1
|
Namekata I, Seki M, Saito T, Odaka R, Hamaguchi S, Tanaka H. Automaticity of the Pulmonary Vein Myocardium and the Effect of Class I Antiarrhythmic Drugs. Int J Mol Sci 2024; 25:12367. [PMID: 39596432 PMCID: PMC11595185 DOI: 10.3390/ijms252212367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/11/2024] [Accepted: 11/16/2024] [Indexed: 11/28/2024] Open
Abstract
The pulmonary vein wall contains a myocardial layer whose ectopic automaticity is the major cause of atrial fibrillation. This review summarizes the results obtained in isolated pulmonary vein myocardium from small experimental animals, focusing on the studies with the guinea pig. The diversity in the action potential waveform reflects the difference in the repolarizing potassium channel currents involved. The diastolic depolarization, the trigger of automatic action potentials, is caused by multiple membrane currents, including the Na+-Ca2+ exchanger current and late INa. The action potential waveform and automaticity are affected differentially by α- and β-adrenoceptor stimulation. Class I antiarrhythmic drugs block the propagation of ectopic electrical activity of the pulmonary vein myocardium through blockade of the peak INa. Some of the class I antiarrhythmic drugs block the late INa and inhibit pulmonary vein automaticity. The negative inotropic and chronotropic effects of class I antiarrhythmic drugs could be largely attributed to their blocking effect on the Ca2+ channel rather than the Na+ channel. Such a comprehensive understanding of pulmonary vein automaticity and class I antiarrhythmic drugs would lead to an improvement in pharmacotherapy and the development of novel therapeutic agents for atrial fibrillation.
Collapse
Affiliation(s)
| | | | | | | | | | - Hikaru Tanaka
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Toho University, 2-2-1 Miyama Funabashi, Chiba 274-8510, Japan; (I.N.); (M.S.); (T.S.); (R.O.); (S.H.)
| |
Collapse
|
2
|
Saito T, Suzuki M, Ohba A, Hamaguchi S, Namekata I, Tanaka H. Enhanced Late I Na Induces Intracellular Ion Disturbances and Automatic Activity in the Guinea Pig Pulmonary Vein Cardiomyocytes. Int J Mol Sci 2024; 25:8688. [PMID: 39201376 PMCID: PMC11354854 DOI: 10.3390/ijms25168688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 07/30/2024] [Accepted: 08/07/2024] [Indexed: 09/02/2024] Open
Abstract
The effects of enhanced late INa, a persistent component of the Na+ channel current, on the intracellular ion dynamics and the automaticity of the pulmonary vein cardiomyocytes were studied with fluorescent microscopy. Anemonia viridis toxin II (ATX- II), an enhancer of late INa, caused increases in the basal Na+ and Ca2+ concentrations, increases in the number of Ca2+ sparks and Ca2+ waves, and the generation of repetitive Ca2+ transients. These phenomena were inhibited by eleclazine, a blocker of the late INa; SEA0400, an inhibitor of the Na+/Ca2+ exchanger (NCX); H89, a protein kinase A (PKA) inhibitor; and KN-93, a Ca2+/calmodulin-dependent protein kinase II (CaMKII) inhibitor. These results suggest that enhancement of late INa in the pulmonary vein cardiomyocytes causes disturbance of the intracellular ion environment through activation of the NCX and Ca2+-dependent enzymes. Such mechanisms are probably involved in the ectopic electrical activity of the pulmonary vein myocardium.
Collapse
Affiliation(s)
| | | | | | | | - Iyuki Namekata
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Toho University, 2-2-1 Miyama Funabashi, Chiba 274-8510, Japan; (T.S.); (M.S.); (A.O.); (S.H.); (H.T.)
| | | |
Collapse
|
3
|
Dzhumaniiazova I, Vornanen M, Pustovit OB, Voronkov YI, Abramochkin DV. Effects of Tetrodotoxin and Ranolazine on the Late INa of Zebrafish Ventricular Myocytes. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022070031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
4
|
Namekata I, Hiiro H, Odaka R, Saito T, Hamaguchi S, Tsukamoto T, Ishikawa R, Katayama Y, Kondo Y, Tanaka H. Inhibitory Effect of a Late Sodium Current Blocker, NCC-3902, on the Automaticity of the Guinea Pig Pulmonary Vein Myocardium. Biol Pharm Bull 2022; 45:1644-1652. [DOI: 10.1248/bpb.b22-00362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Iyuki Namekata
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Toho University
| | - Haruhito Hiiro
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Toho University
| | - Ryosuke Odaka
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Toho University
| | - Taro Saito
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Toho University
| | - Shogo Hamaguchi
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Toho University
| | - Tadaaki Tsukamoto
- Pharmacologicals Group, Medicinal Research Department, Biological Research Laboratories, Nissan Chemical Corporation
| | - Ryutaro Ishikawa
- Pharmacologicals Group, Medicinal Research Department, Biological Research Laboratories, Nissan Chemical Corporation
| | - Yoshimi Katayama
- Pharmacologicals Group, Medicinal Research Department, Biological Research Laboratories, Nissan Chemical Corporation
| | | | - Hikaru Tanaka
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Toho University
| |
Collapse
|
5
|
Lu YY, Cheng CC, Huang SY, Chen YC, Kao YH, Lin YK, Higa S, Chen SA, Chen YJ. Fibroblast Growth Factor 1 Reduces Pulmonary Vein and Atrium Arrhythmogenesis via Modification of Oxidative Stress and Sodium/Calcium Homeostasis. Front Cardiovasc Med 2022; 8:813589. [PMID: 35118146 PMCID: PMC8804298 DOI: 10.3389/fcvm.2021.813589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/21/2021] [Indexed: 11/13/2022] Open
Abstract
Rationale Atrial fibrillation is a critical health burden. Targeting calcium (Ca2+) dysregulation and oxidative stress are potential upstream therapeutic strategies. Fibroblast growth factor (FGF) 1 can modulate Ca2+ homeostasis and has antioxidant activity. The aim of this study was to investigate whether FGF1 has anti-arrhythmic potential through modulating Ca2+ homeostasis and antioxidant activity of pulmonary vein (PV) and left atrium (LA) myocytes. Methods Patch clamp, western blotting, confocal microscopy, cellular and mitochondrial oxidative stress studies were performed in isolated rabbit PV and LA myocytes treated with or without FGF1 (1 and 10 ng/mL). Conventional microelectrodes were used to record electrical activity in isolated rabbit PV and LA tissue preparations with and without FGF1 (3 μg/kg, i.v.). Results FGF1-treated rabbits had a slower heart rate than that observed in controls. PV and LA tissues in FGF1-treated rabbits had slower beating rates and longer action potential duration than those observed in controls. Isoproterenol (1 μM)-treated PV and LA tissues in the FGF1-treated rabbits showed less changes in the increased beating rate and a lower incidence of tachypacing (20 Hz)-induced burst firing than those observed in controls. FGF1 (10 ng/mL)-treated PV and LA myocytes had less oxidative stress and Ca2+ transient than those observed in controls. Compared to controls, FGF1 (10 ng/mL) decreased INa−L in PV myocytes and lowered Ito, IKr−tail in LA myocytes. Protein kinase C (PKC)ε inhibition abolished the effects of FGF1 on the ionic currents of LA and PV myocytes. Conclusion FGF1 changes PV and LA electrophysiological characteristics possibly via modulating oxidative stress, Na+/Ca2+ homeostasis, and the PKCε pathway.
Collapse
Affiliation(s)
- Yen-Yu Lu
- Division of Cardiology, Department of Internal Medicine, Sijhih Cathay General Hospital, New Taipei City, Taiwan
- School of Medicine, College of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Chen-Chuan Cheng
- Division of Cardiology, Chi-Mei Medical Center, Tainan City, Taiwan
| | - Shih-Yu Huang
- School of Medicine, College of Medicine, Fu-Jen Catholic University, New Taipei City, Taiwan
- Division of Cardiac Electrophysiology, Cardiovascular Center, Cathay General Hospital, Taipei, Taiwan
- Post-Baccalaureate Medicine, College of Life Science, National Tsing Hua University, Hsinchu City, Taiwan
| | - Yao-Chang Chen
- Department of Biomedical Engineering, National Defense Medical Center, Taipei, Taiwan
| | - Yu-Hsun Kao
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yung-Kuo Lin
- Cardiovascular Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Satoshi Higa
- Cardiac Electrophysiology and Pacing Laboratory, Division of Cardiovascular Medicine, Makiminato Central Hospital, Okinawa, Japan
| | - Shih-Ann Chen
- Heart Rhythm Center and Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Cardiovascular Center, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yi-Jen Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- Cardiovascular Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
- *Correspondence: Yi-Jen Chen
| |
Collapse
|
6
|
Ranolazine: An Old Drug with Emerging Potential; Lessons from Pre-Clinical and Clinical Investigations for Possible Repositioning. Pharmaceuticals (Basel) 2021; 15:ph15010031. [PMID: 35056088 PMCID: PMC8777683 DOI: 10.3390/ph15010031] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 12/16/2021] [Accepted: 12/20/2021] [Indexed: 02/07/2023] Open
Abstract
Ischemic heart disease is a significant public health problem with high mortality and morbidity. Extensive scientific investigations from basic sciences to clinics revealed multilevel alterations from metabolic imbalance, altered electrophysiology, and defective Ca2+/Na+ homeostasis leading to lethal arrhythmias. Despite the recent identification of numerous molecular targets with potential therapeutic interest, a pragmatic observation on the current pharmacological R&D output confirms the lack of new therapeutic offers to patients. By contrast, from recent trials, molecules initially developed for other fields of application have shown cardiovascular benefits, as illustrated with some anti-diabetic agents, regardless of the presence or absence of diabetes, emphasizing the clear advantage of “old” drug repositioning. Ranolazine is approved as an antianginal agent and has a favorable overall safety profile. This drug, developed initially as a metabolic modulator, was also identified as an inhibitor of the cardiac late Na+ current, although it also blocks other ionic currents, including the hERG/Ikr K+ current. The latter actions have been involved in this drug’s antiarrhythmic effects, both on supraventricular and ventricular arrhythmias (VA). However, despite initial enthusiasm and promising development in the cardiovascular field, ranolazine is only authorized as a second-line treatment in patients with chronic angina pectoris, notwithstanding its antiarrhythmic properties. A plausible reason for this is the apparent difficulty in linking the clinical benefits to the multiple molecular actions of this drug. Here, we review ranolazine’s experimental and clinical knowledge on cardiac metabolism and arrhythmias. We also highlight advances in understanding novel effects on neurons, the vascular system, skeletal muscles, blood sugar control, and cancer, which may open the way to reposition this “old” drug alone or in combination with other medications.
Collapse
|
7
|
Kaplan AD, Joca HC, Boyman L, Greiser M. Calcium Signaling Silencing in Atrial Fibrillation: Implications for Atrial Sodium Homeostasis. Int J Mol Sci 2021; 22:10513. [PMID: 34638854 PMCID: PMC8508839 DOI: 10.3390/ijms221910513] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 09/14/2021] [Accepted: 09/14/2021] [Indexed: 12/19/2022] Open
Abstract
Atrial fibrillation (AF) is the most common type of cardiac arrhythmia, affecting more than 33 million people worldwide. Despite important advances in therapy, AF's incidence remains high, and treatment often results in recurrence of the arrhythmia. A better understanding of the cellular and molecular changes that (1) trigger AF and (2) occur after the onset of AF will help to identify novel therapeutic targets. Over the past 20 years, a large body of research has shown that intracellular Ca2+ handling is dramatically altered in AF. While some of these changes are arrhythmogenic, other changes counteract cellular arrhythmogenic mechanisms (Calcium Signaling Silencing). The intracellular Na+ concentration ([Na+])i is a key regulator of intracellular Ca2+ handling in cardiac myocytes. Despite its importance in the regulation of intracellular Ca2+ handling, little is known about [Na+]i, its regulation, and how it might be changed in AF. Previous work suggests that there might be increases in the late component of the atrial Na+ current (INa,L) in AF, suggesting that [Na+]i levels might be high in AF. Indeed, a pharmacological blockade of INa,L has been suggested as a treatment for AF. Here, we review calcium signaling silencing and changes in intracellular Na+ homeostasis during AF. We summarize the proposed arrhythmogenic mechanisms associated with increases in INa,L during AF and discuss the evidence from clinical trials that have tested the pharmacological INa,L blocker ranolazine in the treatment of AF.
Collapse
Affiliation(s)
- Aaron D. Kaplan
- Center for Biomedical Engineering and Technology, Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (A.D.K.); (H.C.J.); (L.B.)
- Division of Cardiovascular Medicine, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Humberto C. Joca
- Center for Biomedical Engineering and Technology, Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (A.D.K.); (H.C.J.); (L.B.)
| | - Liron Boyman
- Center for Biomedical Engineering and Technology, Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (A.D.K.); (H.C.J.); (L.B.)
| | - Maura Greiser
- Center for Biomedical Engineering and Technology, Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (A.D.K.); (H.C.J.); (L.B.)
| |
Collapse
|
8
|
Eleutheroside B, a selective late sodium current inhibitor, suppresses atrial fibrillation induced by sea anemone toxin II in rabbit hearts. Acta Pharmacol Sin 2021; 42:209-217. [PMID: 32612277 DOI: 10.1038/s41401-020-0453-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 06/01/2020] [Indexed: 11/09/2022]
Abstract
Eleutheroside B (EB) is the main active constituent derived from the Chinese herb Acanthopanax senticosus (AS) that has been reported to possess cardioprotective effects. In this study we investigated the effects of EB on cardiac electrophysiology and its suppression on atrial fibrillation (AF). Whole-cell recording was conducted in isolated rabbit atrial myocytes. The intracellular calcium ([Ca2+]i) concentration was measured using calcium indicator Fura-2/AM fluorescence. Monophasic action potential (MAP) and electrocardiogram (ECG) synchronous recordings were conducted in Langendorff-perfused rabbit hearts using ECG signal sampling and analysis system. We showed that EB dose-dependently inhibited late sodium current (INaL), transient sodium current (INaT), and sea anemone toxin II (ATX II)-increased INaL with IC50 values of 167, 1582, and 181 μM, respectively. On the other hand, EB (800 μM) did not affect L-type calcium current (ICaL), inward rectifier potassium channel current (IK), and action potential duration (APD). Furthermore, EB (300 μM) markedly decreased ATX II-prolonged the APD at 90% repolarization (APD90) and eliminated ATX II-induced early afterdepolarizations (EADs), delayed afterdepolarizations (DADs), and triggered activities (TAs). Moreover, EB (200 μM) significantly suppressed ATX II-induced Na+-dependent [Ca2+]i overload in atrial myocytes. In the Langendorff-perfused rabbit hearts, application of EB (200 μM) or TTX (2 μM) substantially decreased ATX II-induced incidences of atrial fibrillation (AF), ventricular fibrillation (VF), and heart death. These results suggest that augmented INaL alone is sufficient to induce AF, and EB exerts anti-AF actions mainly via blocking INaL, which put forward the basis of pharmacology for new clinical application of EB.
Collapse
|
9
|
Irie M, Hiiro H, Hamaguchi S, Namekata I, Tanaka H. Involvement of the persistent Na+ current in the diastolic depolarization and automaticity of the guinea pig pulmonary vein myocardium. J Pharmacol Sci 2019; 141:9-16. [DOI: 10.1016/j.jphs.2019.08.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 07/31/2019] [Accepted: 08/13/2019] [Indexed: 02/07/2023] Open
|
10
|
Sicouri S, Belardinelli L, Antzelevitch C. Effect of autonomic influences to induce triggered activity in muscular sleeves extending into the coronary sinus of the canine heart and its suppression by ranolazine. J Cardiovasc Electrophysiol 2018; 30:230-238. [PMID: 30302862 DOI: 10.1111/jce.13770] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Revised: 10/01/2018] [Accepted: 10/05/2018] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Extrasystoles arising from the muscular sleeves associated with the pulmonary veins (PV), superior vena cava (SVC), and coronary sinus (CS) are known to precipitate atrial fibrillation (AF). The late sodium channel current (INa ) inhibitor ranolazine has been reported to exert antiarrhythmic effects in canine PV and SVC sleeves by suppressing late phase 3 early and delayed after depolarization (EAD and DAD)-induced triggered activity induced by parasympathetic and/or sympathetic stimulation. The current study was designed to extend our existing knowledge of the electrophysiological and pharmacologic properties of canine CS preparations and assess their response to inhibition of late INa following autonomic stimulation. METHODS Transmembrane action potentials were recorded from canine superfused CS using standard microelectrode techniques. Acetylcholine (ACh, 1 µM), isoproterenol (Iso, 1 µM), high calcium ([Ca2+ ]o = 5.4 mM), or a combination were used to induce EADs, DADs, and triggered activity. RESULTS Action potentials (AP) recorded from the CS displayed short and long AP durations (APD), with and without phase 4 depolarization (n = 19). Iso induced DAD-mediated triggered activity. The combination of sympathetic and parasympathetic agonists resulted in late phase 3 EAD-induced triggered activity in all CS preparations. Ranolazine (5-10 µM) suppressed late phase 3 EAD- and DAD-induced triggered activity in 8 of 8 preparations. Subthreshold stimulation induced a prominent hyperpolarization that could be suppressed by atropine. CONCLUSIONS Our results suggest the important role of parasympathetic innervation in the activity of the CS. Autonomic influences promote DAD- and late phase-3-EAD-mediated triggered activity in canine CS, thus generating extrasystolic activity capable of initiating atrial arrhythmias. Ranolazine effectively suppresses these triggers.
Collapse
Affiliation(s)
- Serge Sicouri
- Department of Experimental Cardiology, Masonic Medical Research Laboratory, Utica, New York.,Cardiovascular Research Program, Lankenau Institute for Medical Research, Wynnewood, Pennsylvania
| | | | - Charles Antzelevitch
- Department of Experimental Cardiology, Masonic Medical Research Laboratory, Utica, New York.,Cardiovascular Research Program, Lankenau Institute for Medical Research, Wynnewood, Pennsylvania.,Lankenau Heart Institute, Wynnewood, Pennsylvania.,Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
11
|
Late sodium current associated cardiac electrophysiological and mechanical dysfunction. Pflugers Arch 2017; 470:461-469. [DOI: 10.1007/s00424-017-2079-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 09/27/2017] [Accepted: 10/09/2017] [Indexed: 12/19/2022]
|
12
|
Azam MA, Zamiri N, Massé S, Kusha M, Lai PFH, Nair GK, Tan NS, Labos C, Nanthakumar K. Effects of Late Sodium Current Blockade on Ventricular Refibrillation in a Rabbit Model. Circ Arrhythm Electrophysiol 2017; 10:CIRCEP.116.004331. [PMID: 28314848 DOI: 10.1161/circep.116.004331] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 01/19/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND After defibrillation of initial ventricular fibrillation (VF), it is crucial to prevent refibrillation to ensure successful resuscitation outcomes. Inability of the late Na+ current to inactivate leads to intracellular Ca2+ dysregulation and arrhythmias. Our aim was to determine the effects of ranolazine and GS-967, inhibitors of the late Na+ current, on ventricular refibrillation. METHODS AND RESULTS Long-duration VF was induced electrically in Langendorff-perfused rabbit hearts (n=22) and terminated with a defibrillator after 6 minutes. Fibrillating hearts were randomized into 3 groups: treatment with ranolazine, GS-967, or nontreated controls. In the treated groups, hearts were perfused with ranolazine or GS-967 at 2 minutes of VF. In control experiments, perfusion solution was supplemented with isotonic saline in lieu of a drug. Inducibility of refibrillation was assessed after initial long-duration VF by attempting to reinduce VF. Sustained refibrillation was successful in fewer ranolazine-treated (29.17%; P=0.005) or GS-967-treated (45.83%, P=0.035) hearts compared with that in nontreated control hearts (84.85%). In GS-967-treated hearts, significantly more spontaneous termination of initial long-duration VF was observed (66.67%; P=0.01). Ca2+ transient duration was reduced in ranolazine-treated hearts compared with that in controls (P=0.05) and also Ca2+ alternans (P=0.03). CONCLUSIONS Late Na+ current inhibition during long-duration VF reduces the susceptibility to subsequent refibrillation, partially by mitigating dysregulation of intracellular Ca2+. These results suggest the potential therapeutic use of ranolazine and GS-967 and call for further testing in cardiac arrest models.
Collapse
Affiliation(s)
- Mohammed Ali Azam
- From the Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Ontario, Canada (M.A.A., N.Z., S.M., M.K., P.F.H.L., G.K.N., N.S.T., K.N.); and Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada (C.L.)
| | - Nima Zamiri
- From the Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Ontario, Canada (M.A.A., N.Z., S.M., M.K., P.F.H.L., G.K.N., N.S.T., K.N.); and Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada (C.L.)
| | - Stéphane Massé
- From the Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Ontario, Canada (M.A.A., N.Z., S.M., M.K., P.F.H.L., G.K.N., N.S.T., K.N.); and Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada (C.L.)
| | - Marjan Kusha
- From the Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Ontario, Canada (M.A.A., N.Z., S.M., M.K., P.F.H.L., G.K.N., N.S.T., K.N.); and Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada (C.L.)
| | - Patrick F H Lai
- From the Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Ontario, Canada (M.A.A., N.Z., S.M., M.K., P.F.H.L., G.K.N., N.S.T., K.N.); and Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada (C.L.)
| | - Govind K Nair
- From the Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Ontario, Canada (M.A.A., N.Z., S.M., M.K., P.F.H.L., G.K.N., N.S.T., K.N.); and Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada (C.L.)
| | - Nigel S Tan
- From the Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Ontario, Canada (M.A.A., N.Z., S.M., M.K., P.F.H.L., G.K.N., N.S.T., K.N.); and Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada (C.L.)
| | - Christopher Labos
- From the Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Ontario, Canada (M.A.A., N.Z., S.M., M.K., P.F.H.L., G.K.N., N.S.T., K.N.); and Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada (C.L.)
| | - Kumaraswamy Nanthakumar
- From the Hull Family Cardiac Fibrillation Management Laboratory, Toronto General Hospital, University Health Network, Ontario, Canada (M.A.A., N.Z., S.M., M.K., P.F.H.L., G.K.N., N.S.T., K.N.); and Institute for Clinical Evaluative Sciences, Toronto, Ontario, Canada (C.L.).
| |
Collapse
|
13
|
Chadda KR, Jeevaratnam K, Lei M, Huang CLH. Sodium channel biophysics, late sodium current and genetic arrhythmic syndromes. Pflugers Arch 2017; 469:629-641. [PMID: 28265756 PMCID: PMC5438422 DOI: 10.1007/s00424-017-1959-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 02/14/2017] [Indexed: 12/11/2022]
Abstract
Arrhythmias arise from breakdown of orderly action potential (AP) activation, propagation and recovery driven by interactive opening and closing of successive voltage-gated ion channels, in which one or more Na+ current components play critical parts. Early peak, Na+ currents (INa) reflecting channel activation drive the AP upstroke central to cellular activation and its propagation. Sustained late Na+ currents (INa-L) include contributions from a component with a delayed inactivation timecourse influencing AP duration (APD) and refractoriness, potentially causing pro-arrhythmic phenotypes. The magnitude of INa-L can be analysed through overlaps or otherwise in the overall voltage dependences of the steady-state properties and kinetics of activation and inactivation of the Na+ conductance. This was useful in analysing repetitive firing associated with paramyotonia congenita in skeletal muscle. Similarly, genetic cardiac Na+ channel abnormalities increasing INa-L are implicated in triggering phenomena of automaticity, early and delayed afterdepolarisations and arrhythmic substrate. This review illustrates a wide range of situations that may accentuate INa-L. These include (1) overlaps between steady-state activation and inactivation increasing window current, (2) kinetic deficiencies in Na+ channel inactivation leading to bursting phenomena associated with repetitive channel openings and (3) non-equilibrium gating processes causing channel re-opening due to more rapid recoveries from inactivation. All these biophysical possibilities were identified in a selection of abnormal human SCN5A genotypes. The latter presented as a broad range of clinical arrhythmic phenotypes, for which effective therapeutic intervention would require specific identification and targeting of the diverse electrophysiological abnormalities underlying their increased INa-L.
Collapse
Affiliation(s)
- Karan R Chadda
- Faculty of Health and Medical Sciences, University of Surrey, VSM Building, Guildford, GU2 7AL, UK
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge, CB2 3EG, UK
| | - Kamalan Jeevaratnam
- Faculty of Health and Medical Sciences, University of Surrey, VSM Building, Guildford, GU2 7AL, UK
- School of Medicine, Perdana University-Royal College of Surgeons Ireland, 43400, Serdang, Selangor Darul Ehsan, Malaysia
| | - Ming Lei
- Department of Pharmacology, University of Oxford, Oxford, OX1 3QT, UK
| | - Christopher L-H Huang
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge, CB2 3EG, UK.
- Department of Biochemistry, University of Cambridge, Hopkins Building, Cambridge, CB2 1QW, UK.
| |
Collapse
|
14
|
Hwang HR, Tai BY, Cheng PY, Chen PN, Sung PJ, Wen ZH, Hsu CH. Excavatolide B Modulates the Electrophysiological Characteristics and Calcium Homeostasis of Atrial Myocytes. Mar Drugs 2017; 15:md15020025. [PMID: 28125029 PMCID: PMC5334606 DOI: 10.3390/md15020025] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 01/10/2017] [Accepted: 01/18/2017] [Indexed: 12/19/2022] Open
Abstract
Severe bacterial infections caused by sepsis always result in profound physiological changes, including fever, hypotension, arrhythmia, necrosis of tissue, systemic multi-organ dysfunction, and finally death. The lipopolysaccharide (LPS) provokes an inflammatory response under sepsis, which may increase propensity to arrhythmogenesis. Excavatolide B (EXCB) possesses potent anti-inflammatory effects. However, it is not clear whether EXCB could modulate the electrophysiological characteristics and calcium homeostasis of atrial myocytes. This study investigated the effects of EXCB on the atrial myocytes exposed to lipopolysaccharide. A whole-cell patch clamp and indo-1 fluorimetric ratio technique was employed to record the action potential (AP), ionic currents, and intracellular calcium ([Ca2+]i) in single, isolated rabbit left atrial (LA) cardiomyocytes, with and without LPS (1 μg/mL) and LPS + EXCB administration (10 μM) for 6 ± 1 h, in order to investigate the role of EXCB on atrial electrophysiology. In the presence of LPS, EXCB-treated LA myocytes (n = 13) had a longer AP duration at 20% (29 ± 2 vs. 20 ± 2 ms, p < 0.05), 50% (52 ± 4 vs. 40 ± 3 ms, p < 0.05), and 90% (85 ± 5 vs. 68 ± 3 ms, p < 0.05), compared to the LPS-treated cells (n = 12). LPS-treated LA myocytes showed a higher late sodium current, Na+/Ca2+ exchanger current, transient outward current, and delayed rectifier potassium current, but a lower l-type Ca2+ current, than the control LA myocytes. Treatment with EXCB reversed the LPS-induced alterations of the ionic currents. LPS-treated, EXCB-treated, and control LA myocytes exhibited similar Na+ currents. In addition, the LPS-treated LA myocytes exhibited a lower [Ca2+]i content and higher sarcoplasmic reticulum calcium content, than the controls. EXCB reversed the LPS-induced calcium alterations. In conclusion, EXCB modulates LPS-induced LA electrophysiological characteristics and calcium homeostasis, which may contribute to attenuating LPS-induced arrhythmogenesis.
Collapse
Affiliation(s)
- Hwong-Ru Hwang
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 804, Taiwan.
- Division of Cardiology, Department of Medicine, E-Da Hospital, Kaohsiung 824, Taiwan.
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan.
| | - Buh-Yuan Tai
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 804, Taiwan.
- Department of Traditional Medicine, Jianan Mental Hospital, Tainan 717, Taiwan.
| | - Pao-Yun Cheng
- Department of Physiology and Biophysics and Graduate Institute of Physiology, National Defense Medical Center, Taipei 114, Taiwan.
| | - Ping-Nan Chen
- Department of Biomedical Engineering, National Defense Medical Center, Taipei 114, Taiwan.
| | - Ping-Jyun Sung
- National Museum of Marine Biology and Aquarium, Pingtung 944, Taiwan.
- Chinese Medicine Research and Development Center, China Medical University Hospital, Taichung 404, Taiwan.
- Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 804, Taiwan.
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-Sen University and Academia Sinica, Kaohsiung 804, Taiwan.
| | - Chih-Hsueng Hsu
- Division of Cardiology, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan.
| |
Collapse
|
15
|
Epigallocatechin-3-gallate modulates arrhythmogenic activity and calcium homeostasis of left atrium. Int J Cardiol 2017; 236:174-180. [PMID: 28117139 DOI: 10.1016/j.ijcard.2017.01.090] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 01/10/2017] [Accepted: 01/13/2017] [Indexed: 11/20/2022]
Abstract
BACKGROUND Atrial fibrillation (AF) is the commonest sustained arrhythmia, and increases the risk of stroke, heart failure, and mortality. Calcium (Ca2+) overload and oxidative stress are thought to participate in the pathogenesis of AF. Epigallocatechin-3-gallate (EGCG) has an antioxidative effect and been shown to be beneficial in promoting cardiovascular health. However, it is not clear if EGCG directly modulates the electrophysiological characteristics and Ca2+ homeostasis of the left atrium (LA). METHODS AND RESULTS Conventional microelectrodes, whole-cell patch-clamp, and Fluo-3 fluorometric ratio technique were performed using the isolated rabbit LA preparations or isolated single LA cardiomyocytes before and after EGCG treatment. EGCG (0.01, 0.1, 1, and 10μM) which concentration-dependently decreased the APD20 by 13±8%, 25±5%, 31±6%, and 37±5%, APD50 by 9±8%, 22±6%, 32±7%, and 40±4%, and APD90 by 2±12%, 9±8%, 24±10%, and 34±5% in LA preparations. EGCG (0.1μM) decreased the late sodium (Na+) current, L-type Ca2+ current, nickel-sensitive Na+-Ca2+ exchanger current, and transient outward current, but did not change the Na+ current and ultra-rapid delayed rectifier potassium current in LA cardiomyocytes. EGCG decreased intracellular Ca2+ transient and sarcoplasmic reticulum Ca2+ content in LA cardiomyocytes. Furthermore, EGCG decreased isoproterenol (ISO, 1μM)-induced burst firing. KT5823 (1μM) or KN93 (1μM) decreased the incidences of ISO-induced LA burst firing, which became lower with EGCG treatment. H89 (10μM) and KN92 (1μM) did not suppress the incidence of ISO-induced LA burst firing. However, EGCG decreased the incidences of ISO-induced LA burst firing in the presence of H89 or KN92. CONCLUSION EGCG directly regulates LA electrophysiological characteristics and Ca2+ homeostasis, and suppresses ISO-induced atrial arrhythmogenesis through inhibiting Ca2+/calmodulin or cGMP-dependent protein kinases.
Collapse
|
16
|
Mason FE, Sossalla S. The Significance of the Late Na+ Current for Arrhythmia Induction and the Therapeutic Antiarrhythmic Potential of Ranolazine. J Cardiovasc Pharmacol Ther 2016; 22:40-50. [DOI: 10.1177/1074248416644989] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The purpose of this article is to review the basis of arrhythmogenesis, the functional and clinical role of the late Na current, and its therapeutic inhibition. Under pathological conditions such as ischemia and heart failure this current is abnormally enhanced and influences cellular electrophysiology as a proarrhythmic substrate in myocardial pathology. Ranolazine the only approved late Na current blocker has been demonstrated to produce antiarrhythmic effects in the atria and the ventricle. We summarize recent experimental and clinical studies of ranolazine and other experimental late Na current blockers and discuss the significance of the available data.
Collapse
Affiliation(s)
- Fleur E. Mason
- Department of Cardiology and Pneumology, Georg-August-University Göttingen, Göttingen, Germany
| | - Samuel Sossalla
- Department of Cardiology and Pneumology, Georg-August-University Göttingen, Göttingen, Germany
- Department of Internal Medicine III (Cardiology and Angiology), University Hospital Schleswig-Holstein, Kiel, Germany
- German Centre for Cardiovascular Research (DZHK), Göttingen & Kiel, Germany
| |
Collapse
|
17
|
Hartmann N, Mason FE, Braun I, Pabel S, Voigt N, Schotola H, Fischer TH, Dobrev D, Danner BC, Renner A, Gummert J, Belardinelli L, Frey N, Maier LS, Hasenfuss G, Sossalla S. The combined effects of ranolazine and dronedarone on human atrial and ventricular electrophysiology. J Mol Cell Cardiol 2016; 94:95-106. [PMID: 27056421 DOI: 10.1016/j.yjmcc.2016.03.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 02/20/2016] [Accepted: 03/23/2016] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Pharmacological rhythm control of atrial fibrillation (AF) in patients with structural heart disease is limited. Ranolazine in combination with low dose dronedarone remarkably reduced AF-burden in the phase II HARMONY trial. We thus aimed to investigate the possible mechanisms underlying these results. METHODS AND RESULTS Patch clamp experiments revealed that ranolazine (5μM), low-dose dronedarone (0.3μM), and the combination significantly prolonged action potential duration (APD90) in atrial myocytes from patients in sinus rhythm (prolongation by 23.5±0.1%, 31.7±0.1% and 25.6±0.1% respectively). Most importantly, in atrial myocytes from patients with AF ranolazine alone, but more the combination with dronedarone, also prolonged the typically abbreviated APD90 (prolongation by 21.6±0.1% and 31.9±0.1% respectively). It was clearly observed that neither ranolazine, dronedarone nor the combination significantly changed the APD or contractility and twitch force in ventricular myocytes or trabeculae from patients with heart failure (HF). Interestingly ranolazine, and more so the combination, but not dronedarone alone, caused hyperpolarization of the resting membrane potential in cardiomyocytes from AF. As measured by confocal microscopy (Fluo-3), ranolazine, dronedarone and the combination significantly suppressed diastolic sarcoplasmic reticulum (SR) Ca(2+) leak in myocytes from sinus rhythm (reduction by ranolazine: 89.0±30.7%, dronedarone: 75.6±27.4% and combination: 78.0±27.2%), in myocytes from AF (reduction by ranolazine: 67.6±33.7%, dronedarone: 86.5±31.7% and combination: 81.0±33.3%), as well as in myocytes from HF (reduction by ranolazine: 64.8±26.5% and dronedarone: 65.9±29.3%). CONCLUSIONS Electrophysiological measurements during exposure to ranolazine alone or in combination with low-dose dronedarone showed APD prolongation, cellular hyperpolarization and reduced SR Ca(2+) leak in human atrial myocytes. The combined inhibitory effects on various currents, in particular Na(+) and K(+) currents, may explain the anti-AF effects observed in the HARMONY trial. Therefore, the combination of ranolazine and dronedarone, but also ranolazine alone, may be promising new treatment options for AF, especially in patients with HF, and merit further clinical investigation.
Collapse
Affiliation(s)
- Nico Hartmann
- Department of Cardiology and Pneumology, Georg August University Göttingen, Göttingen, Germany
| | - Fleur E Mason
- Department of Cardiology and Pneumology, Georg August University Göttingen, Göttingen, Germany
| | - Inga Braun
- Department of Cardiology and Pneumology, Georg August University Göttingen, Göttingen, Germany
| | - Steffen Pabel
- Department of Cardiology and Pneumology, Georg August University Göttingen, Göttingen, Germany
| | - Niels Voigt
- Institute of Pharmacology, West German Heart and Vascular Center, Faculty of Medicine, University Duisburg-Essen, Essen, Germany
| | - Hanna Schotola
- Department of Anesthesiology, Emergency and Intensive Care Medicine, Georg August University Göttingen, Göttingen, Germany
| | - Thomas H Fischer
- Department of Cardiology and Pneumology, Georg August University Göttingen, Göttingen, Germany
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, Faculty of Medicine, University Duisburg-Essen, Essen, Germany
| | - Bernhard C Danner
- Department of Thoracic and Cardiovascular Surgery, Georg August University Göttingen, Göttingen, Germany
| | - André Renner
- Department of Thoracic and Cardiovascular Surgery, Heart and Diabetes Center NRW, Ruhr University Bochum, Bad Oeynhausen, Germany
| | - Jan Gummert
- Department of Thoracic and Cardiovascular Surgery, Heart and Diabetes Center NRW, Ruhr University Bochum, Bad Oeynhausen, Germany
| | - Luiz Belardinelli
- Department of Biology, Cardiovascular, Therapeutic Area, Gilead Sciences, Foster, City, CA, USA
| | - Norbert Frey
- Department of Internal Medicine III: Cardiology and Angiology, University of Kiel, Germany
| | - Lars S Maier
- Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany
| | - Gerd Hasenfuss
- Department of Cardiology and Pneumology, Georg August University Göttingen, Göttingen, Germany; DZHK (German Centre for Cardiovascular Research), partner site Göttingen, Germany
| | - Samuel Sossalla
- Department of Cardiology and Pneumology, Georg August University Göttingen, Göttingen, Germany; Department of Internal Medicine III: Cardiology and Angiology, University of Kiel, Germany; DZHK (German Centre for Cardiovascular Research), partner site Göttingen, Germany.
| |
Collapse
|
18
|
Aldasoro M, Guerra-Ojeda S, Aguirre-Rueda D, Mauricio MD, Vila JM, Marchio P, Iradi A, Aldasoro C, Jorda A, Obrador E, Valles SL. Effects of Ranolazine on Astrocytes and Neurons in Primary Culture. PLoS One 2016; 11:e0150619. [PMID: 26950436 PMCID: PMC4780741 DOI: 10.1371/journal.pone.0150619] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 02/17/2016] [Indexed: 12/15/2022] Open
Abstract
Ranolazine (Rn) is an antianginal agent used for the treatment of chronic angina pectoris when angina is not adequately controlled by other drugs. Rn also acts in the central nervous system and it has been proposed for the treatment of pain and epileptic disorders. Under the hypothesis that ranolazine could act as a neuroprotective drug, we studied its effects on astrocytes and neurons in primary culture. We incubated rat astrocytes and neurons in primary cultures for 24 hours with Rn (10-7, 10-6 and 10-5 M). Cell viability and proliferation were measured using trypan blue exclusion assay, MTT conversion assay and LDH release assay. Apoptosis was determined by Caspase 3 activity assay. The effects of Rn on pro-inflammatory mediators IL-β and TNF-α was determined by ELISA technique, and protein expression levels of Smac/Diablo, PPAR-γ, Mn-SOD and Cu/Zn-SOD by western blot technique. In cultured astrocytes, Rn significantly increased cell viability and proliferation at any concentration tested, and decreased LDH leakage, Smac/Diablo expression and Caspase 3 activity indicating less cell death. Rn also increased anti-inflammatory PPAR-γ protein expression and reduced pro-inflammatory proteins IL-1 β and TNFα levels. Furthermore, antioxidant proteins Cu/Zn-SOD and Mn-SOD significantly increased after Rn addition in cultured astrocytes. Conversely, Rn did not exert any effect on cultured neurons. In conclusion, Rn could act as a neuroprotective drug in the central nervous system by promoting astrocyte viability, preventing necrosis and apoptosis, inhibiting inflammatory phenomena and inducing anti-inflammatory and antioxidant agents.
Collapse
Affiliation(s)
- Martin Aldasoro
- Department of Physiology, School of Medicine, University of Valencia, Spain
| | - Sol Guerra-Ojeda
- Department of Physiology, School of Medicine, University of Valencia, Spain
| | | | | | - Jose Mª Vila
- Department of Physiology, School of Medicine, University of Valencia, Spain
| | - Patricia Marchio
- Department of Physiology, School of Medicine, University of Valencia, Spain
| | - Antonio Iradi
- Department of Physiology, School of Medicine, University of Valencia, Spain
| | - Constanza Aldasoro
- Department of Physiology, School of Medicine, University of Valencia, Spain
| | - Adrian Jorda
- Department of Physiology, School of Medicine, University of Valencia, Spain
| | - Elena Obrador
- Department of Physiology, School of Medicine, University of Valencia, Spain
| | - Soraya L. Valles
- Department of Physiology, School of Medicine, University of Valencia, Spain
- * E-mail:
| |
Collapse
|
19
|
Latrunculin B modulates electrophysiological characteristics and arrhythmogenesis in pulmonary vein cardiomyocytes. Clin Sci (Lond) 2016; 130:721-32. [PMID: 26839418 DOI: 10.1042/cs20150593] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 02/02/2016] [Indexed: 01/28/2023]
Abstract
AF (atrial fibrillation) is the most common sustained arrhythmia, and the PVs (pulmonary veins) play a critical role in triggering AF. Stretch causes structural remodelling, including cytoskeleton rearrangement, which may play a role in the genesis of AF. Lat-B (latrunculin B), an inhibitor of actin polymerization, is involved in Ca(2+) regulation. However, it is unclear whether Lat-B directly modulates the electrophysiological characteristics and Ca(2+) homoeostasis of the PVs. Conventional microelectrodes, whole-cell patch-clamp, and the fluo-3 fluorimetric ratio technique were used to record ionic currents and intracellular Ca(2+) within isolated rabbit PV preparations, or within isolated single PV cardiomyocytes, before and after administration of Lat-B (100 nM). Langendorff-perfused rabbit hearts were exposed to acute and continuous atrial stretch, and we studied PV electrical activity. Lat-B (100 nM) decreased the spontaneous electrical activity by 16±4% in PV preparations. Lat-B (100 nM) decreased the late Na(+) current, L-type Ca(2+) current, Na(+)/Ca(2+) exchanger current, and stretch-activated BKCa current, but did not affect the Na(+) current in PV cardiomyocytes. Lat-B reduced the transient outward K(+) current and ultra-rapid delayed rectifier K(+) current, but increased the delayed rectifier K(+) current in isolated PV cardiomyocytes. In addition, Lat-B (100 nM) decreased intracellular Ca(2+) transient and sarcoplasmic reticulum Ca(2+) content in PV cardiomyocytes. Moreover, Lat-B attenuated stretch-induced increased spontaneous electrical activity and trigger activity. The effects of Lat-B on the PV spontaneous electrical activity were attenuated in the presence of Y-27632 [10 μM, a ROCK (Rho-associated kinase) inhibitor] and cytochalasin D (10 μM, an actin polymerization inhibitor). In conclusion, Lat-B regulates PV electrophysiological characteristics and attenuates stretch-induced arrhythmogenesis.
Collapse
|
20
|
Mesubi OO, Anderson ME. Atrial remodelling in atrial fibrillation: CaMKII as a nodal proarrhythmic signal. Cardiovasc Res 2016; 109:542-57. [PMID: 26762270 DOI: 10.1093/cvr/cvw002] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 01/05/2016] [Indexed: 01/10/2023] Open
Abstract
CaMKII is a serine-threonine protein kinase that is abundant in myocardium. Emergent evidence suggests that CaMKII may play an important role in promoting atrial fibrillation (AF) by targeting a diverse array of proteins involved in membrane excitability, cell survival, calcium homeostasis, matrix remodelling, inflammation, and metabolism. Furthermore, CaMKII inhibition appears to protect against AF in animal models and correct proarrhythmic, defective intracellular Ca(2+) homeostasis in fibrillating human atrial cells. This review considers current concepts and evidence from animal and human studies on the role of CaMKII in AF.
Collapse
Affiliation(s)
- Olurotimi O Mesubi
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA Department of Medicine, The Johns Hopkins University School of Medicine, 1830 E. Monument Street, Suite 9026, Baltimore, MD 21287, USA
| | - Mark E Anderson
- Division of Cardiology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA Department of Medicine, The Johns Hopkins University School of Medicine, 1830 E. Monument Street, Suite 9026, Baltimore, MD 21287, USA Department of Physiology and the Program in Cellular and Molecular Medicine, The Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
21
|
Reiffel JA, Camm AJ, Belardinelli L, Zeng D, Karwatowska-Prokopczuk E, Olmsted A, Zareba W, Rosero S, Kowey P. The HARMONY Trial: Combined Ranolazine and Dronedarone in the Management of Paroxysmal Atrial Fibrillation: Mechanistic and Therapeutic Synergism. Circ Arrhythm Electrophysiol 2015; 8:1048-56. [PMID: 26226999 DOI: 10.1161/circep.115.002856] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 07/09/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND Atrial fibrillation (AF) requires arrhythmogenic changes in atrial ion channels/receptors and usually altered atrial structure. AF is commonly treated with antiarrhythmic drugs; the most effective block many ion channels/receptors. Modest efficacy, intolerance, and safety concerns limit current antiarrhythmic drugs. We hypothesized that combining agents with multiple anti-AF mechanisms at reduced individual drug doses might produce synergistic efficacy plus better tolerance/safety. METHODS AND RESULTS HARMONY tested midrange ranolazine (750 mg BID) combined with 2 reduced dronedarone doses (150 mg BID and 225 mg BID; chosen to reduce dronedarone's negative inotropic effect-see text below) over 12 weeks in 134 patients with paroxysmal AF and implanted pacemakers where AF burden (AFB) could be continuously assessed. Patients were randomized double-blind to placebo, ranolazine alone (750 mg BID), dronedarone alone (225 mg BID), or one of the combinations. Neither placebo nor either drugs alone significantly reduced AFB. Conversely, ranolazine 750 mg BID/dronedarone 225 mg BID reduced AFB by 59% versus placebo (P=0.008), whereas ranolazine 750 mg BID/dronedarone 150 mg BID reduced AFB by 43% (P=0.072). Both combinations were well tolerated. CONCLUSIONS HARMONY showed synergistic AFB reduction by moderate dose ranolazine plus reduced dose dronedarone, with good tolerance/safety, in the population enrolled. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov; Unique identifier: NCT01522651.
Collapse
Affiliation(s)
- James A Reiffel
- From the Division of Cardiology, Department of Medicine, Columbia University, New York, NY (J.A.R.); Department of Cardiovascular Sciences, St Georges University of London, London, United Kingdom (A.J.C.); Cardiovascular Clinical Research, Gilead Sciences, Inc, Foster City, CA (L.B., D.Z., E.K.-P., A.O.); Division of Cardiology, Department of Medicine, University of Rochester Medical Center, NY (W.Z., S.R.); Lankenau Institute for Medical Research, Wynnewood, PA (P.K.); and Division of Cardiovascular Diseases, Department of Medicine, Thomas Jefferson University, Philadelphia, PA (P.K.).
| | - A John Camm
- From the Division of Cardiology, Department of Medicine, Columbia University, New York, NY (J.A.R.); Department of Cardiovascular Sciences, St Georges University of London, London, United Kingdom (A.J.C.); Cardiovascular Clinical Research, Gilead Sciences, Inc, Foster City, CA (L.B., D.Z., E.K.-P., A.O.); Division of Cardiology, Department of Medicine, University of Rochester Medical Center, NY (W.Z., S.R.); Lankenau Institute for Medical Research, Wynnewood, PA (P.K.); and Division of Cardiovascular Diseases, Department of Medicine, Thomas Jefferson University, Philadelphia, PA (P.K.)
| | - Luiz Belardinelli
- From the Division of Cardiology, Department of Medicine, Columbia University, New York, NY (J.A.R.); Department of Cardiovascular Sciences, St Georges University of London, London, United Kingdom (A.J.C.); Cardiovascular Clinical Research, Gilead Sciences, Inc, Foster City, CA (L.B., D.Z., E.K.-P., A.O.); Division of Cardiology, Department of Medicine, University of Rochester Medical Center, NY (W.Z., S.R.); Lankenau Institute for Medical Research, Wynnewood, PA (P.K.); and Division of Cardiovascular Diseases, Department of Medicine, Thomas Jefferson University, Philadelphia, PA (P.K.)
| | - Dewan Zeng
- From the Division of Cardiology, Department of Medicine, Columbia University, New York, NY (J.A.R.); Department of Cardiovascular Sciences, St Georges University of London, London, United Kingdom (A.J.C.); Cardiovascular Clinical Research, Gilead Sciences, Inc, Foster City, CA (L.B., D.Z., E.K.-P., A.O.); Division of Cardiology, Department of Medicine, University of Rochester Medical Center, NY (W.Z., S.R.); Lankenau Institute for Medical Research, Wynnewood, PA (P.K.); and Division of Cardiovascular Diseases, Department of Medicine, Thomas Jefferson University, Philadelphia, PA (P.K.)
| | - Ewa Karwatowska-Prokopczuk
- From the Division of Cardiology, Department of Medicine, Columbia University, New York, NY (J.A.R.); Department of Cardiovascular Sciences, St Georges University of London, London, United Kingdom (A.J.C.); Cardiovascular Clinical Research, Gilead Sciences, Inc, Foster City, CA (L.B., D.Z., E.K.-P., A.O.); Division of Cardiology, Department of Medicine, University of Rochester Medical Center, NY (W.Z., S.R.); Lankenau Institute for Medical Research, Wynnewood, PA (P.K.); and Division of Cardiovascular Diseases, Department of Medicine, Thomas Jefferson University, Philadelphia, PA (P.K.)
| | - Ann Olmsted
- From the Division of Cardiology, Department of Medicine, Columbia University, New York, NY (J.A.R.); Department of Cardiovascular Sciences, St Georges University of London, London, United Kingdom (A.J.C.); Cardiovascular Clinical Research, Gilead Sciences, Inc, Foster City, CA (L.B., D.Z., E.K.-P., A.O.); Division of Cardiology, Department of Medicine, University of Rochester Medical Center, NY (W.Z., S.R.); Lankenau Institute for Medical Research, Wynnewood, PA (P.K.); and Division of Cardiovascular Diseases, Department of Medicine, Thomas Jefferson University, Philadelphia, PA (P.K.)
| | - Wojciech Zareba
- From the Division of Cardiology, Department of Medicine, Columbia University, New York, NY (J.A.R.); Department of Cardiovascular Sciences, St Georges University of London, London, United Kingdom (A.J.C.); Cardiovascular Clinical Research, Gilead Sciences, Inc, Foster City, CA (L.B., D.Z., E.K.-P., A.O.); Division of Cardiology, Department of Medicine, University of Rochester Medical Center, NY (W.Z., S.R.); Lankenau Institute for Medical Research, Wynnewood, PA (P.K.); and Division of Cardiovascular Diseases, Department of Medicine, Thomas Jefferson University, Philadelphia, PA (P.K.)
| | - Spencer Rosero
- From the Division of Cardiology, Department of Medicine, Columbia University, New York, NY (J.A.R.); Department of Cardiovascular Sciences, St Georges University of London, London, United Kingdom (A.J.C.); Cardiovascular Clinical Research, Gilead Sciences, Inc, Foster City, CA (L.B., D.Z., E.K.-P., A.O.); Division of Cardiology, Department of Medicine, University of Rochester Medical Center, NY (W.Z., S.R.); Lankenau Institute for Medical Research, Wynnewood, PA (P.K.); and Division of Cardiovascular Diseases, Department of Medicine, Thomas Jefferson University, Philadelphia, PA (P.K.)
| | - Peter Kowey
- From the Division of Cardiology, Department of Medicine, Columbia University, New York, NY (J.A.R.); Department of Cardiovascular Sciences, St Georges University of London, London, United Kingdom (A.J.C.); Cardiovascular Clinical Research, Gilead Sciences, Inc, Foster City, CA (L.B., D.Z., E.K.-P., A.O.); Division of Cardiology, Department of Medicine, University of Rochester Medical Center, NY (W.Z., S.R.); Lankenau Institute for Medical Research, Wynnewood, PA (P.K.); and Division of Cardiovascular Diseases, Department of Medicine, Thomas Jefferson University, Philadelphia, PA (P.K.)
| | | |
Collapse
|
22
|
Fischer TH, Herting J, Mason FE, Hartmann N, Watanabe S, Nikolaev VO, Sprenger JU, Fan P, Yao L, Popov AF, Danner BC, Schöndube F, Belardinelli L, Hasenfuss G, Maier LS, Sossalla S. Late INa increases diastolic SR-Ca2+-leak in atrial myocardium by activating PKA and CaMKII. Cardiovasc Res 2015; 107:184-96. [PMID: 25990311 PMCID: PMC4476413 DOI: 10.1093/cvr/cvv153] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 04/16/2015] [Indexed: 12/19/2022] Open
Abstract
Aims Enhanced cardiac late Na current (late INa) and increased sarcoplasmic reticulum (SR)-Ca2+-leak are both highly arrhythmogenic. This study seeks to identify signalling pathways interconnecting late INa and SR-Ca2+-leak in atrial cardiomyocytes (CMs). Methods and results In murine atrial CMs, SR-Ca2+-leak was increased by the late INa enhancer Anemonia sulcata toxin II (ATX-II). An inhibition of Ca2+/calmodulin-dependent protein kinase II (Autocamide-2-related inhibitory peptide), protein kinase A (H89), or late INa (Ranolazine or Tetrodotoxin) all prevented ATX-II-dependent SR-Ca2+-leak. The SR-Ca2+-leak induction by ATX-II was not detected when either the Na+/Ca2+ exchanger was inhibited (KBR) or in CaMKIIδc-knockout mice. FRET measurements revealed increased cAMP levels upon ATX-II stimulation, which could be prevented by inhibition of adenylyl cyclases (ACs) 5 and 6 (NKY 80) but not by inhibition of phosphodiesterases (IBMX), suggesting PKA activation via an AC-dependent increase of cAMP levels. Western blots showed late INa-dependent hyperphosphorylation of CaMKII as well as PKA target sites at ryanodine receptor type-2 (-S2814 and -S2808) and phospholamban (-Thr17, -S16). Enhancement of late INa did not alter Ca2+-transient amplitude or SR-Ca2+-load. However, upon late INa activation and simultaneous CaMKII inhibition, Ca2+-transient amplitude and SR-Ca2+-load were increased, whereas PKA inhibition reduced Ca2+-transient amplitude and load and additionally slowed Ca2+ elimination. In atrial CMs from patients with atrial fibrillation, inhibition of late INa, CaMKII, or PKA reduced the SR-Ca2+-leak. Conclusion Late INa exerts distinct effects on Ca2+ homeostasis in atrial myocardium through activation of CaMKII and PKA. Inhibition of late INa represents a potential approach to attenuate CaMKII activation and decreases SR-Ca2+-leak in atrial rhythm disorders. The interconnection with the cAMP/PKA system further increases the antiarrhythmic potential of late INa inhibition.
Collapse
Affiliation(s)
- Thomas H Fischer
- Klinik für Kardiologie und Pneumologie/Herzzentrum, Georg-August-Universität Göttingen, Robert-Koch-Str. 40, 37075 Göttingen, Germany
| | - Jonas Herting
- Klinik für Kardiologie und Pneumologie/Herzzentrum, Georg-August-Universität Göttingen, Robert-Koch-Str. 40, 37075 Göttingen, Germany
| | - Fleur E Mason
- Klinik für Kardiologie und Pneumologie/Herzzentrum, Georg-August-Universität Göttingen, Robert-Koch-Str. 40, 37075 Göttingen, Germany
| | - Nico Hartmann
- Klinik für Kardiologie und Pneumologie/Herzzentrum, Georg-August-Universität Göttingen, Robert-Koch-Str. 40, 37075 Göttingen, Germany
| | - Saera Watanabe
- Klinik für Kardiologie und Pneumologie/Herzzentrum, Georg-August-Universität Göttingen, Robert-Koch-Str. 40, 37075 Göttingen, Germany
| | - Viacheslav O Nikolaev
- Klinik für Kardiologie und Pneumologie/Herzzentrum, Georg-August-Universität Göttingen, Robert-Koch-Str. 40, 37075 Göttingen, Germany DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany
| | - Julia U Sprenger
- Klinik für Kardiologie und Pneumologie/Herzzentrum, Georg-August-Universität Göttingen, Robert-Koch-Str. 40, 37075 Göttingen, Germany
| | - Peidong Fan
- Department of Biology, Cardiovascular, Therapeutic Area, Gilead Sciences, Foster City, CA, USA
| | - Lina Yao
- Department of Biology, Cardiovascular, Therapeutic Area, Gilead Sciences, Foster City, CA, USA
| | - Aron-Frederik Popov
- Klinik für Thorax-, Herz-, Gefäßchirurgie/Herzzentrum, Georg-August-Universität Göttingen, Göttingen, Germany
| | - Bernhard C Danner
- Klinik für Thorax-, Herz-, Gefäßchirurgie/Herzzentrum, Georg-August-Universität Göttingen, Göttingen, Germany
| | - Friedrich Schöndube
- Klinik für Thorax-, Herz-, Gefäßchirurgie/Herzzentrum, Georg-August-Universität Göttingen, Göttingen, Germany
| | - Luiz Belardinelli
- Department of Biology, Cardiovascular, Therapeutic Area, Gilead Sciences, Foster City, CA, USA
| | - Gerd Hasenfuss
- Klinik für Kardiologie und Pneumologie/Herzzentrum, Georg-August-Universität Göttingen, Robert-Koch-Str. 40, 37075 Göttingen, Germany DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany
| | - Lars S Maier
- Innere Medizin II - Kardiologie, Universitätsklinikum Regensburg, Regensburg, Germany
| | - Samuel Sossalla
- Klinik für Kardiologie und Pneumologie/Herzzentrum, Georg-August-Universität Göttingen, Robert-Koch-Str. 40, 37075 Göttingen, Germany DZHK (German Center for Cardiovascular Research), partner site Göttingen, Göttingen, Germany
| |
Collapse
|
23
|
Cardiac late Na+ current: Proarrhythmic effects, roles in long QT syndromes, and pathological relationship to CaMKII and oxidative stress. Heart Rhythm 2015; 12:440-8. [DOI: 10.1016/j.hrthm.2014.11.009] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Indexed: 12/16/2022]
|
24
|
Abstract
Late I Na is an integral part of the sodium current, which persists long after the fast-inactivating component. The magnitude of the late I Na is relatively small in all species and in all types of cardiomyocytes as compared with the amplitude of the fast sodium current, but it contributes significantly to the shape and duration of the action potential. This late component had been shown to increase in several acquired or congenital conditions, including hypoxia, oxidative stress, and heart failure, or due to mutations in SCN5A, which encodes the α-subunit of the sodium channel, as well as in channel-interacting proteins, including multiple β subunits and anchoring proteins. Patients with enhanced late I Na exhibit the type-3 long QT syndrome (LQT3) characterized by high propensity for the life-threatening ventricular arrhythmias, such as Torsade de Pointes (TdP), as well as for atrial fibrillation. There are several distinct mechanisms of arrhythmogenesis due to abnormal late I Na, including abnormal automaticity, early and delayed after depolarization-induced triggered activity, and dramatic increase of ventricular dispersion of repolarization. Many local anesthetic and antiarrhythmic agents have a higher potency to block late I Na as compared with fast I Na. Several novel compounds, including ranolazine, GS-458967, and F15845, appear to be the most selective inhibitors of cardiac late I Na reported to date. Selective inhibition of late I Na is expected to be an effective strategy for correcting these acquired and congenital channelopathies.
Collapse
|
25
|
Luo A, Ma J, Song Y, Qian C, Wu Y, Zhang P, Wang L, Fu C, Cao Z, Shryock JC. Larger late sodium current density as well as greater sensitivities to ATX II and ranolazine in rabbit left atrial than left ventricular myocytes. Am J Physiol Heart Circ Physiol 2013; 306:H455-61. [PMID: 24322614 DOI: 10.1152/ajpheart.00727.2013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
An increase of cardiac late sodium current (INa.L) is arrhythmogenic in atrial and ventricular tissues, but the densities of INa.L and thus the potential relative contributions of this current to sodium ion (Na(+)) influx and arrhythmogenesis in atria and ventricles are unclear. In this study, whole-cell and cell-attached patch-clamp techniques were used to measure INa.L in rabbit left atrial and ventricular myocytes under identical conditions. The density of INa.L was 67% greater in left atrial (0.50 ± 0.09 pA/pF, n = 20) than in left ventricular cells (0.30 ± 0.07 pA/pF, n = 27, P < 0.01) when elicited by step pulses from -120 to -20 mV at a rate of 0.2 Hz. Similar results were obtained using step pulses from -90 to -20 mV. Anemone toxin II (ATX II) increased INa.L with an EC50 value of 14 ± 2 nM and a Hill slope of 1.4 ± 0.1 (n = 9) in atrial myocytes and with an EC50 of 21 ± 5 nM and a Hill slope of 1.2 ± 0.1 (n = 12) in ventricular myocytes. Na(+) channel open probability (but not mean open time) was greater in atrial than in ventricular cells in the absence and presence of ATX II. The INa.L inhibitor ranolazine (3, 6, and 9 μM) reduced INa.L more in atrial than ventricular myocytes in the presence of 40 nM ATX II. In summary, rabbit left atrial myocytes have a greater density of INa.L and higher sensitivities to ATX II and ranolazine than rabbit left ventricular myocytes.
Collapse
Affiliation(s)
- Antao Luo
- Cardio-Electrophysiological Research Laboratory, Medical College of Wuhan University of Science and Technology, Wuhan, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Shryock JC, Song Y, Rajamani S, Antzelevitch C, Belardinelli L. The arrhythmogenic consequences of increasing late INa in the cardiomyocyte. Cardiovasc Res 2013; 99:600-11. [PMID: 23752976 DOI: 10.1093/cvr/cvt145] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
This review presents the roles of cardiac sodium channel NaV1.5 late current (late INa) in generation of arrhythmic activity. The assumption of the authors is that proper Na(+) channel function is necessary to the maintenance of the transmembrane electrochemical gradient of Na(+) and regulation of cardiac electrical activity. Myocyte Na(+) channels' openings during the brief action potential upstroke contribute to peak INa and initiate excitation-contraction coupling. Openings of Na(+) channels outside the upstroke contribute to late INa, a depolarizing current that persists throughout the action potential plateau. The small, physiological late INa does not appear to be critical for normal electrical or contractile function in the heart. Late INa does, however, reduce the net repolarizing current, prolongs action potential duration, and increases cellular Na(+) loading. An increase of late INa, due to acquired conditions (e.g. heart failure) or inherited Na(+) channelopathies, facilitates the formation of early and delayed afterpolarizations and triggered arrhythmias, spontaneous diastolic depolarization, and cellular Ca(2+) loading. These in turn increase the spatial and temporal dispersion of repolarization time and may lead to reentrant arrhythmias.
Collapse
Affiliation(s)
- John C Shryock
- Department of Biology, Cardiovascular Therapeutic Area, Gilead Sciences, Foster City, CA, USA
| | | | | | | | | |
Collapse
|
27
|
Lu YY, Chen YC, Kao YH, Chen SA, Chen YJ. Extracellular matrix of collagen modulates arrhythmogenic activity of pulmonary veins through p38 MAPK activation. J Mol Cell Cardiol 2013; 59:159-66. [PMID: 23524328 DOI: 10.1016/j.yjmcc.2013.03.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Revised: 02/05/2013] [Accepted: 03/13/2013] [Indexed: 11/28/2022]
Abstract
Atrial fibrillation (AF) is the most common sustained arrhythmia. Cardiac fibrosis with enhanced extracellular collagen plays a critical role in the pathophysiology of AF through structural and electrical remodeling. Pulmonary veins (PVs) are important foci for AF genesis. The purpose of this study was to evaluate whether collagen can directly modulate PV arrhythmogenesis. Action potentials and ionic currents were investigated in isolated male New Zealand rabbit PV cardiomyocytes with and without collagen incubation (10μg/ml, 5-7h) using the whole-cell patch-clamp technique. Compared to control PV cardiomyocytes (n=25), collagen-treated PV cardiomyocytes (n=22) had a faster beating rate (3.2±04 vs. 1.9±0.2Hz, p<0.005) and a larger amplitude of delayed afterdepolarization (16±2 vs. 10±1mV, p<0.01). Moreover, collagen-treated PV cardiomyocytes showed a larger transient outward potassium current, small-conductance Ca(2+)-activated K(+) current, inward rectifier potassium current, pacemaker current, and late sodium current than control PV cardiomyocytes, but amplitudes of the sodium current, sustained outward potassium current, and L-type calcium current were similar. Collagen increased the p38 MAPK phosphorylation in PV cardiomyocytes as compared to control. The change of the spontaneous activity and action potential morphology were ameliorated by SB203580 (the p38 MAPK catalytic activity inhibitor), indicating that collagen can directly increase PV cardiomyocyte arrhythmogenesis through p38 MAPK activation, which may contribute to the pathogenesis of AF.
Collapse
Affiliation(s)
- Yen-Yu Lu
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | | | | | | | | |
Collapse
|
28
|
Sicouri S, Belardinelli L, Antzelevitch C. Antiarrhythmic effects of the highly selective late sodium channel current blocker GS-458967. Heart Rhythm 2013; 10:1036-43. [PMID: 23524321 DOI: 10.1016/j.hrthm.2013.03.023] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2013] [Indexed: 12/14/2022]
Abstract
BACKGROUND Previous studies have shown that late sodium channel current (INa) blockers such as ranolazine can exert antiarrhythmic effects by suppressing early and delayed afterdepolarization (EAD and DAD)-induced triggered activity. OBJECTIVE To evaluate the electrophysiological properties of GS-458967 (GS967), a potent and highly selective late INa blocker, in canine Purkinje fibers (PFs) and pulmonary vein (PV) and superior vena cava (SVC) sleeve preparations. METHODS Transmembrane action potentials were recorded from canine PFs and PV and SVC sleeve preparations by using standard microelectrode techniques. The rapidly activating delayed rectifier potassium channel current blocker E-4031 (2.5-5 µM) and the late INa agonist ATX-II (10 nM) were used to induce EADs in PFs. Isoproterenol (1 µM), high calcium ([Ca(2+)]o = 5.4 mM), or their combination was used to induce DADs and triggered activity. RESULTS In PFs, GS967 (10-300 nM) caused a significant concentration-dependent reduction in action potential duration without altering the maximum rate of rise of the action potential upstroke, action potential amplitude, or resting membrane potential at any rate studied (basic cycle lengths of 1000, 500, and 300 ms) or concentration evaluated (n = 5; P < .05). GS967 (30-100 nM) abolished EADs and EAD-induced triggered activity elicited in PFs by exposure to E-4031 (n = 4) or ATX-II (n = 4). In addition, GS967 reduced or abolished DADs and suppressed DAD-induced triggered activity elicited in PFs (n = 4) and PV (n = 4) and SVC (n = 3) sleeve preparations by exposure to isoproterenol, high calcium, or their combination. CONCLUSIONS Our data suggest that the selective inhibition of late INa with GS967 can exert antiarrhythmic effects by suppressing EAD- and DAD-mediated extrasystolic activity in PFs and PV and SVC sleeve preparations.
Collapse
Affiliation(s)
- Serge Sicouri
- Masonic Medical Research Laboratory, Utica, New York 13501-1787, USA.
| | | | | |
Collapse
|
29
|
Abstract
Reactive oxygen species (ROS) have been associated with various human diseases, and considerable attention has been paid to investigate their physiological effects. Various ROS are synthesized in the mitochondria and accumulate in the cytoplasm if the cellular antioxidant defense mechanism fails. The critical balance of this ROS synthesis and antioxidant defense systems is termed the redox system of the cell. Various cardiovascular diseases have also been affected by redox to different degrees. ROS have been indicated as both detrimental and protective, via different cellular pathways, for cardiac myocyte functions, electrophysiology, and pharmacology. Mostly, the ROS functions depend on the type and amount of ROS synthesized. While the literature clearly indicates ROS effects on cardiac contractility, their effects on cardiac excitability are relatively under appreciated. Cardiac excitability depends on the functions of various cardiac sarcolemal or mitochondrial ion channels carrying various depolarizing or repolarizing currents that also maintain cellular ionic homeostasis. ROS alter the functions of these ion channels to various degrees to determine excitability by affecting the cellular resting potential and the morphology of the cardiac action potential. Thus, redox balance regulates cardiac excitability, and under pathological regulation, may alter action potential propagation to cause arrhythmia. Understanding how redox affects cellular excitability may lead to potential prophylaxis or treatment for various arrhythmias. This review will focus on the studies of redox and cardiac excitation.
Collapse
Affiliation(s)
- Nitin T Aggarwal
- Division of Cardiovascular Medicine, Department of Medicine, University of Wisconsin, Madison, WI 53792, USA
| | | |
Collapse
|
30
|
Aidonidis I, Doulas K, Hatziefthimiou A, Tagarakis G, Simopoulos V, Rizos I, Tsilimingas N, Molyvdas PA. Ranolazine-Induced Postrepolarization Refractoriness Suppresses Induction of Atrial Flutter and Fibrillation in Anesthetized Rabbits. J Cardiovasc Pharmacol Ther 2012; 18:94-101. [DOI: 10.1177/1074248412453874] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Ranolazine (Ran) is a novel anti-ischemic agent with electrophysiologic properties mainly attributed to the inhibition of late Na+ current and atrial-selective early Na+ current. However, there are only limited data regarding its efficacy and mechanism of action against atrial flutter (Afl) and atrial fibrillation (AF) in intact animals. Therefore, we aimed to investigate the electrophysiologic mechanism of Ran in a rabbit model of inducible atrial tachyarrhythmias elicited by acetylcholine (ACh). Arrhythmias were produced in 19 rabbits by rapid atrial burst pacing during control, after intravenous ACh and after Ran + ACh administration. Recording of right atrial monophasic action potentials (MAPs) and programmed stimulation were utilized to determine the duration of atrial repolarization at various cycle lengths and voltage levels of action potential, including 75% of total MAP duration (MAPD75), effective refractory period (ERP), and postrepolarization refractoriness (PRR = ERP − MAPD75) prior to and after Ran. Control stimulation yielded no arrhythmias or maximal nonsustained runs of Afl/AF. Upon ACh, 17 of 19 rabbits exhibited sustained Afl and AF as well as mixed forms of Afl/AF, while 2 animals revealed none or short runs of nonsustained arrhythmias and were excluded from the study. High-frequency burst pacing during the first 30 minutes after Ran + ACh failed to induce any arrhythmia in 13 of 17 rabbits (76%), while 2 animals displayed sustained Afl/AF and 2 other animals nonsustained Afl/AF. At basic stimulation cycle length of 250 milliseconds, Ran prolonged baseline atrial ERP (80 ± 8 vs 120 ± 9 milliseconds, P < .001) much more than MAPD75 (65 ± 7 vs 85 ± 7 milliseconds, P < .001), leading to atrial PRR which was more pronounced after Ran compared with control measurements (35 ± 11 vs 15 ± 10 milliseconds, P < .001). This in vivo study demonstrates that Ran exerts antiarrhythmic activity by suppressing inducibility of ACh-mediated Afl/AF in intact rabbits. Its action may predominantly be related to a significant increase in atrial PRR, resulting in depressed electrical excitability and impediment of arrhythmia initiation.
Collapse
Affiliation(s)
- Isaac Aidonidis
- Department of Physiology, Medical School of Larissa & Thoracic and Cardiovascular Surgery of the University Hospital of Larissa, University of Thessaly, Larissa, Greece
| | - Konstantinos Doulas
- Department of Physiology, Medical School of Larissa & Thoracic and Cardiovascular Surgery of the University Hospital of Larissa, University of Thessaly, Larissa, Greece
| | - Apostolia Hatziefthimiou
- Department of Physiology, Medical School of Larissa & Thoracic and Cardiovascular Surgery of the University Hospital of Larissa, University of Thessaly, Larissa, Greece
| | - Georgios Tagarakis
- Department of Physiology, Medical School of Larissa & Thoracic and Cardiovascular Surgery of the University Hospital of Larissa, University of Thessaly, Larissa, Greece
| | - Vassilios Simopoulos
- Department of Physiology, Medical School of Larissa & Thoracic and Cardiovascular Surgery of the University Hospital of Larissa, University of Thessaly, Larissa, Greece
| | - Ioannis Rizos
- Department of Cardiology, Attikon University Hospital of Athens, Greece
| | - Nikolaos Tsilimingas
- Department of Physiology, Medical School of Larissa & Thoracic and Cardiovascular Surgery of the University Hospital of Larissa, University of Thessaly, Larissa, Greece
| | - Paschalis-Adam Molyvdas
- Department of Physiology, Medical School of Larissa & Thoracic and Cardiovascular Surgery of the University Hospital of Larissa, University of Thessaly, Larissa, Greece
| |
Collapse
|
31
|
Lu YY, Cheng CC, Chen YC, Chen SA, Chen YJ. ATX-II-induced pulmonary vein arrhythmogenesis related to atrial fibrillation and long QT syndrome. Eur J Clin Invest 2012; 42:823-31. [PMID: 22339387 DOI: 10.1111/j.1365-2362.2012.02655.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND Long QT syndrome (LQTS) is associated with a high incidence of atrial fibrillation (AF), but the underlying mechanisms are unclear. Pulmonary veins (PVs) play a critical role in AF genesis. Type 3 LQTS increases late sodium current (I(Na,L) ), which may increase PV arrhythmogenesis and AF. Therefore, this study examines PV arrhythmogenesis in anemonia sulcata toxin II (ATX-II)-induced type 3 LQTS and evaluates whether the I(Na,L) inhibitor ranolazine can suppress PV arrhythmogenesis. MATERIALS AND METHODS Conventional microelectrodes were used to record the action potentials (AP) and contractility in isolated rabbit PV specimens before and after ATX-II administration with or without ranolazine. RESULTS Anemonia sulcata toxin II (100 nM) increased the PV spontaneous rates from 2·0 ± 0·1 to 2·9 ± 0·2 Hz (n = 7), induced PV burst firing (100%) with the genesis of early afterdepolarization (EAD) (86%) and prolonged the AP duration. Ranolazine (0·1, 1 and 10 μM) dose dependently reduced the PV spontaneous rates from 2·5 ± 0·2 to 2·3 ± 0·2 Hz, 1·9 ± 0·2 and 1·5 ± 0·3 Hz (P < 0·05) and decreased the diastolic tension by 40 ± 19%, 87 ± 26% and 113 ± 28%. In the presence of ranolazine (10 μM), ATX-II (100 nM) further increased the AP duration. However, ATX-II neither increased the PV spontaneous rates (1·6 ± 0·1 vs. 1·7 ± 0·2 Hz, n = 7) nor induced PV burst firing or EAD. Moreover, ranolazine (10 μM) reduced ATX-II-induced PV acceleration and EAD. CONCLUSIONS The I(Na,L) enhancer ATX-II can increase PV arrhythmogenesis, which can be attenuated or blocked by ranolazine. This suggests that AF may be related to type 3 LQTS through increased I(Na,L) .
Collapse
Affiliation(s)
- Yen-Yu Lu
- Division of Cardiology, Sijhih Cathay General Hospital, Sijhih, Taiwan
| | | | | | | | | |
Collapse
|
32
|
Lowe JS, Stroud DM, Yang T, Hall L, Atack TC, Roden DM. Increased late sodium current contributes to long QT-related arrhythmia susceptibility in female mice. Cardiovasc Res 2012; 95:300-7. [PMID: 22562703 DOI: 10.1093/cvr/cvs160] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
AIMS Female gender is a risk factor for long QT-related arrhythmias, but the underlying mechanisms remain uncertain. Here, we tested the hypothesis that gender-dependent function of the post-depolarization 'late' sodium current (I(Na-L)) contributes. METHODS AND RESULTS Studies were conducted in mice in which the canonical cardiac sodium channel Scn5a locus was disrupted, and expression of human wild-type SCN5A cDNA substituted. Baseline QT intervals were similar in male and female mice, but exposure to the sodium channel opener anemone toxin ATX-II elicited polymorphic ventricular tachycardia in 0/9 males vs. 6/9 females. Ventricular I(Na-L) and action potential durations were increased in myocytes isolated from female mice compared with those from males before and especially after treatment with ATX-II. Further, ATX-II elicited potentially arrhythmogenic early afterdepolarizations in myocytes from 0/5 male mice and 3/5 female mice. CONCLUSION These data identify variable late I(Na) as a modulator of gender-dependent arrhythmia susceptibility.
Collapse
Affiliation(s)
- John S Lowe
- Department of Medicine, Vanderbilt University School of Medicine, 2215B Garland Avenue, Nashville, TN 37232-0575, USA
| | | | | | | | | | | |
Collapse
|
33
|
Sicouri S, Blazek J, Belardinelli L, Antzelevitch C. Electrophysiological characteristics of canine superior vena cava sleeve preparations: effect of ranolazine. Circ Arrhythm Electrophysiol 2012; 5:371-9. [PMID: 22407414 DOI: 10.1161/circep.111.969493] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND In addition to extrasystoles of pulmonary vein (PV) origin, those arising from the superior vena cava (SVC) can precipitate atrial fibrillation (AF). The present study evaluates the electrophysiological properties of canine SVC sleeve preparations and the effect of ranolazine on late phase 3 early and delayed afterdepolarization (EAD and DAD)-induced triggered activity in SVC sleeves and compares SVC and PV sleeve electrophysiological properties. METHODS AND RESULTS Action potentials (APs) were recorded from superfused SVC and PV sleeves using microelectrode techniques. Acetylcholine (1 μmol/L), isoproterenol (1 μmol/L), high calcium ([Ca(2+)](o)=5.4 mmol/L), or a combination were used to induce EADs, DADs, and triggered activity. A marked diversity of action potential characteristics was observed in the SVC sleeve, including action potentials with short and long APs, with and without phase 4 depolarization. Rapid pacing induced hyperpolarization, accentuating the slope of phase 4 depolarization. Phase 4 depolarization and rapid pacing-induced hyperpolarization were reduced or eliminated after atropine (10 μmol/L) or ranolazine (10 μmol/L). APs displaying phase 4 depolarization (n=19) had longer APDs, smaller amplitude and V(max), and a more positive take-off potential than APs lacking phase 4 depolarization (n=15). Ranolazine (5-10 μmol/L) eliminated late phase 3 EAD- and DAD-induced triggered activity as well as isoproterenol-induced automaticity elicited in SVC sleeves. Compared with PV, SVC sleeves display phase 4 depolarization, smaller V(max), and longer APs. CONCLUSIONS Autonomic influences promote spontaneous automaticity and triggered activity in SVC sleeves, thus generating extrasystolic activity capable of initiating atrial arrhythmias. Ranolazine can effectively suppress these triggers.
Collapse
Affiliation(s)
- Serge Sicouri
- Masonic Medical Research Laboratory, Utica, NY 13501-1787, USA.
| | | | | | | |
Collapse
|
34
|
Burashnikov A, Antzelevitch C. Ranolazine versus amiodarone for prevention of postoperative atrial fibrillation. Future Cardiol 2011; 7:733-7. [PMID: 22050058 PMCID: PMC4737707 DOI: 10.2217/fca.11.67] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Postoperative atrial fibrillation (AF) is a major complication of cardiothoracic surgery, leading to significant consequences, including a higher rate of stroke, longer hospital stays and increased costs. Amiodarone is among the most widely used agents for prevention of postoperative AF. Ranolazine, a US FDA-approved antianginal agent, has been shown to effectively, safely prevent and terminate nonpostoperative AF in both experimental and clinical studies. In a recent publication, Miles and colleagues directly compared the efficacy and safety of amiodarone and ranolazine for prevention of postoperative AF in 393 patients. The patients were pretreated with amiodarone and ranolaizne for >1 week and 1 day, respectively, and the treatment continued for 10-14 days after surgery. Following coronary artery bypass grafting (CABG), AF occurred in 26.5% of patients taking amiodarone and in 17.5% of patients taking ranolazine (34% reduction; p < 0.035). No differences in adverse events between the two groups of patients were recorded. The results of this retrospective nonrandomized single-center study indicate that ranolazine may be used to effectively and safely prevent postoperative AF. These results need to be confirmed in a larger randomized study. If confirmed, ranolazine may be a good choice for preventing AF in patients undergoing CABG.
Collapse
Affiliation(s)
- Alexander Burashnikov
- Gordon K Moe Scholar, Masonic Medical Research Laboratory, 2150 Bleecker St, Utica, NY 13501, USA
| | - Charles Antzelevitch
- Gordon K Moe Scholar, Masonic Medical Research Laboratory, 2150 Bleecker St, Utica, NY 13501, USA
| |
Collapse
|
35
|
Antzelevitch C, Burashnikov A, Sicouri S, Belardinelli L. Electrophysiologic basis for the antiarrhythmic actions of ranolazine. Heart Rhythm 2011; 8:1281-90. [PMID: 21421082 DOI: 10.1016/j.hrthm.2011.03.045] [Citation(s) in RCA: 183] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Accepted: 03/11/2011] [Indexed: 12/19/2022]
Abstract
Ranolazine is a Food and Drug Administration-approved antianginal agent. Experimental and clinical studies have shown that ranolazine has antiarrhythmic effects in both ventricles and atria. In the ventricles, ranolazine can suppress arrhythmias associated with acute coronary syndrome, long QT syndrome, heart failure, ischemia, and reperfusion. In atria, ranolazine effectively suppresses atrial tachyarrhythmias and atrial fibrillation (AF). Recent studies have shown that the drug may be effective and safe in suppressing AF when used as a pill-in-the pocket approach, even in patients with structurally compromised hearts, warranting further study. The principal mechanism underlying ranolazine's antiarrhythmic actions is thought to be primarily via inhibition of late I(Na) in the ventricles and via use-dependent inhibition of peak I(Na) and I(Kr) in the atria. Short- and long-term safety of ranolazine has been demonstrated in the clinic, even in patients with structural heart disease. This review summarizes the available data regarding the electrophysiologic actions and antiarrhythmic properties of ranolazine in preclinical and clinical studies.
Collapse
|
36
|
Sossalla S, Kallmeyer B, Wagner S, Mazur M, Maurer U, Toischer K, Schmitto JD, Seipelt R, Schöndube FA, Hasenfuss G, Belardinelli L, Maier LS. Altered Na+Currents in Atrial Fibrillation. J Am Coll Cardiol 2010; 55:2330-42. [DOI: 10.1016/j.jacc.2009.12.055] [Citation(s) in RCA: 188] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2009] [Revised: 11/16/2009] [Accepted: 12/07/2009] [Indexed: 12/19/2022]
|