1
|
Hartmann N, Knierim M, Maurer W, Dybkova N, Zeman F, Hasenfuß G, Sossalla S, Streckfuss-Bömeke K. Na V1.8 as Proarrhythmic Target in a Ventricular Cardiac Stem Cell Model. Int J Mol Sci 2024; 25:6144. [PMID: 38892333 PMCID: PMC11172914 DOI: 10.3390/ijms25116144] [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: 03/03/2024] [Revised: 05/25/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
The sodium channel NaV1.8, encoded by the SCN10A gene, has recently emerged as a potential regulator of cardiac electrophysiology. We have previously shown that NaV1.8 contributes to arrhythmogenesis by inducing a persistent Na+ current (late Na+ current, INaL) in human atrial and ventricular cardiomyocytes (CM). We now aim to further investigate the contribution of NaV1.8 to human ventricular arrhythmogenesis at the CM-specific level using pharmacological inhibition as well as a genetic knockout (KO) of SCN10A in induced pluripotent stem cell CM (iPSC-CM). In functional voltage-clamp experiments, we demonstrate that INaL was significantly reduced in ventricular SCN10A-KO iPSC-CM and in control CM after a specific pharmacological inhibition of NaV1.8. In contrast, we did not find any effects on ventricular APD90. The frequency of spontaneous sarcoplasmic reticulum Ca2+ sparks and waves were reduced in SCN10A-KO iPSC-CM and control cells following the pharmacological inhibition of NaV1.8. We further analyzed potential triggers of arrhythmias and found reduced delayed afterdepolarizations (DAD) in SCN10A-KO iPSC-CM and after the specific inhibition of NaV1.8 in control cells. In conclusion, we show that NaV1.8-induced INaL primarily impacts arrhythmogenesis at a subcellular level, with minimal effects on systolic cellular Ca2+ release. The inhibition or knockout of NaV1.8 diminishes proarrhythmic triggers in ventricular CM. In conjunction with our previously published results, this work confirms NaV1.8 as a proarrhythmic target that may be useful in an anti-arrhythmic therapeutic strategy.
Collapse
Affiliation(s)
- Nico Hartmann
- Clinic for Cardiology and Pneumology, University Medical Center, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen and Rhein Main, 61231 Bad Nauheim, Germany
| | - Maria Knierim
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen and Rhein Main, 61231 Bad Nauheim, Germany
- Clinic for Cardio-Thoracic and Vascular Surgery, University Medical Center, 37075 Göttingen, Germany
| | - Wiebke Maurer
- Clinic for Cardiology and Pneumology, University Medical Center, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen and Rhein Main, 61231 Bad Nauheim, Germany
| | - Nataliya Dybkova
- Clinic for Cardiology and Pneumology, University Medical Center, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen and Rhein Main, 61231 Bad Nauheim, Germany
| | - Florian Zeman
- Center for Clinicial Trials, University of Regensburg, 93042 Regensburg, Germany
| | - Gerd Hasenfuß
- Clinic for Cardiology and Pneumology, University Medical Center, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen and Rhein Main, 61231 Bad Nauheim, Germany
| | - Samuel Sossalla
- Clinic for Cardiology and Pneumology, University Medical Center, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen and Rhein Main, 61231 Bad Nauheim, Germany
- Medical Clinic I, Cardiology and Angiology, Giessen and Department of Cardiology at Kerckhoff Heart and Lung Center, Justus-Liebig-University, 61231 Bad Nauheim, Germany
| | - Katrin Streckfuss-Bömeke
- Clinic for Cardiology and Pneumology, University Medical Center, 37075 Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Göttingen and Rhein Main, 61231 Bad Nauheim, Germany
- Institute of Pharmacology and Toxicology, University of Würzburg, 97078 Würzburg, Germany
| |
Collapse
|
2
|
Mar PL, Horbal P, Chung MK, Dukes JW, Ezekowitz M, Lakkireddy D, Lip GYH, Miletello M, Noseworthy PA, Reiffel JA, Tisdale JE, Olshansky B, Gopinathannair R. Drug Interactions Affecting Antiarrhythmic Drug Use. Circ Arrhythm Electrophysiol 2022; 15:e007955. [PMID: 35491871 DOI: 10.1161/circep.121.007955] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Antiarrhythmic drugs (AAD) play an important role in the management of arrhythmias. Drug interactions involving AAD are common in clinical practice. As AADs have a narrow therapeutic window, both pharmacokinetic as well as pharmacodynamic interactions involving AAD can result in serious adverse drug reactions ranging from arrhythmia recurrence, failure of device-based therapy, and heart failure, to death. Pharmacokinetic drug interactions frequently involve the inhibition of key metabolic pathways, resulting in accumulation of a substrate drug. Additionally, over the past 2 decades, the P-gp (permeability glycoprotein) has been increasingly cited as a significant source of drug interactions. Pharmacodynamic drug interactions involving AADs commonly involve additive QT prolongation. Amiodarone, quinidine, and dofetilide are AADs with numerous and clinically significant drug interactions. Recent studies have also demonstrated increased morbidity and mortality with the use of digoxin and other AAD which interact with P-gp. QT prolongation is an important pharmacodynamic interaction involving mainly Vaughan-Williams class III AAD as many commonly used drug classes, such as macrolide antibiotics, fluoroquinolone antibiotics, antipsychotics, and antiemetics prolong the QT interval. Whenever possible, serious drug-drug interactions involving AAD should be avoided. If unavoidable, patients will require closer monitoring and the concomitant use of interacting agents should be minimized. Increasing awareness of drug interactions among clinicians will significantly improve patient safety for patients with arrhythmias.
Collapse
Affiliation(s)
- Philip L Mar
- Department of Medicine, Division of Cardiology, St. Louis University, St. Louis, MO (P.L.M., P.H.)
| | - Piotr Horbal
- Department of Medicine, Division of Cardiology, St. Louis University, St. Louis, MO (P.L.M., P.H.)
| | - Mina K Chung
- Department of Cardiovascular Medicine, Heart, Vascular & Thoracic Institute (M.K.C.), Cleveland Clinic, OH
| | | | - Michael Ezekowitz
- Lankenau Heart Institute, Bryn Mawr Hospital & Sidney Kimmel Medical College (M.E.)
| | | | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool & Liverpool Heart & Chest Hospital, Liverpool, United Kingdom (G.Y.H.L.).,Department of Clinical Medicine, Aalborg, Denmark (G.Y.H.L.)
| | | | - Peter A Noseworthy
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN (P.A.N.)
| | - James A Reiffel
- Division of Cardiology, Department of Medicine, Columbia University, New York, NY (J.A.R.)
| | - James E Tisdale
- College of Pharmacy, Purdue University (J.E.T.).,School of Medicine, Indiana University, Indianapolis (J.E.T.)
| | - Brian Olshansky
- Division of Cardiology, Department of Medicine, University of Iowa, Iowa City (B.O.)
| | | | | |
Collapse
|
3
|
Horváth B, Szentandrássy N, Almássy J, Dienes C, Kovács ZM, Nánási PP, Banyasz T. Late Sodium Current of the Heart: Where Do We Stand and Where Are We Going? Pharmaceuticals (Basel) 2022; 15:ph15020231. [PMID: 35215342 PMCID: PMC8879921 DOI: 10.3390/ph15020231] [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: 01/13/2022] [Revised: 02/07/2022] [Accepted: 02/09/2022] [Indexed: 02/05/2023] Open
Abstract
Late sodium current has long been linked to dysrhythmia and contractile malfunction in the heart. Despite the increasing body of accumulating information on the subject, our understanding of its role in normal or pathologic states is not complete. Even though the role of late sodium current in shaping action potential under physiologic circumstances is debated, it’s unquestioned role in arrhythmogenesis keeps it in the focus of research. Transgenic mouse models and isoform-specific pharmacological tools have proved useful in understanding the mechanism of late sodium current in health and disease. This review will outline the mechanism and function of cardiac late sodium current with special focus on the recent advances of the area.
Collapse
Affiliation(s)
- Balázs Horváth
- Department of Physiology, University of Debrecen, 4032 Debrecen, Hungary; (B.H.); (N.S.); (J.A.); (C.D.); (Z.M.K.); (P.P.N.)
| | - Norbert Szentandrássy
- Department of Physiology, University of Debrecen, 4032 Debrecen, Hungary; (B.H.); (N.S.); (J.A.); (C.D.); (Z.M.K.); (P.P.N.)
- Department of Basic Medical Sciences, Faculty of Dentistry, University of Debrecen, 4032 Debrecen, Hungary
| | - János Almássy
- Department of Physiology, University of Debrecen, 4032 Debrecen, Hungary; (B.H.); (N.S.); (J.A.); (C.D.); (Z.M.K.); (P.P.N.)
| | - Csaba Dienes
- Department of Physiology, University of Debrecen, 4032 Debrecen, Hungary; (B.H.); (N.S.); (J.A.); (C.D.); (Z.M.K.); (P.P.N.)
| | - Zsigmond Máté Kovács
- Department of Physiology, University of Debrecen, 4032 Debrecen, Hungary; (B.H.); (N.S.); (J.A.); (C.D.); (Z.M.K.); (P.P.N.)
| | - Péter P. Nánási
- Department of Physiology, University of Debrecen, 4032 Debrecen, Hungary; (B.H.); (N.S.); (J.A.); (C.D.); (Z.M.K.); (P.P.N.)
- Department of Dental Physiology and Pharmacology, University of Debrecen, 4032 Debrecen, Hungary
| | - Tamas Banyasz
- Department of Physiology, University of Debrecen, 4032 Debrecen, Hungary; (B.H.); (N.S.); (J.A.); (C.D.); (Z.M.K.); (P.P.N.)
- Correspondence: ; Tel.: +36-(52)-255-575; Fax: +36-(52)-255-116
| |
Collapse
|
4
|
Nguyen HX, Wu T, Needs D, Zhang H, Perelli RM, DeLuca S, Yang R, Pan M, Landstrom AP, Henriquez C, Bursac N. Engineered bacterial voltage-gated sodium channel platform for cardiac gene therapy. Nat Commun 2022; 13:620. [PMID: 35110560 PMCID: PMC8810800 DOI: 10.1038/s41467-022-28251-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 01/11/2022] [Indexed: 12/19/2022] Open
Abstract
Therapies for cardiac arrhythmias could greatly benefit from approaches to enhance electrical excitability and action potential conduction in the heart by stably overexpressing mammalian voltage-gated sodium channels. However, the large size of these channels precludes their incorporation into therapeutic viral vectors. Here, we report a platform utilizing small-size, codon-optimized engineered prokaryotic sodium channels (BacNav) driven by muscle-specific promoters that significantly enhance excitability and conduction in rat and human cardiomyocytes in vitro and adult cardiac tissues from multiple species in silico. We also show that the expression of BacNav significantly reduces occurrence of conduction block and reentrant arrhythmias in fibrotic cardiac cultures. Moreover, functional BacNav channels are stably expressed in healthy mouse hearts six weeks following intravenous injection of self-complementary adeno-associated virus (scAAV) without causing any adverse effects on cardiac electrophysiology. The large diversity of prokaryotic sodium channels and experimental-computational platform reported in this study should facilitate the development and evaluation of BacNav-based gene therapies for cardiac conduction disorders.
Collapse
Affiliation(s)
- Hung X Nguyen
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Tianyu Wu
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Daniel Needs
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Hengtao Zhang
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Robin M Perelli
- Department of Pediatrics, Division of Cardiology, Duke University School of Medicine, Durham, NC, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA
| | - Sophia DeLuca
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA
| | - Rachel Yang
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Michael Pan
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Andrew P Landstrom
- Department of Pediatrics, Division of Cardiology, Duke University School of Medicine, Durham, NC, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA
| | - Craig Henriquez
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
5
|
Kistamás K, Hézső T, Horváth B, Nánási PP. Late sodium current and calcium homeostasis in arrhythmogenesis. Channels (Austin) 2021; 15:1-19. [PMID: 33258400 PMCID: PMC7757849 DOI: 10.1080/19336950.2020.1854986] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 10/26/2020] [Accepted: 11/19/2020] [Indexed: 12/19/2022] Open
Abstract
The cardiac late sodium current (INa,late) is the small sustained component of the sodium current active during the plateau phase of the action potential. Several studies demonstrated that augmentation of the current can lead to cardiac arrhythmias; therefore, INa,late is considered as a promising antiarrhythmic target. Fundamentally, enlarged INa,late increases Na+ influx into the cell, which, in turn, is converted to elevated intracellular Ca2+ concentration through the Na+/Ca2+ exchanger. The excessive Ca2+ load is known to be proarrhythmic. This review describes the behavior of the voltage-gated Na+ channels generating INa,late in health and disease and aims to discuss the physiology and pathophysiology of Na+ and Ca2+ homeostasis in context with the enhanced INa,late demonstrating also the currently accessible antiarrhythmic choices.
Collapse
Affiliation(s)
- Kornél Kistamás
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tamás Hézső
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Balázs Horváth
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Péter P Nánási
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Department of Dental Physiology, Faculty of Dentistry, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
6
|
Ton AT, Nguyen W, Sweat K, Miron Y, Hernandez E, Wong T, Geft V, Macias A, Espinoza A, Truong K, Rasoul L, Stafford A, Cotta T, Mai C, Indersmitten T, Page G, Miller PE, Ghetti A, Abi-Gerges N. Arrhythmogenic and antiarrhythmic actions of late sustained sodium current in the adult human heart. Sci Rep 2021; 11:12014. [PMID: 34103608 PMCID: PMC8187365 DOI: 10.1038/s41598-021-91528-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 05/27/2021] [Indexed: 12/19/2022] Open
Abstract
Late sodium current (late INa) inhibition has been proposed to suppress the incidence of arrhythmias generated by pathological states or induced by drugs. However, the role of late INa in the human heart is still poorly understood. We therefore investigated the role of this conductance in arrhythmias using adult primary cardiomyocytes and tissues from donor hearts. Potentiation of late INa with ATX-II (anemonia sulcata toxin II) and E-4031 (selective blocker of the hERG channel) slowed the kinetics of action potential repolarization, impaired Ca2+ homeostasis, increased contractility, and increased the manifestation of arrhythmia markers. These effects could be reversed by late INa inhibitors, ranolazine and GS-967. We also report that atrial tissues from donor hearts affected by atrial fibrillation exhibit arrhythmia markers in the absence of drug treatment and inhibition of late INa with GS-967 leads to a significant reduction in arrhythmic behaviour. These findings reveal a critical role for the late INa in cardiac arrhythmias and suggest that inhibition of this conductance could provide an effective therapeutic strategy. Finally, this study highlights the utility of human ex-vivo heart models for advancing cardiac translational sciences.
Collapse
Affiliation(s)
- Anh Tuan Ton
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA, 92109, USA
| | - William Nguyen
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA, 92109, USA
| | - Katrina Sweat
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA, 92109, USA
| | - Yannick Miron
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA, 92109, USA
| | - Eduardo Hernandez
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA, 92109, USA
| | - Tiara Wong
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA, 92109, USA
| | - Valentyna Geft
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA, 92109, USA
| | - Andrew Macias
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA, 92109, USA
| | - Ana Espinoza
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA, 92109, USA
| | - Ky Truong
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA, 92109, USA
| | - Lana Rasoul
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA, 92109, USA
| | - Alexa Stafford
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA, 92109, USA
| | - Tamara Cotta
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA, 92109, USA
| | - Christina Mai
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA, 92109, USA
| | - Tim Indersmitten
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA, 92109, USA
| | - Guy Page
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA, 92109, USA
| | - Paul E Miller
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA, 92109, USA
| | - Andre Ghetti
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA, 92109, USA
| | - Najah Abi-Gerges
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA, 92109, USA.
| |
Collapse
|
7
|
Keseroglu BB, Ozer E, Karakan T, Ozgur BC, Surer H, Ogus E, Hucemenoglu S, Yuceturk CN, Agras K. Protective effects of Ranolazine on testicular torsion and detorsion injury in rats. Andrologia 2020; 52:e13616. [DOI: 10.1111/and.13616] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 04/02/2020] [Accepted: 04/03/2020] [Indexed: 01/24/2023] Open
Affiliation(s)
- Bilge Bugra Keseroglu
- Department of Urology Ankara Training and Research Hospital University of Health Sciences Ankara Turkey
| | - Elif Ozer
- Department of Pathology Ankara Training and Research Hospital University of Health Sciences Ankara Turkey
| | - Tolga Karakan
- Department of Urology Ankara Training and Research Hospital University of Health Sciences Ankara Turkey
| | - Berat Cem Ozgur
- Department of Urology Ankara Training and Research Hospital University of Health Sciences Ankara Turkey
| | - Hatice Surer
- Department of Biochemistry Ankara Training and Research Hospital University of Health Sciences Ankara Turkey
| | - Elmas Ogus
- Department of Biochemistry Ankara Training and Research Hospital University of Health Sciences Ankara Turkey
| | - Sema Hucemenoglu
- Department of Pathology Ankara Training and Research Hospital University of Health Sciences Ankara Turkey
| | - Cem Nedim Yuceturk
- Department of Urology Ankara Training and Research Hospital University of Health Sciences Ankara Turkey
| | - Koray Agras
- Department of Urology Ankara Training and Research Hospital University of Health Sciences Ankara Turkey
| |
Collapse
|
8
|
Isaac E, Cooper SM, Jones SA, Loubani M. Do age-associated changes of voltage-gated sodium channel isoforms expressed in the mammalian heart predispose the elderly to atrial fibrillation? World J Cardiol 2020; 12:123-135. [PMID: 32431783 PMCID: PMC7215965 DOI: 10.4330/wjc.v12.i4.123] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 01/18/2020] [Accepted: 03/15/2020] [Indexed: 02/06/2023] Open
Abstract
Atrial fibrillation (AF) is the most common cardiac arrhythmia worldwide. The prevalence of the disease increases with age, strongly implying an age-related process underlying the pathology. At a time when people are living longer than ever before, an exponential increase in disease prevalence is predicted worldwide. Hence unraveling the underlying mechanics of the disease is paramount for the development of innovative treatment and prevention strategies. The role of voltage-gated sodium channels is fundamental in cardiac electrophysiology and may provide novel insights into the arrhythmogenesis of AF. Nav1.5 is the predominant cardiac isoform, responsible for the action potential upstroke. Recent studies have demonstrated that Nav1.8 (an isoform predominantly expressed within the peripheral nervous system) is responsible for cellular arrhythmogenesis through the enhancement of pro-arrhythmogenic currents. Animal studies have shown a decline in Nav1.5 leading to a diminished action potential upstroke during phase 0. Furthermore, the study of human tissue demonstrates an inverse expression of sodium channel isoforms; reduction of Nav1.5 and increase of Nav1.8 in both heart failure and ventricular hypertrophy. This strongly suggests that the expression of voltage-gated sodium channels play a crucial role in the development of arrhythmias in the diseased heart. Targeting aberrant sodium currents has led to novel therapeutic approaches in tackling AF and continues to be an area of emerging research. This review will explore how voltage-gated sodium channels may predispose the elderly heart to AF through the examination of laboratory and clinical based evidence.
Collapse
Affiliation(s)
- Emmanuel Isaac
- Department of Cardiothoracic Surgery, Hull University Teaching Hospitals, Cottingham HU16 5JQ, United Kingdom
| | - Stephanie M Cooper
- Department of Biomedical Sciences, University of Hull, Hull HU6 7RX, United Kingdom
| | - Sandra A Jones
- Department of Biomedical Sciences, University of Hull, Hull HU6 7RX, United Kingdom
| | - Mahmoud Loubani
- Department of Cardiothoracic Surgery, Hull University Teaching Hospitals, Cottingham HU16 5JQ, United Kingdom
| |
Collapse
|
9
|
Horváth B, Hézső T, Kiss D, Kistamás K, Magyar J, Nánási PP, Bányász T. Late Sodium Current Inhibitors as Potential Antiarrhythmic Agents. Front Pharmacol 2020; 11:413. [PMID: 32372952 PMCID: PMC7184885 DOI: 10.3389/fphar.2020.00413] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 03/18/2020] [Indexed: 12/19/2022] Open
Abstract
Based on recent findings, an increased late sodium current (INa,late) plays an important pathophysiological role in cardiac diseases, including rhythm disorders. The article first describes what is INa,late and how it functions under physiological circumstances. Next, it shows the wide range of cellular mechanisms that can contribute to an increased INa,late in heart diseases, and also discusses how the upregulated INa,late can play a role in the generation of cardiac arrhythmias. The last part of the article is about INa,late inhibiting drugs as potential antiarrhythmic agents, based on experimental and preclinical data as well as in the light of clinical trials.
Collapse
Affiliation(s)
- Balázs Horváth
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Tamás Hézső
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Dénes Kiss
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Kornél Kistamás
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - János Magyar
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Division of Sport Physiology, University of Debrecen, Debrecen, Hungary
| | - Péter P. Nánási
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Department of Dental Physiology and Pharmacology, Faculty of Dentistry, University of Debrecen, Debrecen, Hungary
| | - Tamás Bányász
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
10
|
Ranolazine in High-Risk Patients With Implanted Cardioverter-Defibrillators: The RAID Trial. J Am Coll Cardiol 2019; 72:636-645. [PMID: 30071993 DOI: 10.1016/j.jacc.2018.04.086] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 04/27/2018] [Accepted: 04/30/2018] [Indexed: 12/19/2022]
Abstract
BACKGROUND Ventricular tachycardia (VT) and ventricular fibrillation (VF) remain a challenging problem in patients with implantable cardioverter-defibrillators (ICDs). OBJECTIVES This study aimed to determine whether ranolazine administration decreases the likelihood of VT, VF, or death in patients with an ICD. METHODS This was double-blind, placebo-controlled clinical trial in which high-risk ICD patients with ischemic or nonischemic cardiomyopathy were randomized to 1,000 mg ranolazine twice a day or placebo. The primary endpoint was VT or VF requiring appropriate ICD therapy or death, whichever occurred first. Pre-specified secondary endpoints included ICD shock for VT, VF, or death and recurrent VT or VF requiring ICD therapy. RESULTS Among 1,012 ICD patients (510 randomized to ranolazine and 502 to placebo) the mean age was 64 ± 10 years and 18% were women. During 28 ± 16 months of follow-up there were 372 (37%) patients with primary endpoint, 270 (27%) patients with VT or VF, and 148 (15%) deaths. The blinded study drug was discontinued in 199 (39.6%) patients receiving placebo and in 253 (49.6%) patients receiving ranolazine (p = 0.001). The hazard ratio for ranolazine versus placebo was 0.84 (95% confidence interval: 0.67 to 1.05; p = 0.117) for VT, VF, or death. In a pre-specified secondary analysis, patients randomized to ranolazine had a marginally significant lower risk of ICD therapies for recurrent VT or VF (hazard ratio: 0.70; 95% confidence interval: 0.51 to 0.96; p = 0.028). There were no other significant treatment effects in other pre-specified secondary analyses, which included individual components of the primary endpoint, inappropriate shocks, cardiac hospitalizations, and quality of life. CONCLUSIONS In high-risk ICD patients, treatment with ranolazine did not significantly reduce the incidence of the first VT or VF, or death. However, the study was underpowered to detect a difference in the primary endpoint. In prespecified secondary endpoint analyses, ranolazine administration was associated with a significant reduction in recurrent VT or VF requiring ICD therapy without evidence for increased mortality. (Ranolazine Implantable Cardioverter-Defibrillator Trial [RAID]; NCT01215253).
Collapse
|
11
|
Wu M, Tran PN, Sheng J, Randolph AL, Wu WW. Drug potency on inhibiting late Na + current is sensitive to gating modifier and current region where drug effects were measured. J Pharmacol Toxicol Methods 2019; 100:106605. [PMID: 31255744 DOI: 10.1016/j.vascn.2019.106605] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 06/18/2019] [Accepted: 06/24/2019] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Cardiac late Na+ current (INaL) contributes to ventricular action potential duration. Pathological increase in INaL is arrhythmogenic, and inhibition of INaL offers protection against ventricular repolarization disturbance. Recently, two INaL datasets generated by different laboratories that assessed current inhibition by a panel of clinical drugs as a part of the Comprehensive in vitro Proarrhythmia Assay (CiPA) initiative were published. The results revealed a surprising degree of data variability despite of the use of a standardized voltage protocol. This study investigated whether remaining procedural differences related to experimental methods and data analysis associated with these datasets can produce differences in INaL pharmacology. METHODS Whole cell voltage clamp recordings were performed on cells expressing NaV1.5 α- and β1-subunits to study: 1) the impact of gating modifiers used to augment INaL (ATX-II vs. veratridine), internal solution composition (with vs. without ATP and GTP), and recording temperature (23 °C vs 37 °C) on stability of INaL measured across the duration of a patch clamp experiment; 2) mechanisms of each gating modifier on Na+ channels; and 3) effects of six drugs (lidocaine, mexiletine, chloroquine, ranolazine, ritonavir, and verapamil) on INaL induced by either gating modifier. RESULTS Stability of INaL is affected by the choice of gating modifier, presence of nucleotides in the internal solution, and recording temperature. ATX-II and veratridine produced different changes in Na+ channel gating, inducing mechanistically distinct INaL. Drug potencies on inhibiting INaL were dependent on the choice of gating modifier and current region where drug effects were measured. DISCUSSION INaL pharmacology can be impacted by all experimental factors examined in this study. The effect of gating modifier and current region used to quantify drug inhibition alone led to 30× difference in half inhibitory concentration (IC50) for ritonavir, demonstrating that substantial difference in drug inhibition can be produced. Drug potencies on inhibiting INaL derived from different patch clamp studies may thus not be generalizable. For INaL pharmacology to be useful for in silico modeling or interpreting drug-induced changes in cardiac action potentials or ECG, standardizing INaL experimental procedures including data analysis methods is necessary to minimize data variability.
Collapse
Affiliation(s)
- Min Wu
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Center for Drug Evaluation and Research, The US Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD 20993, United States of America
| | - Phu N Tran
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Center for Drug Evaluation and Research, The US Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD 20993, United States of America
| | - Jiansong Sheng
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Center for Drug Evaluation and Research, The US Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD 20993, United States of America
| | - Aaron L Randolph
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Center for Drug Evaluation and Research, The US Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD 20993, United States of America
| | - Wendy W Wu
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Center for Drug Evaluation and Research, The US Food and Drug Administration, 10903 New Hampshire Avenue, Silver Spring, MD 20993, United States of America.
| |
Collapse
|
12
|
Kroncke BM, Yang T, Roden DM. Multiple mechanisms underlie increased cardiac late sodium current. Heart Rhythm 2019; 16:1091-1097. [PMID: 30677491 DOI: 10.1016/j.hrthm.2019.01.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Indexed: 01/08/2023]
Abstract
BACKGROUND We recently reported a quantitative relationship between the degree of functional perturbation reported in the literature for 356 variants in the cardiac sodium channel gene SCN5A and the penetrance of resulting arrhythmia phenotypes. In the course of that work, we identified multiple SCN5A variants, including R1193Q, that are common in populations but are reported in human embryonic kidney (HEK) cells to generate large late sodium current (INa-L). OBJECTIVE The purpose of this study was to compare the functional properties of R1193Q with those of the well-studied type 3 long QT syndrome mutation ΔKPQ. METHODS We compared functional properties of SCN5A R1193Q with those of ΔKPQ in Chinese hamster ovary (CHO) cells at baseline and after exposure to intracellular phosphatidylinositol (3,4,5)-trisphosphate (PIP3), which inhibits INa-L generated by decreased Phosphoinositide 3-kinase (PI3K) activity. We also used CRISPR/Cas9 editing to generate R1193Q in human-induced pluripotent stem cells differentiated to cardiomyocytes (hiPSC-CMs). RESULTS Both R1193Q and ΔKPQ generated robust INa-L in CHO cells. PIP3 abrogated the late current phenotype in R1193Q cells but had no effect on ΔKPQ. Homozygous R1193Q hiPSC-CMs displayed increased INa-L and long action potentials with frequent triggered beats, which were reversed with the addition of PIP3. CONCLUSION The consistency between the late current produced in HEK cells, CHO cells, and hiPSC-CMs suggests that the late current is a feature of the SCN5A R1193Q variant in human cardiomyocytes but that the mechanism by which the late current is produced is distinct and indirect, as compared with the more highly penetrant ΔKPQ. These data suggest that observing a late current in an in vitro setting does not necessarily translate to highly pathogenic type 3 long QT syndrome phenotype but depends on the underlying mechanism.
Collapse
Affiliation(s)
- Brett M Kroncke
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.
| | - Tao Yang
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Dan M Roden
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
13
|
Han D, Tan H, Sun C, Li G. Dysfunctional Nav1.5 channels due to SCN5A mutations. Exp Biol Med (Maywood) 2018; 243:852-863. [PMID: 29806494 DOI: 10.1177/1535370218777972] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The voltage-gated sodium channel 1.5 (Nav1.5), encoded by the SCN5A gene, is responsible for the rising phase of the action potential of cardiomyocytes. The sodium current mediated by Nav1.5 consists of peak and late components (INa-P and INa-L). Mutant Nav1.5 causes alterations in the peak and late sodium current and is associated with an increasingly wide range of congenital arrhythmias. More than 400 mutations have been identified in the SCN5A gene. Although the mechanisms of SCN5A mutations leading to a variety of arrhythmias can be classified according to the alteration of INa-P and INa-L as gain-of-function, loss-of-function and both, few researchers have summarized the mechanisms in this way before. In this review article, we aim to review the mechanisms underlying dysfunctional Nav1.5 due to SCN5A mutations and to provide some new insights into further approaches in the treatment of arrhythmias. Impact statement The field of ion channelopathy caused by dysfunctional Nav1.5 due to SCN5A mutations is rapidly evolving as novel technologies of electrophysiology are introduced and our understanding of the mechanisms of various arrhythmias develops. In this review, we focus on the dysfunctional Nav1.5 related to arrhythmias and the underlying mechanisms. We update SCN5A mutations in a precise way since 2013 and presents novel classifications of SCN5A mutations responsible for the dysfunction of the peak (INa-P) and late (INa-L) sodium channels based on their phenotypes, including loss-, gain-, and coexistence of gain- and loss-of function mutations in INa-P, INa-L, respectively. We hope this review will provide a new comprehensive way to better understand the electrophysiological mechanisms underlying arrhythmias from cell to bedside, promoting the management of various arrhythmias in practice.
Collapse
Affiliation(s)
- Dan Han
- 1 Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Hui Tan
- 2 Department of Respiratory Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Chaofeng Sun
- 1 Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| | - Guoliang Li
- 1 Department of Cardiovascular Medicine, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, P.R. China
| |
Collapse
|
14
|
Mezincescu A, Karthikeyan VJ, Nadar SK. Ranolazine: A true pluripotent cardiovascular drug or jack of all trades, master of none? Sultan Qaboos Univ Med J 2018; 18:e13-e23. [PMID: 29666676 DOI: 10.18295/squmj.2018.18.01.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 12/18/2017] [Accepted: 01/08/2018] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease (CVD) is a leading cause of morbidity and mortality worldwide. Although the majority of patients with CVD are treated with interventional procedures, a substantial number require medical therapy in terms of both prognosis and symptomatic relief. However, commonly used agents such as β-blockers and calcium channel blockers reduce blood pressure in patients whose resting pressures are often already low. Ranolazine is a promising agent that does not have significant effects on blood pressure or heart rate. Use of this drug has been documented in various cardiovascular conditions, including ischaemic heart disease, heart failure and arrhythmias. This review article aimed to examine current evidence on the use of ranolazine in various cardiovascular conditions in order to determine whether it is a true pluripotent cardiovascular agent or, on the other hand, a "jack of all trades, master of none."
Collapse
Affiliation(s)
- Alice Mezincescu
- Cardiovascular & Diabetes Research Unit, University of Aberdeen, Aberdeen, UK
| | - V J Karthikeyan
- Royal Albert Edward Infirmary, Wrightington Wigan & Leigh NHS Foundation Trust, Wigan, UK
| | - Sunil K Nadar
- Department of Medicine, Sultan Qaboos University Hospital, Muscat, Oman
| |
Collapse
|
15
|
Song Y, Belardinelli L. Basal late sodium current is a significant contributor to the duration of action potential of guinea pig ventricular myocytes. Physiol Rep 2018; 5:e13295. [PMID: 28554967 PMCID: PMC5449569 DOI: 10.14814/phy2.13295] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 05/01/2017] [Accepted: 05/02/2017] [Indexed: 01/17/2023] Open
Abstract
In cardiac myocytes, an enhancement of late sodium current (INaL) under pathological conditions is known to cause prolongation of action potential duration (APD). This study investigated the contribution of INaL under basal, physiological conditions to the APD Whole-cell INaL and the APD of ventricular myocytes isolated from healthy adult guinea pigs were measured at 36°C. The INaL inhibitor GS967 or TTX was applied to block INaL The amplitude of basal INaL and the APD at 50% repolarization in myocytes stimulated at a frequency of 0.17 Hz were -0.24 ± 0.02 pA/pF and 229 ± 6 msec, respectively. GS967 (0.01-1 μmol/L) concentration dependently reduced the basal INaL by 18 ± 3-82 ± 4%. At the same concentrations, GS967 shortened the APD by 9 ± 2 to 25 ± 1%. Similarly, TTX at 0.1-10 μmol/L decreased the basal INaL by 13 ± 1-94 ± 1% and APD by 8 ± 1-31 ± 2%. There was a close correlation (R2 = 0.958) between the percentage inhibition of INaL and the percentage shortening of APD caused by either GS967 or TTX MTSEA (methanethiosulfonate ethylammonium, 2 mmol/L), a NaV1.5 channel blocker, reduced the INaL by 90 ± 5%, suggesting that the NaV1.5 channel isoform is the major contributor to the basal INaL KN-93 (10 μmol/L) and AIP (2 μmol/L), blockers of CaMKII, moderately reduced the basal INaL Thus, this study provides strong evidence that basal endogenous INaL is a significant contributor to the APD of cardiac myocytes. In addition, the basal INaL of guinea pig ventricular myocytes is mainly generated from NaV1.5 channel isoform and is regulated by CaMKII.
Collapse
Affiliation(s)
- Yejia Song
- University of Florida, Gainesville, Florida
| | | |
Collapse
|
16
|
|
17
|
Runte KE, Bell SP, Selby DE, Häußler TN, Ashikaga T, LeWinter MM, Palmer BM, Meyer M. Relaxation and the Role of Calcium in Isolated Contracting Myocardium From Patients With Hypertensive Heart Disease and Heart Failure With Preserved Ejection Fraction. Circ Heart Fail 2017; 10:CIRCHEARTFAILURE.117.004311. [PMID: 28784688 DOI: 10.1161/circheartfailure.117.004311] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 07/06/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND Relaxation characteristics and Ca2+ homeostasis have not been studied in isolated myocardium from patients with hypertensive heart disease (HHD) and heart failure with preserved ejection fraction (HFpEF). Prolonged myocardial relaxation is believed to play an important role in the pathophysiology of these conditions. In this study, we evaluated relaxation parameters, myocardial calcium (Ca2+), and sodium (Na+) handling, as well as ion transporter expression and tested the effect of Na+-influx inhibitors on relaxation in isolated myocardium from patients with HHD and HFpEF. METHODS AND RESULTS Relaxation characteristics were studied in myocardial strip preparations under physiological conditions at stimulation rates of 60 and 180 per minute. Intracellular Ca2+ and Na+ were simultaneously assessed using Fura-2 and AsanteNATRIUMGreen-2, whereas elemental analysis was used to measure total myocardial concentrations of Ca, Na, and other elements. Quantitative polymerase chain reaction was used to measure expression levels of key ion transport proteins. The lusitropic effect of Na+-influx inhibitors ranolazine, furosemide, and amiloride was evaluated. Myocardial left ventricular biopsies were obtained from 36 control patients, 29 HHD and 19 HHD+HFpEF. When compared with control patients, half maximal relaxation time (RT50) at 60 per minute was prolonged by 13% in HHD and by 18% in HHD+HFpEF (both P<0.05). Elevated resting Ca2+ levels and a tachycardia-induced increase in diastolic Ca2+ were associated with incomplete relaxation and an increase in diastolic tension in HHD and HHD+HFpEF. Na+ levels were not increased, and expression levels of Ca2+- or Na+-handling proteins were not altered. Na+-influx inhibitors did not improve relaxation or prevent incomplete relaxation at high stimulation rates. CONCLUSIONS Contraction and relaxation are prolonged in isolated myocardium from patients with HHD and HHD+HFpEF. This leads to incomplete relaxation at higher rates. Elevated calcium levels in HFpEF are neither a result of an impaired Na+ gradient nor expression changes in key ion transporters and regulatory proteins.
Collapse
Affiliation(s)
- K Elisabeth Runte
- From the Division of Cardiology, Department of Medicine (K.E.R., S.P.B., D.E.S., T.N.H., M.M.L., M.M.), Biostatistics Unit (T.A.), and Department of Molecular Physiology and Biophysics (M.M.L., B.M.P.), Larner College of Medicine at the University of Vermont, Burlington
| | - Stephen P Bell
- From the Division of Cardiology, Department of Medicine (K.E.R., S.P.B., D.E.S., T.N.H., M.M.L., M.M.), Biostatistics Unit (T.A.), and Department of Molecular Physiology and Biophysics (M.M.L., B.M.P.), Larner College of Medicine at the University of Vermont, Burlington
| | - Donald E Selby
- From the Division of Cardiology, Department of Medicine (K.E.R., S.P.B., D.E.S., T.N.H., M.M.L., M.M.), Biostatistics Unit (T.A.), and Department of Molecular Physiology and Biophysics (M.M.L., B.M.P.), Larner College of Medicine at the University of Vermont, Burlington
| | - Tim N Häußler
- From the Division of Cardiology, Department of Medicine (K.E.R., S.P.B., D.E.S., T.N.H., M.M.L., M.M.), Biostatistics Unit (T.A.), and Department of Molecular Physiology and Biophysics (M.M.L., B.M.P.), Larner College of Medicine at the University of Vermont, Burlington
| | - Takamuru Ashikaga
- From the Division of Cardiology, Department of Medicine (K.E.R., S.P.B., D.E.S., T.N.H., M.M.L., M.M.), Biostatistics Unit (T.A.), and Department of Molecular Physiology and Biophysics (M.M.L., B.M.P.), Larner College of Medicine at the University of Vermont, Burlington
| | - Martin M LeWinter
- From the Division of Cardiology, Department of Medicine (K.E.R., S.P.B., D.E.S., T.N.H., M.M.L., M.M.), Biostatistics Unit (T.A.), and Department of Molecular Physiology and Biophysics (M.M.L., B.M.P.), Larner College of Medicine at the University of Vermont, Burlington
| | - Bradley M Palmer
- From the Division of Cardiology, Department of Medicine (K.E.R., S.P.B., D.E.S., T.N.H., M.M.L., M.M.), Biostatistics Unit (T.A.), and Department of Molecular Physiology and Biophysics (M.M.L., B.M.P.), Larner College of Medicine at the University of Vermont, Burlington
| | - Markus Meyer
- From the Division of Cardiology, Department of Medicine (K.E.R., S.P.B., D.E.S., T.N.H., M.M.L., M.M.), Biostatistics Unit (T.A.), and Department of Molecular Physiology and Biophysics (M.M.L., B.M.P.), Larner College of Medicine at the University of Vermont, Burlington.
| |
Collapse
|
18
|
Curnis A, Salghetti F, Cerini M, Vizzardi E, Sciatti E, Vassanelli F, Villa C, Inama L, Raweh A, Giacopelli D, Bontempi L. Ranolazine therapy in drug-refractory ventricular arrhythmias. J Cardiovasc Med (Hagerstown) 2017; 18:534-538. [PMID: 28368882 DOI: 10.2459/jcm.0000000000000521] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
AIMS Ranolazine is an antiischemic and antianginal agent, but experimental and preclinical data provided evidence of additional antiarrhythmic properties. The aim of this study was to evaluate the safety and efficacy of ranolazine in reducing episodes of ventricular arrhythmias in patients with recurrent antiarrhythmic drug-refractory ventricular arrhythmias or with chronic angina. METHODS Seventeen implantable cardioverter defibrillator (ICD) recipients, who had experienced a worsening of their ventricular arrhythmia burden, and 12 ICD recipients with angina were enrolled. Patients were followed up for 6 months after the addition of ranolazine (postranolazine). Data were compared with before its administration (preranolazine). RESULTS In the Arrhythmias group, a significant reduction was found in the median number of ventricular tachycardia episodes per patient (4 vs. 0, P = 0.01), and in ICD interventions in terms of both antitachycardia pacing (2 vs. 0, P = 0.04) and shock delivery (2 vs. 0, P = 0.02) after the addition of ranolazine. Moreover, fewer patients experienced episodes of nonsustained ventricular tachycardia (71 vs. 41%, P = 0.04), ventricular tachycardia (76 vs. 24%, P = 0.01), ICD antitachycardia pacing (47 vs. 18%, P = 0.02), and ICD shocks (47 vs. 6%, P = 0.03). In the Angina group, none of the patients developed major ventricular arrhythmias while on ranolazine treatment. No adverse effects were observed. CONCLUSION In this small study, ranolazine proved to be effective, well tolerated, and safe in reducing ventricular arrhythmia episodes and ICD interventions in patients with recurrent antiarrhythmic drug-refractory events. In addition, none of the patients with chronic angina developed major ventricular arrhythmias.
Collapse
Affiliation(s)
- Antonio Curnis
- aDivision of Cardiology, Spedali Civili Hospital, Brescia, Italy bCardiac Surgery Department, L.U.de.S. University, Lugano, Switzerland cClinical Research, Biotronik Italia, Milano, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Hortigon-Vinagre MP, Zamora V, Burton FL, Green J, Gintant GA, Smith GL. The Use of Ratiometric Fluorescence Measurements of the Voltage Sensitive Dye Di-4-ANEPPS to Examine Action Potential Characteristics and Drug Effects on Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes. Toxicol Sci 2016; 154:320-331. [PMID: 27621282 PMCID: PMC5139069 DOI: 10.1093/toxsci/kfw171] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM) and higher throughput platforms have emerged as potential tools to advance cardiac drug safety screening. This study evaluated the use of high bandwidth photometry applied to voltage-sensitive fluorescent dyes (VSDs) to assess drug-induced changes in action potential characteristics of spontaneously active hiPSC-CM. Human iPSC-CM from 2 commercial sources (Cor.4U and iCell Cardiomyocytes) were stained with the VSD di-4-ANEPPS and placed in a specialized photometry system that simultaneously monitors 2 wavebands of emitted fluorescence, allowing ratiometric measurement of membrane voltage. Signals were acquired at 10 kHz and analyzed using custom software. Action potential duration (APD) values were normally distributed in cardiomyocytes (CMC) from both sources though the mean and variance differed significantly (APD90: 229 ± 15 ms vs 427 ± 49 ms [mean ± SD, P < 0.01]; average spontaneous cycle length: 0.99 ± 0.02 s vs 1.47 ± 0.35 s [mean ± SD, P < 0.01], Cor.4U vs iCell CMC, respectively). The 10-90% rise time of the AP (Trise) was ∼6 ms and was normally distributed when expressed as 1/[Formula: see text] in both cell preparations. Both cell types showed a rate dependence analogous to that of adult human cardiac cells. Furthermore, nifedipine, ranolazine, and E4031 had similar effects on cardiomyocyte electrophysiology in both cell types. However, ranolazine and E4031 induced early after depolarization-like events and high intrinsic firing rates at lower concentrations in iCell CMC. These data show that VSDs provide a minimally invasive, quantitative, and accurate method to assess hiPSC-CM electrophysiology and detect subtle drug-induced effects for drug safety screening while highlighting a need to standardize experimental protocols across preparations.
Collapse
Affiliation(s)
- M P Hortigon-Vinagre
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Science, University of Glasgow 126 University Place, Glasgow G12 8TA, United Kingdom
- Clyde Biosciences Ltd, BioCity Scotland, Bo'Ness Road, Newhouse, Lanarkshire, Scotland ML1 5UH, United Kingdom
| | - V Zamora
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Science, University of Glasgow 126 University Place, Glasgow G12 8TA, United Kingdom
- Clyde Biosciences Ltd, BioCity Scotland, Bo'Ness Road, Newhouse, Lanarkshire, Scotland ML1 5UH, United Kingdom
| | - F L Burton
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Science, University of Glasgow 126 University Place, Glasgow G12 8TA, United Kingdom
- Clyde Biosciences Ltd, BioCity Scotland, Bo'Ness Road, Newhouse, Lanarkshire, Scotland ML1 5UH, United Kingdom
| | - J Green
- AbbVie, 1 North Waukegan Road, Department ZR-13, Building AP-9A, North Chicago, Illinois 60064-6119
| | - G A Gintant
- AbbVie, 1 North Waukegan Road, Department ZR-13, Building AP-9A, North Chicago, Illinois 60064-6119
| | - G L Smith
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Science, University of Glasgow 126 University Place, Glasgow G12 8TA, United Kingdom
- Clyde Biosciences Ltd, BioCity Scotland, Bo'Ness Road, Newhouse, Lanarkshire, Scotland ML1 5UH, United Kingdom
| |
Collapse
|
20
|
Dempsey GT, Chaudhary KW, Atwater N, Nguyen C, Brown BS, McNeish JD, Cohen AE, Kralj JM. Cardiotoxicity screening with simultaneous optogenetic pacing, voltage imaging and calcium imaging. J Pharmacol Toxicol Methods 2016; 81:240-50. [DOI: 10.1016/j.vascn.2016.05.003] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 05/02/2016] [Accepted: 05/09/2016] [Indexed: 12/23/2022]
|
21
|
|
22
|
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: 0.8] [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
|
23
|
Hale SL, Kloner RA. Ranolazine, an Inhibitor of the Late Sodium Channel Current, Reduces Postischemic Myocardial Dysfunction in the Rabbit. J Cardiovasc Pharmacol Ther 2016; 11:249-55. [PMID: 17220471 DOI: 10.1177/1074248406294607] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Ranolazine is a selective inhibitor of the late sodium current relative to peak sodium channel current, and via this mechanism, it may decrease sodium-dependent intracellular calcium overload during ischemia and reperfusion. Ranolazine reduces the frequency of angina attacks, but there is little information on its effects on myocardial stunning after short-term ischemia. The objective of this study was to test the effects of ranolazine on left ventricular (LV) function and myocardial stunning after ischemia/reperfusion in rabbits. Myocardial stunning was induced in rabbits by 15 minutes of coronary artery occlusion (CAO) followed by 3 hours reperfusion. Ten minutes before CAO, rabbits were randomly assigned to vehicle (n = 15) or ranolazine (2 mg/kg bolus plus 60 μg/kg/min infusion, IV, n = 15). Myocardial stunning was assessed by LV 2-dimensional echocardiography using, as a marker of severity, ischemic free-wall fractional thickening (FWft; systolic wall thickness – diastolic wall thickness/diastolic wall thickness). Regional ejection fraction (EF) was also assessed. During CAO, FWft was depressed in both groups, indicating an ischemic insult (FWft was reduced from 0.62 ± 0.05 at baseline to 0.10 ± 0.04 in vehicle and from 0.73 ± 0.05 to 0.26 ± 0.07 in ranolazine, P < 0.05, ranolazine vs vehicle). After reperfusion, previously ischemic myocardium remained stunned; however, FWft recovered significantly better in ranolazine (0.51 ± 0.05) than in vehicle (0.35 ± 0.04, P = .027). Baseline EF was 0.65 ± 0.02 in the ranolazine and 0.68 ± 0.02 in vehicle ( P = ns). During CAO, EF was reduced by 36% ± 6% in vehicle versus only 20% ± 6% in ranolazine ( P < .05). At the end of reperfusion, EF remained depressed in both groups, but the reduction in the vehicle group (25% ± 5%) was significantly worse than in ranolazine (9% ± 4%, P = .017). Improvement in function was independent of necrosis (negligible) or differences in hemodynamics (no differences between groups). Ranolazine treatment reduced myocardial stunning following brief ischemia/reperfusion suggesting that inhibiting the late sodium channel current may be a novel approach to treating stunning independent of effects on hemodynamics.
Collapse
Affiliation(s)
- Sharon L Hale
- Heart Institute of Good Samaritan Hospital and the Keck School of Medicine, Division of Cardiovascular Medicine, University of Southern California, Los Angeles 90017, USA.
| | | |
Collapse
|
24
|
Potet F, Vanoye CG, George AL. Use-Dependent Block of Human Cardiac Sodium Channels by GS967. Mol Pharmacol 2016; 90:52-60. [PMID: 27136942 DOI: 10.1124/mol.116.103358] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 04/29/2016] [Indexed: 01/22/2023] Open
Abstract
GS-458967, 6-(4-(Trifluoromethoxy)phenyl)-3-(trifluoromethyl)-[1,2,4]triazolo[4,3-a]pyridine (GS967) is a recently described, novel, sodium channel inhibitor exhibiting potent antiarrhythmic effects in various in vitro and in vivo models. The antiarrhythmic mechanism has been attributed to preferential suppression of late sodium current. However, there has been no reported systematic investigation of the effects of this compound on isolated sodium channels. Here, we examined the effects of GS967 on peak (INaP) and late (INaL) sodium current recorded from cells that heterologously expressed human cardiac voltage-gated sodium channel, the principle cardiac sodium channel. As previously described, we observed that GS967 exerted tonic block of INaL (63%) to a significantly greater extent than INaP (19%). However, GS967 also caused a reduction of INaP in a frequency-dependent manner, consistent with use-dependent block (UDB). GS967 evoked more potent UDB of INaP (IC50 = 0.07 µM) than ranolazine (16 µM) and lidocaine (17 µM). Use-dependent block was best explained by a significant slowing of recovery from fast and slow inactivation with a significant enhancement of slow inactivation in the presence of GS967. Furthermore, GS967 was found to exert these same effects on a prototypical long QT syndrome mutation (delKPQ). An engineered mutation at an interaction site for local anesthetic agents (F1760A) partially attenuated the effect of GS967 on UDB, but had no effect on tonic INaL block. We conclude that GS967 is a preferential inhibitor of INaL, but it also exerts previously unreported strong effects on slow inactivation and recovery from inactivation, resulting in substantial UDB that is not entirely dependent on a known interaction site for local anesthetic agents.
Collapse
Affiliation(s)
- Franck Potet
- Department of Pharmacology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | - Carlos G Vanoye
- Department of Pharmacology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| | - Alfred L George
- Department of Pharmacology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
25
|
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: 1.9] [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
|
26
|
Cacciapuoti F. Ranolazine and Ivabradine: two different modalities to act against ischemic heart disease. Ther Adv Cardiovasc Dis 2016; 10:98-102. [PMID: 26944071 PMCID: PMC5933631 DOI: 10.1177/1753944716636042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Among the innovative drugs recently introduced for the management of chronic stable angina, Ranolazine and ivabradine represent two most true innovations. In fact, even if both drugs act by reducing myocardial work and thus oxygen consumption, this happens by a peculiar mechanism unlike that of conventional antischemic drugs. Ranolazine mediates its antianginal effects by the inhibition of cardiac late sodium current. This improves myocardial relaxation favoring myocardial perfusion. Ivabradine is a selective If channel blocker and acts by reducing firing rate of pacemaker cells in the sinoatrial node, without affecting the duration of action potential. The reduction of heart rate causes a reduction of left ventricular end diastolic pressure and increases the time useful to coronary flow by a prolongation of the diastole. A body of evidence found that two drugs are useful in ischemic patients whether at rest or during exercise. In addition, they can be used in monotherapy or in association with other conventional anti-ischemic drugs. The two medications could be used with advantage also in microvascular angina when standard therapy is ineffective. Thus, the two drugs represent an adjunctive and powerful therapeutic modality for the treatment of chronic stable angina, especially when conventional antianginal drugs were insufficient or inadequate.
Collapse
Affiliation(s)
- Federico Cacciapuoti
- Department of Internal Medicine, Second University of Naples, Piazza L. Miraglia, 2, 80138-Naples, Italy
| |
Collapse
|
27
|
Ambrosio G, Tamargo J, Grant PJ. Non-haemodynamic anti-anginal agents in the management of patients with stable coronary artery disease and diabetes: A review of the evidence. Diab Vasc Dis Res 2016; 13:98-112. [PMID: 26873904 DOI: 10.1177/1479164115609028] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Patients with coronary artery disease and concomitant diabetes mellitus tend to have more extensive vessel disease than non-diabetes mellitus coronary artery disease patients, are at high risk of adverse cardiovascular events and suffer from a great anginal burden. Very few trials have specifically addressed the issue of optimal anti-anginal therapy in coronary artery disease patients who also have diabetes mellitus. Among 'classical' anti-anginal agents, recent guidelines do not specifically recommend any molecule over others; however, European Society of Cardiology guidelines acknowledge that favourable data in patients with concomitant diabetes mellitus and coronary artery disease are available for trimetazidine and ranolazine, two anti-anginal agents with a non-haemodynamic mechanism of action. The aim of this article is to review available evidence supporting the anti-anginal efficacy of these two drugs in the difficult-to-treat population of diabetes mellitus patients, including their effects on glycated haemoglobin (HbA1c), a measure of medium-term glycaemic control. Although direct head-to-head comparisons have not been performed, available evidence favours ranolazine as an effective anti-anginal agent over trimetazidine in this population. In addition, ranolazine lowers HbA1c, indicating that it may improve glycaemic control in patients with diabetes mellitus. Conversely, scanty data are available on the metabolic effects of trimetazidine in this cohort of patients. Thus, ranolazine may represent a valuable therapeutic option in stable coronary artery disease patients with diabetes mellitus.
Collapse
Affiliation(s)
- Giuseppe Ambrosio
- Division of Cardiology, School of Medicine, University of Perugia, Perugia, Italy
| | - Juan Tamargo
- Department of Pharmacology, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
| | - Peter J Grant
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| |
Collapse
|
28
|
Unudurthi SD, Hund TJ. Late sodium current dysregulation as a causal factor in arrhythmia. Expert Rev Cardiovasc Ther 2016; 14:545-7. [PMID: 26886049 DOI: 10.1586/14779072.2016.1155451] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Sathya D Unudurthi
- a The Dorothy M. Davis Heart and Lung Research Institute.,b Department of Biomedical Engineering , College of Engineering, The Ohio State University , Columbus , OH , USA
| | - Thomas J Hund
- a The Dorothy M. Davis Heart and Lung Research Institute.,c Department of Biomedical Engineering , College of Engineering.,d Department of Internal Medicine , The Ohio State University.,e Wexner Medical Center, The Ohio State University , Columbus , OH , USA
| |
Collapse
|
29
|
Ranolazine for the symptomatic treatment of patients with chronic angina pectoris in Greece: a cost-utility study. BMC Health Serv Res 2015; 15:566. [PMID: 26684327 PMCID: PMC4683812 DOI: 10.1186/s12913-015-1228-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 12/10/2015] [Indexed: 12/19/2022] Open
Abstract
Background To conduct an economic evaluation comparing ranolazine as add-on therapy to standard-of-care (SoC) with SoC alone in patients with stable angina who did not respond adequately to first line therapy, in Greece. Methods A decision tree model was locally adapted in the Greek setting to evaluate the cost-utility of ranolazine during a 6-month period. The analysis was conducted from a third-party payer perspective. The clinical inputs were extracted from the published literature. The cost inputs considered in the model reflect drug acquisition, hospitalizations, vascular interventions and monitoring of patients. The resource utilization data were obtained from 3 local experts. All costs refer to the year 2014. Cost-effectiveness was assessed by means of the incremental cost per quality adjusted life year (QALY) gained with the ranolazine as add-on therapy relative to SoC alone (ICER). Probabilistic sensitivity analysis (PSA) was performed. Results Ranolazine as add-on therapy was more costly compared to SoC alone, as the 6-month total cost per patient was €1170 and € 984, respectively. Patients received ranolazine plus SoC and SoC alone gained 0.3155 QALYs and 0.2752 QALYs, respectively. Ranolazine plus SoC resulted in an ICER equal to €4620 per QALY gained, well below the threshold of €34,000 per QALY gained. The PSA showed that the likelihood of ranolazine plus SoC being cost-effective at the threshold of €34,000 per QALY gained was 100 %. Conclusions Τhe results suggest that ranolazine as add–on treatment may be a cost-effective alternative for the symptomatic treatment of patients with chronic stable angina in Greece. Electronic supplementary material The online version of this article (doi:10.1186/s12913-015-1228-y) contains supplementary material, which is available to authorized users.
Collapse
|
30
|
Clancy CE, Chen-Izu Y, Bers DM, Belardinelli L, Boyden PA, Csernoch L, Despa S, Fermini B, Hool LC, Izu L, Kass RS, Lederer WJ, Louch WE, Maack C, Matiazzi A, Qu Z, Rajamani S, Rippinger CM, Sejersted OM, O'Rourke B, Weiss JN, Varró A, Zaza A. Deranged sodium to sudden death. J Physiol 2015; 593:1331-45. [PMID: 25772289 DOI: 10.1113/jphysiol.2014.281204] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 10/14/2014] [Indexed: 12/19/2022] Open
Abstract
In February 2014, a group of scientists convened as part of the University of California Davis Cardiovascular Symposium to bring together experimental and mathematical modelling perspectives and discuss points of consensus and controversy on the topic of sodium in the heart. This paper summarizes the topics of presentation and discussion from the symposium, with a focus on the role of aberrant sodium channels and abnormal sodium homeostasis in cardiac arrhythmias and pharmacotherapy from the subcellular scale to the whole heart. Two following papers focus on Na(+) channel structure, function and regulation, and Na(+)/Ca(2+) exchange and Na(+)/K(+) ATPase. The UC Davis Cardiovascular Symposium is a biannual event that aims to bring together leading experts in subfields of cardiovascular biomedicine to focus on topics of importance to the field. The focus on Na(+) in the 2014 symposium stemmed from the multitude of recent studies that point to the importance of maintaining Na(+) homeostasis in the heart, as disruption of homeostatic processes are increasingly identified in cardiac disease states. Understanding how disruption in cardiac Na(+)-based processes leads to derangement in multiple cardiac components at the level of the cell and to then connect these perturbations to emergent behaviour in the heart to cause disease is a critical area of research. The ubiquity of disruption of Na(+) channels and Na(+) homeostasis in cardiac disorders of excitability and mechanics emphasizes the importance of a fundamental understanding of the associated mechanisms and disease processes to ultimately reveal new targets for human therapy.
Collapse
Affiliation(s)
- Colleen E Clancy
- Department of Pharmacology, University of California, Davis, Genome Building Rm 3503, Davis, CA, 95616-8636, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Antagonism of Nav channels and α1-adrenergic receptors contributes to vascular smooth muscle effects of ranolazine. Sci Rep 2015; 5:17969. [PMID: 26655634 PMCID: PMC4674695 DOI: 10.1038/srep17969] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 11/10/2015] [Indexed: 12/19/2022] Open
Abstract
Ranolazine is a recently developed drug used for the treatment of patients with chronic stable angina. It is a selective inhibitor of the persistent cardiac Na+ current (INa), and is known to reduce the Na+-dependent Ca2+ overload that occurs in cardiomyocytes during ischemia. Vascular effects of ranolazine, such as vasorelaxation,have been reported and may involve multiple pathways. As voltage-gated Na+ channels (Nav) present in arteries play a role in contraction, we hypothesized that ranolazine could target these channels. We studied the effects of ranolazine in vitro on cultured aortic smooth muscle cells (SMC) and ex vivo on rat aortas in conditions known to specifically activate or promote INa. We observed that in the presence of the Nav channel agonist veratridine, ranolazine inhibited INa and intracellular Ca2+ calcium increase in SMC, and arterial vasoconstriction. In arterial SMC, ranolazine inhibited the activity of tetrodotoxin-sensitive voltage-gated Nav channels and thus antagonized contraction promoted by low KCl depolarization. Furthermore, the vasorelaxant effects of ranolazine, also observed in human arteries and independent of the endothelium, involved antagonization of the α1-adrenergic receptor. Combined α1-adrenergic antagonization and inhibition of SMCs Nav channels could be involved in the vascular effects of ranolazine.
Collapse
|
32
|
Obejero-Paz CA, Bruening-Wright A, Kramer J, Hawryluk P, Tatalovic M, Dittrich HC, Brown AM. Quantitative Profiling of the Effects of Vanoxerine on Human Cardiac Ion Channels and its Application to Cardiac Risk. Sci Rep 2015; 5:17623. [PMID: 26616666 PMCID: PMC4663487 DOI: 10.1038/srep17623] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 11/03/2015] [Indexed: 12/19/2022] Open
Abstract
Vanoxerine has been in clinical trials for Parkinsonism, depression and cocaine addiction but lacked efficacy. Although a potent blocker of hERG, it produced no serious adverse events. We attributed the unexpected result to offsetting Multiple Ion Channel Effects (MICE). Vanoxerine’s effects were strongly frequency-dependent and we repositioned it for treatment of atrial fibrillation and flutter. Vanoxerine terminated AF/AFL in an animal model and a dose-ranging clinical trial. Reversion to normal rhythm was associated with QT prolongation yet absent proarrhythmia markers for Torsade de Pointes (TdP). To understand the QT/TdP discordance, we used quantitative profiling and compared vanoxerine with dofetilide, a selective hERG-blocking torsadogen used for intractable AF, verapamil, a non-torsadogenic MICE comparator and bepridil, a torsadogenic MICE comparator. At clinically relevant concentrations, verapamil blocked hCav1.2 and hERG, as did vanoxerine and bepridil both of which also blocked hNav1.5. In acute experiments and simulations, dofetilide produced early after depolarizations (EADs) and arrhythmias, whereas verapamil, vanoxerine and bepridil produced no proarrhythmia markers. Of the MICE drugs only bepridil inhibited hERG trafficking following overnight exposure. The results are consistent with the emphasis on MICE of the CiPA assay. Additionally we propose that trafficking inhibition of hERG be added to CiPA.
Collapse
Affiliation(s)
- Carlos A Obejero-Paz
- ChanTest Corporation, a Charles River Company, Discovery Services, 14656 Neo Parkway, Cleveland, OH 44128, USA
| | - Andrew Bruening-Wright
- ChanTest Corporation, a Charles River Company, Discovery Services, 14656 Neo Parkway, Cleveland, OH 44128, USA
| | - James Kramer
- ChanTest Corporation, a Charles River Company, Discovery Services, 14656 Neo Parkway, Cleveland, OH 44128, USA
| | - Peter Hawryluk
- ChanTest Corporation, a Charles River Company, Discovery Services, 14656 Neo Parkway, Cleveland, OH 44128, USA
| | - Milos Tatalovic
- ChanTest Corporation, a Charles River Company, Discovery Services, 14656 Neo Parkway, Cleveland, OH 44128, USA
| | - Howard C Dittrich
- Laguna Pharmaceuticals, 4225 Executive Square, Suite 960, La Jolla, CA 92037, USA
| | - Arthur M Brown
- ChanTest Corporation, a Charles River Company, Discovery Services, 14656 Neo Parkway, Cleveland, OH 44128, USA.,Laguna Pharmaceuticals, 4225 Executive Square, Suite 960, La Jolla, CA 92037, USA
| |
Collapse
|
33
|
Saad M, Mahmoud A, Elgendy IY, Richard Conti C. Ranolazine in Cardiac Arrhythmia. Clin Cardiol 2015; 39:170-8. [PMID: 26459200 DOI: 10.1002/clc.22476] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 08/20/2015] [Accepted: 08/25/2015] [Indexed: 12/19/2022] Open
Abstract
Ranolazine utilization in the management of refractory angina has been established by multiple randomized clinical studies. However, there is growing evidence showing an evolving role in the field of cardiac arrhythmias. Multiple experimental and clinical studies have evaluated the role of ranolazine in prevention and management of atrial fibrillation, with ongoing studies on its role in ventricular arrhythmias. In this review, we will discuss the pharmacological, experimental, and clinical evidence behind ranolazine use in the management of various cardiac arrhythmias.
Collapse
Affiliation(s)
- Marwan Saad
- Department of Medicine, Seton Hall University School of Health and Medical Sciences, Trinitas Regional Medical Center, Elizabeth, New Jersey
| | - Ahmed Mahmoud
- Department of Medicine, University of Florida, Gainesville, Florida
| | - Islam Y Elgendy
- Division of Cardiovascular Medicine, University of Florida, Gainesville, Florida
| | - C Richard Conti
- Division of Cardiovascular Medicine, University of Florida, Gainesville, Florida
| |
Collapse
|
34
|
Abstract
Despite improvements in the therapy of underlying heart disease, sudden cardiac death is a major cause of death worldwide. Disturbed Na and Ca handling is known to be a major predisposing factor for life-threatening tachyarrhythmias. In cardiomyocytes, many ion channels and transporters, including voltage-gated Na and Ca channels, cardiac ryanodine receptors, Na/Ca-exchanger, and SR Ca-ATPase are involved in this regulation. We have learned a lot about the pathophysiological relevance of disturbed ion channel function from monogenetic disorders. Changes in the gating of a single ion channel and the activity of an ion pump suffice to dramatically increase the propensity for arrhythmias even in structurally normal hearts. Nevertheless, patients with heart failure with acquired dysfunction in many ion channels and transporters exhibit profound dysregulation of Na and Ca handling and Ca/calmodulin-dependent protein kinase and are especially prone to arrhythmias. A deeper understanding of the underlying arrhythmic principles is mandatory if we are to improve their outcome. This review addresses basic tachyarrhythmic mechanisms, the underlying ionic mechanisms and the consequences for ion homeostasis, and the situation in complex diseases like heart failure.
Collapse
Affiliation(s)
- Stefan Wagner
- From the Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany (S.W., L.S.M.); and Department of Pharmacology, University of California, Davis, CA (D.M.B.)
| | - Lars S Maier
- From the Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany (S.W., L.S.M.); and Department of Pharmacology, University of California, Davis, CA (D.M.B.).
| | - Donald M Bers
- From the Department of Internal Medicine II, University Hospital Regensburg, Regensburg, Germany (S.W., L.S.M.); and Department of Pharmacology, University of California, Davis, CA (D.M.B.)
| |
Collapse
|
35
|
Glynn P, Musa H, Wu X, Unudurthi SD, Little S, Qian L, Wright PJ, Radwanski PB, Gyorke S, Mohler PJ, Hund TJ. Voltage-Gated Sodium Channel Phosphorylation at Ser571 Regulates Late Current, Arrhythmia, and Cardiac Function In Vivo. Circulation 2015; 132:567-77. [PMID: 26187182 DOI: 10.1161/circulationaha.114.015218] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 06/12/2015] [Indexed: 12/19/2022]
Abstract
BACKGROUND Voltage-gated Na(+) channels (Nav) are essential for myocyte membrane excitability and cardiac function. Nav current (INa) is a large-amplitude, short-duration spike generated by rapid channel activation followed immediately by inactivation. However, even under normal conditions, a small late component of INa (INa,L) persists because of incomplete/failed inactivation of a subpopulation of channels. Notably, INa,L is directly linked with both congenital and acquired disease states. The multifunctional Ca(2+)/calmodulin-dependent kinase II (CaMKII) has been identified as an important activator of INa,L in disease. Several potential CaMKII phosphorylation sites have been discovered, including Ser571 in the Nav1.5 DI-DII linker, but the molecular mechanism underlying CaMKII-dependent regulation of INa,L in vivo remains unknown. METHODS AND RESULTS To determine the in vivo role of Ser571, 2 Scn5a knock-in mouse models were generated expressing either: (1) Nav1.5 with a phosphomimetic mutation at Ser571 (S571E), or (2) Nav1.5 with the phosphorylation site ablated (S571A). Electrophysiology studies revealed that Ser571 regulates INa,L but not other channel properties previously linked to CaMKII. Ser571-mediated increases in INa,L promote abnormal repolarization and intracellular Ca(2+) handling and increase susceptibility to arrhythmia at the cellular and animal level. Importantly, Ser571 is required for maladaptive remodeling and arrhythmias in response to pressure overload. CONCLUSIONS Our data provide the first in vivo evidence for the molecular mechanism underlying CaMKII activation of the pathogenic INa,L. Relevant for improved rational design of potential therapies, our findings demonstrate that Ser571-dependent regulation of Nav1.5 specifically tunes INa,L without altering critical physiological components of the current.
Collapse
Affiliation(s)
- Patric Glynn
- From Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (P.G., H.M., X.W., S.D.U., S.L., L.Q., P.J.W., P.B.R., S.G., P.J.M., T.J.H.); Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus (P.G., X.W., S.D.U., L.Q., T.J.H.); Departments of Physiology & Cell Biology (H.M., S.L., P.J.W., P.B.R., S.G., P.J.M.) and Internal Medicine (P.J.M., T.J.H.), The Ohio State University Wexner Medical Center, Columbus; and Division of Pharmacy Practice and Administration, College of Pharmacy, The Ohio State University, Columbus (P.B.R.)
| | - Hassan Musa
- From Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (P.G., H.M., X.W., S.D.U., S.L., L.Q., P.J.W., P.B.R., S.G., P.J.M., T.J.H.); Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus (P.G., X.W., S.D.U., L.Q., T.J.H.); Departments of Physiology & Cell Biology (H.M., S.L., P.J.W., P.B.R., S.G., P.J.M.) and Internal Medicine (P.J.M., T.J.H.), The Ohio State University Wexner Medical Center, Columbus; and Division of Pharmacy Practice and Administration, College of Pharmacy, The Ohio State University, Columbus (P.B.R.)
| | - Xiangqiong Wu
- From Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (P.G., H.M., X.W., S.D.U., S.L., L.Q., P.J.W., P.B.R., S.G., P.J.M., T.J.H.); Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus (P.G., X.W., S.D.U., L.Q., T.J.H.); Departments of Physiology & Cell Biology (H.M., S.L., P.J.W., P.B.R., S.G., P.J.M.) and Internal Medicine (P.J.M., T.J.H.), The Ohio State University Wexner Medical Center, Columbus; and Division of Pharmacy Practice and Administration, College of Pharmacy, The Ohio State University, Columbus (P.B.R.)
| | - Sathya D Unudurthi
- From Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (P.G., H.M., X.W., S.D.U., S.L., L.Q., P.J.W., P.B.R., S.G., P.J.M., T.J.H.); Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus (P.G., X.W., S.D.U., L.Q., T.J.H.); Departments of Physiology & Cell Biology (H.M., S.L., P.J.W., P.B.R., S.G., P.J.M.) and Internal Medicine (P.J.M., T.J.H.), The Ohio State University Wexner Medical Center, Columbus; and Division of Pharmacy Practice and Administration, College of Pharmacy, The Ohio State University, Columbus (P.B.R.)
| | - Sean Little
- From Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (P.G., H.M., X.W., S.D.U., S.L., L.Q., P.J.W., P.B.R., S.G., P.J.M., T.J.H.); Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus (P.G., X.W., S.D.U., L.Q., T.J.H.); Departments of Physiology & Cell Biology (H.M., S.L., P.J.W., P.B.R., S.G., P.J.M.) and Internal Medicine (P.J.M., T.J.H.), The Ohio State University Wexner Medical Center, Columbus; and Division of Pharmacy Practice and Administration, College of Pharmacy, The Ohio State University, Columbus (P.B.R.)
| | - Lan Qian
- From Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (P.G., H.M., X.W., S.D.U., S.L., L.Q., P.J.W., P.B.R., S.G., P.J.M., T.J.H.); Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus (P.G., X.W., S.D.U., L.Q., T.J.H.); Departments of Physiology & Cell Biology (H.M., S.L., P.J.W., P.B.R., S.G., P.J.M.) and Internal Medicine (P.J.M., T.J.H.), The Ohio State University Wexner Medical Center, Columbus; and Division of Pharmacy Practice and Administration, College of Pharmacy, The Ohio State University, Columbus (P.B.R.)
| | - Patrick J Wright
- From Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (P.G., H.M., X.W., S.D.U., S.L., L.Q., P.J.W., P.B.R., S.G., P.J.M., T.J.H.); Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus (P.G., X.W., S.D.U., L.Q., T.J.H.); Departments of Physiology & Cell Biology (H.M., S.L., P.J.W., P.B.R., S.G., P.J.M.) and Internal Medicine (P.J.M., T.J.H.), The Ohio State University Wexner Medical Center, Columbus; and Division of Pharmacy Practice and Administration, College of Pharmacy, The Ohio State University, Columbus (P.B.R.)
| | - Przemyslaw B Radwanski
- From Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (P.G., H.M., X.W., S.D.U., S.L., L.Q., P.J.W., P.B.R., S.G., P.J.M., T.J.H.); Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus (P.G., X.W., S.D.U., L.Q., T.J.H.); Departments of Physiology & Cell Biology (H.M., S.L., P.J.W., P.B.R., S.G., P.J.M.) and Internal Medicine (P.J.M., T.J.H.), The Ohio State University Wexner Medical Center, Columbus; and Division of Pharmacy Practice and Administration, College of Pharmacy, The Ohio State University, Columbus (P.B.R.)
| | - Sandor Gyorke
- From Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (P.G., H.M., X.W., S.D.U., S.L., L.Q., P.J.W., P.B.R., S.G., P.J.M., T.J.H.); Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus (P.G., X.W., S.D.U., L.Q., T.J.H.); Departments of Physiology & Cell Biology (H.M., S.L., P.J.W., P.B.R., S.G., P.J.M.) and Internal Medicine (P.J.M., T.J.H.), The Ohio State University Wexner Medical Center, Columbus; and Division of Pharmacy Practice and Administration, College of Pharmacy, The Ohio State University, Columbus (P.B.R.)
| | - Peter J Mohler
- From Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (P.G., H.M., X.W., S.D.U., S.L., L.Q., P.J.W., P.B.R., S.G., P.J.M., T.J.H.); Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus (P.G., X.W., S.D.U., L.Q., T.J.H.); Departments of Physiology & Cell Biology (H.M., S.L., P.J.W., P.B.R., S.G., P.J.M.) and Internal Medicine (P.J.M., T.J.H.), The Ohio State University Wexner Medical Center, Columbus; and Division of Pharmacy Practice and Administration, College of Pharmacy, The Ohio State University, Columbus (P.B.R.)
| | - Thomas J Hund
- From Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus (P.G., H.M., X.W., S.D.U., S.L., L.Q., P.J.W., P.B.R., S.G., P.J.M., T.J.H.); Department of Biomedical Engineering, College of Engineering, The Ohio State University, Columbus (P.G., X.W., S.D.U., L.Q., T.J.H.); Departments of Physiology & Cell Biology (H.M., S.L., P.J.W., P.B.R., S.G., P.J.M.) and Internal Medicine (P.J.M., T.J.H.), The Ohio State University Wexner Medical Center, Columbus; and Division of Pharmacy Practice and Administration, College of Pharmacy, The Ohio State University, Columbus (P.B.R.).
| |
Collapse
|
36
|
Gupta T, Khera S, Kolte D, Aronow WS, Iwai S. Antiarrhythmic properties of ranolazine: A review of the current evidence. Int J Cardiol 2015; 187:66-74. [PMID: 25828315 DOI: 10.1016/j.ijcard.2015.03.324] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 03/20/2015] [Indexed: 12/19/2022]
|
37
|
Trenor B, Gomis-Tena J, Cardona K, Romero L, Rajamani S, Belardinelli L, Giles WR, Saiz J. In silico assessment of drug safety in human heart applied to late sodium current blockers. Channels (Austin) 2015; 7:249-62. [PMID: 23696033 DOI: 10.4161/chan.24905] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Drug-induced action potential (AP) prolongation leading to Torsade de Pointes is a major concern for the development of anti-arrhythmic drugs. Nevertheless the development of improved anti-arrhythmic agents, some of which may block different channels, remains an important opportunity. Partial block of the late sodium current (I(NaL)) has emerged as a novel anti-arrhythmic mechanism. It can be effective in the settings of free radical challenge or hypoxia. In addition, this approach can attenuate pro-arrhythmic effects of blocking the rapid delayed rectifying K(+) current (I(Kr)). The main goal of our computational work was to develop an in-silico tool for preclinical anti-arrhythmic drug safety assessment, by illustrating the impact of I(Kr)/I(NaL) ratio of steady-state block of drug candidates on "torsadogenic" biomarkers. The O'Hara et al. AP model for human ventricular myocytes was used. Biomarkers for arrhythmic risk, i.e., AP duration, triangulation, reverse rate-dependence, transmural dispersion of repolarization and electrocardiogram QT intervals, were calculated using single myocyte and one-dimensional strand simulations. Predetermined amounts of block of I(NaL) and I(Kr) were evaluated. "Safety plots" were developed to illustrate the value of the specific biomarker for selected combinations of IC(50)s for I(Kr) and I(NaL) of potential drugs. The reference biomarkers at baseline changed depending on the "drug" specificity for these two ion channel targets. Ranolazine and GS967 (a novel potent inhibitor of I(NaL)) yielded a biomarker data set that is considered safe by standard regulatory criteria. This novel in-silico approach is useful for evaluating pro-arrhythmic potential of drugs and drug candidates in the human ventricle.
Collapse
|
38
|
Sossalla S, Wallisch N, Toischer K, Sohns C, Vollmann D, Seegers J, Lüthje L, Maier LS, Zabel M. Effects of ranolazine on torsades de pointes tachycardias in a healthy isolated rabbit heart model. Cardiovasc Ther 2015; 32:170-7. [PMID: 24785406 PMCID: PMC4285941 DOI: 10.1111/1755-5922.12078] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
PURPOSE Torsades de pointes (TdP) tachycardias are triggered, polymorphic ventricular arrhythmias arising from early afterdepolarizations (EADs) and increased dispersion of repolarization. Ranolazine is a new agent which reduces pathologically elevated late INa but also IKr . Aim of this study was to evaluate the effects of ranolazine in a validated isolated Langendorff-perfused rabbit heart model. METHODS TdP was reproducibly induced with d-sotalol (10(-4) mol/L) and low potassium (K) (1.0 mmol/L for 5 min, pacing at CL 1000 ms). In 10 hearts, ECG and 8 epi- and endocardial monophasic action potentials were recorded. Action potential duration (APD) was measured at 90% repolarization and dispersion defined as APD max-min. RESULTS D-sotalol prolonged APD90 and increased dispersion of APD90 , simultaneously causing EADs and induction of TdP. The combination of d-sotalol and two concentrations of ranolazine did not increase dispersion of ventricular APD90 as compared to vehicle. Ranolazine at 5 μmol/L did not cause additional induction of EADs and/or TdP but also did not significantly suppress arrhythmogenic triggers. The higher concentration of ranolazine (10 μmol/L) in combination with d-sotalol caused further prolongation of APD90 , at the same time reduction in APD90 dispersion. In parallel, the incidence of EADs was reduced and an antitorsadogenic effect was seen. CONCLUSIONS In the healthy isolated rabbit heart (where late INa is not elevated), ranolazine does not cause proarrhythmia but exerts antiarrhythmic effects in a dose-dependent manner against d-sotalol/low K-induced TdP. This finding-despite additional APD prolongation-supports the safety of a combined use of both drugs and merits clinical investigation.
Collapse
Affiliation(s)
- Samuel Sossalla
- Klinik für Kardiologie und Pneumologie/Herzzentrum, Georg-August-Universität Göttingen, Göttingen, Germany; DZHK (German Center for Cardiovascular Research), Göttingen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Horvath B, Bers DM. The late sodium current in heart failure: pathophysiology and clinical relevance. ESC Heart Fail 2014; 1:26-40. [PMID: 28834665 DOI: 10.1002/ehf2.12003] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Revised: 07/13/2014] [Accepted: 07/14/2014] [Indexed: 12/19/2022] Open
Abstract
Large and growing body of data suggest that an increased late sodium current (INa,late ) can have a significant pathophysiological role in heart failure and other heart diseases. The first goal of this article is to describe how INa,late functions under physiological circumstances. The second goal is to show the wide range of cellular mechanisms that can increase INa,late in cardiac disease, and also to describe how the up-regulated INa,late contributes to the pathophysiology of heart failure. The final section of the article discusses the possible use of INa,late -modifying drugs in heart failure, on the basis of experimental and preclinical data.
Collapse
Affiliation(s)
- Balazs Horvath
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Faculty of Pharmacy, University of Debrecen, Debrecen, Hungary
| | - Donald M Bers
- Department of Pharmacology, School of Medicine, University of California, Davis, CA, USA
| |
Collapse
|
40
|
Ma J, Song Y, Shryock JC, Hu L, Wang W, Yan X, Zhang P, Belardinelli L. Ranolazine Attenuates Hypoxia- and Hydrogen Peroxide-induced Increases in Sodium Channel Late Openings in Ventricular Myocytes. J Cardiovasc Pharmacol 2014; 64:60-8. [DOI: 10.1097/fjc.0000000000000090] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
41
|
Abstract
Mortality rates attributable to coronary heart disease have declined in recent years, possibly related to changes in clinical presentation patterns and use of proven secondary prevention strategies. Chronic stable angina (CSA) remains prevalent, and the goal of treatment is control of symptoms and reduction in cardiovascular events. Ranolazine is a selective inhibitor of the late sodium current in myocytes with anti-ischemic and metabolic properties. It was approved by the US Food and Drug Administration in 2006 for use in patients with CSA. Multiple, randomized, placebo-controlled trials have shown that ranolazine improves functional capacity and decreases anginal episodes in CSA patients, despite a lack of a significant hemodynamic effect. Ranolazine did not improve cardiovascular mortality or affect incidence of myocardial infarction in the MERLIN (Metabolic Efficiency with Ranolazine for Less Ischemia in Non-ST-Elevation Acute Coronary Syndrome)-TIMI (Thrombolysis In Myocardial Infarction) 36 trial, but significantly decreased the incidence of recurrent angina. More recently, ranolazine has been shown to have beneficial and potent antiarrhythmic effects, both on supraventricular and ventricular tachyarrhythmias, largely due to its inhibition of the late sodium current. Randomized controlled trials testing these effects are underway. Lastly, ranolazine appears to be cost-effective due to its ability to decrease angina-related hospitalizations and improve quality of life.
Collapse
Affiliation(s)
- J Nicolás Codolosa
- Einstein Center for Heart and Vascular Health, Einstein Medical Center, Philadelphia, PA, USA
| | - Subroto Acharjee
- Einstein Center for Heart and Vascular Health, Einstein Medical Center, Philadelphia, PA, USA
| | - Vincent M Figueredo
- Einstein Center for Heart and Vascular Health, Einstein Medical Center, Philadelphia, PA, USA
- Jefferson Medical College, Philadelphia, PA, USA
| |
Collapse
|
42
|
Selective Inhibition of the Late Sodium Current has No Adverse Effect on Electrophysiological or Contractile Function of the Normal Heart. J Cardiovasc Pharmacol 2014; 63:512-9. [DOI: 10.1097/fjc.0000000000000075] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
43
|
Driessen HE, Bourgonje VJA, van Veen TAB, Vos MA. New antiarrhythmic targets to control intracellular calcium handling. Neth Heart J 2014; 22:198-213. [PMID: 24733689 PMCID: PMC4016334 DOI: 10.1007/s12471-014-0549-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Sudden cardiac death due to ventricular arrhythmias is a major problem. Drug therapies to prevent SCD do not provide satisfying results, leading to the demand for new antiarrhythmic strategies. New targets include Ca2+/Calmodulin-dependent protein kinase II (CaMKII), the Na/Ca exchanger (NCX), the Ryanodine receptor (RyR, and its associated protein FKBP12.6 (Calstabin)) and the late component of the sodium current (INa-Late), all related to intracellular calcium (Ca2+) handling. In this review, drugs interfering with these targets (SEA-0400, K201, KN-93, W7, ranolazine, sophocarpine, and GS-967) are evaluated and their future as clinical compounds is considered. These new targets prove to be interesting; however more insight into long-term drug effects is necessary before clinical applicability becomes reality.
Collapse
Affiliation(s)
- H E Driessen
- Department of Medical Physiology, Division of Heart & Lungs, University Medical Center Utrecht, Yalelaan 50, 3584 CM, Utrecht, the Netherlands,
| | | | | | | |
Collapse
|
44
|
Abstract
Myocardial ischemia is a metabolic problem involving reduced delivery of oxygen to cardiac mitochondria, resulting in less ATP formation, acceleration of glycolysis and production of lactate and H+ by the cell. Traditional therapies for ischemia aim at restoring the balance between mitochondrial ATP production and breakdown by reducing the need for ATP via suppression of heart rate, blood pressure and cardiac contractility, or by increasing oxygen delivery via increased myocardial blood flow. Despite optimal treatment with traditional hemodynamically oriented drugs (beta-adrenergic receptor antagonist, Ca2+ channel antagonist and nitrates), many patients continue to suffer from angina. Thus, there is a need for anti-anginal drugs that act directly on cardiomyocytes to lessen the metabolic abnormalities induced by ischemia and reduce the symptoms (chest pain and exercise intolerance). Ranolazine has been demonstrated to improve exercise time to angina or 1 mm of ST-segment depression in a manner similar to currently approved drugs, but without any significant effects on heart rate or blood pressure at rest or during exercise. In two Phase III trials, ranolazine improved exercise tolerance and reduced the frequency of angina attacks in chronic severe angina patients when administered either as monotherapy or on a background of atenolol, amlodinine or diltiazem. At present, ranolazine is under review for US Food and Drug Administration approval and, if approved, it will represent the first drug of its class in the USA.
Collapse
Affiliation(s)
- William C Stanley
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH 44106-4970, USA.
| |
Collapse
|
45
|
Reffelmann T, Kloner RA. Ranolazine: an anti-anginal drug with further therapeutic potential. Expert Rev Cardiovasc Ther 2014; 8:319-29. [DOI: 10.1586/erc.09.178] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
46
|
Vaishnav A, Vaishnav A, Lokhandwala Y. Refractory atrial fibrillation effectively treated with ranolazine. Indian Heart J 2014; 66:115-8. [PMID: 24581108 PMCID: PMC3946441 DOI: 10.1016/j.ihj.2013.12.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 08/11/2013] [Accepted: 12/05/2013] [Indexed: 12/19/2022] Open
Abstract
Atrial fibrillation is the most common sustained cardiac arrhythmia which is often troublesome to manage. Currently, rhythm and rate control medications are the mainstays of therapy. In 2 amiodarone-refractory highly symptomatic patients, an innovative approach using ranolazine, which selectively acts on Na+ channels and delays atrial depolarization, was tried successfully.
Collapse
Affiliation(s)
- Aditi Vaishnav
- Medical Student, Dr. D.Y. Patil Medical College, Navi Mumbai, India
| | - Avani Vaishnav
- Medical Student, Dr. D.Y. Patil Medical College, Navi Mumbai, India
| | | |
Collapse
|
47
|
Wang XJ, Wang LL, Fu C, Zhang PH, Wu Y, Ma JH. Ranolazine Attenuates the Enhanced Reverse Na+-Ca2+ Exchange Current via Inhibiting Hypoxia-Increased Late Sodium Current in Ventricular Myocytes. J Pharmacol Sci 2014; 124:365-73. [DOI: 10.1254/jphs.13202fp] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
48
|
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
|
49
|
Coppini R, Ferrantini C, Mazzoni L, Sartiani L, Olivotto I, Poggesi C, Cerbai E, Mugelli A. Regulation of intracellular Na(+) in health and disease: pathophysiological mechanisms and implications for treatment. Glob Cardiol Sci Pract 2013; 2013:222-42. [PMID: 24689024 PMCID: PMC3963757 DOI: 10.5339/gcsp.2013.30] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 09/01/2013] [Indexed: 12/19/2022] Open
Abstract
Transmembrane sodium (Na+) fluxes and intracellular sodium homeostasis are central players in the physiology of the cardiac myocyte, since they are crucial for both cell excitability and for the regulation of the intracellular calcium concentration. Furthermore, Na+ fluxes across the membrane of mitochondria affect the concentration of protons and calcium in the matrix, regulating mitochondrial function. In this review we first analyze the main molecular determinants of sodium fluxes across the sarcolemma and the mitochondrial membrane and describe their role in the physiology of the healthy myocyte. In particular we focus on the interplay between intracellular Ca2+ and Na+. A large part of the review is dedicated to discuss the changes of Na+ fluxes and intracellular Na+ concentration([Na+]i) occurring in cardiac disease; we specifically focus on heart failure and hypertrophic cardiomyopathy, where increased intracellular [Na+]i is an established determinant of myocardial dysfunction. We review experimental evidence attributing the increase of [Na+]i to either decreased Na+ efflux (e.g. via the Na+/K+ pump) or increased Na+ influx into the myocyte (e.g. via Na+ channels). In particular, we focus on the role of the “late sodium current” (INaL), a sustained component of the fast Na+ current of cardiac myocytes, which is abnormally enhanced in cardiac diseases and contributes to both electrical and contractile dysfunction. We analyze the pathophysiological role of INaL enhancement in heart failure and hypertrophic cardiomyopathy and the consequences of its pharmacological modulation, highlighting the clinical implications. The central role of Na+ fluxes and intracellular Na+ physiology and pathophysiology of cardiac myocytes has been highlighted by a large number of recent works. The possibility of modulating Na+ inward fluxes and [Na+]i with specific INaL inhibitors, such as ranolazine, has made Na+a novel suitable target for cardiac therapy, potentially capable of addressing arrhythmogenesis and diastolic dysfunction in severe conditions such as heart failure and hypertrophic cardiomyopathy.
Collapse
Affiliation(s)
- Raffaele Coppini
- Department NeuroFarBa, Division of Pharmacology, University of Florence, Italy
| | - Cecilia Ferrantini
- Department of Clinical and Experimental Medicine, division of Physiology, University of Florence, Italy
| | - Luca Mazzoni
- Department NeuroFarBa, Division of Pharmacology, University of Florence, Italy
| | - Laura Sartiani
- Department NeuroFarBa, Division of Pharmacology, University of Florence, Italy
| | - Iacopo Olivotto
- Referral Center for Cardiomyopathies, Careggi University Hospital, Florence, Italy
| | - Corrado Poggesi
- Department of Clinical and Experimental Medicine, division of Physiology, University of Florence, Italy
| | - Elisabetta Cerbai
- Department NeuroFarBa, Division of Pharmacology, University of Florence, Italy
| | - Alessandro Mugelli
- Department NeuroFarBa, Division of Pharmacology, University of Florence, Italy
| |
Collapse
|
50
|
Toischer K, Hartmann N, Wagner S, Fischer TH, Herting J, Danner BC, Sag CM, Hund TJ, Mohler PJ, Belardinelli L, Hasenfuss G, Maier LS, Sossalla S. Role of late sodium current as a potential arrhythmogenic mechanism in the progression of pressure-induced heart disease. J Mol Cell Cardiol 2013; 61:111-22. [PMID: 23570977 PMCID: PMC3720777 DOI: 10.1016/j.yjmcc.2013.03.021] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Revised: 03/08/2013] [Accepted: 03/29/2013] [Indexed: 12/19/2022]
Abstract
The aim of the study was to determine the characteristics of the late Na current (INaL) and its arrhythmogenic potential in the progression of pressure-induced heart disease. Transverse aortic constriction (TAC) was used to induce pressure overload in mice. After one week the hearts developed isolated hypertrophy with preserved systolic contractility. In patch-clamp experiments both, INaL and the action potential duration (APD90) were unchanged. In contrast, after five weeks animals developed heart failure with prolonged APDs and slowed INaL decay time which could be normalized by addition of the INaL inhibitor ranolazine (Ran) or by the Ca/calmodulin-dependent protein kinase II (CaMKII) inhibitor AIP. Accordingly the APD90 could be significantly abbreviated by Ran, tetrodotoxin and the CaMKII inhibitor AIP. Isoproterenol increased the number of delayed afterdepolarizations (DAD) in myocytes from failing but not sham hearts. Application of either Ran or AIP prevented the occurrence of DADs. Moreover, the incidence of triggered activity was significantly increased in TAC myocytes and was largely prevented by Ran and AIP. Western blot analyses indicate that increased CaMKII activity and a hyperphosphorylation of the Nav1.5 at the CaMKII phosphorylation site (Ser571) paralleled our functional observations five weeks after TAC surgery. In pressure overload-induced heart failure a CaMKII-dependent augmentation of INaL plays a crucial role in the AP prolongation and generation of cellular arrhythmogenic triggers, which cannot be found in early and still compensated hypertrophy. Inhibition of INaL and CaMKII exerts potent antiarrhythmic effects and might therefore be of potential therapeutic interest. This article is part of a Special Issue entitled "Na(+) Regulation in Cardiac Myocytes".
Collapse
Affiliation(s)
- Karl Toischer
- Abt. Kardiologie und Pneumologie / Herzzentrum, Georg-August-Universität Göttingen, Germany
| | - Nico Hartmann
- Abt. Kardiologie und Pneumologie / Herzzentrum, Georg-August-Universität Göttingen, Germany
| | - Stefan Wagner
- Abt. Kardiologie und Pneumologie / Herzzentrum, Georg-August-Universität Göttingen, Germany
| | - Thomas H. Fischer
- Abt. Kardiologie und Pneumologie / Herzzentrum, Georg-August-Universität Göttingen, Germany
| | - Jonas Herting
- Abt. Kardiologie und Pneumologie / Herzzentrum, Georg-August-Universität Göttingen, Germany
| | - Bernhard C. Danner
- Abt. Herzund Thoraxchirurgie, Georg-August-Universität Göttingen, Germany
| | - Can M. Sag
- Abt. Kardiologie und Pneumologie / Herzzentrum, Georg-August-Universität Göttingen, Germany
| | - Thomas J. Hund
- Dorothy M. Davis Heart and Lung Research Institute, Dept. of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Peter J. Mohler
- Dorothy M. Davis Heart and Lung Research Institute, Dept. of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH
| | | | - Gerd Hasenfuss
- Abt. Kardiologie und Pneumologie / Herzzentrum, Georg-August-Universität Göttingen, Germany
| | - Lars S. Maier
- Abt. Kardiologie und Pneumologie / Herzzentrum, Georg-August-Universität Göttingen, Germany
| | - Samuel Sossalla
- Abt. Kardiologie und Pneumologie / Herzzentrum, Georg-August-Universität Göttingen, Germany
| |
Collapse
|