Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
PLoS One ; 15(11): e0242747, 2020.
Article in English | MEDLINE | ID: mdl-33232375

ABSTRACT

BACKGROUND: J wave syndromes (JWS), including Brugada (BrS) and early repolarization syndromes (ERS), are associated with increased risk for life-threatening ventricular arrhythmias. Pharmacologic approaches to therapy are currently very limited. Here, we evaluate the effects of the natural flavone acacetin. METHODS: The effects of acacetin on action potential (AP) morphology and transient outward current (Ito) were first studied in isolated canine RV epicardial myocytes using whole-cell patch clamp techniques. Acacetin's effects on transmembrane APs, unipolar electrograms and transmural ECGs were then studied in isolated coronary-perfused canine RV and LV wedge preparations as well as in whole-heart, Langendorff-perfused preparations from which we recorded a 12 lead ECG and unipolar electrograms. Using floating glass microelectrodes we also recorded transmembrane APs from the RVOT of the whole-heart model. The Ito agonist NS5806, sodium channel blocker ajmaline, calcium channel blocker verapamil or hypothermia (32°C) were used to pharmacologically mimic the genetic defects and conditions associated with JWS, thus eliciting prominent J waves and provoking VT/VF. RESULTS: Acacetin (5-10 µM) reduced Ito density, AP notch and J wave area and totally suppressed the electrocardiographic and arrhythmic manifestation of both BrS and ERS, regardless of the experimental model used. In wedge and whole-heart models of JWS, increasing Ito with NS5806, decreasing INa or ICa (with ajmaline or verapamil) or hypothermia all resulted in accentuation of epicardial AP notch and ECG J waves, resulting in characteristic BrS and ERS phenotypes. Phase 2-reentrant extrasystoles originating from the RVOT triggered VT/VF. The J waves in leads V1 and V2 were never associated with a delay of RVOT activation and always coincided with the appearance of the AP notch recorded from RVOT epicardium. All repolarization defects giving rise to VT/VF in the BrS and ERS models were reversed by acacetin, resulting in total suppression of VT/VF. CONCLUSIONS: We present experimental models of BrS and ERS capable of recapitulating all of the ECG and arrhythmic manifestations of the JWS. Our findings provide definitive support for the repolarization but not the depolarization hypothesis proposed to underlie BrS and point to acacetin as a promising new pharmacologic treatment for JWS.


Subject(s)
Brugada Syndrome , Electrocardiography , Flavones/pharmacology , Myocytes, Cardiac/metabolism , Pericardium/metabolism , Ajmaline/pharmacology , Animals , Brugada Syndrome/chemically induced , Brugada Syndrome/drug therapy , Brugada Syndrome/metabolism , Brugada Syndrome/physiopathology , Disease Models, Animal , Dogs , Drug Evaluation, Preclinical , HEK293 Cells , Humans , Hypothermia/metabolism , Hypothermia/pathology , Hypothermia/physiopathology , Phenylurea Compounds/pharmacology , Tetrazoles/pharmacology , Verapamil/pharmacology
2.
Int J Cardiol ; 207: 326-34, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26820362

ABSTRACT

INTRODUCTION: Atrial-selective inhibition of cardiac sodium channel current (INa) and INa-dependent parameters has been shown to contribute to the safe and effective management of atrial fibrillation. The present study was designed to examine the basis for the atrial-selective actions of Wenxin Keli. METHODS: Whole cell INa was recorded at room temperature in canine atrial and ventricular myocytes. Trains of 40 pulses were elicited over a range of pulse durations and interpulse intervals to determine tonic and use-dependent block. A Markovian model for INa that incorporates interaction of Wenxin Keli with different states of the channel was developed to examine the basis for atrial selectivity of the drug. RESULTS: Our data indicate that Wenxin Keli does not bind significantly to either closed or open states of the sodium channel, but binds very rapidly to the inactivated state of the channel and dissociates rapidly from the closed state. Action potentials recorded from atrial and ventricular preparations in the presence of 5g/L Wenxin Keli were introduced into the computer model in current clamp mode to simulate the effects on maximum upstroke velocity (Vmax). The model predicted much greater inhibition of Vmax in atrial vs. ventricular cells at rapid stimulation rates. CONCLUSION: Our findings suggest that atrial selectivity of Wenxin Keli to block INa is due to more negative steady-state inactivation, less negative resting membrane potential, and shorter diastolic intervals in atrial vs. ventricular cells at rapid activation rates. These actions of Wenxin Keli account for its relatively safe and effective suppression of atrial fibrillation.


Subject(s)
Drugs, Chinese Herbal/pharmacology , Heart Atria/drug effects , Models, Theoretical , Sodium Channel Blockers/pharmacology , Sodium Channels/physiology , Action Potentials/drug effects , Action Potentials/physiology , Animals , Cells, Cultured , Dogs , HEK293 Cells , Heart Atria/cytology , Heart Rate/drug effects , Heart Rate/physiology , Humans , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology
3.
Heart Rhythm ; 9(1): 125-31, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21884675

ABSTRACT

BACKGROUND: Wenxin Keli is a Chinese herb extract reported to be of benefit in the treatment of cardiac arrhythmias, cardiac inflammation, and heart failure. METHODS AND RESULTS: We evaluated the electrophysiologic effects of Wenxin Keli in isolated canine arterially perfused right atrial preparations with a rim of right ventricular tissue (n = 11). Transmembrane action potentials and a pseudoelectrocardiogram were simultaneously recorded. Acetylcholine (1 µM) was used to induce atrial fibrillation (AF) and to test the anti-AF potential of Wenxin Keli (5 g/L). Wenxin Keli produced preferential abbreviation of action potential duration measured at 90% repolarization (APD(90)) in atria, but caused atrial-selective prolongation of the effective refractory period, due to the development of postrepolarization refractoriness. The maximum rate of rise of the action potential upstroke was preferentially reduced in atria. The diastolic threshold of excitation increased in both atria and ventricles, but much more in atria. The duration of the "P wave" (index of atrial conduction time) was prolonged to a much greater extent than the duration of the "QRS complex" (index of ventricular conduction time). Wenxin Keli significantly reduced I(Na) and shifted steady-state inactivation to more negative potentials in HEK293 cells stably expressing SCN5A. Wenxin Keli prevented the induction of persistent AF in 100% atria (6/6) and, in another experimental series, was found to terminate persistent acetylcholine-mediated AF in 100% of atria (3/3). CONCLUSION: Wenxin Keli produces atrial-selective depression of I(Na)-dependent parameters in canine isolated coronary-perfused preparations via a unique mechanism and is effective in suppressing AF and preventing its induction, with minimal effects on the ventricular electrophysiology.


Subject(s)
Atrial Fibrillation/drug therapy , Drugs, Chinese Herbal/pharmacology , Heart Atria/drug effects , Membrane Potentials/drug effects , Sodium Channel Blockers/pharmacology , Action Potentials/drug effects , Animals , Atrial Fibrillation/physiopathology , Dogs , Drugs, Chinese Herbal/therapeutic use , Electrophysiological Phenomena/drug effects , HEK293 Cells , Heart Atria/physiopathology , Humans , Sodium Channel Blockers/therapeutic use
4.
Heart Rhythm ; 8(8): 1281-90, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21421082

ABSTRACT

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


Subject(s)
Acetanilides/pharmacology , Anti-Arrhythmia Agents/pharmacology , Arrhythmias, Cardiac/drug therapy , Heart/physiopathology , Piperazines/pharmacology , Action Potentials , Animals , Arrhythmias, Cardiac/physiopathology , Calcium Channels/drug effects , Electrophysiologic Techniques, Cardiac , Heart Atria/drug effects , Heart Atria/physiopathology , Heart Ventricles/cytology , Heart Ventricles/drug effects , Humans , Ion Channels/drug effects , Membrane Potentials , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Potassium Channels/drug effects , Ranolazine , Species Specificity
5.
Heart Rhythm ; 7(9): 1273-9, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20478403

ABSTRACT

BACKGROUND: Dronedarone is approved by the U.S. Food and Drug Administration for the treatment of patients with atrial fibrillation (AF) as a safe alternative to amiodarone. There are no full-length published reports describing the effectiveness of acute dronedarone use against AF in experimental or clinical studies. OBJECTIVE: The purpose of this study was to determine the effect of acute dronedarone and amiodarone on electrophysiological parameters, and their anti-AF efficacy in canine isolated arterially perfused right atria. METHODS: Transmembrane action potentials and pseudoelectrocardiograms were recorded. Acetylcholine (ACh, 1.0 muM) was used to induce persistent AF. RESULTS: Amiodarone-induced changes were much more pronounced than those of dronedarone on (1) action potential duration (DeltaAPD(90), +51 +/- 17 ms vs. 4 +/- 6 ms, P >.01), (2) effective refractory period (DeltaERP, +84 +/- 23 ms vs. 18 +/- 9 ms, P <.001), (3) diastolic threshold of excitation (DeltaDTE, +0.32 +/- 0.11 mA vs. 0.03 +/- 0.02 mA, P <.001), and (4) V(max) (DeltaV(max), -43 +/- 14% vs. -11 +/- 4%, P <.01, n = 5 to 6; all recorded at 10 muM, cycle length = 500 ms). Persistent AF was induced in 10 of 10 atria exposed to ACh alone; subsequent addition of dronedarone or amiodarone terminated AF in 1 of 7 and 4 of 5 atria, respectively. Persistent ACh-mediated AF was induced in 5 of 6 and 0 of 5 atria pretreated with dronedarone and amiodarone, respectively. CONCLUSION: The electrophysiological effects and anti-AF efficacy of acute dronedarone are much weaker than those of amiodarone in a canine model of AF. The efficacy of acute dronedarone to prevent induction of acetylcholine-mediated AF as well as to terminate persistent AF in canine right atria is relatively poor. Our data suggest that acute dronedarone is a poor substitute for amiodarone for acute cardioversion of AF or prevention of AF recurrence.


Subject(s)
Amiodarone/analogs & derivatives , Amiodarone/administration & dosage , Anti-Arrhythmia Agents/administration & dosage , Atrial Fibrillation/prevention & control , Heart Atria/drug effects , Heart Conduction System/drug effects , Animals , Atrial Fibrillation/drug therapy , Atrial Fibrillation/physiopathology , Disease Models, Animal , Dogs , Dose-Response Relationship, Drug , Dronedarone , Electrophysiologic Techniques, Cardiac , Heart Atria/physiopathology , Heart Conduction System/physiopathology , Treatment Outcome
6.
Circ Arrhythm Electrophysiol ; 3(1): 88-95, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19952329

ABSTRACT

BACKGROUND: Amiodarone and ranolazine have been characterized as inactivated- and activated-state blockers of cardiac sodium channel current (I(Na)), respectively, and shown to cause atrial-selective depression of I(Na)-related parameters. This study tests the hypothesis that their combined actions synergistically depress I(Na)-dependent parameters in atria but not ventricles. METHODS AND RESULTS: The effects of acute ranolazine (5 to 10 micromol/L) were studied in coronary-perfused right atrial and left ventricular wedge preparations and superfused left atrial pulmonary vein sleeves isolated from chronic amiodarone-treated (40 mg/kg daily for 6 weeks) and untreated dogs. Floating and standard microelectrode techniques were used to record transmembrane action potentials. When studied separately, acute ranolazine and chronic amiodarone caused atrial-predominant depression of I(Na)-dependent parameters. Ranolazine produced a much greater reduction in V(max) and much greater increase in diastolic threshold of excitation and effective refractory period in atrial preparations isolated from amiodarone-treated versus untreated dogs, leading to a marked increase in postrepolarization refractoriness. The drug combination effectively suppressed triggered activity in pulmonary vein sleeves but produced relatively small changes in I(Na)-dependent parameters in the ventricle. Acetylcholine (0.5 micromol/L) and burst pacing induced atrial fibrillation in 100% of control atria, 75% of ranolazine-treated (5 micromol/L) atria, 16% of atria from amiodarone-treated dogs, and in 0% of atria from amiodarone-treated dogs exposed to 5 micromol/L ranolazine. CONCLUSIONS: The combination of chronic amiodarone and acute ranolazine produces a synergistic use-dependent depression of I(Na)-dependent parameters in isolated canine atria, leading to a potent effect of the drug combination to prevent the induction of atrial fibrillation.


Subject(s)
Acetanilides/pharmacology , Amiodarone/pharmacology , Anti-Arrhythmia Agents/pharmacology , Atrial Fibrillation/prevention & control , Electrophysiologic Techniques, Cardiac , Piperazines/pharmacology , Sodium Channel Blockers/pharmacology , Animals , Dogs , Drug Synergism , Enzyme Inhibitors/pharmacology , In Vitro Techniques , Membrane Potentials/drug effects , Ranolazine , Refractory Period, Electrophysiological/drug effects
7.
Circ Arrhythm Electrophysiol ; 1(3): 202-8, 2008 Aug.
Article in English | MEDLINE | ID: mdl-19649147

ABSTRACT

BACKGROUND: The arrhythmogenic effects of hyperthermia have been highlighted in the Brugada syndrome but remain largely unexplored in other arrhythmic syndromes. The present study examines the effect of hyperthermia on transmural dispersion of action potential duration (TD-APD), early afterdepolarization (EAD) activity, and torsade de pointes (TdP) under long-QT conditions. METHODS AND RESULTS: Standard and floating glass microelectrodes were used to record action potentials from epicardial, M cell, and endocardial regions of the arterially perfused left ventricle wedge, from tissue slices isolated from these regions, and from isolated Purkinje fibers. A transmural ECG was simultaneously recorded across the wedge. Under baseline conditions and in the presence of I(Ks) block (chromanol 293B), hyperthermia (39 degrees C to 40 degrees C) abbreviated APD in tissue slices from all 3 regions. In the presence of I(Kr) block (E-4031), hyperthermia prolonged APD and induced or augmented EADs in M cell and Purkinje preparations at pacing cycle lengths > or = 800 ms but abbreviated APD in epicardium and endocardium, resulting in a marked accentuation of TD-APD. Ryanodine prevented the hyperthermia- induced EAD. In perfused wedge preparations, hyperthermia abbreviated APD throughout both in the absence or presence of I(Kr) or I(Ks) block and did not induce EADs or TdP. Combined I(Kr) and I(Ks) block increased TD-APD and induced EADs (4/12) and spontaneous TdP (3/12) at 36 degrees C to 37 degrees C; hyperthermia (39 degrees C to 40 degrees C) further accentuated TD-APD and facilitated the development of EAD activity (9/12) and TdP (6/12). CONCLUSIONS: Our findings suggest that hyperthermia can be associated with an increased arrhythmic risk when the repolarization reserve of the myocardium is compromised.


Subject(s)
Action Potentials/physiology , Fever/complications , Long QT Syndrome/complications , Purkinje Fibers/physiopathology , Torsades de Pointes/etiology , Animals , Disease Models, Animal , Dogs , Electrophysiologic Techniques, Cardiac , Fever/physiopathology , Hyperthermia, Induced/adverse effects , Long QT Syndrome/physiopathology , Torsades de Pointes/physiopathology
8.
Circulation ; 116(13): 1449-57, 2007 Sep 25.
Article in English | MEDLINE | ID: mdl-17785620

ABSTRACT

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 (I(Na)) in myocytes isolated from canine atria and ventricles. The electrophysiological effects of therapeutic concentrations of ranolazine (1 to 10 micromol/L) and lidocaine (2.1 to 21 micromol/L) were evaluated in canine isolated coronary-perfused atrial and ventricular preparations. Half-inactivation voltage of I(Na) was approximately 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.


Subject(s)
Acetanilides/pharmacology , Anti-Arrhythmia Agents/pharmacology , Heart Atria/drug effects , Piperazines/pharmacology , Sodium Channel Blockers/pharmacology , Action Potentials/drug effects , Animals , Atrial Fibrillation/drug therapy , Cardiotonic Agents/pharmacology , Dogs , Drug Evaluation, Preclinical , Heart Ventricles/drug effects , Ion Channels/drug effects , Lidocaine/pharmacology , Myocytes, Cardiac/drug effects , Organ Specificity , Patch-Clamp Techniques , Purkinje Fibers/drug effects , Ranolazine
10.
J Cardiovasc Pharmacol Ther ; 9 Suppl 1: S65-83, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15378132

ABSTRACT

Ranolazine is a novel antianginal agent capable of producing anti-ischemic effects at plasma concentrations of 2 to 6 microM without a significant reduction of heart rate or blood pressure. This review summarizes the electrophysiologic properties of ranolazine. Ranolazine significantly blocks I(Kr) (IC(50) = 12 microM), late I(Na), late I(Ca), peak I(Ca), I(Na-Ca) (IC(50) = 5.9, 50, 296, and 91 microM, respectively) and I(Ks) (17% at 30 microM), but causes little or no inhibition of I(to) or I(K1). 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 I(Kr) 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 after depolarizations, 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 microM. 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 I(Na), I(Kr), I(Ks), and I(Ca)). Ranolazine's actions to reduce TDR and suppress early after depolarization suggest that in addition to its anti-anginal actions, the drug possesses antiarrhythmic activity.


Subject(s)
Angina Pectoris/drug therapy , Long QT Syndrome/prevention & control , Piperazines/therapeutic use , Torsades de Pointes/drug therapy , Acetanilides , Action Potentials/drug effects , Angina Pectoris/classification , Angina Pectoris/physiopathology , Animals , Electrophysiologic Techniques, Cardiac/methods , Humans , Ion Channels/classification , Ion Channels/drug effects , Ion Channels/physiology , Long QT Syndrome/chemically induced , Long QT Syndrome/physiopathology , Piperazines/metabolism , Piperazines/pharmacology , Ranolazine , Stereoisomerism , Torsades de Pointes/chemically induced , Torsades de Pointes/physiopathology
11.
Circulation ; 110(8): 904-10, 2004 Aug 24.
Article in English | MEDLINE | ID: mdl-15302796

ABSTRACT

BACKGROUND: Ranolazine is a novel antianginal agent capable of producing antiischemic effects at plasma concentrations of 2 to 6 micromol/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 I(Kr) (IC50=11.5 micromol/L), late I(Na), late I(Ca), peak I(Ca), and I(Na-Ca) (IC50=5.9, 50, 296, and 91 micromol/L, respectively) and I(Ks) (17% at 30 micromol/L), but caused little or no inhibition of I(to) or I(K1). 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 micromol/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 micromol/L) suppressed early afterdepolarizations (EADs) and reduced the increase in TDR induced by the selective I(Kr) blocker d-sotalol. CONCLUSIONS: Ranolazine produces ion channel effects similar to those observed after chronic amiodarone (reduced I(Kr), I(Ks), late I(Na), and I(Ca)). The actions of ranolazine to suppress EADs and reduce TDR suggest that, in addition to its antianginal actions, the drug may possess antiarrhythmic activity.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Myocytes, Cardiac/drug effects , Piperazines/pharmacology , Acetanilides , Action Potentials , Animals , Calcium/metabolism , Cells, Cultured/drug effects , Delayed Rectifier Potassium Channels , Dogs , Drug Evaluation, Preclinical , Electrocardiography , Heart Ventricles , Ion Channels/drug effects , Ion Transport/drug effects , Organ Culture Techniques , Patch-Clamp Techniques , Piperidines/pharmacology , Potassium/metabolism , Potassium Channels, Voltage-Gated/antagonists & inhibitors , Pyridines/pharmacology , Ranolazine , Sodium/metabolism , Sodium-Calcium Exchanger/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL