardiovascular Research CentreUniversity of MissouriColumbiaMO65211USA
| | - Philip S. Clifford
- College of Applied Health SciencesUniversity of Illinois at ChicagoChicagoIL60612USA
| | - Michael J. Davis
- Dalton Cardiovascular Research CentreUniversity of MissouriColumbiaMO65211USA
- Department of Medical Pharmacology and PhysiologyUniversity of MissouriColumbiaMO65211USA
| | - Gerald A. Meininger
- Dalton Cardiovascular Research CentreUniversity of MissouriColumbiaMO65211USA
- Department of Medical Pharmacology and PhysiologyUniversity of MissouriColumbiaMO65211USA
| | - Michael A. Hill
- Dalton Cardiovascular Research CentreUniversity of MissouriColumbiaMO65211USA
- Department of Medical Pharmacology and PhysiologyUniversity of MissouriColumbiaMO65211USA
| |
Collapse
|
17
|
DuPont JJ, McCurley A, Davel AP, McCarthy J, Bender SB, Hong K, Yang Y, Yoo JK, Aronovitz M, Baur WE, Christou DD, Hill MA, Jaffe IZ. Vascular mineralocorticoid receptor regulates microRNA-155 to promote vasoconstriction and rising blood pressure with aging. JCI Insight 2016; 1:e88942. [PMID: 27683672 DOI: 10.1172/jci.insight.88942] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Hypertension is nearly universal yet poorly controlled in the elderly despite proven benefits of intensive treatment. Mice lacking mineralocorticoid receptors in smooth muscle cells (SMC-MR-KO) are protected from rising blood pressure (BP) with aging, despite normal renal function. Vasoconstriction is attenuated in aged SMC-MR-KO mice, thus they were used to explore vascular mechanisms that may contribute to hypertension with aging. MicroRNA (miR) profiling identified miR-155 as the most down-regulated miR with vascular aging in MR-intact but not SMC-MR-KO mice. The aging-associated decrease in miR-155 in mesenteric resistance vessels was associated with increased mRNA abundance of MR and of predicted miR-155 targets Cav1.2 (L-type calcium channel (LTCC) subunit) and angiotensin type-1 receptor (AgtR1). SMC-MR-KO mice lacked these aging-associated vascular gene expression changes. In HEK293 cells, MR repressed miR-155 promoter activity. In cultured SMCs, miR-155 decreased Cav1.2 and AgtR1 mRNA. Compared to MR-intact littermates, aged SMC-MR-KO mice had decreased systolic BP, myogenic tone, SMC LTCC current, mesenteric vessel calcium influx, LTCC-induced vasoconstriction and angiotensin II-induced vasoconstriction and oxidative stress. Restoration of miR-155 specifically in SMCs of aged MR-intact mice decreased Cav1.2 and AgtR1 mRNA and attenuated LTCC-mediated and angiotensin II-induced vasoconstriction and oxidative stress. Finally, in a trial of MR blockade in elderly humans, changes in serum miR-155 predicted the BP treatment response. Thus, SMC-MR regulation of miR-155, Cav1.2 and AgtR1 impacts vasoconstriction with aging. This novel mechanism identifies potential new treatment strategies and biomarkers to improve and individualize antihypertensive therapy in the elderly.
Collapse
Affiliation(s)
- Jennifer J DuPont
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA
| | - Amy McCurley
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA
| | - Ana P Davel
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA.,Department of Structural and Functional Biology, Institute of Biology, University of Campinas-UNICAMP, São Paulo, Brazil
| | - Joseph McCarthy
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA
| | - Shawn B Bender
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA.,Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, Missouri, USA.,Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, USA
| | - Kwangseok Hong
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA.,Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
| | - Yan Yang
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| | - Jeung-Ki Yoo
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA
| | - Mark Aronovitz
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA
| | - Wendy E Baur
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA
| | - Demetra D Christou
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA
| | - Michael A Hill
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA.,Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
| | - Iris Z Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
18
|
Potassium Channels in Regulation of Vascular Smooth Muscle Contraction and Growth. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2016; 78:89-144. [PMID: 28212804 DOI: 10.1016/bs.apha.2016.07.001] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Potassium channels importantly contribute to the regulation of vascular smooth muscle (VSM) contraction and growth. They are the dominant ion conductance of the VSM cell membrane and importantly determine and regulate membrane potential. Membrane potential, in turn, regulates the open-state probability of voltage-gated Ca2+ channels (VGCC), Ca2+ influx through VGCC, intracellular Ca2+, and VSM contraction. Membrane potential also affects release of Ca2+ from internal stores and the Ca2+ sensitivity of the contractile machinery such that K+ channels participate in all aspects of regulation of VSM contraction. Potassium channels also regulate proliferation of VSM cells through membrane potential-dependent and membrane potential-independent mechanisms. VSM cells express multiple isoforms of at least five classes of K+ channels that contribute to the regulation of contraction and cell proliferation (growth). This review will examine the structure, expression, and function of large conductance, Ca2+-activated K+ (BKCa) channels, intermediate-conductance Ca2+-activated K+ (KCa3.1) channels, multiple isoforms of voltage-gated K+ (KV) channels, ATP-sensitive K+ (KATP) channels, and inward-rectifier K+ (KIR) channels in both contractile and proliferating VSM cells.
Collapse
|
19
|
Zhu Z, Tang J, Zhou X, Xiang S, Zhu X, Li N, Shi R, Zhong Y, Zhang L, Sun M, Xu Z. Roles of ion channels in regulation of acetylcholine-mediated vasoconstrictions in umbilical cords of rabbit/rats. Reprod Toxicol 2016; 65:95-103. [PMID: 27421582 DOI: 10.1016/j.reprotox.2016.07.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 05/31/2016] [Accepted: 07/08/2016] [Indexed: 12/25/2022]
Abstract
We recently demonstrated that acetylcholine (ACh) produced reliable vasoconstrictions in the umbilical cords. This study investigated the possible mechanisms with different antagonists. ACh-mediated vasoconstrictions were decreased by voltage-operated calcium (Ca2+) channels antagonist nifedipine or inositol-1,4,5-trisphosphate-mediated Ca2+ release antagonist 2-aminoethyl diphenylborinate, indicating that both extracellular and intracellular calcium modulated the ACh-stimulated umbilical contraction. Intracellular Ca2+ concentrations were increased simultaneously with vasoconstrictions by ACh in the umbilical vessels. Inhibiting large-conductance calcium-dependent potassium (BK) channels enhanced ACh-mediated contraction, whereas inhibiting voltage dependent potassium (K+), inward rectifier K+ and ATP-sensitive K+ channels had no effects. Incubation with specific K+ channel inhibitors showed that ACh suppressed BK currents rather than 4-aminopyridine-sensitive K+ channels currents. The results suggested that blood vessels in umbilical cords had special characteristics in response to cholinergic signals. ACh-stimulated umbilical vasoconstrictions were mediated via muscarinic receptor subtype 1/3-protein kinase C/cyclooxygenase-BK channel pathways.
Collapse
Affiliation(s)
- Zhoufeng Zhu
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Jiaqi Tang
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Xiuwen Zhou
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Sharon Xiang
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China; Center for Perinatal Biology, Loma Linda University, Loma Linda, CA, USA
| | - Xiaolin Zhu
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Na Li
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Ruixiu Shi
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Yuan Zhong
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Lubo Zhang
- Center for Perinatal Biology, Loma Linda University, Loma Linda, CA, USA
| | - Miao Sun
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China.
| | - Zhice Xu
- Institute for Fetology, the First Affiliated Hospital of Soochow University, Suzhou 215006, China; Center for Perinatal Biology, Loma Linda University, Loma Linda, CA, USA.
| |
Collapse
|
20
|
Lee S, Yang Y, Tanner MA, Li M, Hill MA. Heterogeneity in Kv7 channel function in the cerebral and coronary circulation. Microcirculation 2015; 22:109-121. [PMID: 25476662 DOI: 10.1111/micc.12183] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 12/02/2014] [Indexed: 12/15/2022]
Abstract
OBJECTIVES Kv7 channels are considered important regulators of vascular smooth muscle contractility. The present study aimed to examine the hypotheses that (i) Kv7 channels are present in mouse cerebral and coronary arteries and regulate vascular reactivity and (ii) regional differences exist in the activity of these channels. METHODS AND RESULTS PCR confirmed that basilar, Circle of Willis and LAD arteries express predominantly Kv7.1 and 7.4. Western blot analysis, however, showed greater Kv7.4 protein levels in the cerebral vessels. Relaxation to the Kv7 channel activator, retigabine (1-50 μM) was significantly greater in the basilar artery compared to the LAD artery. Similarly, the Kv7 channel inhibitor, linopirdine (10 μM) caused a stronger contraction of the basilar artery. Furthermore, pre-incubation with linopirdine reduced forskolin (cAMP activator)-induced vasorelaxation in basilar while not altering forskolin-induced vasorelaxation of the LAD, suggesting that Kv7 channels play a more prominent role in the cerebral than in the coronary circulation. Consistent with the vessel data, whole cell Kv7 currents in cerebral VSMCs were potentiated by retigabine and inhibited by linopirdine, while these responses were blunted in coronary VSMCs. CONCLUSIONS This study provides evidence that mouse Kv7 channels may contribute differently to regulating the functional properties of cerebral and coronary arteries. Such heterogeneity has important implications for developing novel therapeutics for cardiovascular dysfunction.
Collapse
Affiliation(s)
- Sewon Lee
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, MO 65211
| | - Yan Yang
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, MO 65211
| | - Miles A Tanner
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, MO 65211
| | - Min Li
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, MO 65211
| | - Michael A Hill
- Dalton Cardiovascular Research Center, University of Missouri-Columbia, MO 65211.,Department of Medical Pharmacology & Physiology, University of Missouri-Columbia, MO 65211
| |
Collapse
|
21
|
Jackson-Weaver O, Osmond JM, Naik JS, Gonzalez Bosc LV, Walker BR, Kanagy NL. Intermittent hypoxia in rats reduces activation of Ca2+ sparks in mesenteric arteries. Am J Physiol Heart Circ Physiol 2015; 309:H1915-22. [PMID: 26408536 DOI: 10.1152/ajpheart.00179.2015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 09/17/2015] [Indexed: 01/25/2023]
Abstract
Ca(+) sparks are vascular smooth muscle cell (VSMC) Ca(2+)-release events that are mediated by ryanodine receptors (RyR) and promote vasodilation by activating large-conductance Ca(2+)-activated potassium channels and inhibiting myogenic tone. We have previously reported that exposing rats to intermittent hypoxia (IH) to simulate sleep apnea augments myogenic tone in mesenteric arteries through loss of hydrogen sulfide (H2S)-induced dilation. Because we also observed that H2S can increase Ca(2+) spark activity, we hypothesized that loss of H2S after IH exposure reduces Ca(2+) spark activity and that blocking Ca(2+) spark generation reduces H2S-induced dilation. Ca(2+) spark activity was lower in VSMC of arteries from IH compared with sham-exposed rats. Furthermore, depolarizing VSMC by increasing luminal pressure (from 20 to 100 mmHg) or by elevating extracellular [K(+)] increased spark activity in VSMC of arteries from sham rats but had no effect in arteries from IH rats. Inhibiting endogenous H2S production in sham arteries prevented these increases. NaHS or phosphodiesterase inhibition increased spark activity to the same extent in sham and IH arteries. Depolarization-induced increases in Ca(2+) spark activity were due to increased sparks per site, whereas H2S increases in spark activity were due to increased spark sites per cell. Finally, inhibiting Ca(2+) spark activity with ryanodine (10 μM) enhanced myogenic tone in arteries from sham but not IH rats and blocked dilation to exogenous H2S in arteries from both sham and IH rats. Our results suggest that H2S regulates RyR activation and that H2S-induced dilation requires Ca(2+) spark activation. IH exposure decreases endogenous H2S-dependent Ca(2+) spark activation to cause membrane depolarization and enhance myogenic tone in mesenteric arteries.
Collapse
Affiliation(s)
- Olan Jackson-Weaver
- Vascular Physiology Group, Department of Cell Biology and Physiology, School of Medicine, University of New Mexico, Albuquerque, New Mexico
| | - Jessica M Osmond
- Vascular Physiology Group, Department of Cell Biology and Physiology, School of Medicine, University of New Mexico, Albuquerque, New Mexico
| | - Jay S Naik
- Vascular Physiology Group, Department of Cell Biology and Physiology, School of Medicine, University of New Mexico, Albuquerque, New Mexico
| | - Laura V Gonzalez Bosc
- Vascular Physiology Group, Department of Cell Biology and Physiology, School of Medicine, University of New Mexico, Albuquerque, New Mexico
| | - Benjimen R Walker
- Vascular Physiology Group, Department of Cell Biology and Physiology, School of Medicine, University of New Mexico, Albuquerque, New Mexico
| | - Nancy L Kanagy
- Vascular Physiology Group, Department of Cell Biology and Physiology, School of Medicine, University of New Mexico, Albuquerque, New Mexico
| |
Collapse
|
22
|
Harraz OF, Brett SE, Zechariah A, Romero M, Puglisi JL, Wilson SM, Welsh DG. Genetic ablation of CaV3.2 channels enhances the arterial myogenic response by modulating the RyR-BKCa axis. Arterioscler Thromb Vasc Biol 2015; 35:1843-51. [PMID: 26069238 DOI: 10.1161/atvbaha.115.305736] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 05/31/2015] [Indexed: 12/31/2022]
Abstract
OBJECTIVE In resistance arteries, there is an emerging view that smooth muscle CaV3.2 channels restrain arterial constriction through a feedback response involving the large-conductance Ca(2+)-activated K(+) channel (BKCa). Here, we used wild-type and CaV3.2 knockout (CaV3.2(-/-)) mice to definitively test whether CaV3.2 moderates myogenic tone in mesenteric arteries via the CaV3.2-ryanodine receptor-BKCa axis and whether this regulatory mechanism influences blood pressure regulation. APPROACH AND RESULTS Using pressurized vessel myography, CaV3.2(-/-) mesenteric arteries displayed enhanced myogenic constriction to pressure but similar K(+)-induced vasoconstriction compared with wild-type C57BL/6 arteries. Electrophysiological and myography experiments subsequently confirmed the inability of micromolar Ni(2+), a CaV3.2 blocker, to either constrict arteries or suppress T-type currents in CaV3.2(-/-) smooth muscle cells. The frequency of BKCa-induced spontaneous transient outward K(+) currents dropped in wild-type but not in knockout arterial smooth muscle cells upon the pharmacological suppression of CaV3.2 channel. Line scan analysis performed on en face arteries loaded with Fluo-4 revealed the presence of Ca(2+) sparks in all arteries, with the subsequent application of Ni(2+) only affecting wild-type arteries. Although CaV3.2 channel moderated myogenic constriction of resistance arteries, the blood pressure measurements of CaV3.2(-/-) and wild-type animals were similar. CONCLUSIONS Overall, our findings establish a negative feedback mechanism of the myogenic response in which CaV3.2 channel modulates downstream ryanodine receptor-BKCa to hyperpolarize and relax arteries.
Collapse
Affiliation(s)
- Osama F Harraz
- From the Department of Physiology and Pharmacology, Hotchkiss Brain and Libin Cardiovascular Institutes, University of Calgary, Calgary, Alberta, Canada (O.F.H., S.E.B., A.Z., D.G.W.); Department of Pharmacology and Toxicology, Alexandria University, Alexandria, Egypt (O.F.H.); Department of Basic Sciences, Division of Pharmacology, Loma Linda University, CA (M.R., S.M.W.); Department of Pharmacology, University of California, Davis (J.L.P.); and Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada (D.G.W.)
| | - Suzanne E Brett
- From the Department of Physiology and Pharmacology, Hotchkiss Brain and Libin Cardiovascular Institutes, University of Calgary, Calgary, Alberta, Canada (O.F.H., S.E.B., A.Z., D.G.W.); Department of Pharmacology and Toxicology, Alexandria University, Alexandria, Egypt (O.F.H.); Department of Basic Sciences, Division of Pharmacology, Loma Linda University, CA (M.R., S.M.W.); Department of Pharmacology, University of California, Davis (J.L.P.); and Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada (D.G.W.)
| | - Anil Zechariah
- From the Department of Physiology and Pharmacology, Hotchkiss Brain and Libin Cardiovascular Institutes, University of Calgary, Calgary, Alberta, Canada (O.F.H., S.E.B., A.Z., D.G.W.); Department of Pharmacology and Toxicology, Alexandria University, Alexandria, Egypt (O.F.H.); Department of Basic Sciences, Division of Pharmacology, Loma Linda University, CA (M.R., S.M.W.); Department of Pharmacology, University of California, Davis (J.L.P.); and Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada (D.G.W.)
| | - Monica Romero
- From the Department of Physiology and Pharmacology, Hotchkiss Brain and Libin Cardiovascular Institutes, University of Calgary, Calgary, Alberta, Canada (O.F.H., S.E.B., A.Z., D.G.W.); Department of Pharmacology and Toxicology, Alexandria University, Alexandria, Egypt (O.F.H.); Department of Basic Sciences, Division of Pharmacology, Loma Linda University, CA (M.R., S.M.W.); Department of Pharmacology, University of California, Davis (J.L.P.); and Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada (D.G.W.)
| | - Jose L Puglisi
- From the Department of Physiology and Pharmacology, Hotchkiss Brain and Libin Cardiovascular Institutes, University of Calgary, Calgary, Alberta, Canada (O.F.H., S.E.B., A.Z., D.G.W.); Department of Pharmacology and Toxicology, Alexandria University, Alexandria, Egypt (O.F.H.); Department of Basic Sciences, Division of Pharmacology, Loma Linda University, CA (M.R., S.M.W.); Department of Pharmacology, University of California, Davis (J.L.P.); and Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada (D.G.W.)
| | - Sean M Wilson
- From the Department of Physiology and Pharmacology, Hotchkiss Brain and Libin Cardiovascular Institutes, University of Calgary, Calgary, Alberta, Canada (O.F.H., S.E.B., A.Z., D.G.W.); Department of Pharmacology and Toxicology, Alexandria University, Alexandria, Egypt (O.F.H.); Department of Basic Sciences, Division of Pharmacology, Loma Linda University, CA (M.R., S.M.W.); Department of Pharmacology, University of California, Davis (J.L.P.); and Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada (D.G.W.)
| | - Donald G Welsh
- From the Department of Physiology and Pharmacology, Hotchkiss Brain and Libin Cardiovascular Institutes, University of Calgary, Calgary, Alberta, Canada (O.F.H., S.E.B., A.Z., D.G.W.); Department of Pharmacology and Toxicology, Alexandria University, Alexandria, Egypt (O.F.H.); Department of Basic Sciences, Division of Pharmacology, Loma Linda University, CA (M.R., S.M.W.); Department of Pharmacology, University of California, Davis (J.L.P.); and Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada (D.G.W.).
| |
Collapse
|
23
|
Al-Brakati AY, Kamishima T, Dart C, Quayle JM. Caveolar disruption causes contraction of rat femoral arteries via reduced basal NO release and subsequent closure of BKCa channels. PeerJ 2015; 3:e966. [PMID: 26038721 PMCID: PMC4451037 DOI: 10.7717/peerj.966] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 04/29/2015] [Indexed: 12/22/2022] Open
Abstract
Background and Purpose. Caveolae act as signalling hubs in endothelial and smooth muscle cells. Caveolar disruption by the membrane cholesterol depleting agent methyl-β-cyclodextrin (M-β-CD) has various functional effects on arteries including (i) impairment of endothelium-dependent relaxation, and (ii) alteration of smooth muscle cell (SMC) contraction independently of the endothelium. The aim of this study was to explore the effects of M-β-CD on rat femoral arteries. Methods. Isometric force was measured in rat femoral arteries stimulated to contract with a solution containing 20 mM K(+) and 200 nM Bay K 8644 (20 K/Bay K) or with one containing 80 mM K(+)(80 K). Results. Incubation of arteries with M-β-CD (5 mM, 60 min) increased force in response to 20 K/Bay K but not that induced by 80 K. Application of cholesterol saturated M-β-CD (Ch-MCD, 5 mM, 50 min) reversed the effects of M-β-CD. After mechanical removal of endothelial cells M-β-CD caused only a small enhancement of contractions to 20 K/Bay K. This result suggests M-β-CD acts via altering release of an endothelial-derived vasodilator or vasoconstrictor. When nitric oxide synthase was blocked by pre-incubation of arteries with L-NAME (250 µM) the contraction of arteries to 20 K/Bay K was enhanced, and this effect was abolished by pre-treatment with M-β-CD. This suggests M-β-CD is inhibiting endothelial NO release. Inhibition of large conductance voltage- and Ca(2+)-activated (BKCa) channels with 2 mM TEA(+) or 100 nM Iberiotoxin (IbTX) enhanced 20 K/Bay K contractions. L-NAME attenuated the contractile effect of IbTX, as did endothelial removal. Conclusions. Our results suggest caveolar disruption results in decreased release of endothelial-derived nitric oxide in rat femoral artery, resulting in a reduced contribution of BKCa channels to the smooth muscle cell membrane potential, causing depolarisation and contraction.
Collapse
Affiliation(s)
- AY Al-Brakati
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - T Kamishima
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - C Dart
- Department of Biochemistry and Cell Biology, Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - JM Quayle
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| |
Collapse
|
24
|
Bhattarai Y, Fernandes R, Kadrofske MM, Lockwood LR, Galligan JJ, Xu H. Western blot analysis of BK channel β1-subunit expression should be interpreted cautiously when using commercially available antibodies. Physiol Rep 2014; 2:2/10/e12189. [PMID: 25355855 PMCID: PMC4254108 DOI: 10.14814/phy2.12189] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Large conductance Ca2+‐activated K+ (BK) channels consist of pore‐forming α‐ and accessory β‐subunits. There are four β‐subunit subtypes (β1–β4), BK β1‐subunit is specific for smooth muscle cells (SMC). Reduced BK β1‐subunit expression is associated with SMC dysfunction in animal models of human disease, because downregulation of BK β1‐subunit reduces channel activity and increases SMC contractility. Several anti‐BK β1‐subunit antibodies are commercially available; however, the specificity of most antibodies has not been tested or confirmed in the tissues from BK β1‐subunit knockout (KO) mice. In this study, we tested the specificity and sensitivity of six commercially available antibodies from five manufacturers. We performed western blot analysis on BK β1‐subunit enriched tissues (mesenteric arteries and colons) and non‐SM tissue (cortex of kidney) from wild‐type (WT) and BK β1‐KO mice. We found that antibodies either detected protein bands of the appropriate molecular weight in tissues from both WT and BK β1‐KO mice or failed to detect protein bands at the appropriate molecular weight in tissues from WT mice, suggesting that these antibodies may lack specificity for the BK β1‐subunit. The absence of BK β1‐subunit mRNA expression in arteries, colons, and kidneys from BK β1‐KO mice was confirmed by RT‐PCR analysis. We conclude that these commercially available antibodies might not be reliable tools for studying BK β1‐subunit expression in murine tissues under the denaturing conditions that we have used. Data obtained using commercially available antibodies should be interpreted cautiously. Our studies underscore the importance of proper negative controls in western blot analyses. Commercially available anti‐BK β1‐subunit antibodies either detected protein bands of the appropriate molecular weight in tissues from both WT and BK β1‐KO mice or failed to detect protein bands at the appropriate molecular weight in tissues from WT mice. These commercially available antibodies are not reliable tools for studying BK β1‐subunit expression in murine tissues. Data obtained using these antibodies should be interpreted cautiously.
Collapse
Affiliation(s)
- Yogesh Bhattarai
- Neuroscience Program, Michigan State University, East Lansing, Michigan
| | - Roxanne Fernandes
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - Mark M Kadrofske
- Department of Pediatrics and Human Development, Michigan State University, East Lansing, Michigan
| | - Lizbeth R Lockwood
- Department of Pediatrics and Human Development, Michigan State University, East Lansing, Michigan
| | - James J Galligan
- Neuroscience Program, Michigan State University, East Lansing, Michigan Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - Hui Xu
- Neuroscience Program, Michigan State University, East Lansing, Michigan Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| |
Collapse
|
25
|
Kyle BD, Braun AP. The regulation of BK channel activity by pre- and post-translational modifications. Front Physiol 2014; 5:316. [PMID: 25202279 PMCID: PMC4141542 DOI: 10.3389/fphys.2014.00316] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 08/02/2014] [Indexed: 11/17/2022] Open
Abstract
Large conductance, Ca2+-activated K+ (BK) channels represent an important pathway for the outward flux of K+ ions from the intracellular compartment in response to membrane depolarization, and/or an elevation in cytosolic free [Ca2+]. They are functionally expressed in a range of mammalian tissues (e.g., nerve and smooth muscles), where they can either enhance or dampen membrane excitability. The diversity of BK channel activity results from the considerable alternative mRNA splicing and post-translational modification (e.g., phosphorylation) of key domains within the pore-forming α subunit of the channel complex. Most of these modifications are regulated by distinct upstream cell signaling pathways that influence the structure and/or gating properties of the holo-channel and ultimately, cellular function. The channel complex may also contain auxiliary subunits that further affect channel gating and behavior, often in a tissue-specific manner. Recent studies in human and animal models have provided strong evidence that abnormal BK channel expression/function contributes to a range of pathologies in nerve and smooth muscle. By targeting the upstream regulatory events modulating BK channel behavior, it may be possible to therapeutically intervene and alter BK channel expression/function in a beneficial manner.
Collapse
Affiliation(s)
- Barry D Kyle
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Research Institute, University of Calgary Calgary, AB, Canada
| | - Andrew P Braun
- Department of Physiology and Pharmacology, Cumming School of Medicine, Libin Cardiovascular Research Institute, University of Calgary Calgary, AB, Canada
| |
Collapse
|
26
|
Large conductance Ca2+-activated K+ channel (BKCa) α-subunit splice variants in resistance arteries from rat cerebral and skeletal muscle vasculature. PLoS One 2014; 9:e98863. [PMID: 24921651 PMCID: PMC4055454 DOI: 10.1371/journal.pone.0098863] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 05/07/2014] [Indexed: 11/19/2022] Open
Abstract
Previous studies report functional differences in large conductance Ca2+ activated-K+ channels (BKCa) of smooth muscle cells (VSMC) from rat cerebral and cremaster muscle resistance arteries. The present studies aimed to determine if this complexity in BKCa activity may, in part, be due to splice variants in the pore-forming α-subunit. BKCa variants in the intracellular C terminus of the α-subunit, and their relative expression to total α-subunit, were examined by qPCR. Sequencing of RT-PCR products showed two α-subunit variants, ZERO and STREX, to be identical in cremaster and cerebral arteries. Levels of STREX mRNA expression were, however, significantly higher in cremaster VSMCs (28.9±4.2% of total α-BKCa) compared with cerebral vessels (16.5±0.9%). Further, a low level of BKCa SS4 α-subunit variant was seen in cerebral arteries, while undetectable in cremaster arteries. Protein biotinylation assays, in expression systems and arterial preparations, were used to determine whether differences in splice variant mRNA expression affect surface membrane/cytosolic location of the channel. In AD-293 and CHO-K1 cells, rat STREX was more likely to be located at the plasma membrane compared to ZERO, although the great majority of channel protein was in the membrane in both cases. Co-expression of β1-BKCa subunit with STREX or ZERO did not influence the dominant membrane expression of α-BKCa subunits, whereas in the absence of α-BKCa, a significant proportion of β1-subunit remained cytosolic. Biotinylation assays of cremaster and cerebral arteries showed that differences in STREX/ZERO expression do not alter membrane/cytosolic distribution of the channel under basal conditions. These data, however, revealed that the amount of α-BKCa in cerebral arteries is approximately 20X higher than in cremaster vessels. Thus, the data support the major functional differences in BKCa activity in cremaster, as compared to cerebral VSMCs, being related to total α-BKCa expression, regardless of differences in splice variant expression.
Collapse
|
27
|
Hayoz S, Bradley V, Boerman EM, Nourian Z, Segal SS, Jackson WF. Aging increases capacitance and spontaneous transient outward current amplitude of smooth muscle cells from murine superior epigastric arteries. Am J Physiol Heart Circ Physiol 2014; 306:H1512-24. [PMID: 24705555 DOI: 10.1152/ajpheart.00492.2013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Large conductance Ca(2+)-activated K(+) channels (BKCa) contribute to negative feedback regulation of smooth muscle cell (SMC) tone. However, the effects of aging on BKCa function are unclear. We tested the hypothesis that aging alters SMC BKCa function in superior epigastric arteries (SEAs) by using perforated patch recording of enzymatically isolated SMCs from 3- to 4-mo-old male C57BL/6 mice (Young) and 24- to 26-mo-old male C57BL/6 mice (Old). SMC capacitance from Young (15.7 ± 0.4 pF; n = 110) was less than Old (17.9 ± 0.5 pF; n = 104) (P < 0.05). SMCs displayed spontaneous transient outward currents (STOCs) at membrane potentials more positive than -30 mV; depolarization increased STOC amplitude and frequency (P < 0.05; n = 19-24). STOC frequency in Young (2.2 ± 0.6 Hz) was less than Old (4.2 ± 0.7 Hz) at -10 mV (P < 0.05, n = 27-30), with no difference in amplitude (1.0 ± 0.1 vs. 0.9 ± 0.1 pA/pF, respectively). At +30 mV, STOC amplitude in Young (3.2 ± 0.3 pA/pF) was less than Old (5.0 ± 0.5 pA/pF; P < 0.05, n = 61-67) with no difference in frequency (3.9 ± 0.4 vs. 3.2 ± 0.3 Hz, respectively). BKCa blockers (1 μM paxilline, 100 nM iberiotoxin, 1 mM tetraethylammonium) or a ryanodine receptor antagonist (100 μM tetracaine) inhibited STOCs (n ≥ 6; P < 0.05 each). Western blots revealed increased expression of BKCa α-subunit protein in Old. Pressure myography revealed no effect of age on SEA maximal diameter, myogenic tone, or paxilline-induced constriction (n = 10-12; P > 0.05). Enhanced functional expression of SMC BKCa-dependent STOCs in Old may represent an adaptation of resistance arteries to maintain functional integrity.
Collapse
Affiliation(s)
- Sebastien Hayoz
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan
| | - Vanessa Bradley
- College of Veterinary Medicine, Michigan State University, East Lansing, Michigan
| | - Erika M Boerman
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri; and
| | - Zahra Nourian
- Dalton Cardiovascular Research Center, Columbia, Missouri
| | - Steven S Segal
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri; and Dalton Cardiovascular Research Center, Columbia, Missouri
| | - William F Jackson
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan;
| |
Collapse
|
28
|
Effect of diet-induced obesity on BKCa function in contraction and dilation of rat isolated middle cerebral artery. Vascul Pharmacol 2014; 61:10-5. [DOI: 10.1016/j.vph.2014.02.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 02/13/2014] [Accepted: 02/16/2014] [Indexed: 01/09/2023]
|
29
|
Pabbidi MR, Mazur O, Fan F, Farley JM, Gebremedhin D, Harder DR, Roman RJ. Enhanced large conductance K+ channel activity contributes to the impaired myogenic response in the cerebral vasculature of Fawn Hooded Hypertensive rats. Am J Physiol Heart Circ Physiol 2014; 306:H989-H1000. [PMID: 24464756 DOI: 10.1152/ajpheart.00636.2013] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Recent studies have indicated that the myogenic response (MR) in cerebral arteries is impaired in Fawn Hooded Hypertensive (FHH) rats and that transfer of a 2.4 megabase pair region of chromosome 1 (RNO1) containing 15 genes from the Brown Norway rat into the FHH genetic background restores MR in a FHH.1(BN) congenic strain. However, the mechanisms involved remain to be determined. The present study examined the role of the large conductance calcium-activated potassium (BK) channel in impairing the MR in FHH rats. Whole-cell patch-clamp studies of cerebral vascular smooth muscle cells (VSMCs) revealed that iberiotoxin (IBTX; BK inhibitor)-sensitive outward potassium (K+) channel current densities are four- to fivefold greater in FHH than in FHH.1(BN) congenic strain. Inside-out patches indicated that the BK channel open probability (NPo) is 10-fold higher and IBTX reduced NPo to a greater extent in VSMCs isolated from FHH than in FHH.1(BN) rats. Voltage sensitivity of the BK channel is enhanced in FHH as compared with FHH.1(BN) rats. The frequency and amplitude of spontaneous transient outward currents are significantly greater in VSMCs isolated from FHH than in FHH.1(BN) rats. However, the expression of the BK-α and -β-subunit proteins in cerebral vessels as determined by Western blot is similar between the two groups. Middle cerebral arteries (MCAs) isolated from FHH rats exhibited an impaired MR, and administration of IBTX restored this response. These results indicate that there is a gene on RNO1 that impairs MR in the MCAs of FHH rats by enhancing BK channel activity.
Collapse
Affiliation(s)
- Mallikarjuna R Pabbidi
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | | | | | | | | | | | | |
Collapse
|
30
|
Enouri S, Monteith G, Johnson R. Effects of acute transmural pressure elevation on endothelium-dependent vasodilation in isolated rat mesenteric veins. J Vasc Res 2013; 51:27-36. [PMID: 24280957 DOI: 10.1159/000356322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Accepted: 09/29/2013] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS The vascular regulatory function of the endothelium can be impaired by increases in transmural pressure (TMP). We tested the hypothesis that increasing TMP impairs the endothelial dilator function of rat mesenteric small veins (MSVs). METHODS In PGF2α-preconstricted MSVs, bradykinin (BK), sodium nitroprusside (SNP) and S-Nitroso-N-acetylpenicillamine (SNAP) concentration-response curves were generated at intermediate (6 mm Hg) and high (12 mm Hg) pressures. BK-induced vasodilation was examined in the absence and presence of nitric oxide synthase inhibitor [N(ω)-nitro-L-arginine (L-NNA), 100 µM], cyclooxygenase inhibitor (indomethacin, 1 µM), and large (BKCa, paxilline, 500 nM) and small (SKCa, apamin, 300 nM) conductance Ca(2+)-activated K(+) channel blockers. RESULTS BK, SNP and SNAP responses were not altered by TMP increases. BK-induced vasodilation was significantly reduced by L-NNA, indomethacin, apamin and paxilline at 6 mm Hg and L-NNA at 12 mm Hg, and was further reduced by coapplication of apamin and/or paxilline with L-NNA compared with responses obtained with either blocker. Endothelium removal completely abolished BK-induced vasodilation. CONCLUSION Venous endothelial dilator function is not affected by TMP elevation. BK-induced vasodilation is completely dependent on the presence of functional endothelial cells and mediated in part by nitric oxide, BKCa and SKCa channels, while the participation of prostacyclin may be important at intermediate pressures.
Collapse
Affiliation(s)
- Saad Enouri
- Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ont., Canada
| | | | | |
Collapse
|
31
|
Kur J, Bankhead P, Scholfield CN, Curtis TM, McGeown JG. Ca(2+) sparks promote myogenic tone in retinal arterioles. Br J Pharmacol 2013; 168:1675-86. [PMID: 23126272 DOI: 10.1111/bph.12044] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2012] [Revised: 10/30/2012] [Accepted: 10/30/2012] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND AND PURPOSE Ca(2+) imaging reveals subcellular Ca(2+) sparks and global Ca(2+) waves/oscillations in vascular smooth muscle. It is well established that Ca(2+) sparks can relax arteries, but we have previously reported that sparks can summate to generate Ca(2+) waves/oscillations in unpressurized retinal arterioles, leading to constriction. We have extended these studies to test the functional significance of Ca(2+) sparks in the generation of myogenic tone in pressurized arterioles. EXPERIMENTAL APPROACH Isolated retinal arterioles (25-40 μm external diameter) were pressurized to 70 mmHg, leading to active constriction. Ca(2+) signals were imaged from arteriolar smooth muscle in the same vessels using Fluo4 and confocal laser microscopy. KEY RESULTS Tone development was associated with an increased frequency of Ca(2+) sparks and oscillations. Vasomotion was observed in 40% of arterioles and was associated with synchronization of Ca(2+) oscillations, quantifiable as an increased cross-correlation coefficient. Inhibition of Ca(2+) sparks with ryanodine, tetracaine, cyclopiazonic acid or nimodipine, or following removal of extracellular Ca(2+) , resulted in arteriolar relaxation. Cyclopiazonic acid-induced dilatation was associated with decreased Ca(2+) sparks and oscillations but with a sustained rise in the mean global cytoplasmic [Ca(2+) ] ([Ca(2+) ]c ), as measured using Fura2 and microfluorimetry. CONCLUSIONS AND IMPLICATIONS This study provides direct evidence that Ca(2+) sparks can play an excitatory role in pressurized arterioles, promoting myogenic tone. This contrasts with the generally accepted model in which sparks promote relaxation of vascular smooth muscle. Changes in vessel tone in the presence of cyclopiazonic acid correlated more closely with changes in spark and oscillation frequency than global [Ca(2+) ]c , underlining the importance of frequency-modulated signalling in vascular smooth muscle.
Collapse
Affiliation(s)
- J Kur
- Centre for Vision and Vascular Science, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | | | | | | | | |
Collapse
|
32
|
Saldanha PA, Cairrão E, Maia CJ, Verde I. Long- and short-term effects of androgens in human umbilical artery smooth muscle. Clin Exp Pharmacol Physiol 2013; 40:181-9. [PMID: 23278339 DOI: 10.1111/1440-1681.12047] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 11/25/2012] [Accepted: 12/12/2012] [Indexed: 12/19/2022]
Abstract
The aim of the present study was to determine the effects of androgens in the regulation of human umbilical artery (HUA) contractility. The short-term effects of testosterone on the tone of the HUA were investigated, as were the long-term effects of dihydrotestosterone (DHT) on the expression of some proteins involved in the contractile process. Endothelium-denuded HUA were treated for 24 h with DHT (2 μmol/L) or the vehicle control (ethanol) to analyse the genomic effects of androgens. Twenty-four hour treatment of HUA with DHT increased the mRNA expression of the β(1)-subunit of the large-conductance Ca(2+)-activated (BK(Ca)) channel and decreased expression of the α-subunit of L-type calcium channels. In organ bath studies, testosterone (1-100 μmol/L) produced similar relaxant responses in DHT- and vehicle-treated HUA rings precontracted with 5-HT, histamine and KCl. However, the relaxation response obtained by the combined application of testosterone (100 μmol/L) and nifedipine (10 μmol/L) was significantly greater in DHT- compared with vehicle-treated HUA. The results indicate that the rapid vasorelaxant effects of testosterone that are dependent on both BK(Ca) and voltage-sensitive potassium (K(V)) channel activity in control arteries become dependent solely on K(V) channel activity in DHT-treated HUA. Thus, the present study reveals the importance of the investigation of both the short- and long-term effects of androgens in human arteries.
Collapse
Affiliation(s)
- Paulo A Saldanha
- Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | | | | | | |
Collapse
|
33
|
Zheng YM, Park SW, Stokes L, Tang Q, Xiao JH, Wang YX. Distinct activity of BK channel β1-subunit in cerebral and pulmonary artery smooth muscle cells. Am J Physiol Cell Physiol 2013; 304:C780-9. [PMID: 23426969 DOI: 10.1152/ajpcell.00006.2012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
This study was designed to test a hypothesis that the functional activity of big-conductance, Ca(2+)-activated K(+) (BK) channels is different in cerebral and pulmonary artery smooth muscle cells (CASMCs and PASMCs). Using patch-clamp recordings, we found that the activity of whole cell and single BK channels were significantly higher in CASMCs than in PASMCs. The voltage and Ca(2+) sensitivity of BK channels were greater in CASMCs than in PASMCs. Targeted gene knockout of β(1)-subunits significantly reduced BK currents in CASMCs but had no effect in PASMCs. Western blotting experiments revealed that BK channel α-subunit protein expression level was comparable in CASMCs and PASMCs; however, β(1)-subunit protein expression level was higher in CASMCs than in PASMCs. Inhibition of BK channels by the specific blocker iberiotoxin enhanced norepinephrine-induced increase in intracellular calcium concentration in CASMCs but not in PASMCs. Systemic artery blood pressure was elevated in β(1)(-/-) mice. In contrast, pulmonary artery blood pressure was normal in β(1)(-/-) mice. These findings provide the first evidence that the activity of BK channels is higher in cerebral than in PASMCs. This heterogeneity is primarily determined by the differential β(1)-subunit function and contributes to diverse cellular responses in these two distinct types of cells.
Collapse
Affiliation(s)
- Yun-Min Zheng
- Center for Cardiovascular Sciences, Albany Medical College, 47 New Scotland Ave., Albany, NY 12208, USA.
| | | | | | | | | | | |
Collapse
|
34
|
Yang Y, Sohma Y, Nourian Z, Ella SR, Li M, Stupica A, Korthuis RJ, Davis MJ, Braun AP, Hill MA. Mechanisms underlying regional differences in the Ca2+ sensitivity of BK(Ca) current in arteriolar smooth muscle. J Physiol 2013; 591:1277-93. [PMID: 23297302 DOI: 10.1113/jphysiol.2012.241562] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Abstract β1-Subunits enhance the gating properties of large-conductance Ca(2+)-activated K(+) channels (BKCa) formed by α-subunits. In arterial vascular smooth muscle cells (VSMCs), β1-subunits are vital in coupling SR-generated Ca(2+) sparks to BKCa activation, affecting contractility and blood pressure. Studies in cremaster and cerebral VSMCs show heterogeneity of BKCa activity due to apparent differences in the functional β1-subunit:α-subunit ratio. To define these differences, studies were conducted at the single-channel level while siRNA was used to manipulate specific subunit expression. β1 modulation of the α-subunit Ca(2+) sensitivity was studied using patch-clamp techniques. BKCa channel normalized open probability (NPo) versus membrane potential (Vm) curves were more left-shifted in cerebral versus cremaster VSMCs as cytoplasmic Ca(2+) was raised from 0.5 to 100 μm. Calculated V1/2 values of channel activation decreased from 72.0 ± 6.1 at 0.5 μm Ca(2+)i to -89 ± 9 mV at 100 μm Ca(2+)i in cerebral compared with 101 ± 10 to -63 ± 7 mV in cremaster VSMCs. Cremaster BKCa channels thus demonstrated an ∼2.5-fold weaker apparent Ca(2+) sensitivity such that at a value of Vm of -30 mV, a mean value of [Ca(2+)]i of 39 μm was required to open half of the channels in cremaster versus 16 μm [Ca(2+)]i in cerebral VSMCs. Further, shortened mean open and longer mean closed times were evident in BKCa channel events from cremaster VSMCs at either -30 or 30 mV at any given [Ca(2+)]. β1-Subunit-directed siRNA decreased both the apparent Ca(2+) sensitivity of BKCa in cerebral VSMCs and the appearance of spontaneous transient outward currents. The data are consistent with a higher ratio of β1-subunit:α-subunit of BKCa channels in cerebral compared with cremaster VSMCs. Functionally, this leads both to higher Ca(2+) sensitivity and NPo for BKCa channels in the cerebral vasculature relative to that of skeletal muscle.
Collapse
Affiliation(s)
- Yan Yang
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Moreno-Domínguez A, Colinas O, El-Yazbi A, Walsh EJ, Hill MA, Walsh MP, Cole WC. Ca2+ sensitization due to myosin light chain phosphatase inhibition and cytoskeletal reorganization in the myogenic response of skeletal muscle resistance arteries. J Physiol 2012; 591:1235-50. [PMID: 23230233 DOI: 10.1113/jphysiol.2012.243576] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Abstract The myogenic response of resistance arteries to intravascular pressure elevation is a fundamental physiological mechanism of crucial importance for blood pressure regulation and organ-specific control of blood flow. The importance of Ca(2+) entry via voltage-gated Ca(2+) channels leading to phosphorylation of the 20 kDa myosin regulatory light chains (LC20) in the myogenic response is well established. Recent studies, however, have suggested a role for Ca(2+) sensitization via activation of the RhoA/Rho-associated kinase (ROK) pathway in the myogenic response. The possibility that enhanced actin polymerization is also involved in myogenic vasoconstriction has been suggested. Here, we have used pressurized resistance arteries from rat gracilis and cremaster skeletal muscles to assess the contribution to myogenic constriction of Ca(2+) sensitization due to: (1) phosphorylation of the myosin targeting subunit of myosin light chain phosphatase (MYPT1) by ROK; (2) phosphorylation of the 17 kDa protein kinase C (PKC)-potentiated protein phosphatase 1 inhibitor protein (CPI-17) by PKC; and (3) dynamic reorganization of the actin cytoskeleton evoked by ROK and PKC. Arterial diameter, MYPT1, CPI-17 and LC20 phosphorylation, and G-actin content were determined at varied intraluminal pressures ± H1152, GF109203X or latrunculin B to suppress ROK, PKC and actin polymerization, respectively. The myogenic response was associated with an increase in MYPT1 and LC20 phosphorylation that was blocked by H1152. No change in phospho-CPI-17 content was detected although the PKC inhibitor, GF109203X, suppressed myogenic constriction. Basal LC20 phosphorylation at 10 mmHg was high at ∼40%, increased to a maximal level of ∼55% at 80 mmHg, and exhibited no additional change on further pressurization to 120 and 140 mmHg. Myogenic constriction at 80 mmHg was associated with a decline in G-actin content by ∼65% that was blocked by inhibition of ROK or PKC. Taken together, our findings indicate that two mechanisms of Ca(2+) sensitization (ROK-mediated phosphorylation of MYPT1-T855 with augmentation of LC20 phosphorylation, and a ROK- and PKC-evoked increase in actin polymerization) contribute to force generation in the myogenic response of skeletal muscle arterioles.
Collapse
Affiliation(s)
- Alejandro Moreno-Domínguez
- The Smooth Muscle Research Group, Department of Physiology and Pharmacology, Libin Cardiovascular Institute and Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, 3330 Hospital Drive N.W., Calgary, Alberta, Canada T2N 4N1
| | | | | | | | | | | | | |
Collapse
|
36
|
McCurley A, Pires PW, Bender SB, Aronovitz M, Zhao MJ, Metzger D, Chambon P, Hill MA, Dorrance AM, Mendelsohn ME, Jaffe IZ. Direct regulation of blood pressure by smooth muscle cell mineralocorticoid receptors. Nat Med 2012; 18:1429-33. [PMID: 22922412 PMCID: PMC3491085 DOI: 10.1038/nm.2891] [Citation(s) in RCA: 268] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Accepted: 06/28/2012] [Indexed: 02/07/2023]
Affiliation(s)
- Amy McCurley
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Howitt L, Grayson TH, Morris MJ, Sandow SL, Murphy TV. Dietary obesity increases NO and inhibits BKCa-mediated, endothelium-dependent dilation in rat cremaster muscle artery: association with caveolins and caveolae. Am J Physiol Heart Circ Physiol 2012; 302:H2464-76. [DOI: 10.1152/ajpheart.00965.2011] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Obesity is a risk factor for hypertension and other vascular disease. The aim of this study was to examine the effect of diet-induced obesity on endothelium-dependent dilation of rat cremaster muscle arterioles. Male Sprague-Dawley rats (213 ± 1 g) were fed a cafeteria-style high-fat or control diet for 16–20 wk. Control rats weighed 558 ± 7 g compared with obese rats 762 ± 12 g ( n = 52–56; P < 0.05). Diet-induced obesity had no effect on acetylcholine (ACh)-induced dilation of isolated, pressurized (70 mmHg) arterioles, but sodium nitroprusside (SNP)-induced vasodilation was enhanced. ACh-induced dilation of arterioles from control rats was abolished by a combination of the KCa blockers apamin, 1-[(2-chlorophenyl)diphenylmethyl]-1H-pyrazole (TRAM-34), and iberiotoxin (IBTX; all 0.1 μmol/l), with no apparent role for nitric oxide (NO). In arterioles from obese rats, however, IBTX had no effect on responses to ACh while the NO synthase (NOS)/guanylate cyclase inhibitors Nω-nitro-l-arginine methyl ester (l-NAME; 100 μmol/l)/1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ; 10 μmol/l) partially inhibited ACh-induced dilation. Furthermore, NOS activity (but not endothelial NOS expression) was increased in arteries from obese rats. l-NAME/ODQ alone or removal of the endothelium constricted arterioles from obese but not control rats. Expression of caveolin-1 and -2 oligomers (but not monomers or caveolin-3) was increased in arterioles from obese rats. The number of caveolae was reduced in the endothelium of arteries, and caveolae density was increased at the ends of smooth muscle cells from obese rats. Diet-induced obesity abolished the contribution of large-conductance Ca2+-activated K+ channel to ACh-mediated endothelium-dependent dilation of rat cremaster muscle arterioles, while increasing NOS activity and inducing an NO-dependent component.
Collapse
Affiliation(s)
- Lauren Howitt
- Department of Physiology, School of Medical Sciences, University of New South Wales, Sydney, Australia; and
| | - T. Hilton Grayson
- Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Margaret J. Morris
- Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Shaun L. Sandow
- Department of Physiology, School of Medical Sciences, University of New South Wales, Sydney, Australia; and
- Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Timothy V. Murphy
- Department of Physiology, School of Medical Sciences, University of New South Wales, Sydney, Australia; and
| |
Collapse
|
38
|
Stewart M, Needham M, Bankhead P, Gardiner TA, Scholfield CN, Curtis TM, McGeown JG. Feedback via Ca²⁺-activated ion channels modulates endothelin 1 signaling in retinal arteriolar smooth muscle. Invest Ophthalmol Vis Sci 2012; 53:3059-66. [PMID: 22427579 DOI: 10.1167/iovs.11-9192] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
PURPOSE To investigate the role of feedback by Ca²⁺-sensitive plasma-membrane ion channels in endothelin 1 (Et1) signaling in vitro and in vivo. Methods. Et1 responses were imaged from Fluo-4-loaded smooth muscle in isolated segments of rat retinal arteriole using two-dimensional (2-D) confocal laser microscopy. Vasoconstrictor responses to intravitreal injections of Et1 were recorded in the absence and presence of appropriate ion channel blockers using fluorescein angiograms imaged using a confocal scanning laser ophthalmoscope. Results. Et1 (10 nM) increased both basal [Ca²⁺](i) and the amplitude and frequency of Ca²⁺-waves in retinal arterioles. The Ca²⁺-activated Cl⁻-channel blockers DIDS and 9-anthracene carboxylic acid (9AC) blocked Et1-induced increases in wave frequency, and 9AC also inhibited the increase in amplitude. Iberiotoxin, an inhibitor of large conductance (BK) Ca²⁺-activated K⁺-channels, increased wave amplitude in the presence of Et1 but had no effect on frequency. None of these drugs affected basal [Ca²⁺](i). The voltage-operated Ca²⁺-channel inhibitor nimodipine inhibited wave frequency and amplitude and also lowered basal [Ca²⁺](i) in the presence of Et1. Intravitreal injection of Et1 caused retinal arteriolar vasoconstriction. This was inhibited by DIDS but not by iberiotoxin or penitrem A, another BK-channel inhibitor. Conclusions. Et1 evokes increases in the frequency of arteriolar Ca²⁺-waves in vitro, resulting in vasoconstriction in vivo. These responses, initiated by release of stored Ca²⁺, also require positive feedback via Ca²⁺-activated Cl⁻-channels and L-type Ca²⁺-channels.
Collapse
Affiliation(s)
- Michael Stewart
- Centre for Vision and Vascular Science, Queen's University of Belfast, Institute of Clinical Sciences, The Royal Victoria Hospital, Belfast, Northern Ireland
| | | | | | | | | | | | | |
Collapse
|
39
|
Asano S, Bratz IN, Berwick ZC, Fancher IS, Tune JD, Dick GM. Penitrem A as a tool for understanding the role of large conductance Ca(2+)/voltage-sensitive K(+) channels in vascular function. J Pharmacol Exp Ther 2012; 342:453-60. [PMID: 22580348 DOI: 10.1124/jpet.111.191072] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Large conductance, Ca(2+)/voltage-sensitive K(+) channels (BK channels) are well characterized, but their physiological roles, often determined through pharmacological manipulation, are less clear. Iberiotoxin is considered the "gold standard" antagonist, but cost and membrane-impermeability limit its usefulness. Economical and membrane-permeable alternatives could facilitate the study of BK channels. Thus, we characterized the effect of penitrem A, a tremorigenic mycotoxin, on BK channels and demonstrate its utility for studying vascular function in vitro and in vivo. Whole-cell currents from human embryonic kidney 293 cells transfected with hSlo α or α + β1 were blocked >95% by penitrem A (IC(50) 6.4 versus 64.4 nM; p < 0.05). Furthermore, penitrem A inhibited BK channels in inside-out and cell-attached patches, whereas iberiotoxin could not. Inhibitory effects of penitrem A on whole-cell K(+) currents were equivalent to iberiotoxin in canine coronary smooth muscle cells. As for specificity, penitrem A had no effect on native delayed rectifier K(+) currents, cloned voltage-dependent Kv1.5 channels, or native ATP-dependent K(ATP) current. Penitrem A enhanced the sensitivity to K(+)-induced contraction in canine coronary arteries by 23 ± 5% (p < 0.05) and increased the blood pressure response to phenylephrine in anesthetized mice by 36 ± 11% (p < 0.05). Our data indicate that penitrem A is a useful tool for studying the role of BK channels in vascular function and is practical for cell and tissue (in vitro) studies as well as anesthetized animal (in vivo) experiments.
Collapse
Affiliation(s)
- Shinichi Asano
- Division of Exercise Physiology, Center for Cardiovascular and Respiratory Sciences, West Virginia University School of Medicine, Morgantown, West Virginia 26506, USA
| | | | | | | | | | | |
Collapse
|
40
|
Abstract
Research over the last decade has uncovered roles for bile acids (BAs) that extend beyond their traditional functions in regulating lipid digestion and cholesterol metabolism. BAs are now recognized as signaling molecules that interact with both plasma membrane and nuclear receptors. Emerging evidence indicates that by interacting with these receptors, BAs regulate their own synthesis, glucose and energy homeostasis, and other important physiological events. Herein, we provide a comprehensive review of the actions of BAs on cardiovascular function. In the heart and the systemic circulation, BAs interact with plasma membrane G-protein-coupled receptors, for example, TGR5 and muscarinic receptors, and nuclear receptors, for example, the farnesoid (FXR) and pregnane (PXR) xenobiotic receptors. BA receptors are expressed in cardiovascular tissue, however, the mechanisms underlying BA-mediated regulation of cardiovascular function remain poorly understood. BAs reduce heart rate by regulating channel conductance and calcium dynamics in sino-atrial and ventricular cardiomyocytes and regulate vascular tone via both endothelium-dependent and -independent mechanisms. End-stage liver disease, obstructive jaundice, and intrahepatic cholestasis of pregnancy are prominent conditions in which elevated serum BAs alter vascular dynamics. This review focuses on BAs as newly recognized signaling molecules that modulate cardiovascular function.
Collapse
Affiliation(s)
- Sandeep Khurana
- Division of Gastroenterology and Hepatology, VA Maryland Health Care System and University of Maryland School of Medicine, Baltimore, MD, USA.
| | | | | |
Collapse
|
41
|
Lifshitz LM, Carmichael JD, Lai FA, Sorrentino V, Bellvé K, Fogarty KE, ZhuGe R. Spatial organization of RYRs and BK channels underlying the activation of STOCs by Ca(2+) sparks in airway myocytes. ACTA ACUST UNITED AC 2011; 138:195-209. [PMID: 21746845 PMCID: PMC3149436 DOI: 10.1085/jgp.201110626] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Short-lived, localized Ca2+ events mediate Ca2+ signaling with high efficiency and great fidelity largely as a result of the close proximity between Ca2+-permeable ion channels and their molecular targets. However, in most cases, direct evidence of the spatial relationship between these two types of molecules is lacking, and, thus, mechanistic understanding of local Ca2+ signaling is incomplete. In this study, we use an integrated approach to tackling this issue on a prototypical local Ca2+ signaling system composed of Ca2+ sparks resulting from the opening of ryanodine receptors (RYRs) and spontaneous transient outward currents (STOCs) caused by the opening of Ca2+-activated K+ (BK) channels in airway smooth muscle. Biophysical analyses of STOCs and Ca2+ sparks acquired at 333 Hz demonstrate that these two events are associated closely in time, and approximately eight RYRs open to give rise to a Ca2+ spark, which activates ∼15 BK channels to generate a STOC at 0 mV. Dual immunocytochemistry and 3-D deconvolution at high spatial resolution reveal that both RYRs and BK channels form clusters and RYR1 and RYR2 (but not RYR3) localize near the membrane. Using the spatial relationship between RYRs and BK channels, the spatial-temporal profile of [Ca2+] resulting from Ca2+ sparks, and the kinetic model of BK channels, we estimate that an average Ca2+ spark caused by the opening of a cluster of RYR1 or RYR2 acts on BK channels from two to three clusters that are randomly distributed within an ∼600-nm radius of RYRs. With this spatial organization of RYRs and BK channels, we are able to model BK channel currents with the same salient features as those observed in STOCs across a range of physiological membrane potentials. Thus, this study provides a mechanistic understanding of the activation of STOCs by Ca2+ sparks using explicit knowledge of the spatial relationship between RYRs (the Ca2+ source) and BK channels (the Ca2+ target).
Collapse
Affiliation(s)
- Lawrence M Lifshitz
- Biomedical Imaging Group, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Role of myosin light chain kinase and myosin light chain phosphatase in the resistance arterial myogenic response to intravascular pressure. Arch Biochem Biophys 2011; 510:160-73. [DOI: 10.1016/j.abb.2011.02.024] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Revised: 02/24/2011] [Accepted: 02/28/2011] [Indexed: 12/19/2022]
|
43
|
Howitt L, Sandow SL, Grayson TH, Ellis ZE, Morris MJ, Murphy TV. Differential effects of diet-induced obesity on BKCa {beta}1-subunit expression and function in rat skeletal muscle arterioles and small cerebral arteries. Am J Physiol Heart Circ Physiol 2011; 301:H29-40. [PMID: 21536854 DOI: 10.1152/ajpheart.00134.2011] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mechanisms underlying obesity-related vascular dysfunction are unclear. This study examined the effect of diet-induced obesity on expression and function of large conductance Ca(2+)-activated potassium channel (BK(Ca)) in rat pressurized small resistance vessels with myogenic tone. Male Sprague-Dawley rats fed a cafeteria-style high fat diet (HFD; ∼30% energy from fat) for 16-20 wk were ∼30% heavier than controls fed standard chow (∼13% fat). Obesity did not alter BK(Ca) α-subunit function or α-subunit protein or mRNA expression in vessels isolated from the cremaster muscle or middle-cerebral circulations. In contrast, BK(Ca) β(1)-subunit protein expression and function were significantly reduced in cremaster muscle arterioles but increased in middle-cerebral arteries from obese animals. Immunohistochemistry showed α- and β(1)-subunits were present exclusively in the smooth muscle of both vessels. Cremaster muscle arterioles from obese animals showed significantly increased medial thickness, and media-to-lumen ratio and pressurized arterioles showed increased myogenic tone at 30 mmHg, but not at 50-120 mmHg. Myogenic tone was not affected by obesity in middle-cerebral arteries. The BK(Ca) antagonist iberiotoxin constricted both cremaster muscle and middle-cerebral arterioles from control rats; this effect of iberiotoxin was abolished in cremaster muscle arteries only from obese rats. Diet-induced obesity has contrasting effects on BK(Ca) function in different vascular beds, through differential effects on β(1)-subunit expression. However, these alterations in BK(Ca) function had little effect on overall myogenic tone, suggesting that the mechanisms controlling myogenic tone can be altered and compensate for altered BK(Ca) expression and function.
Collapse
Affiliation(s)
- Lauren Howitt
- Dept. of Physiology, School of Medical Sciences, Univ. of New South Wales, Sydney, NSW 2052 Australia.
| | | | | | | | | | | |
Collapse
|
44
|
Leffler CW, Parfenova H, Jaggar JH. Carbon monoxide as an endogenous vascular modulator. Am J Physiol Heart Circ Physiol 2011; 301:H1-H11. [PMID: 21498777 DOI: 10.1152/ajpheart.00230.2011] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Carbon monoxide (CO) is produced by heme oxygenase (HO)-catalyzed heme degradation to CO, iron, and biliverdin. HO has two active isoforms, HO-1 (inducible) and HO-2 (constitutive). HO-2, but not HO-1, is highly expressed in endothelial and smooth muscle cells and in adjacent astrocytes in the brain. HO-1 is expressed basally only in the spleen and liver but can be induced to a varying extent in most tissues. Elevating heme, protein phosphorylation, Ca(2+) influx, and Ca(2+)/calmodulin-dependent processes increase HO-2 activity. CO dilates cerebral arterioles and may constrict or dilate skeletal muscle and renal arterioles. Selected vasodilatory stimuli, including seizures, glutamatergic stimulation, hypoxia, hypotension, and ADP, increase CO, and the inhibition of HO attenuates the dilation to these stimuli. Astrocytic HO-2-derived CO causes glutamatergic dilation of pial arterioles. CO dilates by activating smooth muscle cell large-conductance Ca(2+)-activated K(+) (BK(Ca)) channels. CO binds to BK(Ca) channel-bound heme, leading to an increase in Ca(2+) sparks-to-BK(Ca) channel coupling. Also, CO may bind directly to the BK(Ca) channel at several locations. Endothelial nitric oxide and prostacyclin interact with HO/CO in circulatory regulation. In cerebral arterioles in vivo, in contrast to dilation to acute CO, a prolonged exposure of cerebral arterioles to elevated CO produces progressive constriction by inhibiting nitric oxide synthase. The HO/CO system is highly protective to the vasculature. CO suppresses apoptosis and inhibits components of endogenous oxidant-generating pathways. Bilirubin is a potent reactive oxygen species scavenger. Still many questions remain about the physiology and biochemistry of HO/CO in the circulatory system and about the function and dysfunction of this gaseous mediator system.
Collapse
|
45
|
Westcott EB, Jackson WF. Heterogeneous function of ryanodine receptors, but not IP3 receptors, in hamster cremaster muscle feed arteries and arterioles. Am J Physiol Heart Circ Physiol 2011; 300:H1616-30. [PMID: 21357503 DOI: 10.1152/ajpheart.00728.2010] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The roles played by ryanodine receptors (RyRs) and inositol 1,4,5-trisphosphate receptors (IP₃Rs) in vascular smooth muscle in the microcirculation remain unclear. Therefore, the function of both RyRs and IP₃Rs in Ca(²+) signals and myogenic tone in hamster cremaster muscle feed arteries and downstream arterioles were assessed using confocal imaging and pressure myography. Feed artery vascular smooth muscle displayed Ca(²+) sparks and Ca(²+) waves, which were inhibited by the RyR antagonists ryanodine (10 μM) or tetracaine (100 μM). Despite the inhibition of sparks and waves, ryanodine or tetracaine increased global intracellular Ca(²+) and constricted the arteries. The blockade of IP₃Rs with xestospongin D (5 μM) or 2-aminoethoxydiphenyl borate (100 μM) or the inhibition of phospholipase C using U-73122 (10 μM) also attenuated Ca(2+) waves without affecting Ca(²+) sparks. Importantly, the IP₃Rs and phospholipase C antagonists decreased global intracellular Ca(2+) and dilated the arteries. In contrast, cremaster arterioles displayed only Ca(²+) waves: Ca(²+) sparks were not observed, and neither ryanodine (10-50 μM) nor tetracaine (100 μM) affected either Ca(²+) signals or arteriolar tone despite the presence of functional RyRs as assessed by responses to the RyR agonist caffeine (10 mM). As in feed arteries, arteriolar Ca(²+) waves were attenuated by xestospongin D (5 μM), 2-aminoethoxydiphenyl borate (100 μM), and U-73122 (10 μM), accompanied by decreased global intracellular Ca(²+) and vasodilation. These findings highlight the contrasting roles played by RyRs and IP₃Rs in Ca(²+) signals and myogenic tone in feed arteries and demonstrate important differences in the function of RyRs between feed arteries and downstream arterioles.
Collapse
Affiliation(s)
- Erika B Westcott
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, USA.
| | | |
Collapse
|
46
|
Ella SR, Yang Y, Clifford PS, Gulia J, Dora KA, Meininger GA, Davis MJ, Hill MA. Development of an image-based system for measurement of membrane potential, intracellular Ca(2+) and contraction in arteriolar smooth muscle cells. Microcirculation 2011; 17:629-40. [PMID: 21044217 DOI: 10.1111/j.1549-8719.2010.00059.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Changes in smooth muscle cell (SMC) membrane potential (Em) are critical to vasomotor responses. As a fluorescent indicator approach would lessen limitations of glass electrodes in contracting preparations, we aimed to develop a Forster (or fluorescence) resonance energy transfer (FRET)-based measurement for Em. METHODS The FRET pair used in this study (donor CC2-DMPE [excitation 405 nm] and acceptor DisBAC(4) (3)) provide rapid measurements at a sensitivity not achievable with many ratiometric indicators. The method also combined measurement of changes in Ca(2+) (i) using fluo-4 and excitation at 490 nm. RESULTS After establishing loading conditions, a linear relationship was demonstrated between Em and fluorescence signal in FRET dye-loaded HEK cells held under voltage clamp. Over the voltage range from -70 to +30 mV, slope (of FRET signal vs. voltage, m) = 0.49 ± 0.07, r(2) = 0.96 ± 0.025. Similar data were obtained in cerebral artery SMCs, slope (m) = 0.30 ± 0.02, r(2) = 0.98 ± 0.02. Change in FRET emission ratio over the holding potential of -70 to +30 mV was 41.7 ± 4.9% for HEK cells and 30.0 ± 2.3% for arterial SMCs. The FRET signal was also shown to be modulated by KCl-induced depolarization in a concentration-dependent manner. Further, in isolated arterial SMCs, KCl-induced depolarization (60 mM) measurements occurred with increased fluo-4 fluorescence emission (62 ± 9%) and contraction (-27 ± 4.2%). CONCLUSIONS The data support the FRET-based approach for measuring changes in Em in arterial SMCs. Further, image-based measurements of Em can be combined with analysis of temporal changes in Ca(2+) (i) and contraction.
Collapse
Affiliation(s)
- Srikanth R Ella
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Zhong XZ, Abd-Elrahman KS, Liao CH, El-Yazbi AF, Walsh EJ, Walsh MP, Cole WC. Stromatoxin-sensitive, heteromultimeric Kv2.1/Kv9.3 channels contribute to myogenic control of cerebral arterial diameter. J Physiol 2010; 588:4519-37. [PMID: 20876197 DOI: 10.1113/jphysiol.2010.196618] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Cerebral vascular smooth muscle contractility plays a crucial role in controlling arterial diameter and, thereby, blood flow regulation in the brain. A number of K(+) channels have been suggested to contribute to the regulation of diameter by controlling smooth muscle membrane potential (E(m)) and Ca(2+) influx. Previous studies indicate that stromatoxin (ScTx1)-sensitive, Kv2-containing channels contribute to the control of cerebral arterial diameter at 80 mmHg, but their precise role and molecular composition were not determined. Here, we tested if Kv2 subunits associate with 'silent' subunits from the Kv5, Kv6, Kv8 or Kv9 subfamilies to form heterotetrameric channels that contribute to control of diameter of rat middle cerebral arteries (RMCAs) over a range of intraluminal pressure from 10 to 100 mmHg. The predominant mRNAs expressed by RMCAs encode Kv2.1 and Kv9.3 subunits. Co-localization of Kv2.1 and Kv9.3 proteins at the plasma membrane of dissociated single RMCA myocytes was detected by proximity ligation assay. ScTx1-sensitive native current of RMCA myocytes and Kv2.1/Kv9.3 currents exhibited functional identity based on the similarity of their deactivation kinetics and voltage dependence of activation that were distinct from those of homomultimeric Kv2.1 channels. ScTx1 treatment enhanced the myogenic response of pressurized RMCAs between 40 and 100 mmHg, but this toxin also caused constriction between 10 and 40 mmHg that was not previously observed following inhibition of large conductance Ca(2+)-activated K(+) (BK(Ca)) and Kv1 channels. Taken together, this study defines the molecular basis of Kv2-containing channels and contributes to our understanding of the functional significance of their expression in cerebral vasculature. Specifically, our findings provide the first evidence of heteromultimeric Kv2.1/Kv9.3 channel expression in RMCA myocytes and their distinct contribution to control of cerebral arterial diameter over a wider range of E(m) and transmural pressure than Kv1 or BK(Ca) channels owing to their negative range of voltage-dependent activation.
Collapse
Affiliation(s)
- Xi Zoë Zhong
- The Smooth Muscle Research Group, Department of Physiology and Pharmacology, Faculty of Medicine, University of Calgary, 3330 Hospital Drive N.W., Calgary, Alberta, Canada T2N 4N1
| | | | | | | | | | | | | |
Collapse
|
48
|
Zuidema MY, Yang Y, Wang M, Kalogeris T, Liu Y, Meininger CJ, Hill MA, Davis MJ, Korthuis RJ. Antecedent hydrogen sulfide elicits an anti-inflammatory phenotype in postischemic murine small intestine: role of BK channels. Am J Physiol Heart Circ Physiol 2010; 299:H1554-67. [PMID: 20833953 DOI: 10.1152/ajpheart.01229.2009] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The objectives of this study were to determine the role of calcium-activated, small (SK), intermediate (IK), and large (BK) conductance potassium channels in initiating the development of an anti-inflammatory phenotype elicited by preconditioning with an exogenous hydrogen sulfide (H(2)S) donor, sodium hydrosulfide (NaHS). Intravital microscopy was used to visualize rolling and firmly adherent leukocytes in vessels of the small intestine of mice preconditioned with NaHS (in the absence and presence of SK, IK, and BK channel inhibitors, apamin, TRAM-34, and paxilline, respectively) or SK/IK (NS-309) or BK channel activators (NS-1619) 24 h before ischemia-reperfusion (I/R). I/R induced marked increases in leukocyte rolling and adhesion, effects that were largely abolished by preconditioning with NaHS, NS-309, or NS-1619. The postischemic anti-inflammatory effects of NaHS-induced preconditioning were mitigated by BKB channel inhibitor treatment coincident with NaHS, but not by apamin or TRAM-34, 24 h before I/R. Confocal imaging and immunohistochemistry were used to demonstrate the presence of BKα subunit staining in both endothelial and vascular smooth muscle cells of isolated, pressurized mesenteric venules. Using patch-clamp techniques, we found that BK channels in cultured endothelial cells were activated after exposure to NaHS. Bath application of the same concentration of NaHS used in preconditioning protocols led to a rapid increase in a whole cell K(+) current; specifically, the component of K(+) current blocked by the selective BK channel antagonist iberiotoxin. The activation of BK current by NaHS could also be demonstrated in single channel recording mode where it was independent of a change in intracellular Ca(+) concentration. Our data are consistent with the concept that H(2)S induces the development of an anti-adhesive state in I/R in part mediated by a BK channel-dependent mechanism.
Collapse
Affiliation(s)
- Mozow Y Zuidema
- Department of Medical Pharmacology and Physiology, Dalton Cardiovascular Research Center, University of Missouri School of Medicine, Columbia, Missouri 65212, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Zhong XZ, Harhun MI, Olesen SP, Ohya S, Moffatt JD, Cole WC, Greenwood IA. Participation of KCNQ (Kv7) potassium channels in myogenic control of cerebral arterial diameter. J Physiol 2010; 588:3277-93. [PMID: 20624791 PMCID: PMC2976022 DOI: 10.1113/jphysiol.2010.192823] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2010] [Accepted: 07/09/2010] [Indexed: 01/11/2023] Open
Abstract
KCNQ gene expression was previously shown in various rodent blood vessels, where the products of KCNQ4 and KCNQ5, Kv7.4 and Kv7.5 potassium channel subunits, respectively, have an influence on vascular reactivity. The aim of this study was to determine if small cerebral resistance arteries of the rat express KCNQ genes and whether Kv7 channels participate in the regulation of myogenic control of diameter. Quantitative reverse transcription polymerase chain reaction (QPCR) was undertaken using RNA isolated from rat middle cerebral arteries (RMCAs) and immunocytochemistry was performed using Kv7 subunit-specific antibodies and freshly isolated RMCA myocytes. KCNQ4 message was more abundant than KCNQ5 = KCNQ1, but KCNQ2 and KCNQ3 message levels were negligible. Kv7.1, Kv7.4 and Kv7.5 immunoreactivity was present at the sarcolemma of freshly isolated RMCA myocytes. Linopirdine (1 microm) partially depressed, whereas the Kv7 activator S-1 (3 and/or 20 microm) enhanced whole-cell Kv7.4 (in HEK 293 cells), as well as native RMCA myocyte Kv current amplitude. The effects of S-1 were voltage-dependent, with progressive loss of stimulation at potentials of >15 mV. At the concentrations employed linopirdine and S-1 did not alter currents due to recombinant Kv1.2/Kv1.5 or Kv2.1/Kv9.3 channels (in HEK 293 cells) that are also expressed by RMCA myocytes. In contrast, another widely used Kv7 blocker, XE991 (10 microm), significantly attenuated native Kv current and also reduced Kv1.2/Kv1.5 and Kv2.1/Kv9.3 currents. Pressurized arterial myography was performed using RMCAs exposed to intravascular pressures of 10-100 mmHg. Linopirdine (1 microm) enhanced the myogenic response at 20 mmHg, whereas the activation of Kv7 channels with S-1 (20 microm) inhibited myogenic constriction at >20 mmHg and reversed the increased myogenic response produced by suppression of Kv2-containing channels with 30 nm stromatoxin (ScTx1). These data reveal a novel contribution of KCNQ gene products to the regulation of myogenic control of cerebral arterial diameter and suggest that Kv7 channel activating drugs may be appropriate candidates for the development of an effective therapy to ameliorate cerebral vasospasm.
Collapse
Affiliation(s)
- Xi Zoë Zhong
- Ion Channels and Cell Signaling Centre, Division of Basic Medical Sciences, St George's University of London, London SW17 0RE, UK
| | | | | | | | | | | | | |
Collapse
|
50
|
Mufti RE, Brett SE, Tran CHT, Abd El-Rahman R, Anfinogenova Y, El-Yazbi A, Cole WC, Jones PP, Chen SRW, Welsh DG. Intravascular pressure augments cerebral arterial constriction by inducing voltage-insensitive Ca2+ waves. J Physiol 2010; 588:3983-4005. [PMID: 20736418 DOI: 10.1113/jphysiol.2010.193300] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
This study examined whether elevated intravascular pressure stimulates asynchronous Ca(2+) waves in cerebral arterial smooth muscle cells and if their generation contributes to myogenic tone development. The endothelium was removed from rat cerebral arteries, which were then mounted in an arteriograph, pressurized (20-100 mmHg) and examined under a variety of experimental conditions. Diameter and membrane potential (V(M)) were monitored using conventional techniques; Ca(2+) wave generation and myosin light chain (MLC(20))/MYPT1 (myosin phosphatase targeting subunit) phosphorylation were assessed by confocal microscopy and Western blot analysis, respectively. Elevating intravascular pressure increased the proportion of smooth muscle cells firing asynchronous Ca(2+) waves as well as event frequency. Ca(2+) wave augmentation occurred primarily at lower intravascular pressures (<60 mmHg) and ryanodine, a plant alkaloid that depletes the sarcoplasmic reticulum (SR) of Ca(2+), eliminated these events. Ca(2+) wave generation was voltage insensitive as Ca(2+) channel blockade and perturbations in extracellular [K(+)] had little effect on measured parameters. Ryanodine-induced inhibition of Ca(2+) waves attenuated myogenic tone and MLC(20) phosphorylation without altering arterial V(M). Thapsigargin, an SR Ca(2+)-ATPase inhibitor also attenuated Ca(2+) waves, pressure-induced constriction and MLC(20) phosphorylation. The SR-driven component of the myogenic response was proportionally greater at lower intravascular pressures and subsequent MYPT1 phosphorylation measures revealed that SR Ca(2+) waves facilitated pressure-induced MLC(20) phosphorylation through mechanisms that include myosin light chain phosphatase inhibition. Cumulatively, our findings show that mechanical stimuli augment Ca(2+) wave generation in arterial smooth muscle and that these transient events facilitate tone development particularly at lower intravascular pressures by providing a proportion of the Ca(2+) required to directly control MLC(20) phosphorylation.
Collapse
Affiliation(s)
- Rania E Mufti
- Hotchkiss Brain Institute, Libin Cardiovascular Institute, Department of Physiology & Pharmacology, University of Calgary, Alberta, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|