1
|
Heo R, Park M, Mun SY, Zhuang W, Jeong J, Park H, Han ET, Han JH, Chun W, Jung WK, Choi IW, Park WS. Vasorelaxant mechanisms of the antidiabetic anagliptin in rabbit aorta: roles of Kv channels and SERCA pump. Acta Diabetol 2025; 62:241-251. [PMID: 39103505 DOI: 10.1007/s00592-024-02351-9] [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: 04/11/2024] [Accepted: 07/29/2024] [Indexed: 08/07/2024]
Abstract
AIMS The present study investigated the vasorelaxant mechanisms of an oral antidiabetic drug, anagliptin, using phenylephrine (Phe)-induced pre-contracted rabbit aortic rings. METHODS Arterial tone measurement was performed in rabbit thoracic aortic rings. RESULTS Anagliptin induced vasorelaxation in a dose-dependent manner. Pre-treatment with the classical voltagedependent K+ (Kv) channel inhibitors 4-aminopyridine and tetraethylammonium significantly decreased the vasorelaxant effect of anagliptin, whereas pre-treatment with the inwardly rectifying K+ (Kir) channel inhibitor Ba2+, the ATP-sensitive K+ (KATP) channel inhibitor glibenclamide, and the large-conductance Ca2+-activated K+ (BKCa) channel inhibitor paxilline did not attenuate the vasorelaxant effect. Furthermore, the vasorelaxant response of anagliptin was effectively inhibited by pre-treatment with the sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) pump inhibitors thapsigargin and cyclopiazonic acid. Neither cAMP/protein kinase A (PKA)-related signaling pathway inhibitors (adenylyl cyclase inhibitor SQ 22536 and PKA inhibitor KT 5720) nor cGMP/protein kinase G (PKG)-related signaling pathway inhibitors (guanylyl cyclase inhibitor ODQ and PKG inhibitor KT 5823) reduced the vasorelaxant effect of anagliptin. Similarly, the anagliptin-induced vasorelaxation was independent of the endothelium. CONCLUSIONS Based on these results, we suggest that anagliptin-induced vasorelaxation in rabbit aortic smooth muscle occurs by activating Kv channels and the SERCA pump, independent of other vascular K+ channels, cAMP/PKA- or cGMP/PKG-related signaling pathways, and the endothelium.
Collapse
Affiliation(s)
- Ryeon Heo
- Department of Physiology, Seoul National University College of Medicine, Seoul, 03080, South Korea
| | - Minju Park
- Department of Physiology, Institute of Medical Sciences, Kangwon National University School of Medicine, 1 Kangwondaehak-Gil, Chuncheon, 24341, South Korea
| | - Seo-Yeong Mun
- Department of Physiology, Institute of Medical Sciences, Kangwon National University School of Medicine, 1 Kangwondaehak-Gil, Chuncheon, 24341, South Korea
| | - Wenwen Zhuang
- Department of Physiology, Institute of Medical Sciences, Kangwon National University School of Medicine, 1 Kangwondaehak-Gil, Chuncheon, 24341, South Korea
| | - Junsu Jeong
- Department of Physiology, Institute of Medical Sciences, Kangwon National University School of Medicine, 1 Kangwondaehak-Gil, Chuncheon, 24341, South Korea
| | - Hongzoo Park
- Department of Urology, Institute of Medical Sciences, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Jin-Hee Han
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Wanjoo Chun
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Won-Kyo Jung
- Department of Biomedical Engineering, and Center for Marine-Integrated Biomedical Technology (BK21 Plus), Pukyong National University, Busan, 48513, South Korea
| | - Il-Whan Choi
- Department of Microbiology, College of Medicine, Inje University, Busan, 48516, South Korea
| | - Won Sun Park
- Department of Physiology, Institute of Medical Sciences, Kangwon National University School of Medicine, 1 Kangwondaehak-Gil, Chuncheon, 24341, South Korea.
| |
Collapse
|
2
|
Casado-Arroyo R, Bernardi M, Sabouret P, Franculli G, Tamargo J, Spadafora L, Lellouche N, Biondi-Zoccai G, Toth PP, Banach M. Investigative agents for atrial fibrillation: agonists and stimulants, progress and expectations. Expert Opin Investig Drugs 2024; 33:967-978. [PMID: 39096248 DOI: 10.1080/13543784.2024.2388583] [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: 05/26/2024] [Revised: 07/10/2024] [Accepted: 08/01/2024] [Indexed: 08/05/2024]
Abstract
INTRODUCTION Atrial fibrillation (AF) is the most common type of cardiac arrhythmia. Its prevalence has increased due to worldwide populations that are aging in combination with the growing incidence of risk factors associated. Recent advances in our understanding of AF pathophysiology and the identification of nodal players involved in AF-promoting atrial remodeling highlights potential opportunities for new therapeutic approaches. AREAS COVERED This detailed review summarizes recent developments in the field antiarrhythmic drugs in the field AF. EXPERT OPINION The current situation is far than optimal. Despite clear unmet needs in drug development in the field of AF treatment, the current development of new drugs is absent. The need for a molecule with absence of cardiac and non-cardiac toxicity in the short and long term is a limitation in the field. Improvement in the understanding of AF genetics, pathophysiology, molecular alterations, big data and artificial intelligence with the objective to provide a personalized AF treatment will be the cornerstone of AF treatment in the coming years.
Collapse
Affiliation(s)
- Ruben Casado-Arroyo
- Department of Cardiology, H.U.B.-Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Marco Bernardi
- Department of Clinical, Internal Medicine, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Pierre Sabouret
- Heart Institute, ACTION Study Group-CHU Pitié-Salpétrière Paris, Paris, France
- Collège National des Cardiologues Français (CNCF), Paris, France
| | - Giuseppe Franculli
- Department of Clinical, Internal Medicine, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Juan Tamargo
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense, Instituto De Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Luigi Spadafora
- Department of Clinical, Internal Medicine, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Nicolas Lellouche
- Service de Cardiologie, AP-HP, University Hospital Henri Mondor, Créteil, France
| | - Giuseppe Biondi-Zoccai
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, Italy
| | - Peter P Toth
- CGH Medical Center, Sterling, IL, USA
- Cicarrone Center for the Prevention of Cardiovascular Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Maciej Banach
- Department of Preventive Cardiology and Lipidology, Medical University of Lodz Lodz Poland, Lodz, Poland
- Department of Cardiology and Congenital Diseases of Adults, Polish Mother's Memorial Hospital Research Institute Lodz Poland, Lodz, Poland
| |
Collapse
|
3
|
Mazhar F, Bartolucci C, Regazzoni F, Paci M, Dedè L, Quarteroni A, Corsi C, Severi S. A detailed mathematical model of the human atrial cardiomyocyte: integration of electrophysiology and cardiomechanics. J Physiol 2024; 602:4543-4583. [PMID: 37641426 DOI: 10.1113/jp283974] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 08/02/2023] [Indexed: 08/31/2023] Open
Abstract
Mechano-electric regulations (MER) play an important role in the maintenance of cardiac performance. Mechano-calcium and mechano-electric feedback (MCF and MEF) pathways adjust the cardiomyocyte contractile force according to mechanical perturbations and affects electro-mechanical coupling. MER integrates all these regulations in one unit resulting in a complex phenomenon. Computational modelling is a useful tool to accelerate the mechanistic understanding of complex experimental phenomena. We have developed a novel model that integrates the MER loop for human atrial cardiomyocytes with proper consideration of feedforward and feedback pathways. The model couples a modified version of the action potential (AP) Koivumäki model with the contraction model by Quarteroni group. The model simulates iso-sarcometric and isometric twitches and the feedback effects on AP and Ca2+-handling. The model showed a biphasic response of Ca2+ transient (CaT) peak to increasing pacing rates and highlights the possible mechanisms involved. The model has shown a shift of the threshold for AP and CaT alternans from 4.6 to 4 Hz under post-operative atrial fibrillation, induced by depressed SERCA activity. The alternans incidence was dependent on a chain of mechanisms including RyRs availability time, MCF coupling, CaMKII phosphorylation, and the stretch levels. As a result, the model predicted a 10% slowdown of conduction velocity for a 20% stretch, suggesting a role of stretch in creation of substrate formation for atrial fibrillation. Overall, we conclude that the developed model provides a physiological CaT followed by a physiological twitch. This model can open pathways for the future studies of human atrial electromechanics. KEY POINTS: With the availability of human atrial cellular data, interest in atrial-specific model integration has been enhanced. We have developed a detailed mathematical model of human atrial cardiomyocytes including the mechano-electric regulatory loop. The model has gone through calibration and evaluation phases against a wide collection of available human in-vitro data. The usefulness of the model for analysing clinical problems has been preliminaryly tested by simulating the increased incidence of Ca2+ transient and action potential alternans at high rates in post-operative atrial fibrillation condition. The model determines the possible role of mechano-electric feedback in alternans incidence, which can increase vulnerability to atrial arrhythmias by varying stretch levels. We found that our physiologically accurate description of Ca2+ handling can reproduce many experimental phenomena and can help to gain insights into the underlying pathophysiological mechanisms.
Collapse
Affiliation(s)
- Fazeelat Mazhar
- Department of Electrical, Electronic and Information Engineering 'Guglielmo Marconi', University of Bologna, Cesena, Italy
| | - Chiara Bartolucci
- Department of Electrical, Electronic and Information Engineering 'Guglielmo Marconi', University of Bologna, Cesena, Italy
| | | | - Michelangelo Paci
- Department of Electrical, Electronic and Information Engineering 'Guglielmo Marconi', University of Bologna, Cesena, Italy
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Luca Dedè
- MOX - Dipartimento di Matematica, Politecnico di Milano, Milan, Italy
| | - Alfio Quarteroni
- MOX - Dipartimento di Matematica, Politecnico di Milano, Milan, Italy
- Mathematics Institute, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Cristiana Corsi
- Department of Electrical, Electronic and Information Engineering 'Guglielmo Marconi', University of Bologna, Cesena, Italy
| | - Stefano Severi
- Department of Electrical, Electronic and Information Engineering 'Guglielmo Marconi', University of Bologna, Cesena, Italy
| |
Collapse
|
4
|
Schulz C, Sönmez M, Krause J, Schwedhelm E, Bangfen P, Alihodzic D, Hansen A, Eschenhagen T, Christ T. A critical role of retinoic acid concentration for the induction of a fully human-like atrial action potential phenotype in hiPSC-CM. Stem Cell Reports 2023; 18:2096-2107. [PMID: 37922915 PMCID: PMC10679650 DOI: 10.1016/j.stemcr.2023.10.006] [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: 07/27/2022] [Revised: 10/05/2023] [Accepted: 10/09/2023] [Indexed: 11/07/2023] Open
Abstract
Retinoic acid (RA) induces an atrial phenotype in human induced pluripotent stem cells (hiPSCs), but expression of atrium-selective currents such as the ultrarapid (IKur) and acetylcholine-stimulated K+ current is variable and less than in the adult human atrium. We suspected methodological issues and systematically investigated the concentration dependency of RA. RA treatment increased IKur concentration dependently from 1.1 ± 0.54 pA/pF (0 RA) to 3.8 ± 1.1, 5.8 ± 2.5, and 12.2 ± 4.3 at 0.01, 0.1, and 1 μM, respectively. Only 1 μM RA induced enough IKur to fully reproduce human atrial action potential (AP) shape and a robust shortening of APs upon carbachol. We found that sterile filtration caused substantial loss of RA. We conclude that 1 μM RA seems to be necessary and sufficient to induce a full atrial AP shape in hiPSC-CM in EHT format. RA concentrations are prone to methodological issues and may profoundly impact the success of atrial differentiation.
Collapse
Affiliation(s)
- Carl Schulz
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Muhammed Sönmez
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Julia Krause
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany; Department of Cardiology, University Heart and Vascular Center, Hamburg, Germany
| | - Edzard Schwedhelm
- German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany; Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Pan Bangfen
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Dzenefa Alihodzic
- Hospital Pharmacy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Arne Hansen
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Thomas Eschenhagen
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany.
| | - Torsten Christ
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; German Centre for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany.
| |
Collapse
|
5
|
Här K, Lysenko NN, Dimitrova D, Schlüter T, Zavaritskaya O, Kamkin AG, Mladenov M, Grisk O, Köhler R, Gagov H, Schubert R. Kv2.1 Channels Prevent Vasomotion and Safeguard Myogenic Reactivity in Rat Small Superior Cerebellar Arteries. Cells 2023; 12:1989. [PMID: 37566068 PMCID: PMC10416909 DOI: 10.3390/cells12151989] [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: 07/06/2023] [Revised: 07/27/2023] [Accepted: 07/29/2023] [Indexed: 08/12/2023] Open
Abstract
Vascular smooth muscle voltage-gated potassium (Kv) channels have been proposed to contribute to myogenic autoregulation. Surprisingly, in initial experiments, we observed that the Kv2 channel inhibitor stromatoxin induced vasomotion without affecting myogenic tone. Thus, we tested the hypothesis that Kv2 channels contribute to myogenic autoregulation by fine-tuning the myogenic response. Expression of Kv2 channel mRNA was determined using real-time PCR and 'multiplex' single-cell RT-PCR. Potassium currents were measured using the patch-clamp technique. Contractile responses of intact arteries were studied using isobaric myography. Expression of Kv2.1 but not Kv2.2 channels was detected in intact rat superior cerebellar arteries and in single smooth muscle cells. Stromatoxin, a high-affinity inhibitor of Kv2 channels, reduced smooth muscle Kv currents by 61% at saturating concentrations (EC50 36 nmol/L). Further, stromatoxin (10-100 nmol/L) induced pronounced vasomotion in 48% of the vessels studied. In vessels not exhibiting vasomotion, stromatoxin did not affect myogenic reactivity. Notably, in vessels exhibiting stromatoxin-induced vasomotion, pressure increases evoked two effects: First, they facilitated the occurrence of random vasodilations and/or vasoconstrictions, disturbing the myogenic response (24% of the vessels). Second, they modified the vasomotion by decreasing its amplitude and increasing its frequency, thereby destabilizing myogenic tone (76% of the vessels). Our study demonstrates that (i) Kv2.1 channels are the predominantly expressed Kv channels in smooth muscle cells of rat superior cerebellar arteries, and (ii) Kv2.1 channels provide a novel type of negative feedback mechanism in myogenic autoregulation by preventing vasomotion and thereby safeguarding the myogenic response.
Collapse
Affiliation(s)
- Kristina Här
- European Center of Angioscience (ECAS), Research Division Cardiovascular Physiology, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Natalia N. Lysenko
- European Center of Angioscience (ECAS), Research Division Cardiovascular Physiology, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- Department of Physiology, N. I. Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Daniela Dimitrova
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria
| | - Torsten Schlüter
- Institute of Physiology, Universitätsmedizin Greifswald, 17475 Greifswald, Germany
| | - Olga Zavaritskaya
- European Center of Angioscience (ECAS), Research Division Cardiovascular Physiology, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Andrej G. Kamkin
- Department of Physiology, N. I. Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Mitko Mladenov
- Department of Physiology, N. I. Pirogov Russian National Research Medical University, 117997 Moscow, Russia
- Institute of Biology, Faculty of Natural Sciences and Mathematics, University of Ss. Cyril and Methodius, 1000 Skopje, North Macedonia
| | - Olaf Grisk
- Institute of Physiology, Brandenburg Medical School Theodor Fontane, 16816 Neuruppin, Germany
| | - Ralf Köhler
- ARAID-IACS, UIT University Hospital Miguel Servet, 50009 Zaragoza, Spain
| | - Hristo Gagov
- Department of Animal and Human Physiology, Faculty of Biology, Sofia University ‘St. Kliment Ohridski’, 1164 Sofia, Bulgaria
| | - Rudolf Schubert
- European Center of Angioscience (ECAS), Research Division Cardiovascular Physiology, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
- Physiology, Institute of Theoretical Medicine, Faculty of Medicine, University of Augsburg, Universitätsstrasse 2, 86159 Augsburg, Germany
| |
Collapse
|
6
|
Zhou HF, Li WY, Wu Q, Ren J, Peng LY, Li XN, Zhao QS. Discovery and Biomimetic Semisynthesis of Spirophyllines A-D from Uncaria rhynchophylla. Org Lett 2023; 25:4434-4438. [PMID: 37288843 DOI: 10.1021/acs.orglett.3c01342] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Spirophyllines A-D (1-4), four new spirooxindole alkaloids all characterized by the spiro[pyrrolidin-3,3'-oxindole] core and a rare isoxazolidine ring, were isolated from Uncaria rhynchophylla. Their structures were determined by spectroscopic methods and confirmed by X-ray crystallography. Based on the biomimetic semisynthesis strategy, compounds 1-8 were synthesized in three steps via the key reactions of 1,3-dipolar cycloaddition and Krapcho decarboxylation from corynoxeine. Interestingly, compound 3 showed moderate inhibitory activity against the Kv1.5 potassium channel (IC50 = 9.1 μM).
Collapse
Affiliation(s)
- Hao-Feng Zhou
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, Yunnan 650201, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Wen-Yan Li
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, Yunnan 650201, People's Republic of China
| | - Qi Wu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, Yunnan 650201, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Jian Ren
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, Yunnan 650201, People's Republic of China
| | - Li-Yan Peng
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, Yunnan 650201, People's Republic of China
| | - Xiao-Nian Li
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, Yunnan 650201, People's Republic of China
| | - Qin-Shi Zhao
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, Yunnan 650201, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| |
Collapse
|
7
|
Al-Owais MM, Hettiarachchi NT, Dallas ML, Scragg JL, Lippiat JD, Holden AV, Steele DS, Peers C. Inhibition of the voltage-gated potassium channel Kv1.5 by hydrogen sulfide attenuates remodeling through S-nitrosylation-mediated signaling. Commun Biol 2023; 6:651. [PMID: 37336943 PMCID: PMC10279668 DOI: 10.1038/s42003-023-05016-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 06/05/2023] [Indexed: 06/21/2023] Open
Abstract
The voltage-gated K+ channel plays a key role in atrial excitability, conducting the ultra-rapid rectifier K+ current (IKur) and contributing to the repolarization of the atrial action potential. In this study, we examine its regulation by hydrogen sulfide (H2S) in HL-1 cardiomyocytes and in HEK293 cells expressing human Kv1.5. Pacing induced remodeling resulted in shorting action potential duration, enhanced both Kv1.5 channel and H2S producing enzymes protein expression in HL-1 cardiomyocytes. H2S supplementation reduced these remodeling changes and restored action potential duration through inhibition of Kv1.5 channel. H2S also inhibited recombinant hKv1.5, lead to nitric oxide (NO) mediated S-nitrosylation and activated endothelial nitric oxide synthase (eNOS) by increased phosphorylation of Ser1177, prevention of NO formation precluded these effects. Regulation of Ikur by H2S has important cardiovascular implications and represents a novel and potential therapeutic target.
Collapse
Affiliation(s)
- Moza M Al-Owais
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK.
| | - Nishani T Hettiarachchi
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, University of Leeds, Leeds, LS2 9JT, UK
| | - Mark L Dallas
- Reading School of Pharmacy, University of Reading, Reading, RG6 6UB, UK
| | - Jason L Scragg
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, University of Leeds, Leeds, LS2 9JT, UK
| | - Jonathan D Lippiat
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Arun V Holden
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Derek S Steele
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Chris Peers
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, University of Leeds, Leeds, LS2 9JT, UK
| |
Collapse
|
8
|
Heo R, Kang M, Mun SY, Park M, Han ET, Han JH, Chun W, Park H, Jung WK, Choi IW, Park WS. Antidiabetic omarigliptin dilates rabbit aorta by activating voltage-dependent K + channels and the sarco/endoplasmic reticulum Ca 2+ -ATPase pump. Fundam Clin Pharmacol 2023; 37:75-84. [PMID: 36093990 DOI: 10.1111/fcp.12831] [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: 07/10/2022] [Revised: 09/01/2022] [Accepted: 09/09/2022] [Indexed: 01/25/2023]
Abstract
We investigated the vasodilatory effect of omarigliptin, an oral antidiabetic drug in the dipeptidyl peptidase-4 inhibitor class, and its related mechanisms using phenylephrine (Phe)-induced pre-contracted aortic rings. Omarigliptin dilated aortic rings pre-constricted with Phe in a dose-dependent manner. Pretreatment with the voltage-dependent K+ channel inhibitor 4-aminopyridine significantly attenuated the vasodilatory effect of omarigliptin, whereas pretreatment with the inwardly rectifying K+ channel inhibitor Ba2+ , ATP-sensitive K+ channel inhibitor glibenclamide, and large-conductance Ca2+ -activated K+ channel inhibitor paxilline did not alter its vasodilation. Pretreatment with the sarco/endoplasmic reticulum Ca2+ -ATPase (SERCA) pump inhibitors thapsigargin and cyclopiazonic acid significantly reduced the vasodilatory effect of omarigliptin. Neither cAMP/PKA-related signaling pathway inhibitors nor cGMP/PKG-related signaling pathway inhibitors modulated the vasodilatory effect of omarigliptin. Removal of endothelium did not diminish the vasodilatory effect of omarigliptin. Furthermore, pretreatment with the nitric oxide synthase inhibitor L-NAME or small-conductance Ca2+ -activated K+ channel inhibitor apamin, together with the intermediate-conductance Ca2+ -activated K+ channel inhibitor TRAM-34, did not influence the vasodilatory effect of omarigliptin. In conclusion, omarigliptin induced vasodilation in rabbit aortic smooth muscle by activating voltage-dependent K+ channels and the SERCA pump independently of other K+ channels, cAMP/PKA- and cGMP/PKG-related signaling pathways, and the endothelium.
Collapse
Affiliation(s)
- Ryeon Heo
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Minji Kang
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Seo-Yeong Mun
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Minju Park
- Institute of Medical Sciences, Department of Physiology, 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
| | - Hongzoo Park
- Institute of Medical Sciences, Department of Urology, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Won-Kyo Jung
- Department of Biomedical Engineering, and Center for Marine-Integrated Biomedical Technology (BK21 Plus), Pukyong National University, Busan, South Korea
| | - Il-Whan Choi
- Department of Microbiology, College of Medicine, Inje University, Busan, South Korea
| | - Won Sun Park
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon, South Korea
| |
Collapse
|
9
|
Kang M, Heo R, Park S, Mun SY, Park M, Han ET, Han JH, Chun W, Ha KS, Park H, Jung WK, Choi IW, Park WS. Inhibitory effects of the atypical antipsychotic, clozapine, on voltage-dependent K + channels in rabbit coronary arterial smooth muscle cells. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2022; 26:277-285. [PMID: 35766005 PMCID: PMC9247706 DOI: 10.4196/kjpp.2022.26.4.277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/10/2022] [Accepted: 05/16/2022] [Indexed: 06/15/2023]
Abstract
To investigate the adverse effects of clozapine on cardiovascular ion channels, we examined the inhibitory effect of clozapine on voltage-dependent K+ (Kv) channels in rabbit coronary arterial smooth muscle cells. Clozapine-induced inhibition of Kv channels occurred in a concentration-dependent manner with an half-inhibitory concentration value of 7.84 ± 4.86 µM and a Hill coefficient of 0.47 ± 0.06. Clozapine did not shift the steady-state activation or inactivation curves, suggesting that it inhibited Kv channels regardless of gating properties. Application of train pulses (1 and 2 Hz) progressively augmented the clozapine-induced inhibition of Kv channels in the presence of the drug. Furthermore, the recovery time constant from inactivation was increased in the presence of clozapine, suggesting that clozapine-induced inhibition of Kv channels is use (state)-dependent. Pretreatment of a Kv1.5 subtype inhibitor decreased the Kv current amplitudes, but additional application of clozapine did not further inhibit the Kv current. Pretreatment with Kv2.1 or Kv7 subtype inhibitors partially blocked the inhibitory effect of clozapine. Based on these results, we conclude that clozapine inhibits arterial Kv channels in a concentrationand use (state)-dependent manner. Kv1.5 is the major subtype involved in clozapine-induced inhibition of Kv channels, and Kv2.1 and Kv7 subtypes are partially involved.
Collapse
Affiliation(s)
- Minji Kang
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon 24341, Korea
| | - Ryeon Heo
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon 24341, Korea
| | - Seojin Park
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon 24341, Korea
| | - Seo-Yeong Mun
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon 24341, Korea
| | - Minju Park
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon 24341, Korea
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon 24341, Korea
| | - Jin-Hee Han
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon 24341, Korea
| | - Wanjoo Chun
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon 24341, Korea
| | - Kwon-Soo Ha
- Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon 24341, Korea
| | - Hongzoo Park
- Institute of Medical Sciences, Department of Urology, Kangwon National University School of Medicine, Chuncheon 24341, Korea
| | - Won-Kyo Jung
- Department of Biomedical Engineering and Center for Marine-Integrated Biomedical Technology (BK21 Plus), Pukyong National University, Busan 48513, Korea
| | - Il-Whan Choi
- Department of Microbiology, College of Medicine, Inje University, Busan 48516, Korea
| | - Won Sun Park
- Institute of Medical Sciences, Department of Physiology, Kangwon National University School of Medicine, Chuncheon 24341, Korea
| |
Collapse
|
10
|
Hasan A, Menon SN, Zerin F, Hasan R. Dapagliflozin induces vasodilation in resistance-size mesenteric arteries by stimulating smooth muscle cell K V7 ion channels. Heliyon 2022; 8:e09503. [PMID: 35647331 PMCID: PMC9131249 DOI: 10.1016/j.heliyon.2022.e09503] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 02/17/2022] [Accepted: 05/16/2022] [Indexed: 02/07/2023] Open
Abstract
Dapagliflozin is a sodium-glucose cotransporter 2 (SGLT2) inhibitor that, in addition to glucose reduction, lowers systemic blood pressure. Here, we investigated if dapagliflozin could directly relax small mesenteric arteries that control peripheral vascular resistance and blood pressure, and the underlying molecular mechanism. We used pressurized arterial myography, pharmacological inhibition and Western blotting to investigate the direct effect of dapagliflozin on the contractility of freshly isolated, resistance-size rat mesenteric arteries. Our pressure myography data unveiled that dapagliflozin relaxed small mesenteric arteries in a concentration-dependent manner. Non-selective inhibition of KV channels and selective inhibition of smooth muscle cell voltage-gated K+ channels KV7 attenuated dapagliflozin-induced vasorelaxation. Inhibition of other major KV isoforms such as KV1.3, KV1.5 channels as well as large-conductance Ca2+-activated K+ (BKCa) channels, ATP-sensitive (KATP) channels did not abolish vasodilation. Dapagliflozin-evoked vasodilation remained unaltered by pharmacological inhibition of endothelium-derived nitric oxide (NO) signaling, prostacyclin (PGI2), as well as by endothelium denudation. Our Western blotting data revealed that SGLT2 protein is expressed in rat mesenteric arteries. However, non-selective inhibition of SGLTs did not induce vasodilation, demonstrating that the vasodilatory action is independent of SGLT2 inhibition. Overall, our data suggests that dapagliflozin directly and selectively stimulates arterial smooth muscle cells KV7 channels, leading to vasodilation in resistance-size mesenteric arteries. These findings are significant as it uncovers for the first time a direct vasodilatory action of dapagliflozin in resistance mesenteric arteries, which may lower systemic blood pressure.
Collapse
Affiliation(s)
- Ahasanul Hasan
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA, United States
| | - Sreelakshmi N. Menon
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA, United States
| | - Farzana Zerin
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA, United States
| | - Raquibul Hasan
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA, United States
| |
Collapse
|
11
|
Bae H, Kim T, Lim I. Carbon monoxide activation of delayed rectifier potassium currents of human cardiac fibroblasts through diverse pathways. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2022; 26:25-36. [PMID: 34965993 PMCID: PMC8723981 DOI: 10.4196/kjpp.2022.26.1.25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/28/2021] [Accepted: 09/28/2021] [Indexed: 06/14/2023]
Abstract
To identify the effect and mechanism of carbon monoxide (CO) on delayed rectifier K+ currents (IK) of human cardiac fibroblasts (HCFs), we used the wholecell mode patch-clamp technique. Application of CO delivered by carbon monoxidereleasing molecule-3 (CORM3) increased the amplitude of outward K+ currents, and diphenyl phosphine oxide-1 (a specific IK blocker) inhibited the currents. CORM3- induced augmentation was blocked by pretreatment with nitric oxide synthase blockers (L-NG-monomethyl arginine citrate and L-NG-nitro arginine methyl ester). Pretreatment with KT5823 (a protein kinas G blocker), 1H-[1,-2,-4] oxadiazolo-[4,-3-a] quinoxalin-1-on (ODQ, a soluble guanylate cyclase blocker), KT5720 (a protein kinase A blocker), and SQ22536 (an adenylate cyclase blocker) blocked the CORM3 stimulating effect on IK. In addition, pretreatment with SB239063 (a p38 mitogen-activated protein kinase [MAPK] blocker) and PD98059 (a p44/42 MAPK blocker) also blocked the CORM3's effect on the currents. When testing the involvement of S-nitrosylation, pretreatment of N-ethylmaleimide (a thiol-alkylating reagent) blocked CO-induced IK activation and DL-dithiothreitol (a reducing agent) reversed this effect. Pretreatment with 5,10,15,20-tetrakis(1-methylpyridinium-4-yl)-21H,23H porphyrin manganese (III) pentachloride and manganese (III) tetrakis (4-benzoic acid) porphyrin chloride (superoxide dismutase mimetics), diphenyleneiodonium chloride (an NADPH oxidase blocker), or allopurinol (a xanthine oxidase blocker) also inhibited CO-induced IK activation. These results suggest that CO enhances IK in HCFs through the nitric oxide, phosphorylation by protein kinase G, protein kinase A, and MAPK, S-nitrosylation and reduction/oxidation (redox) signaling pathways.
Collapse
Affiliation(s)
- Hyemi Bae
- Department of Physiology, College of Medicine, Chung-Ang University, Seoul 06974, Korea
| | - Taeho Kim
- Department of Internal Medicine, College of Medicine, Chung-Ang University Hospital, Seoul 06973, Korea
| | - Inja Lim
- Department of Physiology, College of Medicine, Chung-Ang University, Seoul 06974, Korea
| |
Collapse
|
12
|
Seo MS, An JR, Kang M, Heo R, Park H, Han ET, Han JH, Chun W, Park WS. Mechanisms underlying the vasodilatory effects of canagliflozin in the rabbit thoracic aorta: Involvement of the SERCA pump and Kv channels. Life Sci 2021; 287:120101. [PMID: 34715136 DOI: 10.1016/j.lfs.2021.120101] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 10/13/2021] [Accepted: 10/24/2021] [Indexed: 10/20/2022]
Abstract
AIMS Canagliflozin is an anti-diabetic agent and sodium glucose co-transporter-2 inhibitor. Despite numerous clinical trials demonstrating its beneficial effects on blood pressure, the cellular mechanisms underlying the effects of canagliflozin on vascular reactivity have yet to be clarified. We investigated the vasodilatory effect of canagliflozin on aortic rings isolated from rabbits. MAIN METHODS We used rabbit thoracic aortic rings and its arterial tone was tested by using wire myography system. KEY FINDINGS Canagliflozin caused concentration-dependent vasodilation in aortic rings pre-constricted with phenylephrine or high K+. However, the degree of canagliflozin-induced vasodilation of the aortic rings pre-constricted with high K+ was less than that of rings pre-constricted with phenylephrine. Application of 4-aminopyridine, a voltage-dependent K+ (Kv) channel inhibitor, reduced canagliflozin-induced vasodilation. However, pre-incubation of an inwardly rectifying K+ channel inhibitor, a large-conductance Ca2+-activated K+ channel inhibitor, and an ATP-sensitive K+ inhibitor did not modulate the vasodilatory effects of canagliflozin. Indeed, canagliflozin increased Kv currents in aortic smooth muscle cells. Pre-treatment with thapsigargin or cyclopiazonic acid, a sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) pump inhibitors, reduced the vasodilatory effects of canagliflozin. Conversely, pre-treatment with a Ca2+ channel inhibitor, adenylyl cyclase/PKA inhibitors, and guanylyl cyclase/PKG inhibitors did not modulate the vasodilatory effects of canagliflozin. Endothelium removal, and pre-treatment with the nitric oxide synthase inhibitor L-NAME, and small- and intermediate-conductance Ca2+-activated K+ channel inhibitor apamin and TRAM-34, did not diminish the vasodilatory effects of canagliflozin. SIGNIFICANCE Our results indicate that canagliflozin induces vasodilation, which is dependent on the robust SERCA activity and Kv channel activation.
Collapse
Affiliation(s)
- Mi Seon Seo
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon 24341, South Korea
| | - Jin Ryeol An
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon 24341, South Korea
| | - Minji Kang
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon 24341, South Korea
| | - Ryeon Heo
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon 24341, South Korea
| | - Hongzoo Park
- Department of Urology, Kangwon National University School of Medicine, Chuncheon 24341, South Korea
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon 24341, South Korea
| | - Jin-Hee Han
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon 24341, South Korea
| | - Wanjoo Chun
- Department of Pharmacology, Kangwon National University School of Medicine, Chuncheon 24341, South Korea
| | - Won Sun Park
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon 24341, South Korea.
| |
Collapse
|
13
|
Borrego J, Feher A, Jost N, Panyi G, Varga Z, Papp F. Peptide Inhibitors of Kv1.5: An Option for the Treatment of Atrial Fibrillation. Pharmaceuticals (Basel) 2021; 14:1303. [PMID: 34959701 PMCID: PMC8704205 DOI: 10.3390/ph14121303] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/08/2021] [Accepted: 12/09/2021] [Indexed: 12/12/2022] Open
Abstract
The human voltage gated potassium channel Kv1.5 that conducts the IKur current is a key determinant of the atrial action potential. Its mutations have been linked to hereditary forms of atrial fibrillation (AF), and the channel is an attractive target for the management of AF. The development of IKur blockers to treat AF resulted in small molecule Kv1.5 inhibitors. The selectivity of the blocker for the target channel plays an important role in the potential therapeutic application of the drug candidate: the higher the selectivity, the lower the risk of side effects. In this respect, small molecule inhibitors of Kv1.5 are compromised due to their limited selectivity. A wide range of peptide toxins from venomous animals are targeting ion channels, including mammalian channels. These peptides usually have a much larger interacting surface with the ion channel compared to small molecule inhibitors and thus, generally confer higher selectivity to the peptide blockers. We found two peptides in the literature, which inhibited IKur: Ts6 and Osu1. Their affinity and selectivity for Kv1.5 can be improved by rational drug design in which their amino acid sequences could be modified in a targeted way guided by in silico docking experiments.
Collapse
Affiliation(s)
- Jesús Borrego
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem ter 1, H-4032 Debrecen, Hungary; (J.B.); (A.F.); (G.P.); (Z.V.)
| | - Adam Feher
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem ter 1, H-4032 Debrecen, Hungary; (J.B.); (A.F.); (G.P.); (Z.V.)
| | - Norbert Jost
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, 6725 Szeged, Hungary;
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, 6725 Szeged, Hungary
- ELKH-SZTE Research Group for Cardiovascular Pharmacology, Eötvös Loránd Research Network, 6725 Szeged, Hungary
| | - Gyorgy Panyi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem ter 1, H-4032 Debrecen, Hungary; (J.B.); (A.F.); (G.P.); (Z.V.)
| | - Zoltan Varga
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem ter 1, H-4032 Debrecen, Hungary; (J.B.); (A.F.); (G.P.); (Z.V.)
| | - Ferenc Papp
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Egyetem ter 1, H-4032 Debrecen, Hungary; (J.B.); (A.F.); (G.P.); (Z.V.)
| |
Collapse
|
14
|
Hasan A, Hasan R. Empagliflozin Relaxes Resistance Mesenteric Arteries by Stimulating Multiple Smooth Muscle Cell Voltage-Gated K + (K V) Channels. Int J Mol Sci 2021; 22:10842. [PMID: 34639181 PMCID: PMC8509755 DOI: 10.3390/ijms221910842] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/26/2021] [Accepted: 10/04/2021] [Indexed: 12/31/2022] Open
Abstract
The antidiabetic drug empagliflozin is reported to produce a range of cardiovascular effects, including a reduction in systemic blood pressure. However, whether empagliflozin directly modulates the contractility of resistance-size mesenteric arteries remains unclear. Here, we sought to investigate if empagliflozin could relax resistance-size rat mesenteric arteries and the associated underlying molecular mechanisms. We found that acute empagliflozin application produces a concentration-dependent vasodilation in myogenic, depolarized and phenylephrine (PE)-preconstricted mesenteric arteries. Selective inhibition of smooth muscle cell voltage-gated K+ channels KV1.5 and KV7 abolished empagliflozin-induced vasodilation. In contrast, pharmacological inhibition of large-conductance Ca2+-activated K+ (BKCa) channels and ATP-sensitive (KATP) channels did not abolish vasodilation. Inhibition of the vasodilatory signaling axis involving endothelial nitric oxide (NO), smooth muscle cell soluble guanylyl cyclase (sGC) and protein kinase G (PKG) did not abolish empagliflozin-evoked vasodilation. Inhibition of the endothelium-derived vasodilatory molecule prostacyclin (PGI2) had no effect on the vasodilation. Consistently, empagliflozin-evoked vasodilation remained unaltered by endothelium denudation. Overall, our data suggest that empagliflozin stimulates smooth muscle cell KV channels KV1.5 and KV7, resulting in vasodilation in resistance-size mesenteric arteries. This study demonstrates for the first time a novel mechanism whereby empagliflozin regulates arterial contractility, resulting in vasodilation. Due to known antihypertensive properties, treatment with empagliflozin may complement conventional antihypertensive therapy.
Collapse
Affiliation(s)
| | - Raquibul Hasan
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, 3001 Mercer University Drive, Atlanta, GA 30341, USA;
| |
Collapse
|
15
|
Zhang D, Krause BM, Schmalz HG, Wohlfart P, Yard BA, Schubert R. ET-CORM Mediated Vasorelaxation of Small Mesenteric Arteries: Involvement of Kv7 Potassium Channels. Front Pharmacol 2021; 12:702392. [PMID: 34552483 PMCID: PMC8451721 DOI: 10.3389/fphar.2021.702392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 08/24/2021] [Indexed: 11/18/2022] Open
Abstract
Although the vasoactive properties of carbon monoxide (CO) have been extensively studied, the mechanism by which CO mediates vasodilation is not completely understood. Through-out published studies on CO mediated vasodilation there is inconsistency on the type of K+-channels that are activated by CO releasing molecules (CORMs). Since the vasorelaxation properties of enzyme triggered CORMs (ET-CORMs) have not been studied thus far, we first assessed if ET-CORMs can mediate vasodilation of small mesenteric arteries and subsequently addressed the role of soluble guanylate cyclase (sGC) and that of K-channels herein. To this end, 3 different types of ET-CORMs that either contain acetate (rac-1 and rac-4) or pivalate (rac-8) as ester functionality, were tested ex vivo on methoxamine pre-contracted small rat mesenteric arteries in a myograph setting. Pre-contracted mesenteric arteries strongly dilated upon treatment with both types of acetate containing ET-CORMs (rac-1 and rac-4), while treatment with the pivalate containing ET-CORM (rac-8) resulted in no vasodilation. Pre-treatment of mesenteric arteries with the sGC inhibitor ODQ abolished rac-4 mediated vasodilation, similar as for the known sGC activator SNP. Likewise, rac-4 mediated vasodilation did not occur in KCL pretreated mesenteric arteries. Although mesenteric arteries abundantly expressed a variety of K+-channels only Kv7 channels were found to be of functional relevance for rac-4 mediated vasodilation. In conclusion the current results identified Kv7 channels as the main channel by which rac-4 mediates vasodilation. In keeping with the central role of Kv7 in the control of vascular tone and peripheral resistance these promising ex-vivo data warrant further in vivo studies, particularly in models of primary hypertension or cardiac diseases, to assess the potential use of ET-CORMs in these diseases.
Collapse
Affiliation(s)
- Danfeng Zhang
- Department of Nephrology, Endocrinology and Rheumatology, Fifth Medical Department of Medicine, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.,Department of Nephrology, the Second Hospital of Anhui Medical University, Hefei, China
| | | | | | - Paulus Wohlfart
- Diabetes Research, Sanofi Aventis Deutschland GmbH, Frankfurt, Germany
| | - Benito A Yard
- Department of Nephrology, Endocrinology and Rheumatology, Fifth Medical Department of Medicine, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.,European Center of Angioscience (ECAS), Research Division Cardiovascular Physiology, Medical Faculty Mannheim, Heidelberg University, Frankfurt, Germany
| | - Rudolf Schubert
- European Center of Angioscience (ECAS), Research Division Cardiovascular Physiology, Medical Faculty Mannheim, Heidelberg University, Frankfurt, Germany.,Physiology, Institute of Theoretical Medicine, Medical Faculty, University of Augsburg, Augsburg, Germany
| |
Collapse
|
16
|
Jost N, Christ T, Magyar J. New Strategies for the Treatment of Atrial Fibrillation. Pharmaceuticals (Basel) 2021; 14:ph14090926. [PMID: 34577626 PMCID: PMC8466466 DOI: 10.3390/ph14090926] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 09/07/2021] [Accepted: 09/08/2021] [Indexed: 12/19/2022] Open
Abstract
Atrial fibrillation (AF) is the most prevalent cardiac arrhythmia in the clinical practice. It significantly contributes to the morbidity and mortality of the elderly population. Over the past 25-30 years intense effort in basic research has advanced the understanding of the relationship between the pathophysiology of AF and atrial remodelling. Nowadays it is clear that the various forms of atrial remodelling (electrical, contractile and structural) play crucial role in initiating and maintaining the persistent and permanent types of AF. Unlike in ventricular fibrillation, in AF rapid ectopic firing originating from pulmonary veins and re-entry mechanism may induce and maintain (due to atrial remodelling) this complex cardiac arrhythmia. The present review presents and discusses in detail the latest knowledge on the role of remodelling in AF. Special attention is paid to novel concepts and pharmacological targets presumably relevant to the drug treatment of atrial fibrillation.
Collapse
Affiliation(s)
- Norbert Jost
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, 6725 Szeged, Hungary
- Department of Pharmacology and Pharmacotherapy, Interdisciplinary Excellence Centre, University of Szeged, 6725 Szeged, Hungary
- ELKH-SZTE Research Group for Cardiovascular Pharmacology, Eötvös Loránd Research Network, 6725 Szeged, Hungary
- Correspondence:
| | - Torsten Christ
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
- DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, 20246 Hamburg, Germany
| | - János Magyar
- Department of Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary;
- Department of Sport Physiology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
17
|
Honda Y, Li J, Hino A, Tsujimoto S, Lee JK. High-Throughput Drug Screening System Based on Human Induced Pluripotent Stem Cell-Derived Atrial Myocytes ∼ A Novel Platform to Detect Cardiac Toxicity for Atrial Arrhythmias. Front Pharmacol 2021; 12:680618. [PMID: 34413773 PMCID: PMC8369502 DOI: 10.3389/fphar.2021.680618] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 07/13/2021] [Indexed: 12/21/2022] Open
Abstract
Evaluation of proarrhythmic properties is critical for drug discovery. In particular, QT prolongation in electrocardiograms has been utilized as a surrogate marker in many evaluation systems to assess the risk of torsade de pointes and lethal ventricular arrhythmia. Recently, new evaluation systems based on human iPS cell-derived cardiomyocytes have been established. On the other hand, in clinical situations, it has been reported that the incidence of atrial arrhythmias such as atrial fibrillation has been increasing every year, with the prediction of a persistent increase in the near future. As to the increased incidence of atrial arrhythmias, in addition to the increased population of geriatric patients, a wide variety of drug treatments may be related, as an experimental method to detect drug-induced atrial arrhythmia has not been established so far. In the present study, we characterized the atrial-like cardiomyocytes derived from human induced pluripotent stem cells and examined their potential for the evaluation of drug-induced atrial arrhythmia. Atrial-like cardiomyocytes were induced by adding retinoic acid (RA) during the process of myocardial differentiation, and their characteristics were compared to those of RA-free cardiomyocytes. Using gene expression and membrane potential analysis, it was confirmed that the cells with or without RA treatment have atrial or ventricular like cardiomyocytes, respectively. Using the ultra-rapid activating delayed rectifier potassium current (IKur) channel inhibitor, which is specific to atrial cardiomyocytes, Pulse width duration (PWD) 30cF prolongation was confirmed only in atrial-like cardiomyocytes. In addition, ventricular like cardiomyocytes exhibited an early after depolarization by treatment with rapidly activating delayed rectifier potassium current (IKr) channel inhibitor, which induces ventricular arrhythmia in clinical situations. Here, we have established a high-throughput drug evaluation system using human iPS cell-derived atrial-like cardiomyocytes. Based on the obtained data, the system might be a valuable platform to detect potential risks for drug-induced atrial arrhythmias.
Collapse
Affiliation(s)
- Yayoi Honda
- Sumitomo-Dainippon Pharma CO., Ltd., Osaka, Japan.,Bioanalysis Group, Osaka Laboratory, Technical Solution Headquarters, Sumika Chemical Analysis Service, Ltd., Osaka, Japan
| | - Jun Li
- Department of Cardiovascular Medicine, Osaka University Graduate School of Medicine, Suita, Japan.,Department of Cardiovascular Regenerative Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | - Aya Hino
- Department of Cardiovascular Regenerative Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| | | | - Jong-Kook Lee
- Department of Cardiovascular Regenerative Medicine, Osaka University Graduate School of Medicine, Suita, Japan
| |
Collapse
|
18
|
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
|
19
|
Verkerk AO, Marchal GA, Zegers JG, Kawasaki M, Driessen AHG, Remme CA, de Groot JR, Wilders R. Patch-Clamp Recordings of Action Potentials From Human Atrial Myocytes: Optimization Through Dynamic Clamp. Front Pharmacol 2021; 12:649414. [PMID: 33912059 PMCID: PMC8072333 DOI: 10.3389/fphar.2021.649414] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 02/18/2021] [Indexed: 11/29/2022] Open
Abstract
Introduction: Atrial fibrillation (AF) is the most common cardiac arrhythmia. Consequently, novel therapies are being developed. Ultimately, the impact of compounds on the action potential (AP) needs to be tested in freshly isolated human atrial myocytes. However, the frequent depolarized state of these cells upon isolation seriously hampers reliable AP recordings. Purpose: We assessed whether AP recordings from single human atrial myocytes could be improved by providing these cells with a proper inward rectifier K+ current (IK1), and consequently with a regular, non-depolarized resting membrane potential (RMP), through “dynamic clamp”. Methods: Single myocytes were enzymatically isolated from left atrial appendage tissue obtained from patients with paroxysmal AF undergoing minimally invasive surgical ablation. APs were elicited at 1 Hz and measured using perforated patch-clamp methodology, injecting a synthetic IK1 to generate a regular RMP. The injected IK1 had strong or moderate rectification. For comparison, a regular RMP was forced through injection of a constant outward current. A wide variety of ion channel blockers was tested to assess their modulatory effects on AP characteristics. Results: Without any current injection, RMPs ranged from −9.6 to −86.2 mV in 58 cells. In depolarized cells (RMP positive to −60 mV), RMP could be set at −80 mV using IK1 or constant current injection and APs could be evoked upon stimulation. AP duration differed significantly between current injection methods (p < 0.05) and was shortest with constant current injection and longest with injection of IK1 with strong rectification. With moderate rectification, AP duration at 90% repolarization (APD90) was similar to myocytes with regular non-depolarized RMP, suggesting that a synthetic IK1 with moderate rectification is the most appropriate for human atrial myocytes. Importantly, APs evoked using each injection method were still sensitive to all drugs tested (lidocaine, nifedipine, E-4031, low dose 4-aminopyridine, barium, and apamin), suggesting that the major ionic currents of the atrial cells remained functional. However, certain drug effects were quantitatively dependent on the current injection approach used. Conclusion: Injection of a synthetic IK1 with moderate rectification facilitates detailed AP measurements in human atrial myocytes. Therefore, dynamic clamp represents a promising tool for testing novel antiarrhythmic drugs.
Collapse
Affiliation(s)
- Arie O Verkerk
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands.,Department of Experimental Cardiology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Gerard A Marchal
- Department of Experimental Cardiology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Jan G Zegers
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Makiri Kawasaki
- Department of Experimental Cardiology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Antoine H G Driessen
- Department of Cardiothoracic Surgery, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Carol Ann Remme
- Department of Experimental Cardiology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Joris R de Groot
- Department of Cardiology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Ronald Wilders
- Department of Medical Biology, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
20
|
Targeting of Potassium Channels in Cardiac Arrhythmias. Trends Pharmacol Sci 2021; 42:491-506. [PMID: 33858691 DOI: 10.1016/j.tips.2021.03.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 03/14/2021] [Accepted: 03/17/2021] [Indexed: 02/07/2023]
Abstract
Cardiomyocytes are endowed with a complex repertoire of ion channels, responsible for the generation of action potentials (APs), travelling waves of electrical excitation, propagating throughout the heart and leading to cardiac contractions. Cardiac AP waveforms are shaped by a striking diversity of K+ channels. The pivotal role of K+ channels in cardiac health and disease is underscored by the dramatic impact that K+ channel dysfunction has on cardiac arrhythmias. The development of drugs targeted to specific K+ channels is expected to provide an optimized approach to antiarrhythmic therapy. Here, we review the functional roles of cardiac potassium channels under normal and diseased states. We survey current antiarrhythmic drugs (AADs) targeted to voltage-gated and Ca2+-activated K+ channels and highlight future research opportunities.
Collapse
|
21
|
Shvetsova AA, Gaynullina DK, Schmidt N, Bugert P, Lukoshkova EV, Tarasova OS, Schubert R. TASK-1 channel blockade by AVE1231 increases vasocontractile responses and BP in 1- to 2-week-old but not adult rats. Br J Pharmacol 2020; 177:5148-5162. [PMID: 32860629 PMCID: PMC7589011 DOI: 10.1111/bph.15249] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 08/14/2020] [Accepted: 08/18/2020] [Indexed: 12/13/2022] Open
Abstract
Background and Purpose The vasomotor role of K2P potassium channels during early postnatal development has never been investigated. We tested the hypothesis that TASK‐1 channel (K2P family member) contribution to arterial vascular tone and BP is higher in the early postnatal period than in adulthood. Experimental Approach We studied 10‐ to 15‐day‐old (“young”) and 2‐ to 3‐month‐old (“adult”) male rats performing digital PCR (dPCR) (using endothelium‐intact saphenous arteries), isometric myography, sharp microelectrode technique, quantitative PCR (qPCR) and Western blotting (using endothelium‐denuded saphenous arteries), and arterial pressure measurements under urethane anaesthesia. Key Results We found mRNA of Kcnk1–Kcnk7, Kcnk12, and Kcnk13 genes to be expressed in rat saphenous artery, and Kcnk3 (TASK‐1) and Kcnk6 (TWIK‐2) were most abundant in both age groups. The TASK‐1 channel blocker AVE1231 (1 μmol·L−1) prominently depolarized arterial smooth muscle and increased basal tone level and contractile responses to methoxamine of arteries from young rats but had almost no effect in adult rats. The level of TASK‐1 mRNA and protein expression was higher in arteries from young compared with adult rats. Importantly, intravenous administration of AVE1231 (4 mg·kg−1) had no effect on mean arterial pressure in adult rats but prominently raised it in young rats. Conclusion and Implications We showed that TASK‐1 channels are important for negative feedback regulation of vasocontraction in young but not adult rats. The influence of TASK‐1 channels most likely contributes to low BP level at perinatal age.
Collapse
Affiliation(s)
- Anastasia A Shvetsova
- Centre for Biomedicine and Medical Technology Mannheim (CBTM) and European Center of Angioscience (ECAS), Research Division Cardiovascular Physiology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany.,Faculty of Biology, M. V. Lomonosov Moscow State University, Moscow, Russia
| | - Dina K Gaynullina
- Faculty of Biology, M. V. Lomonosov Moscow State University, Moscow, Russia
| | - Nadine Schmidt
- Centre for Biomedicine and Medical Technology Mannheim (CBTM) and European Center of Angioscience (ECAS), Research Division Cardiovascular Physiology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Peter Bugert
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Service Baden-Württemberg-Hessen, Mannheim, Germany
| | - Elena V Lukoshkova
- Institute of Experimental Cardiology, National Medical Research Center of Cardiology, Moscow, Russia
| | - Olga S Tarasova
- Faculty of Biology, M. V. Lomonosov Moscow State University, Moscow, Russia.,State Research Center of the Russian Federation-Institute for Biomedical Problems, Russian Academy of Sciences, Moscow, Russia
| | - Rudolf Schubert
- Centre for Biomedicine and Medical Technology Mannheim (CBTM) and European Center of Angioscience (ECAS), Research Division Cardiovascular Physiology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany.,Physiology, Institute of Theoretical Medicine, Medical Faculty, University of Augsburg, Augsburg, Germany
| |
Collapse
|
22
|
Honda Y. [Availability of a novel cardiotoxicity evaluation system using human induced pluripotent stem cell-derived atrial-like myocytes]. Nihon Yakurigaku Zasshi 2020; 155:303-308. [PMID: 32879170 DOI: 10.1254/fpj.20041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
It is reported that the incidence of atrial arrhythmias has been increasing year by year and it might increase from now on. Although not only aging but pharmaceutical drug treatments might relate to atrial arrhythmias, experimental method to detect drug-induced atrial arrhythmias has not been established so far. Therefore, we induced differentiation of atrial-like cardiomyocytes from human induced pluripotent stem (iPS) cell, and clarified their characteristics and verified their dug responsiveness. Atrial-like cardiomyocytes were induced by adding retinoic acid (RA) during the process of myocardial differentiation, and their character was compared to RA-untreated cardiomyocytes. In gene expression and membrane potential analysis, it was confirmed that the cells with or without RA treatment have the characters of atrial or ventricular like cardiomyocytes, respectively. In addition, it was also confirmed that atrial-like cardiomyocytes induced reentry-like conduction disorder, which is atrial arrhythmias. Furthermore, as a result of examining the responsiveness of various ion channel inhibitors using these cells, the inhibition of ultra-rapid delayed rectifier potassium current (IKur) specifically existed in atrial muscle induced prolongation of PWD30cF (membrane potential duration at 30% depolarization corrected by Fridericia formula) only in atrial-like cardiomyocytes. In addition, ventricular-like cardiomyocytes alone exhibited an early after depolarization by treatment of rapid rectifier potassium current (IKr) inhibitor which induced ventricular arrhythmia in clinical situation. Based on above evidences, current evaluation systems using human iPS cell-derived atrial-like cardiomyocytes might be a valuable tool for drug-induced atrial arrhythmias.
Collapse
Affiliation(s)
- Yayoi Honda
- Sumitomo Dainippon Pharma, Co., Ltd.,Bioanalysis Group, Osaka Laboratory, Technical Solution Headquarters, Sumika Chemical Analysis Service, Ltd
| |
Collapse
|
23
|
Ni H, Fogli Iseppe A, Giles WR, Narayan SM, Zhang H, Edwards AG, Morotti S, Grandi E. Populations of in silico myocytes and tissues reveal synergy of multiatrial-predominant K + -current block in atrial fibrillation. Br J Pharmacol 2020; 177:4497-4515. [PMID: 32667679 DOI: 10.1111/bph.15198] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 06/22/2020] [Accepted: 07/03/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND PURPOSE Pharmacotherapy of atrial fibrillation (AF), the most common cardiac arrhythmia, remains unsatisfactory due to low efficacy and safety concerns. New therapeutic strategies target atrial-predominant ion-channels and involve multichannel block (poly)therapy. As AF is characterized by rapid and irregular atrial activations, compounds displaying potent antiarrhythmic effects at fast and minimal effects at slow rates are desirable. We present a novel systems pharmacology framework to quantitatively evaluate synergistic anti-AF effects of combined block of multiple atrial-predominant K+ currents (ultra-rapid delayed rectifier K+ current, IKur , small conductance Ca2+ -activated K+ current, IKCa , K2P 3.1 2-pore-domain K+ current, IK2P ) in AF. EXPERIMENTAL APPROACH We constructed experimentally calibrated populations of virtual atrial myocyte models in normal sinus rhythm and AF-remodelled conditions using two distinct, well-established atrial models. Sensitivity analyses on our atrial populations was used to investigate the rate dependence of action potential duration (APD) changes due to blocking IKur , IK2P or IKCa and interactions caused by blocking of these currents in modulating APD. Block was simulated in both single myocytes and one-dimensional tissue strands to confirm insights from the sensitivity analyses and examine anti-arrhythmic effects of multi-atrial-predominant K+ current block in single cells and coupled tissue. KEY RESULTS In both virtual atrial myocytes and tissues, multiple atrial-predominant K+ -current block promoted favourable positive rate-dependent APD prolongation and displayed positive rate-dependent synergy, that is, increasing synergistic antiarrhythmic effects at fast pacing versus slow rates. CONCLUSION AND IMPLICATIONS Simultaneous block of multiple atrial-predominant K+ currents may be a valuable antiarrhythmic pharmacotherapeutic strategy for AF.
Collapse
Affiliation(s)
- Haibo Ni
- Department of Pharmacology, University of California, Davis, CA, USA
| | - Alex Fogli Iseppe
- Department of Pharmacology, University of California, Davis, CA, USA
| | - Wayne R Giles
- Faculties of Kinesiology and Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Sanjiv M Narayan
- Division of Cardiology, Cardiovascular Institute, Stanford University, Stanford, CA, USA
| | - Henggui Zhang
- Biological Physics Group, School of Physics and Astronomy, The University of Manchester, Manchester, UK
| | - Andrew G Edwards
- Department of Pharmacology, University of California, Davis, CA, USA
| | - Stefano Morotti
- Department of Pharmacology, University of California, Davis, CA, USA
| | - Eleonora Grandi
- Department of Pharmacology, University of California, Davis, CA, USA
| |
Collapse
|
24
|
Jung HS, Seo MS, An JR, Kang M, Heo R, Li H, Jung WK, Choi IW, Cho EH, Park H, Bae YM, Park WS. The vasodilatory effect of gemigliptin via activation of voltage-dependent K + channels and SERCA pumps in aortic smooth muscle. Eur J Pharmacol 2020; 882:173243. [PMID: 32535099 DOI: 10.1016/j.ejphar.2020.173243] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 04/20/2020] [Accepted: 06/03/2020] [Indexed: 02/06/2023]
Abstract
This study investigated the vasodilatory effects and acting mechanism of gemigliptin, a dipeptidyl peptidase-4 (DPP-4) inhibitor. Tests were conducted in aortic rings pre-contracted with phenylephrine. Gemigliptin induced dose-dependent vasodilation of the aortic smooth muscle. Several pre-treatment groups were used to investigate the mechanism of action. While pre-treatment with paxilline, a large-conductance Ca2+-activated K+ channel inhibitor, glibenclamide, an ATP-sensitive K+ channel inhibitor, and Ba2+, an inwardly rectifying K+ channel inhibitor, had no impact on the vasodilatory effect of gemigliptin, pre-treatment with 4-aminopyridine, a voltage-dependent K+ (Kv) channel inhibitor, effectively attenuated the vasodilatory action of gemigliptin. In addition, pre-treatment with sarcoplasmic/endoplasmic reticulum Ca2+-ATPase (SERCA) pump inhibitors thapsigargin and cyclopiazonic acid significantly reduced the vasodilatory effect of gemigliptin. cAMP/PKA-related or cGMP/PKG-related signaling pathway inhibitors, including adenylyl cyclase inhibitor SQ 22536, PKA inhibitor KT 5720, guanylyl cyclase inhibitor ODQ, and PKG inhibitor KT 5823 did not alter the vasodilatory effect of gemigliptin. Similarly, elimination of the endothelium and pre-treatment with a nitric oxide (NO) synthase inhibitor (L-NAME) or small- and intermediate-conductance Ca2+-activated K+ channels (apamin and TRAM-34, respectively) did not change the gemigliptin effect. These findings suggested that gemigliptin induces vasodilation through the activation of Kv channels and SERCA pumps independent of cAMP/PKA-related or cGMP/PKG-related signaling pathways and the endothelium. Therefore, caution is required when prescribing gemigliptin to the patients with hypotension and diabetes.
Collapse
Affiliation(s)
- Hee Seok Jung
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Mi Seon Seo
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Jin Ryeol An
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Minji Kang
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Ryeon Heo
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Hongliang Li
- Institute of Translational Medicine, Medical College, Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment for Senile Diseases, Yangzhou University, Yangzhou, 225001, China
| | - Won-Kyo Jung
- Department of Biomedical Engineering, And Center for Marine-Integrated Biomedical Technology (BK21 Plus), Pukyong National University, Busan, 48513, South Korea
| | - Il-Whan Choi
- Department of Microbiology, Inje University College of Medicine, Busan, 48516, South Korea
| | - Eun-Hee Cho
- Department of Internal Medicine, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Hongzoo Park
- Department of Urology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea
| | - Young Min Bae
- Department of Physiology, Konkuk University School of Medicine, Chungju, 27478, South Korea
| | - Won Sun Park
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon, 24341, South Korea.
| |
Collapse
|
25
|
Zavaritskaya O, Dudem S, Ma D, Rabab KE, Albrecht S, Tsvetkov D, Kassmann M, Thornbury K, Mladenov M, Kammermeier C, Sergeant G, Mullins N, Wouappi O, Wurm H, Kannt A, Gollasch M, Hollywood MA, Schubert R. Vasodilation of rat skeletal muscle arteries by the novel BK channel opener GoSlo is mediated by the simultaneous activation of BK and K v 7 channels. Br J Pharmacol 2020; 177:1164-1186. [PMID: 31658366 PMCID: PMC7042121 DOI: 10.1111/bph.14910] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 09/27/2019] [Accepted: 10/01/2019] [Indexed: 01/17/2023] Open
Abstract
Background and Purpose BK channels play important roles in various physiological and pathophysiological processes and thus have been the target of several drug development programmes focused on creating new efficacious BK channel openers, such as the GoSlo‐SR compounds. However, the effect of GoSlo‐SR compounds on vascular smooth muscle has not been studied. Therefore, we tested the hypothesis that GoSlo‐SR compounds dilate arteries exclusively by activating BK channels. Experimental Approach Experiments were performed on rat Gracilis muscle, saphenous, mesenteric and tail arteries using isobaric and isometric myography, sharp microelectrodes, digital droplet PCR and the patch‐clamp technique. Key Results GoSlo‐SR compounds dilated isobaric and relaxed and hyperpolarised isometric vessel preparations and their effects were abolished after (a) functionally eliminating K+ channels by pre‐constriction with 50 mM KCl or (b) blocking all K+ channels known to be expressed in vascular smooth muscle. However, these effects were not blocked when BK channels were inhibited. Surprisingly, the Kv7 channel inhibitor XE991 reduced their effects considerably, but neither Kv1 nor Kv2 channel blockers altered the inhibitory effects of GoSlo‐SR. However, the combined blockade of BK and Kv7 channels abolished the GoSlo‐SR‐induced relaxation. GoSlo‐SR compounds also activated Kv7.4 and Kv7.5 channels expressed in HEK 293 cells. Conclusion and Implications This study shows that GoSlo‐SR compounds are effective relaxants in vascular smooth muscle and mediate their effects by a combined activation of BK and Kv7.4/Kv7.5 channels. Activation of Kv1, Kv2 or Kv7.1 channels or other vasodilator pathways seems not to be involved.
Collapse
Affiliation(s)
- Olga Zavaritskaya
- Centre for Biomedicine and Medical Technology Mannheim (CBTM), Research Division Cardiovascular Physiology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Srikanth Dudem
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Ireland
| | - Dongyu Ma
- Centre for Biomedicine and Medical Technology Mannheim (CBTM), Research Division Cardiovascular Physiology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Anhui, China
| | - Kaneez E Rabab
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Ireland
| | - Sarah Albrecht
- Centre for Biomedicine and Medical Technology Mannheim (CBTM), Research Division Cardiovascular Physiology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Dmitry Tsvetkov
- Experimental and Clinical Research Center (ECRC), a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Mario Kassmann
- Experimental and Clinical Research Center (ECRC), a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Keith Thornbury
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Ireland.,Ion Channel Biotechnology Centre, Dundalk Institute of Technology, Dundalk, Ireland
| | - Mitko Mladenov
- Centre for Biomedicine and Medical Technology Mannheim (CBTM), Research Division Cardiovascular Physiology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Institute of Biology, Faculty of Natural Sciences and Mathematics, Sts. Cyril and Methodius, University of Skopje, Skopje, Macedonia.,Department of Fundamental and Applied Physiology, Russian National Research Medical University, Moscow, Russia
| | - Claire Kammermeier
- Sanofi Diabetes Research, Industriepark Hoechst, Frankfurt am Main, Germany
| | - Gerard Sergeant
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Ireland.,Ion Channel Biotechnology Centre, Dundalk Institute of Technology, Dundalk, Ireland
| | - Nicholas Mullins
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Ireland
| | - Ornella Wouappi
- Centre for Biomedicine and Medical Technology Mannheim (CBTM), Research Division Cardiovascular Physiology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Hannah Wurm
- Centre for Biomedicine and Medical Technology Mannheim (CBTM), Research Division Cardiovascular Physiology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Aimo Kannt
- Sanofi Diabetes Research, Industriepark Hoechst, Frankfurt am Main, Germany.,Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Maik Gollasch
- Experimental and Clinical Research Center (ECRC), a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Mark A Hollywood
- Smooth Muscle Research Centre, Dundalk Institute of Technology, Dundalk, Ireland.,Ion Channel Biotechnology Centre, Dundalk Institute of Technology, Dundalk, Ireland
| | - Rudolf Schubert
- Centre for Biomedicine and Medical Technology Mannheim (CBTM), Research Division Cardiovascular Physiology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Faculty of Medicine, Department of Physiology, Augsburg University, Augsburg, Germany
| |
Collapse
|
26
|
Abi-Gerges N, Miller PE, Ghetti A. Human Heart Cardiomyocytes in Drug Discovery and Research: New Opportunities in Translational Sciences. Curr Pharm Biotechnol 2019; 21:787-806. [PMID: 31820682 DOI: 10.2174/1389201021666191210142023] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 10/14/2019] [Accepted: 10/28/2019] [Indexed: 12/28/2022]
Abstract
In preclinical drug development, accurate prediction of drug effects on the human heart is critically important, whether in the context of cardiovascular safety or for the purpose of modulating cardiac function to treat heart disease. Current strategies have significant limitations, whereby, cardiotoxic drugs can escape detection or potential life-saving therapies are abandoned due to false positive toxicity signals. Thus, new and more reliable translational approaches are urgently needed to help accelerate the rate of new therapy development. Renewed efforts in the recovery of human donor hearts for research and in cardiomyocyte isolation methods, are providing new opportunities for preclinical studies in adult primary cardiomyocytes. These cells exhibit the native physiological and pharmacological properties, overcoming the limitations presented by artificial cellular models, animal models and have great potential for providing an excellent tool for preclinical drug testing. Adult human primary cardiomyocytes have already shown utility in assessing drug-induced cardiotoxicity risk and helping in the identification of new treatments for cardiac diseases, such as heart failure and atrial fibrillation. Finally, strategies with actionable decision-making trees that rely on data derived from adult human primary cardiomyocytes will provide the holistic insights necessary to accurately predict human heart effects of drugs.
Collapse
Affiliation(s)
- Najah Abi-Gerges
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA 92109, United States
| | - Paul E Miller
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA 92109, United States
| | - Andre Ghetti
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA 92109, United States
| |
Collapse
|
27
|
Wang Y, Zhu R, Tung L. Contribution of potassium channels to action potential repolarization of human embryonic stem cell-derived cardiomyocytes. Br J Pharmacol 2019; 176:2780-2794. [PMID: 31074016 DOI: 10.1111/bph.14704] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 03/11/2019] [Accepted: 03/29/2019] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND AND PURPOSE The electrophysiological properties of human pluripotent stem cell-derived cardiomyocytes (CMs) have not yet been characterized in a syncytial context. This study systematically characterized the contributions of different repolarizing potassium currents in human embryonic stem cell-derived CMs (hESC-CMs) during long-term culture as cell monolayers. EXPERIMENTAL APPROACH The H9 hESC line was differentiated to CMs and plated to form confluent cell monolayers. Optical mapping was used to record the action potentials (APs) and conduction velocity (CV) during electrophysiological and pharmacological experiments. RT-PCR and Western blot were used to detect the presence and expression levels of ion channel subunits. KEY RESULTS Long-term culture of hESC-CMs led to shortened AP duration (APD), faster repolarization rate, and increased CV. Selective block of IKr , IKs , IK1 , and IKur significantly affected AP repolarization and APD in a concentration- and culture time-dependent manner. Baseline variations in APD led to either positive or negative APD dependence of drug response. Chromanol 293B produced greater relative AP prolongation in mid- and late-stage cultures, while DPO-1 had more effect in early-stage cultures. CV in cell monolayers in early- and late-stage cultures was most susceptible to slowing by E-4031 and BaCl2 respectively. CONCLUSIONS AND IMPLICATIONS IKr , IKs , IK1 , and IKur all play an essential role in the regulation of APD and CV in hESC-CMs. During time in culture, increased expression of IKr and IK1 helps to accelerate repolarization, shorten APD, and increase CV. We identified a new pro-arrhythmic parameter, positive APD dependence of ion channel block, which can increase APD and repolarization gradients.
Collapse
Affiliation(s)
- Yin Wang
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, USA.,Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Renjun Zhu
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, USA
| | - Leslie Tung
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
28
|
Kajanus J, Antonsson T, Carlsson L, Jurva U, Pettersen A, Sundell J, Inghardt T. Potassium channel blocking 1,2-bis(aryl)ethane-1,2-diamines active as antiarrhythmic agents. Bioorg Med Chem Lett 2019; 29:1241-1245. [DOI: 10.1016/j.bmcl.2019.03.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/19/2019] [Accepted: 03/05/2019] [Indexed: 10/27/2022]
|
29
|
Shvetsova AA, Gaynullina DK, Tarasova OS, Schubert R. Negative feedback regulation of vasocontraction by potassium channels in 10- to 15-day-old rats: Dominating role of K v 7 channels. Acta Physiol (Oxf) 2019; 225:e13176. [PMID: 30136434 DOI: 10.1111/apha.13176] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 08/11/2018] [Accepted: 08/20/2018] [Indexed: 12/26/2022]
Abstract
AIM Potassium channels are key regulators of smooth muscle membrane potential and arterial tone. However, the roles of potassium channels in vascular tone regulation in the systemic circulation during early postnatal development are poorly understood. Therefore, this study tested the hypothesis that the negative feedback regulation of vasocontraction by potassium channels changes during maturation. METHODS Experiments were performed on endothelium-denuded saphenous arteries from 10- to 15-day-old and 2- to 3-month-old male rats. Isometric force and membrane potential were recorded using wire myography and the sharp microelectrode technique respectively; mRNA and protein contents were determined by qPCR and Western blotting. RESULTS The effects of Kv 1, Kir and Kv 7 channel blockers (DPO-1, BaCl2 , XE991) on methoxamine-induced contraction were larger in arteries of 10- to 15-day-old compared to 2- to 3-month-old animals. In contrast, the BKC a channel blocker iberiotoxin had a stronger influence in 2- to 3- month-old rats. The effects of KATP and Kv 2 channel blockers (glibenclamide, stromatoxin) were not pronounced at both ages. The larger influence of Kv 7 and Kir channel blockade on arterial contraction in 10- to 15-day-old rats was associated with more prominent smooth muscle depolarization. The developmental alterations in potassium channel functioning were generally consistent with their mRNA and protein expression levels in arterial smooth muscle. CONCLUSION The negative feedback regulation of vasocontraction by potassium channels varies during maturation depending on the channel type. A dominating contribution of Kv 7 channels to the regulation of basal tone and agonist-induced contraction was observed in arteries of 10- to 15-day-old animals.
Collapse
Affiliation(s)
- Anastasia A. Shvetsova
- Centre for Biomedicine and Medical Technology Mannheim (CBTM); European Center of Angioscience (ECAS); Research Division Cardiovascular Physiology; Medical Faculty Mannheim; Heidelberg University; Heidelberg Germany
- Faculty of Biology; M.V. Lomonosov Moscow State University; Moscow Russia
- State Research Center of the Russian Federation - Institute for Biomedical Problems; Russian Academy of Sciences; Moscow Russia
| | - Dina K. Gaynullina
- Faculty of Biology; M.V. Lomonosov Moscow State University; Moscow Russia
- State Research Center of the Russian Federation - Institute for Biomedical Problems; Russian Academy of Sciences; Moscow Russia
- Department of Physiology; Russian National Research Medical University; Moscow Russia
| | - Olga S. Tarasova
- Faculty of Biology; M.V. Lomonosov Moscow State University; Moscow Russia
- State Research Center of the Russian Federation - Institute for Biomedical Problems; Russian Academy of Sciences; Moscow Russia
| | - Rudolf Schubert
- Centre for Biomedicine and Medical Technology Mannheim (CBTM); European Center of Angioscience (ECAS); Research Division Cardiovascular Physiology; Medical Faculty Mannheim; Heidelberg University; Heidelberg Germany
| |
Collapse
|
30
|
Mondejar‐Parreño G, Callejo M, Barreira B, Morales‐Cano D, Esquivel‐Ruiz S, Moreno L, Cogolludo A, Perez‐Vizcaino F. miR-1 is increased in pulmonary hypertension and downregulates Kv1.5 channels in rat pulmonary arteries. J Physiol 2019; 597:1185-1197. [PMID: 29717493 PMCID: PMC6375863 DOI: 10.1113/jp276054] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 04/27/2018] [Indexed: 12/30/2022] Open
Abstract
KEY POINTS The expression of miR-1 is increased in lungs from the Hyp/Su5416 PAH rat model. Pulmonary artery smooth muscle cells from this animal model are more depolarized and show decreased expression and activity of voltage-dependent potassium channel (Kv)1.5. miR-1 directly targets Kv1.5 channels, reduces Kv1.5 activity and induces membrane depolarization. Antagomir-1 prevents Kv1.5 channel downregulation and the depolarization induced by hypoxia/Su5416 exposition. ABSTRACT Impairment of the voltage-dependent potassium channel (Kv) plays a central role in the development of cardiovascular diseases, including pulmonary arterial hypertension (PAH). MicroRNAs are non-coding RNAs that regulate gene expression by binding to the 3'-untranslated region region of specific mRNAs. The present study aimed to analyse the effects of miR-1 on Kv channel function in pulmonary arteries (PA). Kv channel activity was studied in PA from healthy animals transfected with miR-1 or scrambled-miR. Kv currents were studied using the whole-cell configuration of the patch clamp technique. The characterization of the Kv1.5 currents was performed with the selective inhibitor DPO-1. miR-1 expression was increased and Kv1.5 channels were decreased in lungs from a rat model of PAH induced by hypoxia and Su5416. miR-1 transfection increased cell capacitance, reduced Kv1.5 currents and induced membrane depolarization in isolated pulmonary artery smooth muscle cells. A luciferase reporter assay indicated that KCNA5, which encodes Kv1.5 channels, is a direct target gene of miR-1. Incubation of PA with Su5416 and hypoxia (3% O2 ) increased miR-1 and induced a decline in Kv1.5 currents, which was prevented by antagomiR-1. In conclusion, these data indicate that miR-1 induces pulmonary artery smooth muscle cell hypertrophy and reduces the activity and expression of Kv channels, suggesting a pathophysiological role in PAH.
Collapse
Affiliation(s)
- Gema Mondejar‐Parreño
- Departamento de Farmacología y Toxicología. Facultad de MedicinaUniversidad Complutense de MadridMadridSpain
- Ciber Enfermedades Respiratorias (Ciberes)MadridSpain
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM)MadridSpain
| | - María Callejo
- Departamento de Farmacología y Toxicología. Facultad de MedicinaUniversidad Complutense de MadridMadridSpain
- Ciber Enfermedades Respiratorias (Ciberes)MadridSpain
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM)MadridSpain
| | - Bianca Barreira
- Departamento de Farmacología y Toxicología. Facultad de MedicinaUniversidad Complutense de MadridMadridSpain
- Ciber Enfermedades Respiratorias (Ciberes)MadridSpain
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM)MadridSpain
| | - Daniel Morales‐Cano
- Departamento de Farmacología y Toxicología. Facultad de MedicinaUniversidad Complutense de MadridMadridSpain
- Ciber Enfermedades Respiratorias (Ciberes)MadridSpain
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM)MadridSpain
| | - Sergio Esquivel‐Ruiz
- Departamento de Farmacología y Toxicología. Facultad de MedicinaUniversidad Complutense de MadridMadridSpain
- Ciber Enfermedades Respiratorias (Ciberes)MadridSpain
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM)MadridSpain
| | - Laura Moreno
- Departamento de Farmacología y Toxicología. Facultad de MedicinaUniversidad Complutense de MadridMadridSpain
- Ciber Enfermedades Respiratorias (Ciberes)MadridSpain
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM)MadridSpain
| | - Angel Cogolludo
- Departamento de Farmacología y Toxicología. Facultad de MedicinaUniversidad Complutense de MadridMadridSpain
- Ciber Enfermedades Respiratorias (Ciberes)MadridSpain
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM)MadridSpain
| | - Francisco Perez‐Vizcaino
- Departamento de Farmacología y Toxicología. Facultad de MedicinaUniversidad Complutense de MadridMadridSpain
- Ciber Enfermedades Respiratorias (Ciberes)MadridSpain
- Instituto de Investigación Sanitaria Gregorio Marañón (IISGM)MadridSpain
| |
Collapse
|
31
|
Yao C, Veleva T, Scott L, Cao S, Li L, Chen G, Jeyabal P, Pan X, Alsina KM, Abu-Taha I, Ghezelbash S, Reynolds CL, Shen YH, LeMaire SA, Schmitz W, Müller FU, El-Armouche A, Eissa NT, Beeton C, Nattel S, Wehrens XH, Dobrev D, Li N. Enhanced Cardiomyocyte NLRP3 Inflammasome Signaling Promotes Atrial Fibrillation. Circulation 2018; 138:2227-2242. [PMID: 29802206 PMCID: PMC6252285 DOI: 10.1161/circulationaha.118.035202] [Citation(s) in RCA: 453] [Impact Index Per Article: 64.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 05/14/2018] [Indexed: 01/22/2023]
Abstract
BACKGROUND Atrial fibrillation (AF) is frequently associated with enhanced inflammatory response. The NLRP3 (NACHT, LRR, and PYD domain containing protein 3) inflammasome mediates caspase-1 activation and interleukin-1β release in immune cells but is not known to play a role in cardiomyocytes (CMs). Here, we assessed the role of CM NLRP3 inflammasome in AF. METHODS NLRP3 inflammasome activation was assessed by immunoblot in atrial whole-tissue lysates and CMs from patients with paroxysmal AF or long-standing persistent (chronic) AF. To determine whether CM-specific activation of NLPR3 is sufficient to promote AF, a CM-specific knockin mouse model expressing constitutively active NLRP3 (CM-KI) was established. In vivo electrophysiology was used to assess atrial arrhythmia vulnerability. To evaluate the mechanism of AF, electric activation pattern, Ca2+ spark frequency, atrial effective refractory period, and morphology of atria were evaluated in CM-KI mice and wild-type littermates. RESULTS NLRP3 inflammasome activity was increased in the atrial CMs of patients with paroxysmal AF and chronic AF. CM-KI mice developed spontaneous premature atrial contractions and inducible AF, which was attenuated by a specific NLRP3 inflammasome inhibitor, MCC950. CM-KI mice exhibited ectopic activity, abnormal sarcoplasmic reticulum Ca2+ release, atrial effective refractory period shortening, and atrial hypertrophy. Adeno-associated virus subtype-9-mediated CM-specific knockdown of Nlrp3 suppressed AF development in CM-KI mice. Finally, genetic inhibition of Nlrp3 prevented AF development in CREM transgenic mice, a well-characterized mouse model of spontaneous AF. CONCLUSIONS Our study establishes a novel pathophysiological role for CM NLRP3 inflammasome signaling, with a mechanistic link to the pathogenesis of AF, and establishes the inhibition of NLRP3 as a potential novel AF therapy approach.
Collapse
Affiliation(s)
- Chunxia Yao
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine (Cardiovascular Research), Baylor College of Medicine, Houston, TX, USA
- Affiliated Hospital, College of Medicine, Hebei University of Engineering, Handan, Hebei Province, China
| | - Tina Veleva
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Larry Scott
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine (Cardiovascular Research), Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Shuyi Cao
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Luge Li
- Department of Medicine (Cardiovascular Research), Baylor College of Medicine, Houston, TX, USA
| | - Gong Chen
- Department of Medicine (Cardiovascular Research), Baylor College of Medicine, Houston, TX, USA
| | - Prince Jeyabal
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine (Cardiovascular Research), Baylor College of Medicine, Houston, TX, USA
| | - Xiaolu Pan
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Katherina M. Alsina
- Integrative Molecular and Biomedical Sciences Program, Baylor College of Medicine, Houston, TX, USA
| | - Issam Abu-Taha
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Shokoufeh Ghezelbash
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Corey L. Reynolds
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA
- Mouse Phenotyping Core, Baylor College of Medicine, Houston, TX, USA
| | - Ying H. Shen
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
- Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Scott A. LeMaire
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
- Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Wilhelm Schmitz
- Department of Pharmacology and Toxicology, University of Münster, Münster, Germany
| | - Frank U. Müller
- Department of Pharmacology and Toxicology, University of Münster, Münster, Germany
| | - Ali El-Armouche
- Department of Pharmacology and Toxicology, Dresden University of Technology, Dresden, Germany
| | - N. Tony Eissa
- Department of Medicine (Pulmonary), Baylor College of Medicine, Houston, TX, USA
| | - Christine Beeton
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Stanley Nattel
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
- Department of Medicine, Montreal Heart Institute and Université de Montréal, Montréal, Québec, Canada
| | - Xander H.T. Wehrens
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine (Cardiology), Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
- Center for Space Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Na Li
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
- Department of Medicine (Cardiovascular Research), Baylor College of Medicine, Houston, TX, USA
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
32
|
Schmid J, Müller B, Heppeler D, Gaynullina D, Kassmann M, Gagov H, Mladenov M, Gollasch M, Schubert R. The Unexpected Role of Calcium-Activated Potassium Channels: Limitation of NO-Induced Arterial Relaxation. J Am Heart Assoc 2018; 7:e007808. [PMID: 29574460 PMCID: PMC5907584 DOI: 10.1161/jaha.117.007808] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 02/14/2018] [Indexed: 11/24/2022]
Abstract
BACKGROUND Multiple studies have shown that an NO-induced activation of vascular smooth muscle BK channels contributes to the NO-evoked dilation in many blood vessels. In vivo, NO is released continuously. NO attenuates vessel constrictions and, therefore, exerts an anticontractile effect. It is unknown whether the anticontractile effect of continuously present NO is mediated by BK channels. METHODS AND RESULTS This study tested the hypothesis that BK channels mediate the vasodilatory effect of continuously present NO. Experiments were performed on rat and mouse tail and rat saphenous arteries using isometric myography and FURA-2 fluorimetry. Continuously present NO donors, as well as endogenous NO, attenuated methoxamine-induced vasoconstrictions. This effect was augmented in the presence of the BK channel blocker iberiotoxin. Moreover, the contractile effect of iberiotoxin was reduced in the presence of NO donors. The effect of the NO donor sodium nitroprusside was abolished by an NO scavenger and by a guanylyl cyclase inhibitor. In addition, the effect of sodium nitroprusside was reduced considerably by a protein kinase G inhibitor, but was not altered by inhibition of H2S generation. Sodium nitroprusside attenuated the intracellular calcium concentration response to methoxamine. Furthermore, sodium nitroprusside strongly reduced methoxamine-induced calcium influx, which depends entirely on L-type calcium channels. It did not affect methoxamine-induced calcium release. CONCLUSIONS In summary, this study demonstrates the following: (1) continuously present NO evokes a strong anticontractile effect on rat and mouse arteries; (2) the iberiotoxin-induced augmentation of the effect of NO is associated with an NO-induced reduction of the effect of iberiotoxin; and (3) NO evoked a reduction of calcium influx via L-type calcium channels.
Collapse
Affiliation(s)
- Johannes Schmid
- Centre for Biomedicine and Medical Technology Mannheim and European Center of Angioscience, Research Division Cardiovascular Physiology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Bettina Müller
- Centre for Biomedicine and Medical Technology Mannheim and European Center of Angioscience, Research Division Cardiovascular Physiology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - David Heppeler
- Centre for Biomedicine and Medical Technology Mannheim and European Center of Angioscience, Research Division Cardiovascular Physiology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Dina Gaynullina
- Centre for Biomedicine and Medical Technology Mannheim and European Center of Angioscience, Research Division Cardiovascular Physiology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Faculty of Biology, M.V. Lomonosov, Moscow State University, Moscow, Russia
- Department of Physiology, Russian National Research Medical University, Moscow, Russia
| | - Mario Kassmann
- Experimental and Clinical Research Center, a joint cooperation between the, Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Hristo Gagov
- Centre for Biomedicine and Medical Technology Mannheim and European Center of Angioscience, Research Division Cardiovascular Physiology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Mitko Mladenov
- Centre for Biomedicine and Medical Technology Mannheim and European Center of Angioscience, Research Division Cardiovascular Physiology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Maik Gollasch
- Experimental and Clinical Research Center, a joint cooperation between the, Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Rudolf Schubert
- Centre for Biomedicine and Medical Technology Mannheim and European Center of Angioscience, Research Division Cardiovascular Physiology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
33
|
Streit J, Kleinlogel S. Dynamic all-optical drug screening on cardiac voltage-gated ion channels. Sci Rep 2018; 8:1153. [PMID: 29348631 PMCID: PMC5773578 DOI: 10.1038/s41598-018-19412-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 12/27/2017] [Indexed: 11/09/2022] Open
Abstract
Voltage-gated ion channels (VGCs) are prime targets for the pharmaceutical industry, but drug profiling on VGCs is challenging, since drug interactions are confined to specific conformational channel states mediated by changes in transmembrane potential. Here we combined various optogenetic tools to develop dynamic, high-throughput drug profiling assays with defined light-step protocols to interrogate VGC states on a millisecond timescale. We show that such light-induced electrophysiology (LiEp) yields high-quality pharmacological data with exceptional screening windows for drugs acting on the major cardiac VGCs, including hNav1.5, hKv1.5 and hERG. LiEp-based screening remained robust when using a variety of optogenetic actuators (ChR2, ChR2(H134R), CatCh, ChR2-EYFP-βArchT) and different types of organic (RH421, Di-4-ANBDQPQ, BeRST1) or genetic voltage sensors (QuasAr1). The tractability of LiEp allows a versatile and precise alternative to state-of-the-art VGC drug screening platforms such as automated electrophysiology or FLIPR readers.
Collapse
Affiliation(s)
- Jonas Streit
- Institute of Physiology, University of Bern, Bühlplatz 5, 3012, Bern, Switzerland
| | - Sonja Kleinlogel
- Institute of Physiology, University of Bern, Bühlplatz 5, 3012, Bern, Switzerland.
| |
Collapse
|
34
|
Zeng H, Balasubramanian B, Lagrutta A, Sannajust F. Response of human induced pluripotent stem cell-derived cardiomyocytes to several pharmacological agents when intrinsic syncytial pacing is overcome by acute external stimulation. J Pharmacol Toxicol Methods 2018; 91:18-26. [PMID: 29330131 DOI: 10.1016/j.vascn.2017.12.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 12/19/2017] [Accepted: 12/19/2017] [Indexed: 01/14/2023]
Abstract
We challenged human induced pluripotent stem cell-derived cardiomyocyte (hiPSC-CM) syncytia, mainly, CDI iCells with several classes of well-characterized pharmacological agents (including hERG blocker, Nav1.5 blocker, Cav1.2 blocker and opener, β-adrenergic agonist, and If blocker) under pacing conditions, utilizing the Cardio-ECR instrument, a non-invasive platform featuring simultaneous and continuous measurement of synchronized beating rate and contractility (both signals were acquired simultaneously and well aligned). We found that: 1) with increasing acute stimulation rates (no pacing; 1, 1.5, and 2Hz), beat interval was gradually shortened mainly in the relaxation phase of each beat cycle; 2) typical responses of iCells hiPSC-CMs to all tested pharmacological agents were either attenuated or even eliminated by pacing, in a concentration- and stimulation rate-dependent manner; and 3) when iCells were influenced by pharmacological agents and cannot follow pacing rates, they still beat regularly at exactly 1/2 or 1/3 of pacing rates. We concluded that when intrinsic syncytial pacing was overcome by faster, external stimulations, beat intervals of hiPSC-CMs were mainly shortened in the relaxation phase, instead of proportionally in each beat cycle, with increasing pacing rates. In addition, in response to pharmacological agents upon pacing, hiPSC-CMs exhibited distinct patterns of refractoriness, manifested by skipped beats in pacing-rate dependent manner, and attenuation (or even abolition) of the typical response evoked under spontaneous beating.
Collapse
Affiliation(s)
- Haoyu Zeng
- Merck & Co., Inc., Safety & Exploratory Pharmacology Department, West Point, PA, USA.
| | | | - Armando Lagrutta
- Merck & Co., Inc., Safety & Exploratory Pharmacology Department, West Point, PA, USA
| | - Frederick Sannajust
- Merck & Co., Inc., Safety & Exploratory Pharmacology Department, West Point, PA, USA
| |
Collapse
|
35
|
Ellinwood N, Dobrev D, Morotti S, Grandi E. In Silico Assessment of Efficacy and Safety of I Kur Inhibitors in Chronic Atrial Fibrillation: Role of Kinetics and State-Dependence of Drug Binding. Front Pharmacol 2017; 8:799. [PMID: 29163179 PMCID: PMC5681918 DOI: 10.3389/fphar.2017.00799] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 10/23/2017] [Indexed: 12/14/2022] Open
Abstract
Current pharmacological therapy against atrial fibrillation (AF), the most common cardiac arrhythmia, is limited by moderate efficacy and adverse side effects including ventricular proarrhythmia and organ toxicity. One way to circumvent the former is to target ion channels that are predominantly expressed in atria vs. ventricles, such as KV1.5, carrying the ultra-rapid delayed-rectifier K+ current (IKur). Recently, we used an in silico strategy to define optimal KV1.5-targeting drug characteristics, including kinetics and state-dependent binding, that maximize AF-selectivity in human atrial cardiomyocytes in normal sinus rhythm (nSR). However, because of evidence for IKur being strongly diminished in long-standing persistent (chronic) AF (cAF), the therapeutic potential of drugs targeting IKur may be limited in cAF patients. Here, we sought to simulate the efficacy (and safety) of IKur inhibitors in cAF conditions. To this end, we utilized sensitivity analysis of our human atrial cardiomyocyte model to assess the importance of IKur for atrial cardiomyocyte electrophysiological properties, simulated hundreds of theoretical drugs to reveal those exhibiting anti-AF selectivity, and compared the results obtained in cAF with those in nSR. We found that despite being downregulated, IKur contributes more prominently to action potential (AP) and effective refractory period (ERP) duration in cAF vs. nSR, with ideal drugs improving atrial electrophysiology (e.g., ERP prolongation) more in cAF than in nSR. Notably, the trajectory of the AP during cAF is such that more IKur is available during the more depolarized plateau potential. Furthermore, IKur block in cAF has less cardiotoxic effects (e.g., AP duration not exceeding nSR values) and can increase Ca2+ transient amplitude thereby enhancing atrial contractility. We propose that in silico strategies such as that presented here should be combined with in vitro and in vivo assays to validate model predictions and facilitate the ongoing search for novel agents against AF.
Collapse
Affiliation(s)
- Nicholas Ellinwood
- Department of Pharmacology, University of California, Davis, Davis, CA, United States
| | - Dobromir Dobrev
- West German Heart and Vascular Center, Institute of Pharmacology, University Duisburg-Essen, Essen, Germany
| | - Stefano Morotti
- Department of Pharmacology, University of California, Davis, Davis, CA, United States
| | - Eleonora Grandi
- Department of Pharmacology, University of California, Davis, Davis, CA, United States
| |
Collapse
|
36
|
Al-Owais MM, Hettiarachchi NT, Boyle JP, Scragg JL, Elies J, Dallas ML, Lippiat JD, Steele DS, Peers C. Multiple mechanisms mediating carbon monoxide inhibition of the voltage-gated K + channel Kv1.5. Cell Death Dis 2017; 8:e3163. [PMID: 29095440 PMCID: PMC5775415 DOI: 10.1038/cddis.2017.568] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 09/19/2017] [Accepted: 09/21/2017] [Indexed: 12/13/2022]
Abstract
The voltage-gated K+ channel has key roles in the vasculature and in atrial excitability and contributes to apoptosis in various tissues. In this study, we have explored its regulation by carbon monoxide (CO), a product of the cytoprotective heme oxygenase enzymes, and a recognized toxin. CO inhibited recombinant Kv1.5 expressed in HEK293 cells in a concentration-dependent manner that involved multiple signalling pathways. CO inhibition was partially reversed by superoxide dismutase mimetics and by suppression of mitochondrial reactive oxygen species. CO also elevated intracellular nitric oxide (NO) levels. Prevention of NO formation also partially reversed CO inhibition of Kv1.5, as did inhibition of soluble guanylyl cyclase. CO also elevated intracellular peroxynitrite levels, and a peroxynitrite scavenger markedly attenuated the ability of CO to inhibit Kv1.5. CO caused nitrosylation of Kv1.5, an effect that was also observed in C331A and C346A mutant forms of the channel, which had previously been suggested as nitrosylation sites within Kv1.5. Augmentation of Kv1.5 via exposure to hydrogen peroxide was fully reversed by CO. Native Kv1.5 recorded in HL-1 murine atrial cells was also inhibited by CO. Action potentials recorded in HL-1 cells were increased in amplitude and duration by CO, an effect mimicked and occluded by pharmacological inhibition of Kv1.5. Our data indicate that Kv1.5 is a target for modulation by CO via multiple mechanisms. This regulation has important implications for diverse cellular functions, including excitability, contractility and apoptosis.
Collapse
Affiliation(s)
- Moza M Al-Owais
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Nishani T Hettiarachchi
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - John P Boyle
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Jason L Scragg
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Jacobo Elies
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Mark L Dallas
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Jon D Lippiat
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Derek S Steele
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Chris Peers
- Division of Cardiovascular and Diabetes Research, LICAMM, Faculty of Medicine and Health, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
37
|
Jeevaratnam K, Chadda KR, Huang CLH, Camm AJ. Cardiac Potassium Channels: Physiological Insights for Targeted Therapy. J Cardiovasc Pharmacol Ther 2017; 23:119-129. [PMID: 28946759 PMCID: PMC5808825 DOI: 10.1177/1074248417729880] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The development of novel drugs specifically directed at the ion channels underlying particular features of cardiac action potential (AP) initiation, recovery, and refractoriness would contribute to an optimized approach to antiarrhythmic therapy that minimizes potential cardiac and extracardiac toxicity. Of these, K+ channels contribute numerous and diverse currents with specific actions on different phases in the time course of AP repolarization. These features and their site-specific distribution make particular K+ channel types attractive therapeutic targets for the development of pharmacological agents attempting antiarrhythmic therapy in conditions such as atrial fibrillation. However, progress in the development of such temporally and spatially selective antiarrhythmic drugs against particular ion channels has been relatively limited, particularly in view of our incomplete understanding of the complex physiological roles and interactions of the various ionic currents. This review summarizes the physiological properties of the main cardiac potassium channels and the way in which they modulate cardiac electrical activity and then critiques a number of available potential antiarrhythmic drugs directed at them.
Collapse
Affiliation(s)
- Kamalan Jeevaratnam
- 1 Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom.,2 School of Medicine, Perdana University-Royal College of Surgeons Ireland, Serdang, Selangor Darul Ehsan, Malaysia
| | - Karan R Chadda
- 1 Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom.,3 Physiological Laboratory, University of Cambridge, Cambridge, United Kingdom
| | - Christopher L-H Huang
- 3 Physiological Laboratory, University of Cambridge, Cambridge, United Kingdom.,4 Division of Cardiovascular Biology, Department of Biochemistry, University of Cambridge, Cambridge, United Kingdom
| | - A John Camm
- 5 Cardiac Clinical Academic Group, St George's Hospital Medical School, University of London, Cranmer Terrace, London, United Kingdom
| |
Collapse
|
38
|
Ellinwood N, Dobrev D, Morotti S, Grandi E. Revealing kinetics and state-dependent binding properties of I Kur-targeting drugs that maximize atrial fibrillation selectivity. CHAOS (WOODBURY, N.Y.) 2017; 27:093918. [PMID: 28964116 PMCID: PMC5573366 DOI: 10.1063/1.5000226] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 07/06/2017] [Indexed: 06/07/2023]
Abstract
The KV1.5 potassium channel, which underlies the ultra-rapid delayed-rectifier current (IKur) and is predominantly expressed in atria vs. ventricles, has emerged as a promising target to treat atrial fibrillation (AF). However, while numerous KV1.5-selective compounds have been screened, characterized, and tested in various animal models of AF, evidence of antiarrhythmic efficacy in humans is still lacking. Moreover, current guidelines for pre-clinical assessment of candidate drugs heavily rely on steady-state concentration-response curves or IC50 values, which can overlook adverse cardiotoxic effects. We sought to investigate the effects of kinetics and state-dependent binding of IKur-targeting drugs on atrial electrophysiology in silico and reveal the ideal properties of IKur blockers that maximize anti-AF efficacy and minimize pro-arrhythmic risk. To this aim, we developed a new Markov model of IKur that describes KV1.5 gating based on experimental voltage-clamp data in atrial myocytes from patient right-atrial samples in normal sinus rhythm. We extended the IKur formulation to account for state-specificity and kinetics of KV1.5-drug interactions and incorporated it into our human atrial cell model. We simulated 1- and 3-Hz pacing protocols in drug-free conditions and with a [drug] equal to the IC50 value. The effects of binding and unbinding kinetics were determined by examining permutations of the forward (kon) and reverse (koff) binding rates to the closed, open, and inactivated states of the KV1.5 channel. We identified a subset of ideal drugs exhibiting anti-AF electrophysiological parameter changes at fast pacing rates (effective refractory period prolongation), while having little effect on normal sinus rhythm (limited action potential prolongation). Our results highlight that accurately accounting for channel interactions with drugs, including kinetics and state-dependent binding, is critical for developing safer and more effective pharmacological anti-AF options.
Collapse
Affiliation(s)
- Nicholas Ellinwood
- Department of Pharmacology, University of California Davis, Davis, California 95616, USA
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Stefano Morotti
- Department of Pharmacology, University of California Davis, Davis, California 95616, USA
| | - Eleonora Grandi
- Department of Pharmacology, University of California Davis, Davis, California 95616, USA
| |
Collapse
|
39
|
Aguilar M, Feng J, Vigmond E, Comtois P, Nattel S. Rate-Dependent Role of I Kur in Human Atrial Repolarization and Atrial Fibrillation Maintenance. Biophys J 2017; 112:1997-2010. [PMID: 28494969 DOI: 10.1016/j.bpj.2017.03.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 03/06/2017] [Accepted: 03/23/2017] [Indexed: 11/24/2022] Open
Abstract
The atrial-specific ultrarapid delayed rectifier K+ current (IKur) inactivates slowly but completely at depolarized voltages. The consequences for IKur rate-dependence have not been analyzed in detail and currently available mathematical action-potential (AP) models do not take into account experimentally observed IKur inactivation dynamics. Here, we developed an updated formulation of IKur inactivation that accurately reproduces time-, voltage-, and frequency-dependent inactivation. We then modified the human atrial cardiomyocyte Courtemanche AP model to incorporate realistic IKur inactivation properties. Despite markedly different inactivation dynamics, there was no difference in AP parameters across a wide range of stimulation frequencies between the original and updated models. Using the updated model, we showed that, under physiological stimulation conditions, IKur does not inactivate significantly even at high atrial rates because the transmembrane potential spends little time at voltages associated with inactivation. Thus, channel dynamics are determined principally by activation kinetics. IKur magnitude decreases at higher rates because of AP changes that reduce IKur activation. Nevertheless, the relative contribution of IKur to AP repolarization increases at higher frequencies because of reduced activation of the rapid delayed-rectifier current IKr. Consequently, IKur block produces dose-dependent termination of simulated atrial fibrillation (AF) in the absence of AF-induced electrical remodeling. The inclusion of AF-related ionic remodeling stabilizes simulated AF and greatly reduces the predicted antiarrhythmic efficacy of IKur block. Our results explain a range of experimental observations, including recently reported positive rate-dependent IKur-blocking effects on human atrial APs, and provide insights relevant to the potential value of IKur as an antiarrhythmic target for the treatment of AF.
Collapse
Affiliation(s)
- Martin Aguilar
- Research Center, Montreal Heart Institute, Université de Montréal, Montreal, Québec, Canada; Department of Pharmacology and Physiology/Institute of Biomedical Engineering, Université de Montréal, Montreal, Québec, Canada
| | | | - Edward Vigmond
- L'Institut de Rythmologie et Modélisation Cardiaque LIRYC, Fondation Université de Bordeaux, Hôpital Xavier-Arnozan, Pessac, France; Institut de Mathématiques de Bordeaux, Université de Bordeaux, Talence, France
| | - Philippe Comtois
- Research Center, Montreal Heart Institute, Université de Montréal, Montreal, Québec, Canada; Department of Pharmacology and Physiology/Institute of Biomedical Engineering, Université de Montréal, Montreal, Québec, Canada
| | - Stanley Nattel
- Research Center, Montreal Heart Institute, Université de Montréal, Montreal, Québec, Canada; Department of Medicine, McGill University, Montreal, Québec, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal, Québec, Canada; Department of Medicine, Université de Montréal, Montreal, Québec, Canada; West German Heart and Vascular Center, Faculty of Medicine, University Duisburg-Essen, Essen, Germany.
| |
Collapse
|
40
|
Shin SE, Li H, Kim HS, Kim HW, Seo MS, Ha KS, Han ET, Hong SH, Firth AL, Choi IW, Bae YM, Park WS. Nortriptyline, a tricyclic antidepressant, inhibits voltage-dependent K + channels in coronary arterial smooth muscle cells. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2017; 21:225-232. [PMID: 28280416 PMCID: PMC5343056 DOI: 10.4196/kjpp.2017.21.2.225] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 12/07/2016] [Accepted: 12/07/2016] [Indexed: 11/30/2022]
Abstract
We demonstrated the effect of nortriptyline, a tricyclic antidepressant drug and serotonin reuptake inhibitor, on voltage-dependent K+ (Kv) channels in freshly isolated rabbit coronary arterial smooth muscle cells using a whole-cell patch clamp technique. Nortriptyline inhibited Kv currents in a concentration-dependent manner, with an apparent IC50 value of 2.86±0.52 µM and a Hill coefficient of 0.77±0.1. Although application of nortriptyline did not change the activation curve, nortriptyline shifted the inactivation current toward a more negative potential. Application of train pulses (1 or 2 Hz) did not change the nortriptyline-induced Kv channel inhibition, suggesting that the effects of nortiprtyline were not use-dependent. Preincubation with the Kv1.5 and Kv2.1/2.2 inhibitors, DPO-1 and guangxitoxin did not affect nortriptyline inhibition of Kv channels. From these results, we concluded that nortriptyline inhibited Kv channels in a concentration-dependent and state-independent manner independently of serotonin reuptake.
Collapse
Affiliation(s)
- Sung Eun Shin
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon 24341, Korea
| | - Hongliang Li
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon 24341, Korea
| | - Han Sol Kim
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon 24341, Korea
| | - Hye Won Kim
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon 24341, Korea
| | - Mi Seon Seo
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon 24341, Korea
| | - Kwon-Soo Ha
- Department of Molecular and Cellular Biochemistry, Kangwon National University School of Medicine, Chuncheon 24341, Korea
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon 24341, Korea
| | - Seok-Ho Hong
- Department of Internal Medicine, Kangwon National University School of Medicine, Chuncheon 24341, Korea
| | - Amy L Firth
- Department of Pulmonary, Critical Care and Sleep Medicine, University of Southern California, Keck School of Medicine, Los Angeles, CA90033, USA
| | - Il-Whan Choi
- Department of Microbiology, Inje University College of Medicine, Busan 48516, Korea
| | - Young Min Bae
- Department of Physiology, Konkuk University School of Medicine, Chungju 27478, Korea
| | - Won Sun Park
- Department of Physiology, Kangwon National University School of Medicine, Chuncheon 24341, Korea
| |
Collapse
|
41
|
Suppression of Kv1.5 protects against endothelial apoptosis induced by palmitate and in type 2 diabetes mice. Life Sci 2017; 168:28-37. [DOI: 10.1016/j.lfs.2015.12.054] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Revised: 11/14/2015] [Accepted: 12/31/2015] [Indexed: 01/09/2023]
|
42
|
Benson M, Iñiguez-Lluhí JA, Martens J. Sumo Modification of Ion Channels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 963:127-141. [PMID: 28197910 DOI: 10.1007/978-3-319-50044-7_8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Recently, a role for SUMO modification outside of the nucleus has emerged. Although the number of extranuclear proteins known to be sumoylated is comparatively small, ion channels represent one important new class of these proteins. Ion channels are responsible for the control of membrane excitability and therefore are critical for fundamental physiological processes such as muscle contraction, neuronal firing, and cellular homeostasis. As such, these ion-conducting proteins are subject to precise regulation. Recently, several studies have identified sumoylation as a novel mechanism of modulating ion channel function. These studies expand the list of known functions of sumoylation and reveal that, in addition to its more established role in the regulation of nuclear proteins, this modification plays important roles at the cytoplasmic face of membranes.
Collapse
Affiliation(s)
- Mark Benson
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | | | - Jeffrey Martens
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
43
|
Nishijima Y, Cao S, Chabowski DS, Korishettar A, Ge A, Zheng X, Sparapani R, Gutterman DD, Zhang DX. Contribution of K V1.5 Channel to Hydrogen Peroxide-Induced Human Arteriolar Dilation and Its Modulation by Coronary Artery Disease. Circ Res 2016; 120:658-669. [PMID: 27872049 DOI: 10.1161/circresaha.116.309491] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 11/11/2016] [Accepted: 11/21/2016] [Indexed: 02/06/2023]
Abstract
RATIONALE Hydrogen peroxide (H2O2) regulates vascular tone in the human microcirculation under physiological and pathophysiological conditions. It dilates arterioles by activating large-conductance Ca2+-activated K+ channels in subjects with coronary artery disease (CAD), but its mechanisms of action in subjects without CAD (non-CAD) when compared with those with CAD remain unknown. OBJECTIVE We hypothesize that H2O2-elicited dilation involves different K+ channels in non-CAD versus CAD, resulting in an altered capacity for vasodilation during disease. METHODS AND RESULTS H2O2 induced endothelium-independent vasodilation in non-CAD adipose arterioles, which was reduced by paxilline, a large-conductance Ca2+-activated K+ channel blocker, and by 4-aminopyridine, a voltage-gated K+ (KV) channel blocker. Assays of mRNA transcripts, protein expression, and subcellular localization revealed that KV1.5 is the major KV1 channel expressed in vascular smooth muscle cells and is abundantly localized on the plasma membrane. The selective KV1.5 blocker diphenylphosphine oxide-1 and the KV1.3/1.5 blocker 5-(4-phenylbutoxy)psoralen reduced H2O2-elicited dilation to a similar extent as 4-aminopyridine, but the selective KV1.3 blocker phenoxyalkoxypsoralen-1 was without effect. In arterioles from CAD subjects, H2O2-induced dilation was significantly reduced, and this dilation was inhibited by paxilline but not by 4-aminopyridine, diphenylphosphine oxide-1, or 5-(4-phenylbutoxy)psoralen. KV1.5 cell membrane localization and diphenylphosphine oxide-1-sensitive K+ currents were markedly reduced in isolated vascular smooth muscle cells from CAD arterioles, although mRNA or total cellular protein expression was largely unchanged. CONCLUSIONS In human arterioles, H2O2-induced dilation is impaired in CAD, which is associated with a transition from a combined large-conductance Ca2+-activated K+- and KV (KV1.5)-mediated vasodilation toward a large-conductance Ca2+-activated K+-predominant mechanism of dilation. Loss of KV1.5 vasomotor function may play an important role in microvascular dysfunction in CAD or other vascular diseases.
Collapse
Affiliation(s)
- Yoshinori Nishijima
- From the Department of Medicine (Y.N., S.C., D.S.C., A.K., A.G., X.Z., D.D.G., D.X.Z.), Cardiovascular Center (Y.N., S.C., D.S.C., A.K., A.G., X.Z., D.D.G., D.X.Z.), Department of Pharmacology and Toxicology (D.S.C., A.K.), Division of Biostatistics (R.S.), Medical College of Wisconsin, and Zablocki Veterans Affairs Medical Center (D.D.G.), Milwaukee, WI
| | - Sheng Cao
- From the Department of Medicine (Y.N., S.C., D.S.C., A.K., A.G., X.Z., D.D.G., D.X.Z.), Cardiovascular Center (Y.N., S.C., D.S.C., A.K., A.G., X.Z., D.D.G., D.X.Z.), Department of Pharmacology and Toxicology (D.S.C., A.K.), Division of Biostatistics (R.S.), Medical College of Wisconsin, and Zablocki Veterans Affairs Medical Center (D.D.G.), Milwaukee, WI
| | - Dawid S Chabowski
- From the Department of Medicine (Y.N., S.C., D.S.C., A.K., A.G., X.Z., D.D.G., D.X.Z.), Cardiovascular Center (Y.N., S.C., D.S.C., A.K., A.G., X.Z., D.D.G., D.X.Z.), Department of Pharmacology and Toxicology (D.S.C., A.K.), Division of Biostatistics (R.S.), Medical College of Wisconsin, and Zablocki Veterans Affairs Medical Center (D.D.G.), Milwaukee, WI
| | - Ankush Korishettar
- From the Department of Medicine (Y.N., S.C., D.S.C., A.K., A.G., X.Z., D.D.G., D.X.Z.), Cardiovascular Center (Y.N., S.C., D.S.C., A.K., A.G., X.Z., D.D.G., D.X.Z.), Department of Pharmacology and Toxicology (D.S.C., A.K.), Division of Biostatistics (R.S.), Medical College of Wisconsin, and Zablocki Veterans Affairs Medical Center (D.D.G.), Milwaukee, WI
| | - Alyce Ge
- From the Department of Medicine (Y.N., S.C., D.S.C., A.K., A.G., X.Z., D.D.G., D.X.Z.), Cardiovascular Center (Y.N., S.C., D.S.C., A.K., A.G., X.Z., D.D.G., D.X.Z.), Department of Pharmacology and Toxicology (D.S.C., A.K.), Division of Biostatistics (R.S.), Medical College of Wisconsin, and Zablocki Veterans Affairs Medical Center (D.D.G.), Milwaukee, WI
| | - Xiaodong Zheng
- From the Department of Medicine (Y.N., S.C., D.S.C., A.K., A.G., X.Z., D.D.G., D.X.Z.), Cardiovascular Center (Y.N., S.C., D.S.C., A.K., A.G., X.Z., D.D.G., D.X.Z.), Department of Pharmacology and Toxicology (D.S.C., A.K.), Division of Biostatistics (R.S.), Medical College of Wisconsin, and Zablocki Veterans Affairs Medical Center (D.D.G.), Milwaukee, WI
| | - Rodney Sparapani
- From the Department of Medicine (Y.N., S.C., D.S.C., A.K., A.G., X.Z., D.D.G., D.X.Z.), Cardiovascular Center (Y.N., S.C., D.S.C., A.K., A.G., X.Z., D.D.G., D.X.Z.), Department of Pharmacology and Toxicology (D.S.C., A.K.), Division of Biostatistics (R.S.), Medical College of Wisconsin, and Zablocki Veterans Affairs Medical Center (D.D.G.), Milwaukee, WI
| | - David D Gutterman
- From the Department of Medicine (Y.N., S.C., D.S.C., A.K., A.G., X.Z., D.D.G., D.X.Z.), Cardiovascular Center (Y.N., S.C., D.S.C., A.K., A.G., X.Z., D.D.G., D.X.Z.), Department of Pharmacology and Toxicology (D.S.C., A.K.), Division of Biostatistics (R.S.), Medical College of Wisconsin, and Zablocki Veterans Affairs Medical Center (D.D.G.), Milwaukee, WI
| | - David X Zhang
- From the Department of Medicine (Y.N., S.C., D.S.C., A.K., A.G., X.Z., D.D.G., D.X.Z.), Cardiovascular Center (Y.N., S.C., D.S.C., A.K., A.G., X.Z., D.D.G., D.X.Z.), Department of Pharmacology and Toxicology (D.S.C., A.K.), Division of Biostatistics (R.S.), Medical College of Wisconsin, and Zablocki Veterans Affairs Medical Center (D.D.G.), Milwaukee, WI.
| |
Collapse
|
44
|
Tsvetkov D, Tano JY, Kassmann M, Wang N, Schubert R, Gollasch M. The Role of DPO-1 and XE991-Sensitive Potassium Channels in Perivascular Adipose Tissue-Mediated Regulation of Vascular Tone. Front Physiol 2016; 7:335. [PMID: 27540364 PMCID: PMC4973012 DOI: 10.3389/fphys.2016.00335] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 07/20/2016] [Indexed: 11/13/2022] Open
Abstract
The anti-contractile effect of perivascular adipose tissue (PVAT) is an important mechanism in the modulation of vascular tone in peripheral arteries. Recent evidence has implicated the XE991-sensitive voltage-gated KV (KCNQ) channels in the regulation of arterial tone by PVAT. However, until now the in vivo pharmacology of the involved vascular KV channels with regard to XE991 remains undetermined, since XE991 effects may involve Ca(2+) activated BKCa channels and/or voltage-dependent KV1.5 channels sensitive to diphenyl phosphine oxide-1 (DPO-1). In this study, we tested whether KV1.5 channels are involved in the control of mesenteric arterial tone and its regulation by PVAT. Our study was also aimed at extending our current knowledge on the in situ vascular pharmacology of DPO-1 and XE991 regarding KV1.5 and BKCa channels, in helping to identify the nature of K(+) channels that could contribute to PVAT-mediated relaxation. XE991 at 30 μM reduced the anti-contractile response of PVAT, but had no effects on vasocontraction induced by phenylephrine (PE) in the absence of PVAT. Similar effects were observed for XE991 at 0.3 μM, which is known to almost completely inhibit mesenteric artery VSMC KV currents. 30 μM XE991 did not affect BKCa currents in VSMCs. Kcna5 (-/-) arteries and wild-type arteries incubated with 1 μM DPO-1 showed normal vasocontractions in response to PE in the presence and absence of PVAT. KV current density and inhibition by 30 μM XE991 were normal in mesenteric artery VSMCs isolated from Kcna5 (-/-) mice. We conclude that KV channels are involved in the control of arterial vascular tone by PVAT. These channels are present in VSMCs and very potently inhibited by the KCNQ channel blocker XE991. BKCa channels and/or DPO-1 sensitive KV1.5 channels in VSMCs are not the downstream mediators of the XE991 effects on PVAT-dependent arterial vasorelaxation. Further studies will need to be undertaken to examine the role of other KV channels in the phenomenon.
Collapse
Affiliation(s)
- Dmitry Tsvetkov
- Experimental and Clinical Research Center, A Joint Cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association of German Research Centres Berlin, Germany
| | - Jean-Yves Tano
- Experimental and Clinical Research Center, A Joint Cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association of German Research Centres Berlin, Germany
| | - Mario Kassmann
- Experimental and Clinical Research Center, A Joint Cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association of German Research Centres Berlin, Germany
| | - Ning Wang
- Experimental and Clinical Research Center, A Joint Cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association of German Research Centres Berlin, Germany
| | - Rudolf Schubert
- Research Division Cardiovascular Physiology, Centre for Biomedicine and Medical Technology Mannheim, Medical Faculty Mannheim of the University Heidelberg Mannheim, Germany
| | - Maik Gollasch
- Experimental and Clinical Research Center, A Joint Cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association of German Research CentresBerlin, Germany; Medical Clinic for Nephrology and Internal Intensive Care, Charité University MedicineBerlin, Germany
| |
Collapse
|
45
|
Wu W, Sanguinetti MC. Molecular Basis of Cardiac Delayed Rectifier Potassium Channel Function and Pharmacology. Card Electrophysiol Clin 2016; 8:275-84. [PMID: 27261821 DOI: 10.1016/j.ccep.2016.01.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Human cardiomyocytes express 3 distinct types of delayed rectifier potassium channels. Human ether-a-go-go-related gene (hERG) channels conduct the rapidly activating current IKr; KCNQ1/KCNE1 channels conduct the slowly activating current IKs; and Kv1.5 channels conduct an ultrarapid activating current IKur. Here the authors provide a general overview of the mechanistic and structural basis of ion selectivity, gating, and pharmacology of the 3 types of cardiac delayed rectifier potassium ion channels. Most blockers bind to S6 residues that line the central cavity of the channel, whereas activators interact with the channel at 4 symmetric binding sites outside the cavity.
Collapse
Affiliation(s)
- Wei Wu
- Department of Medicine, Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, 95 South 2000 East, Salt Lake City, UT 84112, USA
| | - Michael C Sanguinetti
- Department of Medicine, Nora Eccles Harrison Cardiovascular Research and Training Institute, University of Utah, 95 South 2000 East, Salt Lake City, UT 84112, USA.
| |
Collapse
|
46
|
Fancher IS, Butcher JT, Brooks SD, Rottgen TS, Skaff PR, Frisbee JC, Dick GM. Diphenyl phosphine oxide-1-sensitive K(+) channels contribute to the vascular tone and reactivity of resistance arteries from brain and skeletal muscle. Microcirculation 2016; 22:315-25. [PMID: 25808400 DOI: 10.1111/micc.12201] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 03/17/2015] [Indexed: 01/15/2023]
Abstract
OBJECTIVE Many types of vascular smooth muscle cells exhibit prominent KDR currents. These KDR currents may be mediated, at least in part, by KV1.5 channels, which are sensitive to inhibition by DPO-1. We tested the hypothesis that DPO-1-sensitive KDR channels regulate the tone and reactivity of resistance-sized vessels from rat brain (MCA) and skeletal muscle (GA). METHODS Middle cerebral and gracilis arteries were isolated and subjected to three kinds of experimental analysis: (i) western blot/immunocytochemistry; (ii) patch clamp electrophysiology; and (iii) pressure myography. RESULTS Western blot and immunocytochemistry experiments demonstrated KV1.5 immunoreactivity in arteries and smooth muscle cells isolated from them. Whole-cell patch clamp experiments revealed smooth muscle cells from resistance-sized arteries to possess a KDR current that was blocked by DPO-1. Resistance arteries constricted in response to increasing concentrations of DPO-1. DPO-1 enhanced constrictions to PE and serotonin in gracilis and middle cerebral arteries, respectively. When examining the myogenic response, we found that DPO-1 reduced the diameter at any given pressure. Dilations in response to ACh and SNP were reduced by DPO-1. CONCLUSION We suggest that KV1.5, a DPO-1-sensitive KDR channel, plays a major role in determining microvascular tone and the response to vasoconstrictors and vasodilators.
Collapse
Affiliation(s)
- Ibra S Fancher
- Center for Cardiovascular and Respiratory Sciences, West Virginia University School of Medicine, Morgantown, West Virginia, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Ryland KE, Hawkins AG, Weisenberger DJ, Punj V, Borinstein SC, Laird PW, Martens JR, Lawlor ER. Promoter Methylation Analysis Reveals That KCNA5 Ion Channel Silencing Supports Ewing Sarcoma Cell Proliferation. Mol Cancer Res 2015; 14:26-34. [PMID: 26573141 DOI: 10.1158/1541-7786.mcr-15-0343] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 11/04/2015] [Indexed: 02/07/2023]
Abstract
UNLABELLED Polycomb proteins are essential regulators of gene expression in stem cells and development. They function to reversibly repress gene transcription via posttranslational modification of histones and chromatin compaction. In many human cancers, genes that are repressed by polycomb in stem cells are subject to more stable silencing via DNA methylation of promoter CpG islands. Ewing sarcoma is an aggressive bone and soft-tissue tumor that is characterized by overexpression of polycomb proteins. This study investigates the DNA methylation status of polycomb target gene promoters in Ewing sarcoma tumors and cell lines and observes that the promoters of differentiation genes are frequent targets of CpG-island DNA methylation. In addition, the promoters of ion channel genes are highly differentially methylated in Ewing sarcoma compared with nonmalignant adult tissues. Ion channels regulate a variety of biologic processes, including proliferation, and dysfunction of these channels contributes to tumor pathogenesis. In particular, reduced expression of the voltage-gated Kv1.5 channel has been implicated in tumor progression. These data show that DNA methylation of the KCNA5 promoter contributes to stable epigenetic silencing of the Kv1.5 channel. This epigenetic repression is reversed by exposure to the DNA methylation inhibitor decitabine, which inhibits Ewing sarcoma cell proliferation through mechanisms that include restoration of the Kv1.5 channel function. IMPLICATIONS This study demonstrates that promoters of ion channels are aberrantly methylated in Ewing sarcoma and that epigenetic silencing of KCNA5 contributes to tumor cell proliferation, thus providing further evidence of the importance of ion channel dysregulation to tumorigenesis.
Collapse
Affiliation(s)
- Katherine E Ryland
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan. Translational Oncology Program, University of Michigan, Ann Arbor, Michigan. Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
| | - Allegra G Hawkins
- Translational Oncology Program, University of Michigan, Ann Arbor, Michigan. Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
| | - Daniel J Weisenberger
- Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles, California. Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Vasu Punj
- Keck School of Medicine, University of Southern California, Los Angeles, California
| | | | - Peter W Laird
- Van Andel Research Institute, Grand Rapids, Michigan
| | - Jeffrey R Martens
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida
| | - Elizabeth R Lawlor
- Translational Oncology Program, University of Michigan, Ann Arbor, Michigan. Department of Pediatrics, University of Michigan, Ann Arbor, Michigan. Department of Pathology, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
48
|
|
49
|
Lu HR, Whittaker R, Price JH, Vega R, Pfeiffer ER, Cerignoli F, Towart R, Gallacher DJ. High Throughput Measurement of Ca++Dynamics in Human Stem Cell-Derived Cardiomyocytes by Kinetic Image Cytometery: A Cardiac Risk Assessment Characterization Using a Large Panel of Cardioactive and Inactive Compounds. Toxicol Sci 2015; 148:503-16. [DOI: 10.1093/toxsci/kfv201] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
50
|
Kidd MW, Leo MD, Bannister JP, Jaggar JH. Intravascular pressure enhances the abundance of functional Kv1.5 channels at the surface of arterial smooth muscle cells. Sci Signal 2015; 8:ra83. [PMID: 26286025 DOI: 10.1126/scisignal.aac5128] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Voltage-dependent potassium (K(v)) channels are present in various cell types, including smooth muscle cells (myocytes) of resistance-sized arteries that control systemic blood pressure and regional organ blood flow. Intravascular pressure depolarizes arterial myocytes, stimulating calcium (Ca(2+)) influx through voltage-dependent Ca(2+) (Ca(v)) channels that results in vasoconstriction and also K(+) efflux through K(v) channels that oppose vasoconstriction. We hypothesized that pressure-induced depolarization may not only increase the open probability of plasma membrane-resident K(v) channels but also increase the abundance of these channels at the surface of arterial myocytes to limit vasoconstriction. We found that K(v)1.5 and K(v)2.1 proteins were abundant in the myocytes of resistance-sized mesenteric arteries. K(v)1.5, but not K(v)2.1, continuously recycled between the intracellular compartment and the plasma membrane in contractile arterial myocytes. Using ex vivo preparations of intact arteries, we showed that physiological intravascular pressure through membrane depolarization or membrane depolarization in the absence of pressure inhibited the degradation of internalized K(v)1.5 and increased recycling of K(v)1.5 to the plasma membrane. Accordingly, by stimulating the activity of Ca(v)1.2, membrane depolarization increased whole-cell K(v)1.5 current density in myocytes and K(v)1.5 channel activity in pressurized arteries. In contrast, the total amount and cell surface abundance of K(v)2.1 were independent of intravascular pressure or membrane potential. Thus, our data indicate that intravascular pressure-induced membrane depolarization selectively increased K(v)1.5 surface abundance to increase K(v) currents in arterial myocytes, which would limit vasoconstriction.
Collapse
Affiliation(s)
- Michael W Kidd
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - M Dennis Leo
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - John P Bannister
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jonathan H Jaggar
- Department of Physiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| |
Collapse
|