Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Alexander Burashnikov is active.

Publication


Featured researches published by Alexander Burashnikov.


Circulation | 2004

Electrophysiological Effects of Ranolazine, a Novel Antianginal Agent With Antiarrhythmic Properties

Charles Antzelevitch; Luiz Belardinelli; Andrew C. Zygmunt; Alexander Burashnikov; José M. Di Diego; Jeffrey M. Fish; Jonathan M. Cordeiro; George P. Thomas

Background—Ranolazine is a novel antianginal agent capable of producing antiischemic effects at plasma concentrations of 2 to 6 &mgr;mol/L without reducing heart rate or blood pressure. The present study examines its electrophysiological effects in isolated canine ventricular myocytes, tissues, and arterially perfused left ventricular wedge preparations. Methods and Results—Transmembrane action potentials (APs) from epicardial and midmyocardial (M) regions and a pseudo-ECG were recorded simultaneously from wedge preparations. APs were also recorded from epicardial and M tissues. Whole-cell currents were recorded from epicardial and M myocytes. Ranolazine inhibited IKr (IC50=11.5 &mgr;mol/L), late INa, late ICa, peak ICa, and INa-Ca (IC50=5.9, 50, 296, and 91 &mgr;mol/L, respectively) and IKs (17% at 30 &mgr;mol/L), but caused little or no inhibition of Ito or IK1. In tissues and wedge preparations, ranolazine produced a concentration-dependent prolongation of AP duration of epicardial but abbreviation of that of M cells, leading to reduction or no change in transmural dispersion of repolarization (TDR). At [K+]o=4 mmol/L, 10 &mgr;mol/L ranolazine prolonged QT interval by 20 ms but did not increase TDR. Extrasystolic activity and spontaneous torsade de pointes (TdP) were never observed, and stimulation-induced TdP could not be induced at any concentration of ranolazine, either in normal or low [K+]o. Ranolazine (5 to 20 &mgr;mol/L) suppressed early afterdepolarizations (EADs) and reduced the increase in TDR induced by the selective IKr blocker d-sotalol. Conclusions—Ranolazine produces ion channel effects similar to those observed after chronic amiodarone (reduced IKr, IKs, late INa, and ICa). The actions of ranolazine to suppress EADs and reduce TDR suggest that, in addition to its antianginal actions, the drug may possess antiarrhythmic activity.


Journal of Cardiovascular Electrophysiology | 1999

The M cell: its contribution to the ECG and to normal and abnormal electrical function of the heart.

Charles Antzelevitch; Wataru Shimizu; Gan-Xin Yan; Serge Sicouri; Jacques Weissenburger M.D.; Vladislav V. Nesterenko; Alexander Burashnikov; José M Di Diego; Jeferey Saffitz M.D.; George P. Thomas

Characteristics of the M Cell. The discovery and characterization of the M cell, a unique cell type residing in the deep layers of the ventricular myocardium, has opened a new door in our understanding of the electrophysiology and pharmacology of the heart in both health and disease. The hallmark of the M cell is the ability of its action potential to prolong much more than that of other ventricular myocardial cells in response to a slowing of rate and/or in response to agents that act to prolong action potential duration. Our goal in this review is to provide a comprehensive characterization of the M cell, its contribution to transmural heterogeneity, and its role in the normal electrical function of the heart, in the inscription of the ECG (particularly the T wave), and in the development of QT dispersion, T wave alternans, long QT intervals, and cardiac arrhythmias, such as torsades de pointes. Our secondary goal is to address the controversy that has arisen relative to the functional importance of the M cell in the normal heart. The controversy derives largely from the failure of some investigators to demonstrate transmural heterogeneity of repolarization in the dog in vivo under control conditions and after administration of quinidine. The inability to demonstrate transmural heterogeneity under these conditions may he due to the use of bipolar recording techniques that, in our experience, seriously underestimate transmural dispersion of repolarization (TDK). The use of sodium pentobarhital and α‐chloralose as anesthesia also is problematic, because these agents reduce or eliminate TDR by affecting a variety of ion channel currents. Finally, attempts to amplify transmural dispersion of repolarization with an agent such as quinidine must take into account that relatively high concentrations can result in effects opposite to those desired due to drug inhibition of multiple ion channels. These observations may explain the inability of earlier studies to detect the M cell.


Circulation | 2007

Atrium-Selective Sodium Channel Block as a Strategy for Suppression of Atrial Fibrillation: Differences in Sodium Channel Inactivation Between Atria and Ventricles and the Role of Ranolazine

Alexander Burashnikov; José M. Di Diego; Andrew C. Zygmunt; Luiz Belardinelli; Charles Antzelevitch

Background— The development of selective atrial antiarrhythmic agents is a current strategy for suppression of atrial fibrillation (AF). Methods and Results— Whole-cell patch clamp techniques were used to evaluate inactivation of peak sodium channel current (INa) in myocytes isolated from canine atria and ventricles. The electrophysiological effects of therapeutic concentrations of ranolazine (1 to 10 &mgr;mol/L) and lidocaine (2.1 to 21 &mgr;mol/L) were evaluated in canine isolated coronary-perfused atrial and ventricular preparations. Half-inactivation voltage of INa was ≈15 mV more negative in atrial versus ventricular cells under control conditions; this difference increased after exposure to ranolazine. Ranolazine produced a marked use-dependent depression of sodium channel parameters, including the maximum rate of rise of the action potential upstroke, conduction velocity, and diastolic threshold of excitation, and induced postrepolarization refractoriness in atria but not in ventricles. Lidocaine also preferentially suppressed these parameters in atria versus ventricles, but to a much lesser extent than ranolazine. Ranolazine produced a prolongation of action potential duration (APD90) in atria, no effect on APD90 in ventricular myocardium, and an abbreviation of APD90 in Purkinje fibers. Lidocaine abbreviated both atrial and ventricular APD90. Ranolazine was more effective than lidocaine in terminating persistent AF and in preventing the induction of AF. Conclusions— Our study demonstrates important differences in the inactivation characteristics of atrial versus ventricular sodium channels and a striking atrial selectivity for the action of ranolazine to produce use-dependent block of sodium channels, leading to suppression of AF. Our results point to atrium-selective sodium channel block as a novel strategy for the management of AF.


Circulation | 2003

Reinduction of Atrial Fibrillation Immediately After Termination of the Arrhythmia Is Mediated by Late Phase 3 Early Afterdepolarization–Induced Triggered Activity

Alexander Burashnikov; Charles Antzelevitch

Background—Atrial fibrillation (AF) at times recurs immediately after termination of the arrhythmia. The mechanism(s) responsible for the extrasystole that reinduces AF is largely unknown. We hypothesized that abbreviation of action potential duration (APD) would permit very rapid rates of excitation, known to induce intracellular calcium loading, which in turn could promote delayed and/or early afterdepolarizations (EADs). Methods and Results—Acetylcholine (ACh, 1 &mgr;mol/L) was used to abbreviate atrial APD and permit rapid-pacing induction of AF in isolated coronary-perfused canine right atria. Transmembrane action potentials, pseudo-ECG, and tension development were recorded. AF or rapid pacing was associated with an increase in tonic tension. Termination of AF or rapid pacing (cycle length, 150 to 80 ms) resulted in a dramatic rise of phasic tension, prolongation of repolarization of the initial beats at the regular rate (cycle length, 700 ms), and the development of late phase 3 EADs and extrasystoles. These extrasystoles initiated AF in 15 cases (involving 9 right atria) within the first 11 seconds after termination of AF or rapid pacing. This novel EAD mechanism is observed only in association with marked APD abbreviation. The calcium channel blocker nifedipine reduced, and the sarcoplasmic reticulum calcium release blocker ryanodine eliminated, the post–rapid pacing–induced increase in phasic tension, late phase 3 EADs, and extrasystoles that initiate AF. Conclusions—These data suggest that calcium overload conditions present after termination of vagally mediated AF contribute to the development of late phase 3 EAD-induced triggered activity and that this mechanism may be responsible for the extrasystolic activity that reinitiates AF.


Heart Rhythm | 2011

Electrophysiologic basis for the antiarrhythmic actions of ranolazine

Charles Antzelevitch; Alexander Burashnikov; Serge Sicouri; Luiz Belardinelli

Ranolazine is a Food and Drug Administration-approved antianginal agent. Experimental and clinical studies have shown that ranolazine has antiarrhythmic effects in both ventricles and atria. In the ventricles, ranolazine can suppress arrhythmias associated with acute coronary syndrome, long QT syndrome, heart failure, ischemia, and reperfusion. In atria, ranolazine effectively suppresses atrial tachyarrhythmias and atrial fibrillation (AF). Recent studies have shown that the drug may be effective and safe in suppressing AF when used as a pill-in-the pocket approach, even in patients with structurally compromised hearts, warranting further study. The principal mechanism underlying ranolazines antiarrhythmic actions is thought to be primarily via inhibition of late I(Na) in the ventricles and via use-dependent inhibition of peak I(Na) and I(Kr) in the atria. Short- and long-term safety of ranolazine has been demonstrated in the clinic, even in patients with structural heart disease. This review summarizes the available data regarding the electrophysiologic actions and antiarrhythmic properties of ranolazine in preclinical and clinical studies.


Journal of the American College of Cardiology | 2010

Synergistic Effect of the Combination of Ranolazine and Dronedarone to Suppress Atrial Fibrillation

Alexander Burashnikov; Serge Sicouri; José M. Di Diego; Luiz Belardinelli; Charles Antzelevitch

OBJECTIVES The aim of this study was to evaluate the effectiveness of a combination of dronedarone and ranolazine in suppression of atrial fibrillation (AF). BACKGROUND Safe and effective pharmacological management of AF remains one of the greatest unmet medical needs. METHODS The electrophysiological effects of dronedarone (10 μmol/l) and a relatively low concentration of ranolazine (5 μmol/l) separately and in combination were evaluated in canine isolated coronary-perfused right and left atrial and left ventricular preparations as well as in pulmonary vein preparations. RESULTS Ranolazine caused moderate atrial-selective prolongation of action potential duration and atrial-selective depression of sodium channel-mediated parameters, including maximal rate of rise of the action potential upstroke, leading to the development of atrial-specific post-repolarization refractoriness. Dronedarone caused little or no change in electrophysiological parameters in both atrial and ventricular preparations. The combination of dronedarone and ranolazine caused little change in action potential duration in either chamber but induced potent use-dependent atrial-selective depression of the sodium channel-mediated parameters (maximal rate of rise of the action potential upstroke, diastolic threshold of excitation, and the shortest cycle length permitting a 1:1 response) and considerable post-repolarization refractoriness. Separately, dronedarone or a low concentration of ranolazine prevented the induction of AF in 17% and 29% of preparations, respectively. In combination, the 2 drugs suppressed AF and triggered activity and prevented the induction of AF in 9 of 10 preparations (90%). CONCLUSIONS Low concentrations of ranolazine and dronedarone produce relatively weak electrophysiological effects and weak suppression of AF when used separately but when combined exert potent synergistic effects, resulting in atrial-selective depression of sodium channel-dependent parameters and effective suppression of AF.


Journal of Cardiovascular Pharmacology and Therapeutics | 2004

Electrophysiologic properties and antiarrhythmic actions of a novel antianginal agent.

Charles Antzelevitch; Luiz Belardinelli; Lin Wu; Heather Fraser; Andrew C. Zygmunt; Alexander Burashnikov; José M. Di Diego; Jeffrey M. Fish; Jonathan M. Cordeiro; Robert J. Goodrow; Fabiana Scomik; Guillermo J. Pérez

Ranolazine is a novel antianginal agent capable of producing anti-ischemic effects at plasma concentrations of 2 to 6 μM without a significant reduction of heart rate or blood pressure. This review summarizes the electrophysiologic properties of ranolazine. Ranolazine significantly blocks IKr (IC50 = 12 μM), late INa, late ICa, peak ICa, INa-Ca (IC50 = 5.9, 50, 296, and 91 μM, respectively) and IKs (17% at 30,uM), but causes little or no inhibition of Ito or IKl. In left ventricular tissue and wedge preparations, ranolazine produces a concentration-dependent prolongation of action potential duration (APD) in epicardium, but abbreviation of APD of M cells, leading to either no change or a reduction in transmural dispersion of repolarization (TDR). The result is a modest prolongation of the QT interval. Prolongation of APD and QT by ranolazine is fundamentally different from that of other drugs that block IKr and induce torsade de pointes in that APD prolongation is rate-independent (ie, does not display reverse rate-dependent prolongation of APD) and is not associated with early afterdepolarizations, triggered activity, increased spatial dispersion of repolarization, or polymorphic ventricular tachycardia. Torsade de pointes arrhythmias were not observed spontaneously nor could they be induced with programmed electrical stimulation in the presence of ranolazine at concentrations as high as 100 μM. Indeed, ranolazine was found to possess significant antiarrhythmic activity, acting to suppress the arrhythmogenic effects of other QT-prolonging drugs. Ranolazine produces ion channel effects similar to those observed after chronic exposure to amiodarone (reduced late INa, IKs, IKS, and ICa). Ranolazines actions to reduce TDR and suppress early afterdepolarization suggest that in addition to its anti-anginal actions, the drug possesses antiarrhythmic activity.


Journal of Cardiovascular Electrophysiology | 1998

Acceleration‐Induced Action Potential Prolongation and Early Afterdepolarizations

Alexander Burashnikov; Charles Antzelevitch

Acceleration‐Induced Early Afterdepolarizations. Introduction: Precipitation of torsades de pointes (TdP) has been shown to he associated with acceleration of heart rate in both experimental and clinical studies. To gain insight into the cellular mechanism(s) responsible for the initiation of acceleration‐induced TdP, we studied the effect of acceleration of pacing rate in canine left ventricular epicardial, M region, endocardial, and Purkinje fiber preparations pretreated with E‐4031, an IKr blocker known to induce the long QT syndrome and TdP.


Circulation | 2005

Cellular Mechanisms Underlying the Development of Catecholaminergic Ventricular Tachycardia

Gi-Byoung Nam; Alexander Burashnikov; Charles Antzelevitch

Background—Mutations in the ryanodine 2 receptor (RyR2) gene have been identified in patients with catecholaminergic polymorphic ventricular tachycardia. We examined the cellular basis for the ECG features and arrhythmia mechanisms using low-dose caffeine to mimic the defective calcium homeostasis encountered under these conditions. Methods and Results—A transmural ECG and action potentials were recorded simultaneously from epicardial, M, and endocardial cells in arterially perfused canine ventricular wedge preparations. Caffeine alone produced no change (10 to 100 &mgr;mol/L) or a slight abbreviation (300 &mgr;mol/L) of the QT interval and no change in transmural dispersion of repolarization. Isoproterenol (100 nmol/L) alone induced sustained monomorphic ventricular tachycardia (VT) that originated in the epicardium in 3 of 14 wedge preparations. Isoproterenol in the presence of caffeine (100 to 300 &mgr;mol/L) induced epicardial VT in 9 of 16 wedge preparations. Delayed afterdepolarization–induced triggered beats that originated in the epicardium were associated with an increased Tpeak-Tend interval and transmural dispersion of repolarization. Bidirectional VT developed in 11 of 16 wedge preparations as a consequence of alternation in the origin of ectopic activity between endocardial, M, and epicardial regions. Single extrastimuli delivered during sustained epicardial VT induced a rapid polymorphic VT/ventricular fibrillation (VF) in 3 of 9 wedges. Spontaneous polymorphic VT was observed in 3 of 16 preparations. Propranolol (1.0 &mgr;mol/L) or verapamil (1.0 &mgr;mol/L) completely suppressed ectopic activity that arose from the epicardium and prevented induction of polymorphic VT. Conclusions—Our data suggest delayed afterdepolarization–induced extrasystolic activity serves to trigger catecholamine-induced VT/VF under conditions of defective calcium handling. Epicardial origin of the ectopic beats increases transmural dispersion of repolarization, thus providing the substrate for the development of reentrant tachyarrhythmias that underlie rapid polymorphic VT/VF.


Pacing and Clinical Electrophysiology | 2006

Late‐Phase 3 EAD. A Unique Mechanism Contributing to Initiation of Atrial Fibrillation

Alexander Burashnikov; Charles Antzelevitch

Early (EAD) and delayed (DAD) afterdepolarizations‐induced triggered activity is capable of initiating and maintaining cardiac arrhythmias. EAD‐induced triggered responses are traditionally thought to be involved in the generation of ventricular arrhythmias under long QT conditions and are precipitated by bradycardia or long pauses. In contrast, DAD‐induced triggered activity commonly underlies arrhythmias precipitated by tachycardia. Spontaneous release of calcium from the sarcoplasmic reticulum (SR) secondary to cellular calcium overload induces DADs and some forms of EADs. Recent studies from our laboratory have uncovered a novel mechanism giving rise to triggered activity, termed “late‐phase 3 EAD,” which combines properties of both EAD and DAD, but has its own unique character. Late‐phase 3 EAD‐induced triggered extrasystoles represent a new concept of arrhythmogenesis in which abbreviated repolarization permits “normal SR calcium release” to induce an EAD‐mediated closely coupled triggered response, particularly under conditions permitting intracellular calcium loading. This review briefly describes the mechanisms and properties of late‐phase 3 EADs, how they differ from conventional EADs and DADs, as well as their role in the initiation of cardiac arrhythmias, such as atrial fibrillation.

Collaboration


Dive into the Alexander Burashnikov's collaboration.

Top Co-Authors

Avatar

Charles Antzelevitch

Lankenau Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

José M. Di Diego

Lankenau Institute for Medical Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Dewan Zeng

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge