1
|
Moraes RA, Brito DS, Araujo FA, Jesus RLC, Silva LB, Lima GBDC, Sá DS, Silva da Silva CD, Pernomian L, Wenceslau CF, Silva DF. NONO2P, a nitric oxide donor, induces relaxation in coronary artery, negative inotropism and hypotensive effect in rats. Biochem Pharmacol 2025; 236:116918. [PMID: 40158820 DOI: 10.1016/j.bcp.2025.116918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 03/21/2025] [Accepted: 03/27/2025] [Indexed: 04/02/2025]
Abstract
Reduced NO synthesis and/or bioavailability is related with many cardiovascular diseases, such as coronary artery disease and hypertension. This study aimed to evaluate the effects of cis-[Ru(NO)(NO2)(phen)2](PF6)2-(NONO2P) on blood pressure in normotensive and hypertensive rats. Specifically, we wanted to investigate its action on the atrial contractility, mesenteric and coronary arteries function. Male Wistar and spontaneously hypertensive rats (SHR) (13-18 weeks old) were used to assess the NONO2P effects on blood pressure and heart rate. Superior mesenteric and coronary arteries, and atria were isolated for recording to analyze force changes. Cultured endothelial cells were used to measure intracellular reactive oxygen species (ROS) generation using fluorescent dye (dihydroethidium, DHE). Acute administration of NONO2P induced hypotension in non-anesthetized normotensive and hypertensive rats. Moreover, NONO2P caused a negative inotropic effect without altering cardiac rhythmicity. Further, NONO2P displays a vasorelaxant effect on different blood vessels (mesenteric and coronary arteries). For comparison purposes, we observed that NONO2P and NTG presented with a similar potency and maximum response values in inducing relaxation in coronary arteries. On the other hand, mesenteric arteries were more sensitive to both donors, NONO2P and NTG, than the coronary artery. In addition, exposure to NONO2P induced tolerance and increased ROS levels. This is the first evidence that NONO2P induces hypotension, negative cardiac inotropism and coronary artery relaxation. In addition, pre-exposure to NONO2P induces vascular tolerance. Overall, these results may shed light on the potential therapeutic use of NONO2P, particularly in treating angina and hypertensive crises.
Collapse
Affiliation(s)
- Raiana A Moraes
- Laboratory of Cardiovascular Physiology and Pharmacology, Bioregulation Department, Federal University of Bahia, Salvador, Bahia, Brazil; Gonçalo Moniz Institute, FIOCRUZ, Salvador, BA, Brazil
| | - Daniele S Brito
- Laboratory of Cardiovascular Physiology and Pharmacology, Bioregulation Department, Federal University of Bahia, Salvador, Bahia, Brazil; Gonçalo Moniz Institute, FIOCRUZ, Salvador, BA, Brazil
| | - Fênix A Araujo
- Laboratory of Cardiovascular Physiology and Pharmacology, Bioregulation Department, Federal University of Bahia, Salvador, Bahia, Brazil; Gonçalo Moniz Institute, FIOCRUZ, Salvador, BA, Brazil
| | - Rafael L C Jesus
- Laboratory of Cardiovascular Physiology and Pharmacology, Bioregulation Department, Federal University of Bahia, Salvador, Bahia, Brazil
| | - Liliane B Silva
- Laboratory of Cardiovascular Physiology and Pharmacology, Bioregulation Department, Federal University of Bahia, Salvador, Bahia, Brazil
| | - Gabriela B de C Lima
- Laboratory of Cardiovascular Physiology and Pharmacology, Bioregulation Department, Federal University of Bahia, Salvador, Bahia, Brazil
| | - Denise S Sá
- Federal Institute of Bahia, Salvador, BA, Brazil
| | | | - Laena Pernomian
- Department of Cell Biology and Anatomy, University of South Carolina, Columbia, SC, USA; Cardiovascular Translational Research Center, University of South Carolina, Columbia, SC, USA
| | - Camilla F Wenceslau
- Department of Cell Biology and Anatomy, University of South Carolina, Columbia, SC, USA; Cardiovascular Translational Research Center, University of South Carolina, Columbia, SC, USA
| | - Darizy F Silva
- Laboratory of Cardiovascular Physiology and Pharmacology, Bioregulation Department, Federal University of Bahia, Salvador, Bahia, Brazil; Gonçalo Moniz Institute, FIOCRUZ, Salvador, BA, Brazil.
| |
Collapse
|
2
|
Manville RW, Nguyen HA, Abbott GW. Comment on "The quercetin metabolite 4-methylcatechol causes vasodilation via voltage-gated potassium (Kv) channels" by Patrícia Dias, Rudy Salam, Jana Pourová, Marie Vopršalová, Lukáš Konečný, Eduard Jirkovský, Jurjen Duintjer Tebbens and Přemysl Mladěnka, Food & Function, 2024, 15, 11047. Food Funct 2025; 16:2909-2913. [PMID: 40094416 DOI: 10.1039/d4fo06086e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Voltage-gated potassium (Kv) channels in the Kv1 and Kv7 gene families are expressed in vascular smooth muscle, where they can modulate cellular excitability and potentially act as targets for antihypertensive drugs and botanicals. In an article recently published in Food & Function, Dias and colleagues concluded that "in silico reverse docking confirmed that 4-MC [4-methylcatechol] binds to Kv7.4" and that "4-MC exerts vasorelaxation by opening Kv channels with the involvement of Kv7.4". Here, we tested their conclusions using heterologous expression experiments, an important step in target validation, and found that Kv7.4 is in fact insensitive to 4-MC.
Collapse
Affiliation(s)
- Rían W Manville
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA.
| | - Hanh A Nguyen
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA.
| | - Geoffrey W Abbott
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA.
| |
Collapse
|
3
|
Manville RW, Baldwin SN, Schaub OH, Jepps TA, Abbott GW. Vasorelaxant effects of 3-methoxycatechol are not via direct activation of voltage-gated potassium channels. Vascul Pharmacol 2025; 159:107471. [PMID: 39933640 DOI: 10.1016/j.vph.2025.107471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 02/02/2025] [Accepted: 02/04/2025] [Indexed: 02/13/2025]
Affiliation(s)
- Rían W Manville
- Bioelectricity Laboratory, Dept. of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA
| | - Samuel N Baldwin
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Olivia H Schaub
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas A Jepps
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Geoffrey W Abbott
- Bioelectricity Laboratory, Dept. of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, USA.
| |
Collapse
|
4
|
Lemke J, Gollasch M, Tsvetkov D, Schulig L. Advances in the design and development of chemical modulators of the voltage-gated potassium channels K V7.4 and K V7.5. Expert Opin Drug Discov 2025; 20:47-62. [PMID: 39627683 DOI: 10.1080/17460441.2024.2438226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 10/20/2024] [Accepted: 12/02/2024] [Indexed: 12/10/2024]
Abstract
INTRODUCTION Hypertension remains a major public health concern, with significant morbidity and mortality worldwide. Despite the availability of various antihypertensive medications, blood pressure control remains suboptimal in many individuals. During the last decades, KV7.4 and KV7.5, which were already known from the view of neuronal regulation, emerged as possible important players in the regulation of vascular tone and blood pressure. AREAS COVERED This review covers physiological functions and current advancements in the development of KV7.4 and KV7.5 channel modulators. The authors highlight the structural elements likely to be important for the future design of KV7 subtype-selective modulators, underscoring their potential as an innovative hypertension treatment. EXPERT OPINION Extensive research has been focused on targeting neuronal KV7.2 and KV7.3 channels, while KV7.4 and KV7.5 attracted less attention. Many of the developed compounds represent derivatives of flupirtine or retigabine, whereby subtype channel selectivity has only been demonstrated for a handful of individual compounds. Novel substances address additional sites within the binding pocket by incorporating new functional groups. A comprehensive and systematic evaluation of a compound set with significant subtype selectivity should be performed. The discovery of new highly active, less toxic, and selective compounds, therefore, remains the goal of further research in the coming years.
Collapse
Affiliation(s)
- Jana Lemke
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, University of Greifswald, Greifswald, Germany
| | - Maik Gollasch
- Department of Internal Medicine and Geriatrics, University Medicine, Greifswald, Germany
| | - Dmitry Tsvetkov
- Department of Internal Medicine and Geriatrics, University Medicine, Greifswald, Germany
| | - Lukas Schulig
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, University of Greifswald, Greifswald, Germany
| |
Collapse
|
5
|
Dias P, Salam R, Pourová J, Vopršalová M, Konečný L, Jirkovský E, Duintjer Tebbens J, Mladěnka P. The quercetin metabolite 4-methylcatechol causes vasodilation via voltage-gated potassium (K V) channels. Food Funct 2024; 15:11047-11059. [PMID: 39422021 DOI: 10.1039/d3fo04672a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Dietary polyphenols have been associated with many beneficial cardiovascular effects. However, these effects are rather attributed to small phenolic metabolites formed by the gut microbiota, which reach sufficient concentrations in systemic circulation. 4-Methylcatechol (4-MC) is one such metabolite. As it is shown to possess considerable vasorelaxant effects, this study aimed to unravel its mechanism of action. To this end, experimental in vitro and in silico approaches were employed. In the first step, isometric tension recordings were performed on rat aortic rings. 4-MC potentiated the effect of cyclic nucleotides, but the effect was not mediated by either soluble guanylyl cyclase (sGC), modification of cyclic adenosine monophosphate levels, or protein kinase G. Hence, downstream targets such as calcium or potassium channels were considered. Inhibition of voltage-gated K+ channels (KV) markedly decreased the effect of 4-MC, and vasodilation was partly decreased by inhibition of the KV7 isoform. Contrarily, other types of K+ channels or L-type Ca2+ channels were not involved. In silico reverse docking confirmed that 4-MC binds to KV7.4 through hydrogen bonding and hydrophobic interactions. In particular, it interacts with two crucial residues for KV7.4 activation: Trp242 and Phe246. In summary, our findings suggested that 4-MC exerts vasorelaxation by opening KV channels with the involvement of KV7.4.
Collapse
Affiliation(s)
- Patrícia Dias
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University, Hradec Kralove, 500 05, Czech Republic.
- Division of Outcomes & Translational Sciences, Pelotonia Research Center, The Ohio State University, 2255 Kenny Rd, Columbus, OH, USA
| | - Rudy Salam
- Department of Biophysics and Physical Chemistry, Faculty of Pharmacy, Charles University, Hradec Kralove, 500 05, Czech Republic
- Department of Pharmacy, Faculty of Medicine, Brawijaya University, Malang, Indonesia
| | - Jana Pourová
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University, Hradec Kralove, 500 05, Czech Republic.
| | - Marie Vopršalová
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University, Hradec Kralove, 500 05, Czech Republic.
| | - Lukáš Konečný
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University, Hradec Kralove, 500 05, Czech Republic.
| | - Eduard Jirkovský
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University, Hradec Kralove, 500 05, Czech Republic.
| | - Jurjen Duintjer Tebbens
- Department of Biophysics and Physical Chemistry, Faculty of Pharmacy, Charles University, Hradec Kralove, 500 05, Czech Republic
| | - Přemysl Mladěnka
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Charles University, Hradec Kralove, 500 05, Czech Republic.
| |
Collapse
|
6
|
Stott JB, Greenwood IA. G protein βγ regulation of KCNQ-encoded voltage-dependent K channels. Front Physiol 2024; 15:1382904. [PMID: 38655029 PMCID: PMC11035767 DOI: 10.3389/fphys.2024.1382904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 03/25/2024] [Indexed: 04/26/2024] Open
Abstract
The KCNQ family is comprised of five genes and the expression products form voltage-gated potassium channels (Kv7.1-7.5) that have a major impact upon cellular physiology in many cell types. Each functional Kv7 channel forms as a tetramer that often associates with proteins encoded by the KCNE gene family (KCNE1-5) and is critically reliant upon binding of phosphatidylinositol bisphosphate (PIP2) and calmodulin. Other modulators like A-kinase anchoring proteins, ubiquitin ligases and Ca-calmodulin kinase II alter Kv7 channel function and trafficking in an isoform specific manner. It has now been identified that for Kv7.4, G protein βγ subunits (Gβγ) can be added to the list of key regulators and is paramount for channel activity. This article provides an overview of this nascent field of research, highlighting themes and directions for future study.
Collapse
Affiliation(s)
| | - Iain A. Greenwood
- Vascular Biology Research Group, Institute of Molecular and Clinical Sciences, St George’s University of London, London, United Kingdom
| |
Collapse
|
7
|
Banerjee D, Sabe SA, Xing H, Xu C, Sabra M, Harris DD, Broadwin M, Abid MR, Usheva A, Feng J, Sellke FW. Canagliflozin improves coronary microvascular vasodilation and increases absolute blood flow to the myocardium independent of angiogenesis. J Thorac Cardiovasc Surg 2023; 166:e535-e550. [PMID: 37604273 PMCID: PMC10840801 DOI: 10.1016/j.jtcvs.2023.08.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/01/2023] [Accepted: 08/09/2023] [Indexed: 08/23/2023]
Abstract
OBJECTIVES Sodium-glucose cotransporter-2 inhibitor, canagliflozin, improves myocardial perfusion to ischemic territory without accompanying changes in vascular density. We aimed to (1) characterize effects on angiogenic pathways, (2) use multiomics to identify gene expression and metabolite profiles relevant to regulation of myocardial blood flow, and (3) investigate drug effect on coronary microvascular reactivity. METHODS A nondiabetic swine model of chronic myocardial ischemia and nondiabetic rat model were used to study functional and molecular effects of canagliflozin on myocardium and in vitro microvascular reactivity. RESULTS Canagliflozin resulted in increased coronary microvascular vasodilation and decreased vasoconstriction (P < .05) without changes in microvascular density (P > .3). Expression of the angiogenic modulator, endostatin, increased (P = .008), along with its precursor, collagen 18 (P < .001), and factors that increase its production, including cathepsin L (P = .004). Endostatin and collagen 18 levels trended toward an inverse correlation with blood flow to ischemic territory at rest. Proangiogenic fibroblast growth factor receptor was increased (P = .03) and matrix metalloproteinase-9 was decreased (P < .001) with canagliflozin treatment. Proangiogenic vascular endothelial growth factor A (P = .13), Tie-2 (P = .10), and Ras (P = .18) were not significantly altered. Gene expression related to the cardiac renin-angiotensin system was significantly decreased. CONCLUSIONS In chronic myocardial ischemia, canagliflozin increased absolute blood flow to the myocardium without robustly increasing vascular density or proangiogenic signaling. Canagliflozin resulted in altered coronary microvascular reactivity to favor vasodilation, likely through direct effect on vascular smooth muscle. Downregulation of cardiac renin-angiotensin system demonstrated local regulation of perfusion. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Debolina Banerjee
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Sharif A Sabe
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Hang Xing
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Cynthia Xu
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Mohamed Sabra
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Dwight D Harris
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Mark Broadwin
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - M Ruhul Abid
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Anny Usheva
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Jun Feng
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Frank W Sellke
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI.
| |
Collapse
|
8
|
Gharaie S, Lee K, Noller K, Lo EK, Miller B, Jung HJ, Newman-Rivera AM, Kurzhagen JT, Singla N, Welling PA, Fan J, Cahan P, Noel S, Rabb H. Single cell and spatial transcriptomics analysis of kidney double negative T lymphocytes in normal and ischemic mouse kidneys. Sci Rep 2023; 13:20888. [PMID: 38017015 PMCID: PMC10684868 DOI: 10.1038/s41598-023-48213-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 11/23/2023] [Indexed: 11/30/2023] Open
Abstract
T cells are important in the pathogenesis of acute kidney injury (AKI), and TCR+CD4-CD8- (double negative-DN) are T cells that have regulatory properties. However, there is limited information on DN T cells compared to traditional CD4+ and CD8+ cells. To elucidate the molecular signature and spatial dynamics of DN T cells during AKI, we performed single-cell RNA sequencing (scRNA-seq) on sorted murine DN, CD4+, and CD8+ cells combined with spatial transcriptomic profiling of normal and post AKI mouse kidneys. scRNA-seq revealed distinct transcriptional profiles for DN, CD4+, and CD8+ T cells of mouse kidneys with enrichment of Kcnq5, Klrb1c, Fcer1g, and Klre1 expression in DN T cells compared to CD4+ and CD8+ T cells in normal kidney tissue. We validated the expression of these four genes in mouse kidney DN, CD4+ and CD8+ T cells using RT-PCR and Kcnq5, Klrb1, and Fcer1g genes with the NIH human kidney precision medicine project (KPMP). Spatial transcriptomics in normal and ischemic mouse kidney tissue showed a localized cluster of T cells in the outer medulla expressing DN T cell genes including Fcer1g. These results provide a template for future studies in DN T as well as CD4+ and CD8+ cells in normal and diseased kidneys.
Collapse
Affiliation(s)
- Sepideh Gharaie
- Department of Medicine, Johns Hopkins University, School of Medicine, Ross 965, 720 Rutland Ave, Baltimore, MD, 21205, USA
| | - Kyungho Lee
- Department of Medicine, Johns Hopkins University, School of Medicine, Ross 965, 720 Rutland Ave, Baltimore, MD, 21205, USA
| | - Kathleen Noller
- Department of Biomedical Engineering, Johns Hopkins University, School of Medicine, Baltimore, MD, 21205, USA
| | - Emily K Lo
- Department of Biomedical Engineering, Johns Hopkins University, School of Medicine, Baltimore, MD, 21205, USA
| | - Brendan Miller
- Department of Biomedical Engineering, Johns Hopkins University, School of Medicine, Baltimore, MD, 21205, USA
| | - Hyun Jun Jung
- Department of Medicine, Johns Hopkins University, School of Medicine, Ross 965, 720 Rutland Ave, Baltimore, MD, 21205, USA
| | - Andrea M Newman-Rivera
- Department of Medicine, Johns Hopkins University, School of Medicine, Ross 965, 720 Rutland Ave, Baltimore, MD, 21205, USA
| | - Johanna T Kurzhagen
- Department of Medicine, Johns Hopkins University, School of Medicine, Ross 965, 720 Rutland Ave, Baltimore, MD, 21205, USA
| | - Nirmish Singla
- Department of Urology, Johns Hopkins University, School of Medicine, Baltimore, MD, 21205, USA
| | - Paul A Welling
- Department of Medicine, Johns Hopkins University, School of Medicine, Ross 965, 720 Rutland Ave, Baltimore, MD, 21205, USA
- Department of Physiology, Johns Hopkins University, School of Medicine, Baltimore, MD, 21205, USA
| | - Jean Fan
- Department of Biomedical Engineering, Johns Hopkins University, School of Medicine, Baltimore, MD, 21205, USA
| | - Patrick Cahan
- Department of Biomedical Engineering, Johns Hopkins University, School of Medicine, Baltimore, MD, 21205, USA
| | - Sanjeev Noel
- Department of Medicine, Johns Hopkins University, School of Medicine, Ross 965, 720 Rutland Ave, Baltimore, MD, 21205, USA
| | - Hamid Rabb
- Department of Medicine, Johns Hopkins University, School of Medicine, Ross 965, 720 Rutland Ave, Baltimore, MD, 21205, USA.
| |
Collapse
|
9
|
Manville RW, Baldwin SN, Eriksen EØ, Jepps TA, Abbott GW. Medicinal plant rosemary relaxes blood vessels by activating vascular smooth muscle KCNQ channels. FASEB J 2023; 37:e23125. [PMID: 37535015 PMCID: PMC10437472 DOI: 10.1096/fj.202301132r] [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: 06/07/2023] [Revised: 07/18/2023] [Accepted: 07/25/2023] [Indexed: 08/04/2023]
Abstract
The evergreen plant rosemary (Salvia rosmarinus) has been employed medicinally for centuries as a memory aid, analgesic, spasmolytic, vasorelaxant and antihypertensive, with recent preclinical and clinical evidence rationalizing some applications. Voltage-gated potassium (Kv) channels in the KCNQ (Kv7) subfamily are highly influential in the nervous system, muscle and epithelia. KCNQ4 and KCNQ5 regulate vascular smooth muscle excitability and contractility and are implicated as antihypertensive drug targets. Here, we found that rosemary extract potentiates homomeric and heteromeric KCNQ4 and KCNQ5 activity, resulting in membrane hyperpolarization. Two rosemary diterpenes, carnosol and carnosic acid, underlie the effects and, like rosemary, are efficacious KCNQ-dependent vasorelaxants, quantified by myography in rat mesenteric arteries. Sex- and estrous cycle stage-dependence of the vasorelaxation matches sex- and estrous cycle stage-dependent KCNQ expression. The results uncover a molecular mechanism underlying rosemary vasorelaxant effects and identify new chemical spaces for KCNQ-dependent vasorelaxants.
Collapse
Affiliation(s)
- Rían W. Manville
- Bioelectricity Laboratory, Dept. of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, 92697, USA
| | - Samuel N. Baldwin
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Emil Ørnberg Eriksen
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas A. Jepps
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Geoffrey W. Abbott
- Bioelectricity Laboratory, Dept. of Physiology and Biophysics, School of Medicine, University of California, Irvine, CA, 92697, USA
| |
Collapse
|
10
|
Ehlers TS, van der Horst J, Møller S, Piil PK, Gliemann L, Aalkjaer C, Jepps TA, Hellsten Y. Colchicine enhances β adrenoceptor-mediated vasodilation in men with essential hypertension. Br J Clin Pharmacol 2023; 89:2179-2189. [PMID: 36764326 DOI: 10.1111/bcp.15688] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 12/20/2022] [Accepted: 01/05/2023] [Indexed: 02/12/2023] Open
Abstract
AIMS The aim of this study is to examine whether colchicine improves β adrenoceptor-mediated vasodilation in humans by conducting a double-blinded, placebo-controlled intervention study. Colchicine treatment has known beneficial effects on cardiovascular health and reduces the incidence of cardiovascular disease. Studies in isolated rodent arteries have shown that colchicine can enhance β adrenoceptor-mediated vasodilation, but this has not been determined in humans. METHODS Middle-aged men with essential hypertension were randomly assigned firstly to acute treatment with either 0.5 mg colchicine (n = 19) or placebo (n = 12). They were subsequently re-randomized for 3 weeks of treatment with either colchicine 0.5 mg twice daily (n = 16) or placebo (n = 15) followed by a washout period of 48-72 h. The vasodilator responses to isoprenaline, acetylcholine and sodium nitroprusside were determined as well as arterial pressure, arterial compliance and plasma inflammatory markers. RESULTS Acute colchicine treatment increased isoprenaline (by 38% for the highest dose) as well as sodium nitroprusside (by 29% main effect) -induced vasodilation but had no effect on the response to acetylcholine. The 3-week colchicine treatment followed by a washout period did not induce an accumulated or sustained effect on the β adrenoceptor response, and there was no effect on arterial pressure, arterial compliance or the level of measured inflammatory markers. CONCLUSION Colchicine acutely enhances β adrenoceptor- and nitric oxide-mediated changes in vascular conductance in humans, supporting that the mechanism previously demonstrated in rodents, translates to humans. The results provide novel translational evidence for a transient enhancing effect of colchicine on β adrenoceptor-mediated vasodilation in humans with essential hypertension. CONDENSED ABSTRACT Preclinical studies in isolated rodent arteries have shown that colchicine can enhance β adrenoceptor-mediated vasodilation. Here we show that this effect of colchicine can be translated to humans. Acute colchicine treatment was found to increase both isoprenaline- and sodium nitroprusside-induced vasodilation. The study provides the first translational evidence for a transient β adrenoceptor-mediated vasodilatory effect of colchicine in humans. The finding of an acute effect suggests that it may be clinically important to maintain an adequate bioavailability of colchicine.
Collapse
Affiliation(s)
- Thomas S Ehlers
- The August Krogh Section for Human Physiology, Department of Nutrition, Exercise and Sports (Experimental Site), University of Copenhagen, Copenhagen, Denmark
| | - Jennifer van der Horst
- The August Krogh Section for Human Physiology, Department of Nutrition, Exercise and Sports (Experimental Site), University of Copenhagen, Copenhagen, Denmark
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sophie Møller
- The August Krogh Section for Human Physiology, Department of Nutrition, Exercise and Sports (Experimental Site), University of Copenhagen, Copenhagen, Denmark
| | - Peter K Piil
- The August Krogh Section for Human Physiology, Department of Nutrition, Exercise and Sports (Experimental Site), University of Copenhagen, Copenhagen, Denmark
| | - Lasse Gliemann
- The August Krogh Section for Human Physiology, Department of Nutrition, Exercise and Sports (Experimental Site), University of Copenhagen, Copenhagen, Denmark
| | - Christian Aalkjaer
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Thomas A Jepps
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ylva Hellsten
- The August Krogh Section for Human Physiology, Department of Nutrition, Exercise and Sports (Experimental Site), University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
11
|
Seo MS, An JR, Heo R, Kang M, Park S, Mun SY, Park H, Han ET, Han JH, Chun W, Song G, Park WS. The inhibitory effects of pimozide, an antipsychotic drug, on voltage-gated K + channels in rabbit coronary arterial smooth muscle cells. Drug Chem Toxicol 2023; 46:271-280. [PMID: 35317682 DOI: 10.1080/01480545.2021.2021932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Pimozide is an antipsychotic drug used to treat chronic psychosis, such as Tourette's syndrome. Despite its widespread clinical use, pimozide can cause unexpected adverse effects, including arrhythmias. However, the adverse effects of pimozide on vascular K+ channels have not yet been determined. Therefore, we investigated the effects of pimozide on voltage-gated K+ (Kv) channels in rabbit coronary arterial smooth muscle cells. Pimozide concentration-dependently inhibited the Kv currents with an IC50 value of 1.78 ± 0.17 μM and a Hill coefficient of 0.90 ± 0.05. The inhibitory effect on the Kv current by pimozide was highly voltage-dependent in the voltage range of Kv channel activation, and additive inhibition of the Kv current by pimozide was observed in the full activation voltage range. The decay rate of inactivation was significantly accelerated by pimozide. Pimozide shifted the inactivation curve to a more negative potential. The recovery time constant from inactivation increased in the presence of pimozide. Furthermore, pimozide-induced inhibition of the Kv current was augmented by applying train pulses. Although pretreatment with the Kv2.1 subtype inhibitor guangxitoxin and the Kv7 subtype inhibitor linopirdine did not alter the degree of pimozide-induced inhibition of the Kv currents, pretreatment with the Kv1.5 channel inhibitor DPO-1 reduced the inhibitory effects of pimozide on Kv currents. Pimozide induced membrane depolarization. We conclude that pimozide inhibits Kv currents in voltage-, time-, and use (state)-dependent manners. Furthermore, the major Kv channel target of pimozide is the Kv1.5 channel.
Collapse
Affiliation(s)
- Mi Seon Seo
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Jin Ryeol An
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Ryeon Heo
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Minji Kang
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Seojin Park
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Seo-Yeong Mun
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Hongzoo Park
- Department of Urology, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Jin-Hee Han
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Wanjoo Chun
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Geehyun Song
- Department of Urology, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Won Sun Park
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon, South Korea
| |
Collapse
|
12
|
Estacion M, Liu S, Cheng X, Dib-Hajj S, Waxman SG. Kv7-specific activators hyperpolarize resting membrane potential and modulate human iPSC-derived sensory neuron excitability. Front Pharmacol 2023; 14:1138556. [PMID: 36923357 PMCID: PMC10008904 DOI: 10.3389/fphar.2023.1138556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 02/13/2023] [Indexed: 03/02/2023] Open
Abstract
Chronic pain is highly prevalent and remains a significant unmet global medical need. As part of a search for modulatory genes that confer pain resilience, we have studied two family cohorts where one individual reported much less pain than other family members that share the same pathogenic gain-of-function Nav1.7 mutation that confers hyperexcitability on pain-signaling dorsal root ganglion (DRG) neurons. In each of these kindreds, the pain-resilient individual carried a gain-of-function variant in Kv7.2 or Kv7.3, two potassium channels that stabilize membrane potential and reduce excitability. Our observation in this molecular genetic study that these gain-of-function Kv7.2 and 7.3 variants reduce DRG neuron excitability suggests that agents that activate or open Kv7 channels should attenuate sensory neuron firing. In the present study, we assess the effects on sensory neuron excitability of three Kv7 modulators-retigabine (Kv7.2 thru Kv7.5 activator), ICA-110381 (Kv7.2/Kv7.3 specific activator), and as a comparator ML277 (Kv7.1 specific activator)-in a "human-pain-in-a-dish" model (human iPSC-derived sensory neurons, iPSC-SN). Multi-electrode-array (MEA) recordings demonstrated inhibition of firing with retigabine and ICA-110381 (but not with ML277), with the concentration-response curve indicating that retigabine can achieve a 50% reduction of firing with sub-micromolar concentrations. Current-clamp recording demonstrated that retigabine hyperpolarized iPSC-SN resting potential and increased threshold. This study implicates Kv7.2/Kv7.3 channels as effective modulators of sensory neuron excitability, and suggest that compounds that specifically target Kv7.2/Kv7.3 currents in sensory neurons, including human sensory neurons, might provide an effective approach toward pain relief.
Collapse
Affiliation(s)
- Mark Estacion
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, United States
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, United States
| | - Shujun Liu
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, United States
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, United States
| | - Xiaoyang Cheng
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, United States
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, United States
| | - Sulayman Dib-Hajj
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, United States
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, United States
| | - Stephen G. Waxman
- Department of Neurology and Center for Neuroscience and Regeneration Research, Yale University School of Medicine, New Haven, CT, United States
- Rehabilitation Research Center, Veterans Affairs Connecticut Healthcare System, West Haven, CT, United States
| |
Collapse
|
13
|
Emerging mechanisms involving brain Kv7 channel in the pathogenesis of hypertension. Biochem Pharmacol 2022; 206:115318. [PMID: 36283445 DOI: 10.1016/j.bcp.2022.115318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/15/2022] [Accepted: 10/17/2022] [Indexed: 12/14/2022]
Abstract
Hypertension is a prevalent health problem inducing many organ damages. The pathogenesis of hypertension involves a complex integration of different organ systems including the brain. The elevated sympathetic nerve activity is closely related to the etiology of hypertension. Ion channels are critical regulators of neuronal excitability. Several mechanisms have been proposed to contribute to hypothalamic-driven elevated sympathetic activity, including altered ion channel function. Recent findings indicate one of the voltage-gated potassium channels, Kv7 channels (M channels), plays a vital role in regulating cardiovascular-related neurons activity, and the expression of Kv7 channels is downregulated in hypertension. This review highlights recent findings that the Kv7 channels in the brain, blood vessels, and kidneys are emerging targets involved in the pathogenesis of hypertension, suggesting new therapeutic targets for treating drug-resistant, neurogenic hypertension.
Collapse
|
14
|
The KCNQ channel inhibitor XE991 suppresses nicotinic acetylcholine receptor-mediated responses in rat intracardiac ganglion neurons. Pharmacol Rep 2022; 74:745-751. [PMID: 35672575 DOI: 10.1007/s43440-022-00375-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/29/2022] [Accepted: 05/13/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND XE991 (10,10-bis(4-pyridinylmethyl)-9(10H)-anthracenone) is reportedly a potent and selective Kv7 (KCNQ) channel inhibitor. This study aimed to evaluate how XE991 affects nicotinic responses in intracardiac ganglion neurons. METHODS We studied how the KCNQ channel inhibitor XE991 could affect nicotinic responses in acutely isolated rat intracardiac ganglion neurons using a perforated patch-clamp recording configuration and Ca2+ imaging. RESULTS XE991 reversibly and concentration-dependently inhibited the nicotine (10 μM)-induced current with an IC50 of 14.4 μM. The EC50 values for nicotine-induced currents in the absence and presence of 10 μM XE991 were 8.7 and 12.0 μM, respectively. Because XE991 suppressed the maximum response of the nicotine concentration-response curve, the inhibitory effect of this drug appears to be noncompetitive. In addition, linopirdine reduced the amplitude of 10 µM nicotine-induced currents with an IC50 value of 16.9 μM. The inorganic KCNQ channel inhibitor Ba2+ affected neither the nicotine-induced current nor the inhibitory effect of XE991 on the nicotinic response. The KCNQ activator flupirtine at a concentration of 10 μM slightly but markedly inhibited the nicotine-induced current. Finally, XE991 inhibited the nicotine-induced elevation of intracellular calcium concentration and the nicotine-induced firing of action potentials. CONCLUSION We propose that XE991 inhibits nicotinic acetylcholine receptors in intracardiac ganglion neurons, which in turn attenuate nicotine-induced neuronal excitation.
Collapse
|
15
|
Tan YQ, Li J, Chen HW. Epac, a positive or negative signaling molecule in cardiovascular diseases. Pharmacotherapy 2022; 148:112726. [DOI: 10.1016/j.biopha.2022.112726] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 02/08/2023]
|
16
|
Abstract
Since prehistory, human species have depended on plants for both food and medicine. Even in countries with ready access to modern medicines, alternative treatments are still highly regarded and commonly used. Unlike modern pharmaceuticals, many botanical medicines are in widespread use despite a lack of safety and efficacy data derived from controlled clinical trials and often unclear mechanisms of action. Contributing to this are the complex and undefined composition and likely multifactorial mechanisms of action and multiple targets of many botanical medicines. Here, we review the newfound importance of the ubiquitous KCNQ subfamily of voltage-gated potassium channels as targets for botanical medicines, including basil, capers, cilantro, lavender, fennel, chamomile, ginger, and Camellia, Sophora, and Mallotus species. We discuss the implications for the traditional use of these plants for disorders such as seizures, hypertension, and diabetes and the molecular mechanisms of plant secondary metabolite effects on KCNQ channels.
Collapse
Affiliation(s)
- Kaitlyn E Redford
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, California 92697, USA;
| | - Geoffrey W Abbott
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine, California 92697, USA;
| |
Collapse
|
17
|
Baldwin SN, Forrester EA, McEwan L, Greenwood IA. Sexual dimorphism in prostacyclin-mimetic responses within rat mesenteric arteries: A novel role for K V 7.1 in shaping IP-receptor mediated relaxation. Br J Pharmacol 2021; 179:1338-1352. [PMID: 34766649 PMCID: PMC9340493 DOI: 10.1111/bph.15722] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/21/2021] [Accepted: 10/19/2021] [Indexed: 11/29/2022] Open
Abstract
Background and Purpose Prostacyclin mimetics express potent vasoactive effects via prostanoid receptors that are not unequivocally defined, as to date no study has considered sex as a factor. The aim of this study was to determine the contribution of IP and EP3 prostanoid receptors to prostacyclin mimetic iloprost‐mediated responses, whether KV7.1–5 channels represent downstream targets of selective prostacyclin‐IP‐receptor agonist MRE‐269 and the impact of the oestrus cycle on vascular reactivity. Experimental Approach Within second‐order mesenteric arteries from male and female Wistar rats, we determined (1) relative mRNA transcripts for EP1–4 (Ptger1–4), IP (Ptgi) and TXA2 (Tbxa) prostanoid receptors via RT‐qPCR; (2) the effect of iloprost, MRE‐269, isoprenaline and ML277 on precontracted arterial tone in the presence of inhibitors of prostanoid receptors, potassium channels and the molecular interference of KV7.1 via wire‐myograph; (3) oestrus cycle stage via histological changes in cervical cell preparations. Key Results Iloprost evoked a biphasic response in male mesenteric arteries, at concentrations ≤100 nmol·L−1 relaxing, then contracting the vessel at concentration ≥300 nmol·L−1, a process attributed to IP and EP3 receptors respectively. Secondary contraction was absent in the females, which was associated with a reduction in Ptger3. Pharmacological inhibition and molecular interference of KV7.1 significantly attenuated relaxations produced by the selective IP receptor agonist MRE‐269 in male and female Wistar in dioestrus/metoestrus, but not pro‐oestrus/oestrus. Conclusions and Implications Stark sexual dimorphisms in iloprost‐mediated vasoactive responses are present within mesenteric arteries. KV7.1 is implicated in IP receptor‐mediated vasorelaxation and is impaired by the oestrus cycle.
Collapse
Affiliation(s)
- Samuel N Baldwin
- Molecular and clinical sciences research institute, St George's university, Cranmer terrace, London
| | - Elizabeth A Forrester
- Molecular and clinical sciences research institute, St George's university, Cranmer terrace, London
| | - Lauren McEwan
- Molecular and clinical sciences research institute, St George's university, Cranmer terrace, London
| | - Iain A Greenwood
- Molecular and clinical sciences research institute, St George's university, Cranmer terrace, London
| |
Collapse
|
18
|
Jepps TA. Kv7 channel trafficking by the microtubule network in vascular smooth muscle. Acta Physiol (Oxf) 2021; 232:e13692. [PMID: 34021973 PMCID: PMC8365713 DOI: 10.1111/apha.13692] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/17/2021] [Accepted: 05/19/2021] [Indexed: 12/17/2022]
Abstract
In arterial smooth muscle cells, changes in availability of integral membrane proteins influence the regulation of blood flow and blood pressure, which is critical for human health. However, the mechanisms that coordinate the trafficking and membrane expression of specific receptors and ion channels in vascular smooth muscle are poorly understood. In the vasculature, very little is known about microtubules, which form a road network upon which proteins can be transported to and from the cell membrane. This review article summarizes the impact of the microtubule network on arterial contractility, highlighting the importance of the network, with an emphasis on our recent findings regarding the trafficking of the voltage‐dependent Kv7 channels.
Collapse
Affiliation(s)
- Thomas A Jepps
- Vascular Biology Group Department of Biomedical Sciences University of Copenhagen Blegdamsvej 3 2200 Copenhagen N Denmark
| |
Collapse
|
19
|
Zhang X, Zhao Z, Xu C, Zhao F, Yan Z. Allisartan ameliorates vascular remodeling through regulation of voltage-gated potassium channels in hypertensive rats. BMC Pharmacol Toxicol 2021; 22:33. [PMID: 34108047 PMCID: PMC8188709 DOI: 10.1186/s40360-021-00498-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 04/27/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The objective of the present study was to determine the effect of allisartan, a new angiotensin II type 1 receptor antagonist on vascular remodeling through voltage gated potassium channels (Kv7) in hypertensive rats. METHODS The study included a total of 47 Sprague Dawley (SD) rats. The animals were randomized to sham operation (n = 14), untreated hypertensive control group (n = 18) and allisartan treatment group (n = 15). Using renal artery stenosis, hypertension was induced in animals. Single dose of allisartan was administered intra-gastrically to animals in the allisartan treatment group and match placebo in the other 2 groups. Wire myography was used to measure the muscle tension in isolated mesenteric arteries from the animals. Real-time polymerase chain reaction was used to quantify the expression of Kv7 channel mRNA subunits. RESULTS After 4 weeks of treatment, a significant decrease in mean arterial, systolic and diastolic blood pressure (SBP and DBP) was observed in allisartan treatment group compared to hypertension control group. The median arterial wall thickness and area/diameter ratio reduced significantly in treatment group compared to untreated hypertension group (P < 0.05). Wire myography demonstrated increased relaxation of mesenteric artery with increase in concentration of ML213. A significant up-regulation in the expression of all Kv7 mRNA subunits was observed in allisartan group compared to untreated hypertension group. CONCLUSIONS From the results, allisartan was found to lower BP and preserve vascular remodeling through Kv7 channels.
Collapse
Affiliation(s)
- Xiaoqin Zhang
- Department of Cardiology, Southern Medical University affiliated Fengxian Hospital, Shanghai, 201499, China
- Shanghai University of Medicine and Health Sciences Affiliated Sixth People's Hospital South Campus, Nanfeng Road No.6600, Shanghai, 201499, China
| | - Ziying Zhao
- Endoscopy Center, East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Chunfang Xu
- Department of Cardiology, Southern Medical University affiliated Fengxian Hospital, Shanghai, 201499, China
| | - Fengping Zhao
- Department of Cardiology, Southern Medical University affiliated Fengxian Hospital, Shanghai, 201499, China
| | - Zhiqiang Yan
- Department of Cardiology, Southern Medical University affiliated Fengxian Hospital, Shanghai, 201499, China.
| |
Collapse
|
20
|
Shvetsova AA, Gaynullina DK, Tarasova OS, Schubert R. Remodeling of Arterial Tone Regulation in Postnatal Development: Focus on Smooth Muscle Cell Potassium Channels. Int J Mol Sci 2021; 22:ijms22115413. [PMID: 34063769 PMCID: PMC8196626 DOI: 10.3390/ijms22115413] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/15/2021] [Accepted: 05/18/2021] [Indexed: 11/30/2022] Open
Abstract
Maturation of the cardiovascular system is associated with crucial structural and functional remodeling. Thickening of the arterial wall, maturation of the sympathetic innervation, and switching of the mechanisms of arterial contraction from calcium-independent to calcium-dependent occur during postnatal development. All these processes promote an almost doubling of blood pressure from the moment of birth to reaching adulthood. This review focuses on the developmental alterations of potassium channels functioning as key smooth muscle membrane potential determinants and, consequently, vascular tone regulators. We present evidence that the pattern of potassium channel contribution to vascular control changes from Kir2, Kv1, Kv7 and TASK-1 channels to BKCa channels with maturation. The differences in the contribution of potassium channels to vasomotor tone at different stages of postnatal life should be considered in treatment strategies of cardiovascular diseases associated with potassium channel malfunction.
Collapse
Affiliation(s)
- Anastasia A. Shvetsova
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, 119234 Moscow, Russia; (D.K.G.); (O.S.T.)
- Correspondence:
| | - Dina K. Gaynullina
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, 119234 Moscow, Russia; (D.K.G.); (O.S.T.)
- Department of Physiology, Russian National Research Medical University, 117997 Moscow, Russia
| | - Olga S. Tarasova
- Department of Human and Animal Physiology, Faculty of Biology, M.V. Lomonosov Moscow State University, 119234 Moscow, Russia; (D.K.G.); (O.S.T.)
- Laboratory of Exercise Physiology, State Research Center of the Russian Federation-Institute for Biomedical Problems, Russian Academy of Sciences, 123007 Moscow, Russia
| | - Rudolf Schubert
- Physiology, Institute of Theoretical Medicine, Medical Faculty, University of Augsburg, 86159 Augsburg, Germany;
| |
Collapse
|
21
|
Zhang Y, Wernly B, Cao X, Mustafa SJ, Tang Y, Zhou Z. Adenosine and adenosine receptor-mediated action in coronary microcirculation. Basic Res Cardiol 2021; 116:22. [PMID: 33755785 PMCID: PMC7987637 DOI: 10.1007/s00395-021-00859-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 03/08/2021] [Indexed: 12/20/2022]
Abstract
Adenosine is an ubiquitous extracellular signaling molecule and plays a fundamental role in the regulation of coronary microcirculation through activation of adenosine receptors (ARs). Adenosine is regulated by various enzymes and nucleoside transporters for its balance between intra- and extracellular compartments. Adenosine-mediated coronary microvascular tone and reactive hyperemia are through receptors mainly involving A2AR activation on both endothelial and smooth muscle cells, but also involving interaction among other ARs. Activation of ARs further stimulates downstream targets of H2O2, KATP, KV and KCa2+ channels leading to coronary vasodilation. An altered adenosine-ARs signaling in coronary microcirculation has been observed in several cardiovascular diseases including hypertension, diabetes, atherosclerosis and ischemic heart disease. Adenosine as a metabolite and its receptors have been studied for its both therapeutic and diagnostic abilities. The present review summarizes important aspects of adenosine metabolism and AR-mediated actions in the coronary microcirculation.
Collapse
Affiliation(s)
- Ying Zhang
- The International Collaborative Centre On Big Science Plan for Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Bernhard Wernly
- Department of Anaesthesiology, Perioperative Medicine and Intensive Care Medicine, Paracelsus Medical University of Salzburg, Salzburg, Austria
| | - Xin Cao
- The International Collaborative Centre On Big Science Plan for Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - S Jamal Mustafa
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, USA
| | - Yong Tang
- The International Collaborative Centre On Big Science Plan for Purinergic Signalling, Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Acupuncture and Chronobiology Key Laboratory of Sichuan Province, Chengdu, China
| | - Zhichao Zhou
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, 17176, Stockholm, Sweden.
| |
Collapse
|
22
|
Beck L, Pinilla E, Arcanjo DDR, Hernanz R, Prat-Duran J, Petersen AG, Köhler R, Sheykhzade M, Comerma-Steffensen S, Simonsen U. Pirfenidone Is a Vasodilator: Involvement of K V7 Channels in the Effect on Endothelium-Dependent Vasodilatation in Type-2 Diabetic Mice. Front Pharmacol 2021; 11:619152. [PMID: 33643042 PMCID: PMC7906977 DOI: 10.3389/fphar.2020.619152] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 12/30/2020] [Indexed: 12/27/2022] Open
Abstract
Endothelial cell dysfunction and fibrosis are associated with worsening of the prognosis in patients with cardiovascular disease. Pirfenidone has a direct antifibrotic effect, but vasodilatation may also contribute to the effects of pirfenidone. Therefore, in a first study we investigated the mechanisms involved in the relaxant effect of pirfenidone in rat intrapulmonary arteries and coronary arteries from normal mice. Then in a second study, we investigated whether pirfenidone restores endothelial function in the aorta and mesenteric arteries from diabetic animals. From 16–18-week old normal male C57BL/6 mice and normoglycemic (db/db+), and type 2 diabetic (db/db) male and female mice, arteries were mounted in microvascular isometric myographs for functional studies, and immunoblotting was performed. In rat pulmonary arteries and mouse coronary arteries, pirfenidone induced relaxations, which were inhibited in preparations without endothelium. In mouse coronary arteries, pirfenidone relaxation was inhibited in the presence of a nitric oxide (NO) synthase inhibitor, NG-nitro-l-arginine (L-NOARG), a blocker of large-conductance calcium-activated potassium channels (BKCa), iberiotoxin, and a blocker of KV7 channels, XE991. Patch clamp studies in vascular smooth muscle revealed pirfenidone increased iberiotoxin-sensitive current. In the aorta and mesenteric small arteries from diabetic db/db mice relaxations induced by the endothelium-dependent vasodilator, acetylcholine, were markedly reduced compared to db/db + mice. Pirfenidone enhanced the relaxations induced by acetylcholine in the aorta from diabetic male and female db/db mice. An opener of KV7 channels, flupirtine, had the same effect as pirfenidone. XE991 reduced the effect of pirfenidone and flupirtine and further reduced acetylcholine relaxations in the aorta. In the presence of iberiotoxin, pirfenidone still increased acetylcholine relaxation in aorta from db/db mice. Immunoblotting for KV7.4, KV7.5, and BKCa channel subunits were unaltered in aorta from db/db mice. Pirfenidone failed to improve acetylcholine relaxation in mesenteric arteries, and neither changed acetylcholine-induced transient decreases in blood pressure in db/db+ and db/db mice. In conclusion, pirfenidone vasodilates pulmonary and coronary arteries. In coronary arteries from normal mice, pirfenidone induces NO-dependent vasodilatation involving BKCa and KV7 channels. Pirfenidone improves endothelium-dependent vasodilatation in aorta from diabetic animals by a mechanism involving voltage-gated KV7 channels, a mechanism that may contribute to the antifibrotic effect of pirfenidone.
Collapse
Affiliation(s)
- Lilliana Beck
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark
| | - Estéfano Pinilla
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark.,Department of Physiology, Faculty of Pharmacy, Universidad Complutense, Madrid, Spain
| | - Daniel Dias Rufino Arcanjo
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark.,Department of Biophysics and Physiology, Laboratory of Functional and Molecular Studies in Physiopharmacology, Federal University of Piauí, Teresina, Brazil
| | - Raquel Hernanz
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark.,Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos, Alcorcón, Spain
| | - Judit Prat-Duran
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark
| | - Asbjørn Graver Petersen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark
| | - Ralf Köhler
- Aragón Agency for Research and Development (ARAID), Zaragoza, Spain
| | - Majid Sheykhzade
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Simon Comerma-Steffensen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark.,Department of Biomedical Sciences/Animal Physiology, Faculty of Veterinary, Central University of Venezuela, Maracay, Venezuela
| | - Ulf Simonsen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology, Aarhus University, Aarhus, Denmark
| |
Collapse
|
23
|
Abstract
Kv7.1-Kv7.5 (KCNQ1-5) K+ channels are voltage-gated K+ channels with major roles in neurons, muscle cells and epithelia where they underlie physiologically important K+ currents, such as neuronal M current and cardiac IKs. Specific biophysical properties of Kv7 channels make them particularly well placed to control the activity of excitable cells. Indeed, these channels often work as 'excitability breaks' and are targeted by various hormones and modulators to regulate cellular activity outputs. Genetic deficiencies in all five KCNQ genes result in human excitability disorders, including epilepsy, arrhythmias, deafness and some others. Not surprisingly, this channel family attracts considerable attention as potential drug targets. Here we will review biophysical properties and tissue expression profile of Kv7 channels, discuss recent advances in the understanding of their structure as well as their role in various neurological, cardiovascular and other diseases and pathologies. We will also consider a scope for therapeutic targeting of Kv7 channels for treatment of the above health conditions.
Collapse
|
24
|
Baldwin SN, Sandow SL, Mondéjar-Parreño G, Stott JB, Greenwood IA. K V7 Channel Expression and Function Within Rat Mesenteric Endothelial Cells. Front Physiol 2020; 11:598779. [PMID: 33364977 PMCID: PMC7750541 DOI: 10.3389/fphys.2020.598779] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 11/13/2020] [Indexed: 01/04/2023] Open
Abstract
Background and Purpose: Arterial diameter is dictated by the contractile state of the vascular smooth muscle cells (VSMCs), which is modulated by direct and indirect inputs from endothelial cells (ECs). Modulators of KCNQ-encoded kV7 channels have considerable impact on arterial diameter and these channels are known to be expressed in VSMCs but not yet defined in ECs. However, expression of kV7 channels in ECs would add an extra level of vascular control. This study aims to characterize the expression and function of KV7 channels within rat mesenteric artery ECs. Experimental Approach: In rat mesenteric artery, KCNQ transcript and KV7 channel protein expression were determined via RT-qPCR, immunocytochemistry, immunohistochemistry and immunoelectron microscopy. Wire myography was used to determine vascular reactivity. Key Results: KCNQ transcript was identified in isolated ECs and VSMCs. KV7.1, KV7.4 and KV7.5 protein expression was determined in both isolated EC and VSMC and in whole vessels. Removal of ECs attenuated vasorelaxation to two structurally different KV7.2-5 activators S-1 and ML213. KIR2 blockers ML133, and BaCl2 also attenuated S-1 or ML213-mediated vasorelaxation in an endothelium-dependent process. KV7 inhibition attenuated receptor-dependent nitric oxide (NO)-mediated vasorelaxation to carbachol, but had no impact on relaxation to the NO donor, SNP. Conclusion and Implications: In rat mesenteric artery ECs, KV7.4 and KV7.5 channels are expressed, functionally interact with endothelial KIR2.x channels and contribute to endogenous eNOS-mediated relaxation. This study identifies KV7 channels as novel functional channels within rat mesenteric ECs and suggests that these channels are involved in NO release from the endothelium of these vessels.
Collapse
Affiliation(s)
- Samuel N Baldwin
- Vascular Biology Research Centre, Institute of Molecular and Clinical Sciences, St George's University of London, London, United Kingdom
| | - Shaun L Sandow
- Biomedical Science, School of Health and Sports Science, University of the Sunshine Coast, Maroochydore, QLD, Australia
| | - Gema Mondéjar-Parreño
- Department of Pharmacology and Toxicology, School of Medicine, Complutense University of Madrid, Madrid, Spain
| | - Jennifer B Stott
- Vascular Biology Research Centre, Institute of Molecular and Clinical Sciences, St George's University of London, London, United Kingdom
| | - Iain A Greenwood
- Vascular Biology Research Centre, Institute of Molecular and Clinical Sciences, St George's University of London, London, United Kingdom
| |
Collapse
|
25
|
Mondéjar-Parreño G, Barreira B, Callejo M, Morales-Cano D, Barrese V, Esquivel-Ruiz S, Olivencia MA, Macías M, Moreno L, Greenwood IA, Perez-Vizcaino F, Cogolludo A. Uncovered Contribution of Kv7 Channels to Pulmonary Vascular Tone in Pulmonary Arterial Hypertension. Hypertension 2020; 76:1134-1146. [DOI: 10.1161/hypertensionaha.120.15221] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
K
+
channels play a fundamental role regulating membrane potential of pulmonary artery (PA) smooth muscle cells and their impairment is a common feature in pulmonary arterial hypertension (PAH). K
+
voltage-gated channel subfamily Q (
KCNQ1-5
) or Kv7 channels and their regulatory subunits subfamily E (KCNE) regulatory subunits are known to regulate vascular tone, but whether Kv7 channel function is impaired in PAH and how this can affect the rationale for targeting Kv7 channels in PAH remains unknown. Here, we have studied the role of Kv7/KCNE subunits in rat PA and their possible alteration in PAH. Using the patch-clamp technique, we found that the total K
+
current is reduced in PA smooth muscle cells from pulmonary hypertension animals (SU5416 plus hypoxia) and Kv7 currents made a higher contribution to the net K
+
current. Likewise, enhanced vascular responses to Kv7 channel modulators were found in pulmonary hypertension rats. Accordingly, KCNE4 subunit was highly upregulated in lungs from pulmonary hypertension animals and patients. Additionally, Kv7 channel activity was enhanced in the presence of Kv1.5 and TASK-1 channel inhibitors and this was associated with an increased KCNE4 membrane abundance. Compared with systemic arteries, PA showed a poor response to Kv7 channel modulators which was associated with reduced expression and membrane abundance of Kv7.4 and KCNE4. Our data indicate that Kv7 channel function is preserved and KCNE4 is upregulated in PAH. Therefore, compared with other downregulated channels, the contribution of Kv7 channels is increased in PAH resulting in an enhanced sensitivity to Kv7 channel modulators. This study provides insight into the potential usefulness of targeting Kv7 channels in PAH.
Collapse
Affiliation(s)
- Gema Mondéjar-Parreño
- From the Departamento de Farmacología y Toxicología. Facultad de Medicina, Universidad Complutense de Madrid, Spain (G.M.-P., B.B., M.C., S.E.-R., M.A.O., M.M., L.M., F.P.-V., A.C.)
- Ciber Enfermedades Respiratorias (Ciberes), Spain (G.M.-P., B.B., M.C., S.E.-R., M.A.O., M.M., L.M., F.P.-V., A.C.)
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM) (G.M.-P., B.B., M.C., S.E.-R., M.A.O., M.M., L.M., F.P.-V., A.C.)
| | - Bianca Barreira
- From the Departamento de Farmacología y Toxicología. Facultad de Medicina, Universidad Complutense de Madrid, Spain (G.M.-P., B.B., M.C., S.E.-R., M.A.O., M.M., L.M., F.P.-V., A.C.)
- Ciber Enfermedades Respiratorias (Ciberes), Spain (G.M.-P., B.B., M.C., S.E.-R., M.A.O., M.M., L.M., F.P.-V., A.C.)
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM) (G.M.-P., B.B., M.C., S.E.-R., M.A.O., M.M., L.M., F.P.-V., A.C.)
| | - María Callejo
- From the Departamento de Farmacología y Toxicología. Facultad de Medicina, Universidad Complutense de Madrid, Spain (G.M.-P., B.B., M.C., S.E.-R., M.A.O., M.M., L.M., F.P.-V., A.C.)
- Ciber Enfermedades Respiratorias (Ciberes), Spain (G.M.-P., B.B., M.C., S.E.-R., M.A.O., M.M., L.M., F.P.-V., A.C.)
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM) (G.M.-P., B.B., M.C., S.E.-R., M.A.O., M.M., L.M., F.P.-V., A.C.)
| | - Daniel Morales-Cano
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain (D.M.-C.)
| | - Vincenzo Barrese
- Vascular Biology Research Centre, Institute of Molecular and Clinical Sciences, St George’s University of London, United Kingdom (V.B., I.A.G.)
- Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples Federico II, Naples, Italy (V.B.)
| | - Sergio Esquivel-Ruiz
- From the Departamento de Farmacología y Toxicología. Facultad de Medicina, Universidad Complutense de Madrid, Spain (G.M.-P., B.B., M.C., S.E.-R., M.A.O., M.M., L.M., F.P.-V., A.C.)
- Ciber Enfermedades Respiratorias (Ciberes), Spain (G.M.-P., B.B., M.C., S.E.-R., M.A.O., M.M., L.M., F.P.-V., A.C.)
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM) (G.M.-P., B.B., M.C., S.E.-R., M.A.O., M.M., L.M., F.P.-V., A.C.)
| | - Miguel A. Olivencia
- From the Departamento de Farmacología y Toxicología. Facultad de Medicina, Universidad Complutense de Madrid, Spain (G.M.-P., B.B., M.C., S.E.-R., M.A.O., M.M., L.M., F.P.-V., A.C.)
- Ciber Enfermedades Respiratorias (Ciberes), Spain (G.M.-P., B.B., M.C., S.E.-R., M.A.O., M.M., L.M., F.P.-V., A.C.)
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM) (G.M.-P., B.B., M.C., S.E.-R., M.A.O., M.M., L.M., F.P.-V., A.C.)
| | - Miguel Macías
- From the Departamento de Farmacología y Toxicología. Facultad de Medicina, Universidad Complutense de Madrid, Spain (G.M.-P., B.B., M.C., S.E.-R., M.A.O., M.M., L.M., F.P.-V., A.C.)
- Ciber Enfermedades Respiratorias (Ciberes), Spain (G.M.-P., B.B., M.C., S.E.-R., M.A.O., M.M., L.M., F.P.-V., A.C.)
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM) (G.M.-P., B.B., M.C., S.E.-R., M.A.O., M.M., L.M., F.P.-V., A.C.)
| | - Laura Moreno
- From the Departamento de Farmacología y Toxicología. Facultad de Medicina, Universidad Complutense de Madrid, Spain (G.M.-P., B.B., M.C., S.E.-R., M.A.O., M.M., L.M., F.P.-V., A.C.)
- Ciber Enfermedades Respiratorias (Ciberes), Spain (G.M.-P., B.B., M.C., S.E.-R., M.A.O., M.M., L.M., F.P.-V., A.C.)
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM) (G.M.-P., B.B., M.C., S.E.-R., M.A.O., M.M., L.M., F.P.-V., A.C.)
| | - Iain A. Greenwood
- Vascular Biology Research Centre, Institute of Molecular and Clinical Sciences, St George’s University of London, United Kingdom (V.B., I.A.G.)
| | - Francisco Perez-Vizcaino
- From the Departamento de Farmacología y Toxicología. Facultad de Medicina, Universidad Complutense de Madrid, Spain (G.M.-P., B.B., M.C., S.E.-R., M.A.O., M.M., L.M., F.P.-V., A.C.)
- Ciber Enfermedades Respiratorias (Ciberes), Spain (G.M.-P., B.B., M.C., S.E.-R., M.A.O., M.M., L.M., F.P.-V., A.C.)
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM) (G.M.-P., B.B., M.C., S.E.-R., M.A.O., M.M., L.M., F.P.-V., A.C.)
| | - Angel Cogolludo
- From the Departamento de Farmacología y Toxicología. Facultad de Medicina, Universidad Complutense de Madrid, Spain (G.M.-P., B.B., M.C., S.E.-R., M.A.O., M.M., L.M., F.P.-V., A.C.)
- Ciber Enfermedades Respiratorias (Ciberes), Spain (G.M.-P., B.B., M.C., S.E.-R., M.A.O., M.M., L.M., F.P.-V., A.C.)
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM) (G.M.-P., B.B., M.C., S.E.-R., M.A.O., M.M., L.M., F.P.-V., A.C.)
| |
Collapse
|
26
|
Diniz AFA, Ferreira RC, de Souza ILL, da Silva BA. Ionic Channels as Potential Therapeutic Targets for Erectile Dysfunction: A Review. Front Pharmacol 2020; 11:1120. [PMID: 32848741 PMCID: PMC7396897 DOI: 10.3389/fphar.2020.01120] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 07/10/2020] [Indexed: 12/12/2022] Open
Abstract
Erectile dysfunction (ED) is a prevalent condition, especially in men over 40 years old, characterized by the inability to obtain and/or maintain penile erection sufficient for satisfactory sexual intercourse. Several psychological and/or organic factors are involved in the etiopathogenesis of ED. In this context, we gathered evidence of the involvement of Large-conductance, Ca2+-activated K+ channels (BKCa), Small-conductance, Ca2+-activated K+ channels (SKCa), KCNQ-encoded voltage-dependent K+ channels (KV7), Transient Receptor Potential channels (TRP), and Calcium-activated Chloride channels (CaCC) dysfunctions on ED. In addition, the use of modulating agents of these channels are involved in relaxation of the cavernous smooth muscle cell and, consequent penile erection, suggesting that these channels are promising therapeutic targets for the treatment of erectile dysfunction.
Collapse
Affiliation(s)
- Anderson Fellyp Avelino Diniz
- Programa de Pós-Graduação em Produtos Naturais e Sintéticos Bioativos, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Rafael Carlos Ferreira
- Programa de Pós-Graduação em Produtos Naturais e Sintéticos Bioativos, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Iara Leão Luna de Souza
- Departamento de Ciências Biológicas e da Saúde, Universidade Estadual de Roraima, Boa Vista, Brazil
| | - Bagnólia Araújo da Silva
- Programa de Pós-Graduação em Produtos Naturais e Sintéticos Bioativos, Universidade Federal da Paraíba, João Pessoa, Brazil
| |
Collapse
|
27
|
Hansen J, Johnsen J, Nielsen JM, Sørensen CB, Elkjær CC, Jespersen NR, Bøtker HE. Impact of Administration Time and Kv7 Subchannels on the Cardioprotective Efficacy of Kv7 Channel Inhibition. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:2549-2560. [PMID: 32669836 PMCID: PMC7337438 DOI: 10.2147/dddt.s226406] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 04/15/2020] [Indexed: 01/12/2023]
Abstract
Purpose The mechanism of cardioprotection by Kv7.1-5 (KCNQ1-5) channels inhibition by XE991 is unclear. We examined the impact of administration time on the cardioprotective efficacy of XE991, the involvement of key pro-survival kinases, and the importance of the Kv7 subchannels. Methods Isolated perfused rat hearts were divided into five groups: 1) vehicle, 2) pre-, 3) post- or 4) pre- and post-ischemic administration of XE991 or 5) chromanol 293B (Kv7.1 inhibitor) followed by infarct size quantification. HL-1 cells undergoing simulated ischemia/reperfusion were exposed to either a) vehicle, b) pre-, c) per-, d) post-ischemic administration of XE991 or pre-, per- and post-ischemic administration of e) XE991, f) Chromanol 293B or g) HMR1556 (Kv7.1 inhibitor). HL-1 cell injury was evaluated by propidium iodide/Hoechst staining. Pro-survival kinase activation of Akt, Erk and STAT3 in XE991-mediated HL-1 cell protection was evaluated using phosphokinase inhibitors. Kv7 subtype expression was examined by RT-PCR and qPCR. Results XE991, but not Chromanol 293B, reduced infarct size and improved hemodynamic recovery in all isolated heart groups. XE991 protected HL-1 cells when administered during simulated ischemia. Minor activation of the survival kinases was observed in cells exposed to XE991 but pharmacological inhibition of kinase activation did not reduce XE991-mediated protection. Kv7 subchannels 1-5 were all present in rat hearts but predominately Kv7.1 and Kv7.4 were present in HL-1 cells and selective Kv7.1 did not reduce ischemia/reperfusion injury. Conclusion The cardioprotective efficacy of XE991 seems to depend on its presence during ischemia and early reperfusion and do not rely on RISK (p-Akt and p-Erk) and SAFE (p-STAT3) pathway activation. The protective effect of XE991 seems mainly mediated through the Kv7.4 subchannel.
Collapse
Affiliation(s)
- Jan Hansen
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jacob Johnsen
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jan Møller Nielsen
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Charlotte Brandt Sørensen
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Casper Carlsen Elkjær
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Nichlas Riise Jespersen
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Hans Erik Bøtker
- Department of Cardiology, Aarhus University Hospital, Aarhus, Denmark.,Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
28
|
van der Horst J, Greenwood IA, Jepps TA. Cyclic AMP-Dependent Regulation of Kv7 Voltage-Gated Potassium Channels. Front Physiol 2020; 11:727. [PMID: 32695022 PMCID: PMC7338754 DOI: 10.3389/fphys.2020.00727] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 06/04/2020] [Indexed: 01/08/2023] Open
Abstract
Voltage-gated Kv7 potassium channels, encoded by KCNQ genes, have major physiological impacts cardiac myocytes, neurons, epithelial cells, and smooth muscle cells. Cyclic adenosine monophosphate (cAMP), a well-known intracellular secondary messenger, can activate numerous downstream effector proteins, generating downstream signaling pathways that regulate many functions in cells. A role for cAMP in ion channel regulation has been established, and recent findings show that cAMP signaling plays a role in Kv7 channel regulation. Although cAMP signaling is recognized to regulate Kv7 channels, the precise molecular mechanism behind the cAMP-dependent regulation of Kv7 channels is complex. This review will summarize recent research findings that support the mechanisms of cAMP-dependent regulation of Kv7 channels.
Collapse
Affiliation(s)
- Jennifer van der Horst
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Iain A Greenwood
- Molecular and Clinical Sciences Institute, St. George's University of London, London, United Kingdom
| | - Thomas A Jepps
- Vascular Biology Group, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
29
|
van der Horst J, Manville RW, Hayes K, Thomsen MB, Abbott GW, Jepps TA. Acetaminophen (Paracetamol) Metabolites Induce Vasodilation and Hypotension by Activating Kv7 Potassium Channels Directly and Indirectly. Arterioscler Thromb Vasc Biol 2020; 40:1207-1219. [PMID: 32188278 DOI: 10.1161/atvbaha.120.313997] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Intravenous acetaminophen/paracetamol (APAP) is well documented to cause hypotension. Since the patients receiving intravenous APAP are usually critically ill, any severe hemodynamic changes, as with those associated with APAP, can be life-threatening. The mechanism underlying this dangerous iatrogenic effect of APAP was unknown. Approach and Results: Here, we show that intravenous APAP caused transient hypotension in rats, which was attenuated by the Kv7 channel blocker, linopirdine. APAP metabolite N-acetyl-p-benzoquinone imine caused vasodilatation of rat mesenteric arteries ex vivo. This vasodilatation was sensitive to linopirdine and also the calcitonin gene-related peptide antagonist, BIBN 4096. Further investigation revealed N-acetyl-p-benzoquinone imine stimulates calcitonin gene-related peptide release from perivascular nerves, causing a cAMP-dependent activation of Kv7 channels. We also show that N-acetyl-p-benzoquinone imine enhances Kv7.4 and Kv7.5 channels overexpressed in oocytes, suggesting that it can activate Kv7.4 and Kv7.5 channels directly, to elicit vasodilatation. CONCLUSIONS Direct and indirect activation of Kv7 channels by the APAP metabolite N-acetyl-p-benzoquinone imine decreases arterial tone, which can lead to a drop in blood pressure. Our findings provide a molecular mechanism and potential preventive intervention for the clinical phenomenon of intravenous APAP-dependent transient hypotension.
Collapse
Affiliation(s)
- Jennifer van der Horst
- From the Vascular Biology Group, Department of Biomedical Science (J.v.d.H., K.H., T.A.J.), University of Copenhagen, Denmark
| | - Rian W Manville
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine (R.W.M., G.W.A.)
| | - Katie Hayes
- From the Vascular Biology Group, Department of Biomedical Science (J.v.d.H., K.H., T.A.J.), University of Copenhagen, Denmark
| | - Morten B Thomsen
- Cardiac Electrophysiology Group, Department of Biomedical Science (M.B.T.), University of Copenhagen, Denmark
| | - Geoffrey W Abbott
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California, Irvine (R.W.M., G.W.A.)
| | - Thomas A Jepps
- From the Vascular Biology Group, Department of Biomedical Science (J.v.d.H., K.H., T.A.J.), University of Copenhagen, Denmark
| |
Collapse
|
30
|
Manoury B, Idres S, Leblais V, Fischmeister R. Ion channels as effectors of cyclic nucleotide pathways: Functional relevance for arterial tone regulation. Pharmacol Ther 2020; 209:107499. [PMID: 32068004 DOI: 10.1016/j.pharmthera.2020.107499] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 02/05/2020] [Indexed: 02/07/2023]
Abstract
Numerous mediators and drugs regulate blood flow or arterial pressure by acting on vascular tone, involving cyclic nucleotide intracellular pathways. These signals lead to regulation of several cellular effectors, including ion channels that tune cell membrane potential, Ca2+ influx and vascular tone. The characterization of these vasocontrictive or vasodilating mechanisms has grown in complexity due to i) the variety of ion channels that are expressed in both vascular endothelial and smooth muscle cells, ii) the heterogeneity of responses among the various vascular beds, and iii) the number of molecular mechanisms involved in cyclic nucleotide signalling in health and disease. This review synthesizes key data from literature that highlight ion channels as physiologically relevant effectors of cyclic nucleotide pathways in the vasculature, including the characterization of the molecular mechanisms involved. In smooth muscle cells, cation influx or chloride efflux through ion channels are associated with vasoconstriction, whereas K+ efflux repolarizes the cell membrane potential and mediates vasodilatation. Both categories of ion currents are under the influence of cAMP and cGMP pathways. Evidence that some ion channels are influenced by CN signalling in endothelial cells will also be presented. Emphasis will also be put on recent data touching a variety of determinants such as phosphodiesterases, EPAC and kinase anchoring, that complicate or even challenge former paradigms.
Collapse
Affiliation(s)
- Boris Manoury
- Inserm, Umr-S 1180, Université Paris-Saclay, Châtenay-Malabry, France.
| | - Sarah Idres
- Inserm, Umr-S 1180, Université Paris-Saclay, Châtenay-Malabry, France
| | - Véronique Leblais
- Inserm, Umr-S 1180, Université Paris-Saclay, Châtenay-Malabry, France
| | | |
Collapse
|
31
|
Corydon KK, Matchkov V, Fais R, Abramochkin D, Hedegaard ER, Comerma-Steffensen S, Simonsen U. Effect of ischemic preconditioning and a Kv7 channel blocker on cardiac ischemia-reperfusion injury in rats. Eur J Pharmacol 2019; 866:172820. [PMID: 31760069 DOI: 10.1016/j.ejphar.2019.172820] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 11/19/2019] [Accepted: 11/20/2019] [Indexed: 12/30/2022]
Abstract
Recently, we found cardioprotective effects of ischemic preconditioning (IPC), and from a blocker of KCNQ voltage-gated K+ channels (KV7), XE991 (10,10-bis(4-pyridinylmethyl)-9(10H)-anthracenone), in isolated rat hearts. The purpose of the present study was to investigate the cardiovascular effects of IPC and XE991 and whether they are cardioprotective in intact rats. In conscious rats, we measured the effect of the KV7 channel blocker XE991 on heart rate and blood pressure by use of telemetry. In anesthetized rats, cardiac ischemia was induced by occluding the left coronary artery, and the animals received IPC (2 × 5 min of occlusion), XE991, or a combination. After a 2 h reperfusion period, the hearts were excised, and the area at risk and infarct size were determined. In both anesthetized and conscious rats, XE991 increased blood pressure, and the highest dose (7.5 mg/kg) of XE991 also increased heart rate, and 44% of conscious rats died. XE991 induced marked changes in the electrocardiogram (e.g., increased PR interval and prolonged QTC interval) without changing cardiac action potentials. The infarct size to area at risk ratio was reduced from 53 ± 2% (n = 8) in the vehicle compared to 36 ± 3% in the IPC group (P < 0.05, n = 9). XE991 (0.75 mg/kg) treatment alone or on top of IPC failed to reduce myocardial infarct size. Similar to the effect in isolated hearts, locally applied IPC was cardioprotective in intact animals exposed to ischemia-reperfusion. Systemic administration of XE991 failed to protect the heart against ischemia-reperfusion injury suggesting effects on the autonomic nervous system counteracting the cardioprotection in intact animals.
Collapse
Affiliation(s)
- Krestine Kjeldsen Corydon
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology and Physiology, Aarhus University, Wilhelm Meyers Allé 4, 8000, Aarhus C, Denmark
| | - Vladimir Matchkov
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology and Physiology, Aarhus University, Wilhelm Meyers Allé 4, 8000, Aarhus C, Denmark
| | - Rafael Fais
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology and Physiology, Aarhus University, Wilhelm Meyers Allé 4, 8000, Aarhus C, Denmark; Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, São Paulo, Brazil
| | - Denis Abramochkin
- Department of Human and Animal Physiology, Biological Faculty, Lomonosov Moscow State University, Leninskiye Gory, 1, 12, Moscow, Russia; Ural Federal University, Mira 19, Ekaterinburg, Russia; Department of Physiology, Russian National Research Medical University, Ostrovityanova 1, Moscow, Russia
| | - Elise Røge Hedegaard
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology and Physiology, Aarhus University, Wilhelm Meyers Allé 4, 8000, Aarhus C, Denmark
| | - Simon Comerma-Steffensen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology and Physiology, Aarhus University, Wilhelm Meyers Allé 4, 8000, Aarhus C, Denmark; Department of Biomedical Sciences/Animal Physiology, Veterinary Faculty, Central University of Venezuela, Maracay, Aragua, Venezuela
| | - Ulf Simonsen
- Department of Biomedicine, Pulmonary and Cardiovascular Pharmacology and Physiology, Aarhus University, Wilhelm Meyers Allé 4, 8000, Aarhus C, Denmark.
| |
Collapse
|
32
|
Stott JB, Barrese V, Suresh M, Masoodi S, Greenwood IA. Investigating the Role of G Protein βγ in Kv7-Dependent Relaxations of the Rat Vasculature. Arterioscler Thromb Vasc Biol 2019; 38:2091-2102. [PMID: 30002060 DOI: 10.1161/atvbaha.118.311360] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Objective- In renal arteries, inhibitors of G protein βγ subunits (Gβγ) reduce Kv7 activity and inhibit Kv7-dependent receptor-mediated vasorelaxations. However, the mechanisms underlying receptor-mediated relaxation are artery specific. Consequently, the aim of this study was to ascertain the role of Gβγ in Kv7-dependent vasorelaxations of the rat vasculature. Approach and Results- Isometric tension recording was performed in isolated rat renal, mesenteric, and cerebral arteries to study isoproterenol and calcitonin gene-related peptide relaxations. Kv7.4 was knocked down via morpholino transfection while inhibition of Gβγ was investigated with gallein and M119K. Proximity ligation assay was performed on isolated myocytes to study the association between Kv7.4 and G protein β subunits or signaling intermediaries. Isoproterenol or calcitonin gene-related peptide-induced relaxations were attenuated by Kv7.4 knockdown in all arteries studied. Inhibition of Gβγ with gallein or M119K had no effect on isoproterenol-mediated relaxations in mesenteric artery but had a marked effect on calcitonin gene-related peptide-induced responses in mesenteric artery and cerebral artery and isoproterenol responses in renal artery. Isoproterenol increased association with Kv7.4 and Rap1a in mesenteric artery which were not sensitive to gallein, whereas in renal artery, isoproterenol increased Kv7.4-AKAP (A-kinase anchoring protein) associations in a gallein-sensitive manner. Conclusions- The Gβγ-Kv7 relationship differs between vessels and is an essential requirement for AKAP, but not Rap-mediated regulation of the channel.
Collapse
Affiliation(s)
- Jennifer B Stott
- From the Vascular Biology Research Centre, Institute of Molecular and Clinical Sciences, St George's University of London, United Kingdom
| | - Vincenzo Barrese
- From the Vascular Biology Research Centre, Institute of Molecular and Clinical Sciences, St George's University of London, United Kingdom
| | - Malavika Suresh
- From the Vascular Biology Research Centre, Institute of Molecular and Clinical Sciences, St George's University of London, United Kingdom
| | - Shirou Masoodi
- From the Vascular Biology Research Centre, Institute of Molecular and Clinical Sciences, St George's University of London, United Kingdom
| | - Iain A Greenwood
- From the Vascular Biology Research Centre, Institute of Molecular and Clinical Sciences, St George's University of London, United Kingdom
| |
Collapse
|
33
|
Mondéjar-Parreño G, Moral-Sanz J, Barreira B, De la Cruz A, Gonzalez T, Callejo M, Esquivel-Ruiz S, Morales-Cano D, Moreno L, Valenzuela C, Perez-Vizcaino F, Cogolludo A. Activation of K v 7 channels as a novel mechanism for NO/cGMP-induced pulmonary vasodilation. Br J Pharmacol 2019; 176:2131-2145. [PMID: 30883701 DOI: 10.1111/bph.14662] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 02/08/2019] [Accepted: 02/13/2019] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND AND PURPOSE The NO/cGMP pathway represents a major physiological signalling controlling tone in pulmonary arteries (PA), and drugs activating this pathway are used to treat pulmonary arterial hypertension. Kv channels expressed in PA smooth muscle cells (PASMCs) are key determinants of vascular tone. We aimed to analyse the contribution of Kv 1.5 and Kv 7 channels in the electrophysiological and vasodilating effects evoked by NO donors and the GC stimulator riociguat in PA. EXPERIMENTAL APPROACH Kv currents were recorded in isolated rat PASMCs using the patch-clamp technique. Vascular reactivity was assessed in a wire myograph. KEY RESULTS The NO donors diethylamine NONOate diethylammonium (DEA-NO) and sodium nitroprusside hyperpolarized the membrane potential and induced a bimodal effect on Kv currents (augmenting the current between -40 and -10 mV and decreasing it at more depolarized potentials). The hyperpolarization and the enhancement of the current were suppressed by Kv 7 channel inhibitors and by the GC inhibitor ODQ but preserved when Kv 1.5 channels were inhibited. Additionally, DEA-NO enhanced Kv 7.5 currents in COS7 cells expressing the KCNQ5 gene. Riociguat increased Kv currents at all potentials ≥-40 mV and induced membrane hyperpolarization. Both effects were prevented by Kv 7 inhibition. Likewise, PA relaxation induced by NO donors and riociguat was attenuated by Kv 7 inhibitors. CONCLUSIONS AND IMPLICATIONS NO donors and riociguat enhance Kv 7 currents, leading to PASMC hyperpolarization. This mechanism contributes to NO/cGMP-induced PA vasodilation. Our study identifies Kv 7 channels as a novel mechanism of action of vasodilator drugs used in the treatment of pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Gema Mondéjar-Parreño
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Javier Moral-Sanz
- Centres for Discovery Brain Sciences and Cardiovascular Science, College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| | - Bianca Barreira
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Alicia De la Cruz
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Madrid, Spain.,CIBER Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Teresa Gonzalez
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Madrid, Spain.,CIBER Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.,Department of Biochemistry, School of Medicine, Universidad Autónoma de Madrid, Hospital La Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Maria Callejo
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Sergio Esquivel-Ruiz
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Daniel Morales-Cano
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Laura Moreno
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Carmen Valenzuela
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Madrid, Spain.,CIBER Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Francisco Perez-Vizcaino
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Angel Cogolludo
- Department of Pharmacology and Toxicology, School of Medicine, University Complutense of Madrid, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain.,Ciber Enfermedades Respiratorias (CIBERES), Madrid, Spain
| |
Collapse
|
34
|
Khanamiri S, Rhee JW, Paik DT, Chen IY, Liu C, Sayed N. Marked Vascular Dysfunction in a Case of Peripartum Cardiomyopathy. J Vasc Res 2019; 56:11-15. [PMID: 30763932 DOI: 10.1159/000496163] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 12/11/2018] [Indexed: 11/19/2022] Open
Abstract
Peripartum cardiomyopathy (PPCM) is a rare form of congestive heart failure characterized by left ventricular dysfunction that develops towards the end of pregnancy or during the early postpartum phase. Even though the majority of PPCM patients show partial or complete recovery of their heart functions, the mortality rate of PPCM remains high. Previous research has suggested that vascular dysfunction triggered by late-gestational hormones and potent anti-angiogenic factors play key roles in the pathogenesis of PPCM; however, the exact mechanisms remain elusive due to limited patient tissues for characterization. Here, we report a case of PPCM where the coronary vessels from the patient's explanted heart showed marked vascular dysfunction with impaired nitric oxide response. Importantly, these vessels exhibited deficient adenosine-mediated vasorelaxation when subjected to myograph studies, suggesting impaired Kv7 ion channels. Results from this work may lead to new therapeutic strategies for improving Kv7 function in PPCM patients.
Collapse
Affiliation(s)
- Saereh Khanamiri
- Stanford Cardiovascular Institute, Stanford, California, USA.,Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - June-Wha Rhee
- Stanford Cardiovascular Institute, Stanford, California, USA.,Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - David T Paik
- Stanford Cardiovascular Institute, Stanford, California, USA.,Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Ian Y Chen
- Stanford Cardiovascular Institute, Stanford, California, USA.,Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Chun Liu
- Stanford Cardiovascular Institute, Stanford, California, USA.,Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Nazish Sayed
- Stanford Cardiovascular Institute, Stanford, California, USA, .,Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA,
| |
Collapse
|
35
|
Lawson K. Pharmacology and clinical applications of flupirtine: Current and future options. World J Pharmacol 2019; 8:1-13. [DOI: 10.5497/wjp.v8.i1.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 11/17/2018] [Accepted: 01/05/2019] [Indexed: 02/06/2023] Open
Abstract
Flupirtine is the first representative in a class of triaminopyridines that exhibits pharmacological properties leading to the suppression of over-excitability of neuronal and non-neuronal cells. Consequently, this drug has been used as a centrally acting analgesic in patients with a range of acute and persistent pain conditions without the adverse effects characteristic of opioids and non-steroidal anti-inflammatory drug and is well tolerated. The pharmacological profile exhibited involves actions on several cellular targets, including Kv7 channels, G-protein-regulated inwardly rectifying K channels and γ-aminobutyric acid type A receptors, but also there is evidence of additional as yet unidentified mechanisms of action involved in the effects of flupirtine. Flupirtine has exhibited effects in a range of cells and tissues related to the locations of these targets. In additional to analgesia, flupirtine has demonstrated pharmacological properties consistent with use as an anticonvulsant, a neuroprotectant, skeletal and smooth muscle relaxant, in treatment of auditory and visual disorders, and treatment of memory and cognitive impairment. Flupirtine is providing important information and clues regarding novel mechanistic approaches to the treatment of a range of clinical conditions involving hyper-excitability of cells. Identification of molecules exhibiting specificity for the pharmacological targets (e.g., Kv7 isoforms) involved in the actions of flupirtine will provide further insight into clinical applications. Whether the broad-spectrum pharmacology of flupirtine or target-specific actions is preferential to gain benefit, especially in complex clinical conditions, requires further investigation. This review will consider recent advancement in understanding of the pharmacological profile and related clinical applications of flupirtine.
Collapse
Affiliation(s)
- Kim Lawson
- Department of Biosciences and Chemistry, Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield S1 1WB, United Kingdom
| |
Collapse
|
36
|
Jackson WF. K V channels and the regulation of vascular smooth muscle tone. Microcirculation 2018; 25. [PMID: 28985443 DOI: 10.1111/micc.12421] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 10/01/2017] [Indexed: 12/31/2022]
Abstract
VSMCs in resistance arteries and arterioles express a diverse array of KV channels with members of the KV 1, KV 2 and KV 7 families being particularly important. Members of the KV channel family: (i) are highly expressed in VSMCs; (ii) are active at the resting membrane potential of VSMCs in vivo (-45 to -30 mV); (iii) contribute to the negative feedback regulation of VSMC membrane potential and myogenic tone; (iv) are activated by cAMP-related vasodilators, hydrogen sulfide and hydrogen peroxide; (v) are inhibited by increases in intracellular Ca2+ and vasoconstrictors that signal through Gq -coupled receptors; (vi) are involved in the proliferative phenotype of VSMCs; and (vii) are modulated by diseases such as hypertension, obesity, the metabolic syndrome and diabetes. Thus, KV channels participate in every aspect of the regulation of VSMC function in both health and disease.
Collapse
Affiliation(s)
- William F Jackson
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
37
|
Byron KL, Brueggemann LI. Kv7 potassium channels as signal transduction intermediates in the control of microvascular tone. Microcirculation 2018; 25. [PMID: 28976052 DOI: 10.1111/micc.12419] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 09/27/2017] [Indexed: 12/18/2022]
Abstract
Potassium channels are recognized as important regulators of cellular functions in most, if not all cell types. These cellular proteins assemble to form gated pores in the plasma membrane, which serve to regulate the flow of potassium ions (K+ ) from the cytosol to the extracellular space. In VSMCs, the open state of potassium channels enables the efflux of K+ and thereby establishes a negative resting voltage across the plasma membrane that inhibits the opening of VSCCs. Under these conditions, cytosolic Ca2+ concentrations are relatively low and Ca2+ -dependent contraction is inhibited. Recent research has identified Kv7 family potassium channels as important contributors to resting membrane voltage in VSMCs, with much of the research focusing on the effects of drugs that specifically activate or block these channels to produce corresponding effects on VSMC contraction and vascular tone. Increasingly, evidence is emerging that these channels are not just good drug targets-they are also essential intermediates in vascular signal transduction, mediating vasoconstrictor or vasodilator responses to a variety of physiological stimuli. This review will summarize recent research findings that support a crucial function of Kv7 channels in both positive (vasoconstrictive) and negative (vasorelaxant) regulation of microvascular tone.
Collapse
Affiliation(s)
- Kenneth L Byron
- Department of Molecular Pharmacology & Therapeutics, Loyola University Chicago, Maywood, IL, USA
| | - Lyubov I Brueggemann
- Department of Molecular Pharmacology & Therapeutics, Loyola University Chicago, Maywood, IL, USA
| |
Collapse
|
38
|
Idres S, Perrin G, Domergue V, Lefebvre F, Gomez S, Varin A, Fischmeister R, Leblais V, Manoury B. Contribution of BKCa channels to vascular tone regulation by PDE3 and PDE4 is lost in heart failure. Cardiovasc Res 2018; 115:130-144. [DOI: 10.1093/cvr/cvy161] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 06/21/2018] [Indexed: 12/23/2022] Open
Abstract
Abstract
Aims
Regulation of vascular tone by 3′,5′-cyclic adenosine monophosphate (cAMP) involves many effectors including the large conductance, Ca2+-activated, K+ (BKCa) channels. In arteries, cAMP is mainly hydrolyzed by type 3 and 4 phosphodiesterases (PDE3, PDE4). Here, we examined the specific contribution of BKCa channels to tone regulation by these PDEs in rat coronary arteries, and how this is altered in heart failure (HF).
Methods and results
Concomitant application of PDE3 (cilostamide) and PDE4 (Ro-20-1724) inhibitors increased BKCa unitary channel activity in isolated myocytes from rat coronary arteries. Myography was conducted in isolated, U46619-contracted coronary arteries. Cilostamide (Cil) or Ro-20-1724 induced a vasorelaxation that was greatly reduced by iberiotoxin (IBTX), a BKCa channel blocker. Ro-20-1724 and Cil potentiated the relaxation induced by the β-adrenergic agonist isoprenaline (ISO) or the adenylyl cyclase activator L-858051 (L85). IBTX abolished the effect of PDE inhibitors on ISO but did not on L85. In coronary arteries from rats with HF induced by aortic stenosis, contractility and response to acetylcholine were dramatically reduced compared with arteries from sham rats, but relaxation to PDE inhibitors was retained. Interestingly, however, IBTX had no effect on Ro-20-1724- and Cil-induced vasorelaxations in HF. Expression of the BKCa channel α-subunit, of a 98 kDa PDE3A and of a 80 kDa PDE4D were lower in HF compared with sham coronary arteries, while that of a 70 kDa PDE4B was increased. Proximity ligation assays demonstrated that PDE3 and PDE4 were localized in the vicinity of the channel.
Conclusion
BKCa channels mediate the relaxation of coronary artery induced by PDE3 and PDE4 inhibition. This is achieved by co-localization of both PDEs with BKCa channels, enabling tight control of cAMP available for channel opening. Contribution of the channel is prominent at rest and on β-adrenergic stimulation. This coupling is lost in HF.
Collapse
Affiliation(s)
- Sarah Idres
- Signalling and Cardiovascular Pathophysiology—UMR-S 1180, Université Paris-Sud, INSERM, Université Paris-Saclay, 5 rue J-B Clément, Châtenay-Malabry, France
| | - Germain Perrin
- Signalling and Cardiovascular Pathophysiology—UMR-S 1180, Université Paris-Sud, INSERM, Université Paris-Saclay, 5 rue J-B Clément, Châtenay-Malabry, France
| | - Valérie Domergue
- UMS IPSIT, Université Paris-Sud, Université Paris-Saclay, 5 rue J-B Clément, Châtenay-Malabry, France
| | - Florence Lefebvre
- Signalling and Cardiovascular Pathophysiology—UMR-S 1180, Université Paris-Sud, INSERM, Université Paris-Saclay, 5 rue J-B Clément, Châtenay-Malabry, France
| | - Susana Gomez
- Signalling and Cardiovascular Pathophysiology—UMR-S 1180, Université Paris-Sud, INSERM, Université Paris-Saclay, 5 rue J-B Clément, Châtenay-Malabry, France
| | - Audrey Varin
- Signalling and Cardiovascular Pathophysiology—UMR-S 1180, Université Paris-Sud, INSERM, Université Paris-Saclay, 5 rue J-B Clément, Châtenay-Malabry, France
| | - Rodolphe Fischmeister
- Signalling and Cardiovascular Pathophysiology—UMR-S 1180, Université Paris-Sud, INSERM, Université Paris-Saclay, 5 rue J-B Clément, Châtenay-Malabry, France
| | - Véronique Leblais
- Signalling and Cardiovascular Pathophysiology—UMR-S 1180, Université Paris-Sud, INSERM, Université Paris-Saclay, 5 rue J-B Clément, Châtenay-Malabry, France
| | - Boris Manoury
- Signalling and Cardiovascular Pathophysiology—UMR-S 1180, Université Paris-Sud, INSERM, Université Paris-Saclay, 5 rue J-B Clément, Châtenay-Malabry, France
| |
Collapse
|
39
|
Wei X, Zhang Y, Yin B, Wen J, Cheng J, Fu X. The expression and function of KCNQ potassium channels in human chorionic plate arteries from women with normal pregnancies and pre-eclampsia. PLoS One 2018; 13:e0192122. [PMID: 29579054 PMCID: PMC5868761 DOI: 10.1371/journal.pone.0192122] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 01/18/2018] [Indexed: 11/25/2022] Open
Abstract
Pre-eclampsia is associated with altered maternal and placental vascular reactivity. Kv7 channels (encoded by KCNQ 1–5 genes) are a potential contributor to the regulation of vascular tone in CPAs (chorionic plate arteries) during normal pregnancy. The aim of this study is to establish the expression profile of KCNQ subunits in CPAs taken from women with preeclampsia or normotensive women and to examine the functional relevance of the Kv7 channels on an altered expression profile of KCNQ subunits. The effects of Kv7 channel modulators on CPAs were investigated by tension measurement. Quantitative PCR experiments were used to analyze the expression of KCNQ genes. Western blotting and immunofluorescence were both used to analyze the protein expression of Kv7 channels. Finally, in CPAs from normotensive women, the Kv7 channel blocker XE991 increased arterial basal tone and U46619-induced contraction, and pre-contracted CPAs (10−7 M U46619) exhibited significant relaxation following treatment with Retigabine(Kv7.2–7.5 activator) and BMS-204352(Kv7.2–7.5 activator). However, ICA-27243(selective KCNQ2 and KCNQ3 activator) and ML277(selective KV7.1 activator) had no significant effect on tension in the pre-contracted CPAs. Conversely, compared with CPAs from normotensive women, the effects of XE991 on basal tone and agonist (U46619)-induced contractions in CPAs from women with preeclampsia were markedly attenuated. Moreover, the relaxation effects of Retigabine and BMS-204352 on pre-contracted CPA vessels from women with pre-eclampsia were also markedly down-regulated. Interestingly, the relaxation ability of ICA-27243 in pre-contracted CPA vessels in women with pre-eclampsia was enhanced. The mRNA of KCNQ3 was specifically up-regulated, whereas those for KCNQ4 and KCNQ5 were down-regulated in CPAs from women with pre-eclampsia compared with those in normotensive women. Similar observations were found in a subsequent analysis of protein expression of KCNQ genes 3–5. Thus, down-regulated Kv7 channel function in tension regulation of CPAs in women with pre-eclampsia could be associated with considerably altered expression profiles of Kv7 subunits.
Collapse
Affiliation(s)
- Xiaohong Wei
- Department of Gynecology and Obstetrics, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yujiao Zhang
- Department of Gynecology and Obstetrics, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Benlan Yin
- Department of Gynecology and Obstetrics, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Jing Wen
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| | - Jun Cheng
- Key Laboratory of Medical Electrophysiology of Ministry of Education, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China
| | - Xiaodong Fu
- Department of Gynecology and Obstetrics, the Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- * E-mail:
| |
Collapse
|
40
|
Barrese V, Stott JB, Greenwood IA. KCNQ-Encoded Potassium Channels as Therapeutic Targets. Annu Rev Pharmacol Toxicol 2018; 58:625-648. [DOI: 10.1146/annurev-pharmtox-010617-052912] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
| | | | - Iain A. Greenwood
- Vascular Biology Research Centre, Molecular and Clinical Sciences Institute, St George's, University of London, London, SW17 0RE, United Kingdom;, ,
| |
Collapse
|
41
|
Nieves-Cintrón M, Syed AU, Nystoriak MA, Navedo MF. Regulation of voltage-gated potassium channels in vascular smooth muscle during hypertension and metabolic disorders. Microcirculation 2018; 25:10.1111/micc.12423. [PMID: 29044853 PMCID: PMC5760350 DOI: 10.1111/micc.12423] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 10/11/2017] [Indexed: 12/12/2022]
Abstract
Voltage-gated potassium (KV ) channels are key regulators of vascular smooth muscle contractility and vascular tone, and thus have major influence on the microcirculation. KV channels are important determinants of vascular smooth muscle membrane potential (Em ). A number of KV subunits are expressed in the plasma membrane of smooth muscle cells. Each subunit confers distinct kinetics and regulatory properties that allow for fine control of Em to orchestrate vascular tone. Modifications in KV subunit expression and/or channel activity can contribute to changes in vascular smooth muscle contractility in response to different stimuli and in diverse pathological conditions. Consistent with this, a number of studies suggest alterations in KV subunit expression and/or function as underlying contributing mechanisms for small resistance artery dysfunction in pathologies such as hypertension and metabolic disorders, including diabetes. Here, we review our current knowledge on the effects of these pathologies on KV channel expression and function in vascular smooth muscle cells, and the repercussions on (micro)vascular function.
Collapse
Affiliation(s)
| | - Arsalan U. Syed
- Department of Pharmacology, University of California, Davis, CA 95616
| | - Matthew A. Nystoriak
- Diabetes and Obesity Center, Department of Medicine, University of Louisville, Louisville, KY 40202
| | - Manuel F. Navedo
- Department of Pharmacology, University of California, Davis, CA 95616
| |
Collapse
|
42
|
Lindman J, Khammy MM, Lundegaard PR, Aalkjær C, Jepps TA. Microtubule Regulation of Kv7 Channels Orchestrates cAMP-Mediated Vasorelaxations in Rat Arterial Smooth Muscle. Hypertension 2017; 71:336-345. [PMID: 29279314 DOI: 10.1161/hypertensionaha.117.10152] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 09/01/2017] [Accepted: 11/29/2017] [Indexed: 11/16/2022]
Abstract
Microtubules can regulate GPCR (G protein-coupled receptor) signaling in various cell types. In vascular smooth muscle, activation of the β-adrenoceptor leads to production of cAMP to mediate a vasorelaxation. Little is known about the role of microtubules in smooth muscle, and given the importance of this pathway in vascular smooth muscle cells, we investigated the role of microtubule stability on β-adrenoceptor signaling in rat renal and mesenteric arteries. In isometric tension experiments, incubation with the microtubule inhibitors colchicine and nocodazole enhanced isoprenaline-mediated relaxations of renal and mesenteric arteries that the microtubule stabilizer, paclitaxel, prevented. Sharp microelectrode experiments showed that colchicine treatment caused increased hyperpolarization of mesenteric artery segments in response to isoprenaline. Application of the Kv7 channel blocker, XE991, attenuated the effect of colchicine on isoprenaline relaxations, whereas iberiotoxin-a BKCa channel blocker-had no effect. In addition, colchicine improved the relaxations to the Kv7.2 to 7.5 activator, S-1, in both renal and mesenteric artery segments compared with dimethyl sulfoxide incubation. We determined that increased mesenteric artery myocytes treated with colchicine showed increased Kv7.4 membrane expression, but Western blot analysis showed no change in total Kv7.4 protein. This study is the first to show microtubule disruption improves the β-adrenoceptor-mediated relaxations of mesenteric and renal arteries and determine this enhancement to be because of increased membrane expression of the Kv7 voltage-gated potassium channels.
Collapse
Affiliation(s)
- Johanna Lindman
- From the Department of Biomedical Sciences, Ion Channels Group, University of Copenhagen, Denmark (J.L., M.M.K., P.R.L., C.A., T.A.J.); and Department of Biomedicine, Aarhus University, Denmark (M.M.K., C.A.)
| | - Makhala M Khammy
- From the Department of Biomedical Sciences, Ion Channels Group, University of Copenhagen, Denmark (J.L., M.M.K., P.R.L., C.A., T.A.J.); and Department of Biomedicine, Aarhus University, Denmark (M.M.K., C.A.)
| | - Pia R Lundegaard
- From the Department of Biomedical Sciences, Ion Channels Group, University of Copenhagen, Denmark (J.L., M.M.K., P.R.L., C.A., T.A.J.); and Department of Biomedicine, Aarhus University, Denmark (M.M.K., C.A.)
| | - Christian Aalkjær
- From the Department of Biomedical Sciences, Ion Channels Group, University of Copenhagen, Denmark (J.L., M.M.K., P.R.L., C.A., T.A.J.); and Department of Biomedicine, Aarhus University, Denmark (M.M.K., C.A.)
| | - Thomas A Jepps
- From the Department of Biomedical Sciences, Ion Channels Group, University of Copenhagen, Denmark (J.L., M.M.K., P.R.L., C.A., T.A.J.); and Department of Biomedicine, Aarhus University, Denmark (M.M.K., C.A.).
| |
Collapse
|
43
|
Fosmo AL, Skraastad ØB. The Kv7 Channel and Cardiovascular Risk Factors. Front Cardiovasc Med 2017; 4:75. [PMID: 29259974 PMCID: PMC5723334 DOI: 10.3389/fcvm.2017.00075] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 11/21/2017] [Indexed: 12/30/2022] Open
Abstract
Potassium channels play a pivotal role in the regulation of excitability in cells such as neurons, cardiac myocytes, and vascular smooth muscle cells. The KCNQ (Kv7) family of voltage-activated K+ channels hyperpolarizes the cell and stabilizes the membrane potential. Here, we outline how Kv7 channel activity may contribute to the development of the cardiovascular risk factors such as hypertension, diabetes, and obesity. Questions and hypotheses regarding previous and future research have been raised. Alterations in the Kv7 channel may contribute to the development of cardiovascular disease (CVD). Pharmacological modification of Kv7 channels may represent a possible treatment for CVD in the future.
Collapse
Affiliation(s)
- Andreas L Fosmo
- Division of Physiology, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Øyvind B Skraastad
- Division of Physiology, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|
44
|
Zimmer J, Takahashi T, Hofmann AD, Puri P. Downregulation of KCNQ5 expression in the rat pulmonary vasculature of nitrofen-induced congenital diaphragmatic hernia. J Pediatr Surg 2017; 52:702-705. [PMID: 28189443 DOI: 10.1016/j.jpedsurg.2017.01.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 01/23/2017] [Indexed: 12/17/2022]
Abstract
PURPOSE Pulmonary hypertension (PH) is a common complication of congenital diaphragmatic hernia (CDH). Voltage-gated potassium channels KCNQ1, KCNQ4, and KCNQ5 are expressed by rodent pulmonary artery smooth muscle cells, contributing to their vascular tone. We hypothesized that KCNQ1, KCNQ4, and KCNQ5 expression is altered in the pulmonary vasculature of nitrofen-induced CDH rats. METHODS After ethical approval (REC913b), time-pregnant rats received nitrofen or vehicle on gestational day (D)9. D21 fetuses were divided into CDH and control group (n=22). QRT-PCR and western blotting were performed to determine gene and protein expression of KCNQ1, KCNQ4, and KCNQ5. Confocal microscopy was used to detect these proteins in the pulmonary vasculature. RESULTS Relative mRNA level of KCNQ5 (p=0.025) was significantly downregulated in CDH lungs compared to controls. KCNQ1 (p=0.052) and KCNQ4 (p=0.574) expression was not altered. Western blotting confirmed the decreased pulmonary KCNQ5 protein expression in CDH lungs. Confocal-microscopy detected a markedly diminished KCNQ5 expression in pulmonary vasculature of CDH fetuses. CONCLUSIONS Downregulated pulmonary expression of KCNQ5 in CDH lungs suggests that this potassium channel may play an important role in the development of PH in this model. KCNQ5 channel activator drugs may be a potential therapeutic target for the treatment of PH in CDH. LEVEL OF EVIDENCE 2b (Centre for Evidence-Based Medicine, Oxford).
Collapse
Affiliation(s)
- Julia Zimmer
- National Children's Research Centre, Our Lady's Children's Hospital Crumlin, Gate 5, Dublin 12, Dublin, Ireland
| | - Toshiaki Takahashi
- National Children's Research Centre, Our Lady's Children's Hospital Crumlin, Gate 5, Dublin 12, Dublin, Ireland
| | - Alejandro D Hofmann
- Department of Pediatric Surgery, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Prem Puri
- National Children's Research Centre, Our Lady's Children's Hospital Crumlin, Gate 5, Dublin 12, Dublin, Ireland; School of Medicine and Medical Science and Conway Institute of Biomedical Research, University College Dublin, Belfield, Dublin 4, Dublin, Ireland.
| |
Collapse
|
45
|
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: 236] [Impact Index Per Article: 29.5] [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
|
46
|
Gariboldi V, Vairo D, Guieu R, Marlingue M, Ravis E, Lagier D, Mari A, Thery E, Collart F, Gaudry M, Bonello L, Paganelli F, Condo J, Kipson N, Fenouillet E, Ruf J, Mottola G. Expressions of adenosine A 2A receptors in coronary arteries and peripheral blood mononuclear cells are correlated in coronary artery disease patients. Int J Cardiol 2016; 230:427-431. [PMID: 28041708 DOI: 10.1016/j.ijcard.2016.12.089] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 11/15/2016] [Accepted: 12/16/2016] [Indexed: 01/02/2023]
Abstract
BACKGROUND Altered coronary blood flow occurs in patients with coronary artery disease (CAD). Adenosine strongly impacts blood flow mostly via adenosine A2A receptor (A2AR) expressed in coronary tissues. As part of a systemic regulation of the adenosinergic system, we compared A2AR expression in situ, and on peripheral blood mononuclear cells (PBMC) in CAD patients. METHODS AND RESULTS Aortic and coronary tissues, and PBMC were sampled in 20 CAD patients undergoing coronary artery bypass surgery and consecutively included. Controls were PBMC obtained from 15 healthy subjects. Expression and activity of A2AR were studied by Western blotting and cAMP measurement, respectively. A2AR expression on PBMC was lower in patients than in controls (0.83±0.31 vs 1.2±0.35 arbitrary units; p<0.01), and correlated with A2AR expression in coronary and aortic tissues (Pearson's r: 0.77 and 0.59, p<0.01, respectively). Basal and maximal cAMP productions following agonist stimulation of PBMC were significantly lower in patients than in controls (120±42 vs 191±65 and 360±113 vs 560±215pg/106 cells, p<0.05, respectively). In CAD patients, the increase from basal to maximal cAMP production in PBMC and aortic tissues was similar (+300% and +246%, respectively). CONCLUSION Expression of A2AR on PBMC correlated with those measured in coronary artery and aortic tissues in CAD patients, A2AR activity of PBMC matched that observed in aorta, and A2AR expression and activity in PBMC were found reduced as compared to controls. Measuring the expression level of A2AR on PBMC represents a good tool to address in situ expression in coronary tissues of CAD patients.
Collapse
Affiliation(s)
- Vlad Gariboldi
- Department of Cardiac Surgery, Timone University Hospital, Marseille, France
| | - Donato Vairo
- UMR MD2, Aix-Marseille University and IRBA, Marseille, France
| | - Régis Guieu
- UMR MD2, Aix-Marseille University and IRBA, Marseille, France; Laboratory of Biochemistry, Timone University Hospital, Marseille, France.
| | - Marion Marlingue
- Laboratory of Biochemistry, Timone University Hospital, Marseille, France
| | - Eléonore Ravis
- Department of Cardiology, Timone University Hospital, Marseille, France
| | - David Lagier
- Department of Anesthesia, Timone University Hospital, Marseille, France
| | - Alissa Mari
- UMR MD2, Aix-Marseille University and IRBA, Marseille, France
| | - Elsa Thery
- Laboratory of Biochemistry, Timone University Hospital, Marseille, France
| | - Frédéric Collart
- Department of Cardiac Surgery, Timone University Hospital, Marseille, France
| | - Marine Gaudry
- Department of Vascular Surgery, Timone University Hospital, Marseille, France
| | - Laurent Bonello
- Department of Cardiology, North University Hospital, Marseille, France
| | - Franck Paganelli
- Department of Cardiology, North University Hospital, Marseille, France
| | - Jocelyne Condo
- UMR MD2, Aix-Marseille University and IRBA, Marseille, France
| | - Nathalie Kipson
- UMR MD2, Aix-Marseille University and IRBA, Marseille, France
| | | | - Jean Ruf
- UMR MD2, Aix-Marseille University and IRBA, Marseille, France
| | - Giovanna Mottola
- UMR MD2, Aix-Marseille University and IRBA, Marseille, France; Laboratory of Biochemistry, Timone University Hospital, Marseille, France
| |
Collapse
|
47
|
Tsvetkov D, Kaßmann M, Tano JY, Chen L, Schleifenbaum J, Voelkl J, Lang F, Huang Y, Gollasch M. Do K V 7.1 channels contribute to control of arterial vascular tone? Br J Pharmacol 2016; 174:150-162. [PMID: 28000293 DOI: 10.1111/bph.13665] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 10/11/2016] [Accepted: 10/28/2016] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND AND PURPOSE KV 7.1 voltage-gated potassium channels are expressed in vascular smooth muscle cells (VSMC) of diverse arteries, including mesenteric arteries. Based on pharmacological evidence using R-L3 (KV 7.1 channel opener), HMR1556, chromanol 293B (KV 7.1 channel blockers), stimulation of these channels has been suggested to evoke profound relaxation in various vascular beds of rats. However, the specificity of these drugs in vivo is uncertain. EXPERIMENTAL APPROACH We used Kcnq1-/- mice and pharmacological tools to determine whether KV 7.1 channels play a role in the regulation of arterial tone. KEY RESULTS R-L3 produced similar concentration-dependent relaxations (EC50 ~ 1.4 μM) of arteries from wild-type (Kcnq1+/+ ) and Kcnq1-/- mice, pre-contracted with either phenylephrine or 60 mM KCl. This relaxation was not affected by 10 μM chromanol 293B, 10 μM HMR1556 or 30 μM XE991 (pan-KV 7 channel blocker). The anti-contractile effects of the perivascular adipose tissue (PVAT) were normal in Kcnq1-/- arteries. Chromanol 293B and HMR1556 did not affect the anti-contractile effects of (PVAT). Isolated VSMCs from Kcnq1-/- mice exhibited normal peak KV currents. The KV 7.2-5 channel opener retigabine caused similar relaxations in Kcnq1-/- and wild-type vessels. CONCLUSION AND IMPLICATIONS We conclude that KV 7.1 channels were apparently not involved in the control of arterial tone by α1 -adrenoceptor agonists and PVAT. In addition, R-L3 is an inappropriate pharmacological tool for studying the function of native vascular KV 7.1 channels in mice.
Collapse
Affiliation(s)
- Dmitry Tsvetkov
- Experimental and Clinical Research Center (ECRC), A Joint Cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.,Department of Pharmacology and Experimental Therapy, Institute of Experimental and Clinical Pharmacology and Toxicology, Eberhard Karls University Hospitals and Clinics and Interfaculty Center of Pharmacogenomics and Drug Research, University of Tübingen, Tübingen, Germany
| | - Mario Kaßmann
- Experimental and Clinical Research Center (ECRC), A Joint Cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Jean-Yves Tano
- Experimental and Clinical Research Center (ECRC), A Joint Cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Lan Chen
- Experimental and Clinical Research Center (ECRC), A Joint Cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.,Xiamen Zhongshan Hospital, Xiamen University, Xiamen, China
| | - Johanna Schleifenbaum
- Experimental and Clinical Research Center (ECRC), A Joint Cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Jakob Voelkl
- Department of Cardiology, Vascular Medicine and Physiology, University of Tübingen, Tübingen, Germany
| | - Florian Lang
- Department of Cardiology, Vascular Medicine and Physiology, University of Tübingen, Tübingen, Germany
| | - Yu Huang
- School of Biomedical Sciences, 223A, Lo Kwee-Seong Integrated Biomedical Sciences Building, Area 39, Chinese University of Hong Kong, Sha Tin, Hong Kong
| | - Maik Gollasch
- Experimental and Clinical Research Center (ECRC), A Joint Cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.,Medical Clinic for Nephrology and Internal Intensive Care, Campus Virchow, Charité University Medicine, Berlin, Germany
| |
Collapse
|
48
|
Stott JB, Barrese V, Greenwood IA. Kv7 Channel Activation Underpins EPAC-Dependent Relaxations of Rat Arteries. Arterioscler Thromb Vasc Biol 2016; 36:2404-2411. [PMID: 27789473 DOI: 10.1161/atvbaha.116.308517] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 10/11/2016] [Indexed: 01/27/2023]
Abstract
OBJECTIVE To establish the role of Kv7 channels in EPAC (exchange protein directly activated by cAMP)-dependent relaxations of the rat vasculature and to investigate whether this contributes to β-adrenoceptor-mediated vasorelaxations. APPROACH AND RESULTS Isolated rat renal and mesenteric arteries (RA and MA, respectively) were used for isometric tension recording to study the relaxant effects of a specific EPAC activator and the β-adrenoceptor agonist isoproterenol in the presence of potassium channel inhibitors and cell signaling modulators. Isolated myocytes were used in proximity ligation assay studies to detect localization of signaling intermediaries with Kv7.4 before and after cell stimulation. Our studies showed that the EPAC activator (8-pCPT-2Me-cAMP-AM) produced relaxations and enhanced currents of MA and RA that were sensitive to linopirdine (Kv7 inhibitor). Linopirdine also inhibited isoproterenol-mediated relaxations in both RA and MA. In the MA, isoproterenol relaxations were sensitive to EPAC inhibition, but not protein kinase A inhibition. In contrast, isoproterenol relaxations in RA were attenuated by protein kinase A but not by EPAC inhibition. Proximity ligation assay showed a localization of Kv7.4 with A-kinase anchoring protein in both vessels in the basal state, which increased only in the RA with isoproterenol stimulation. In the MA, but not the RA, a localization of Kv7.4 with both Rap1a and Rap2 (downstream of EPAC) increased with isoproterenol stimulation. CONCLUSIONS EPAC-dependent vasorelaxations occur in part via activation of Kv7 channels. This contributes to the isoproterenol-mediated relaxation in mesenteric, but not renal, arteries.
Collapse
Affiliation(s)
- Jennifer B Stott
- From the Vascular Biology Research Group, Institute for Cardiovascular and Cell Sciences, St George's University of London, UK
| | - Vincenzo Barrese
- From the Vascular Biology Research Group, Institute for Cardiovascular and Cell Sciences, St George's University of London, UK
| | - Iain A Greenwood
- From the Vascular Biology Research Group, Institute for Cardiovascular and Cell Sciences, St George's University of London, UK.
| |
Collapse
|
49
|
Ebrahimiadib N, Modjtahedi BS, Davoudi S, Foster CS. Treatment of Serpiginous Choroiditis with Chlorambucil: A Report of 17 Patients. Ocul Immunol Inflamm 2016; 26:228-238. [PMID: 27754769 DOI: 10.1080/09273948.2016.1214737] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
PURPOSE To evaluate the efficacy of chlorambucil in the treatment of serpiginous choroiditis. METHODS Patient records from the Massachusetts Eye Research and Surgery Institution (MERSI) were reviewed from over the past 10 years. In total, 17 patients with the diagnosis of serpiginous choroiditis treated with chlorambucil were identified. QuantiFERON gold was negative in all of them. Chlorambucil was started at 0.15 mg/kg and dosage was titrated up using weekly white blood cell (WBC) count to achieve a target cell number of 3.0-4.5 × 109 cells/L. The goal of therapy was to maintain this value for at least 6-9 months. Adverse effects, recurrence, rate of new choroidal neovascularization (CNVM), and visual acuity before and after treatment were recorded. RESULTS The mean age of the 17 patients with the diagnosis of serpiginous choroiditis treated with chlorambucil was 46 years, and six patients (35%) were male. The mean duration of treatment for chlorambucil was 8.4 months. None of them developed cancer or persistent side-effects, with a mean follow-up of 53 months. Of the patients, 12 (71%) achieved an average of 45 (5-120) months drug-free remission in their last follow-up. Visual acuity of 33 treated eyes remained within two lines of Snellen acuity in 27 eyes (82%), improved in one eye (3%), and deteriorated in five eyes (15%). Leukopenia was the most common side-effect, which was reversible in all cases. CONCLUSIONS Chlorambucil in a relatively short duration of time, with an escalating dose guided by weekly WBC was well tolerated, as well as effective in preventing recurrence and maintaining vision in patients with serpiginous choroiditis.
Collapse
Affiliation(s)
- Nazanin Ebrahimiadib
- a Massachusetts Eye Research and Surgery Institute , Waltham , Massachusetts , USA.,b Ocular Immunology and Uveitis Foundation , Weston , Massachusetts , USA
| | - Bobeck S Modjtahedi
- c Retina Service, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary , Harvard Medical School , Boston , Massachusetts , USA
| | - Samaneh Davoudi
- c Retina Service, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary , Harvard Medical School , Boston , Massachusetts , USA
| | - C Stephen Foster
- a Massachusetts Eye Research and Surgery Institute , Waltham , Massachusetts , USA.,b Ocular Immunology and Uveitis Foundation , Weston , Massachusetts , USA.,d Department of Ophthalmology , Harvard Medical School , Boston , Massachusetts , USA
| |
Collapse
|
50
|
Abbott GW, Jepps TA. Kcne4 Deletion Sex-Dependently Alters Vascular Reactivity. J Vasc Res 2016; 53:138-148. [PMID: 27710966 DOI: 10.1159/000449060] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Accepted: 08/10/2016] [Indexed: 01/06/2023] Open
Abstract
Voltage-gated potassium (Kv) channels formed by Kv7 (KCNQ) α-subunits are recognized as crucial for vascular smooth muscle function, in addition to their established roles in the heart (Kv7.1) and the brain (Kv7.2-5). In vivo, Kv7 α-subunits are often regulated by KCNE subfamily ancillary (β) subunits. We investigated the effects of targeted germline Kcne4 deletion on mesenteric artery reactivity in adult male and female mice. Kcne4 deletion increased mesenteric artery contractility in response to α-adrenoceptor agonist methoxamine, and decreased responses to Kv7.2-7.5 channel activator ML213, in male but not female mice. In contrast, Kcne4 deletion markedly decreased vasorelaxation in response to isoprenaline in both male and female mice. Kcne4 expression was 2-fold lower in the female versus the male mouse mesenteric artery, and Kcne4 deletion elicited only moderate changes of other Kcne transcripts, with no striking sex-specific differences. However, Kv7.4 protein expression in females was twice that in males, and was reduced in both sexes by Kcne4 deletion. Our findings confirm a crucial role for KCNE4 in regulation of Kv7 channel activity to modulate vascular tone, and provide the first known molecular mechanism for sex-specificity of this modulation that has important implications for vascular reactivity and may underlie sex-specific susceptibility to cardiovascular diseases.
Collapse
Affiliation(s)
- Geoffrey W Abbott
- Bioelectricity Laboratory, Departments of Pharmacology and Physiology and Biophysics, School of Medicine, University of California, Irvine, Calif., USA
| | | |
Collapse
|