Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
PLoS Comput Biol ; 11(4): e1004242, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25928268

RESUMO

The traditional cardiac model-building paradigm involves constructing a composite model using data collected from many cells. Equations are derived for each relevant cellular component (e.g., ion channel, exchanger) independently. After the equations for all components are combined to form the composite model, a subset of parameters is tuned, often arbitrarily and by hand, until the model output matches a target objective, such as an action potential. Unfortunately, such models often fail to accurately simulate behavior that is dynamically dissimilar (e.g., arrhythmia) to the simple target objective to which the model was fit. In this study, we develop a new approach in which data are collected via a series of complex electrophysiology protocols from single cardiac myocytes and then used to tune model parameters via a parallel fitting method known as a genetic algorithm (GA). The dynamical complexity of the electrophysiological data, which can only be fit by an automated method such as a GA, leads to more accurately parameterized models that can simulate rich cardiac dynamics. The feasibility of the method is first validated computationally, after which it is used to develop models of isolated guinea pig ventricular myocytes that simulate the electrophysiological dynamics significantly better than does a standard guinea pig model. In addition to improving model fidelity generally, this approach can be used to generate a cell-specific model. By so doing, the approach may be useful in applications ranging from studying the implications of cell-to-cell variability to the prediction of intersubject differences in response to pharmacological treatment.


Assuntos
Potenciais de Ação/fisiologia , Sistema de Condução Cardíaco/fisiologia , Canais Iônicos/fisiologia , Potenciais da Membrana/fisiologia , Modelos Cardiovasculares , Miócitos Cardíacos/fisiologia , Animais , Células Cultivadas , Simulação por Computador , Ativação do Canal Iônico/fisiologia , Modelos Estatísticos , Suínos
2.
J Mol Cell Cardiol ; 64: 79-89, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24035801

RESUMO

BACKGROUND: Developmental changes in the electrical characteristics of the ventricular myocardium are not well defined. This study examines the contribution of inwardly rectifying K(+) current (IK1), transient outward K(+) current (Ito), delayed rectifier K(+) currents (IKr and IKs) and sodium channel current (INa) to repolarization in the canine neonate myocardium. METHODS: Single myocytes isolated from the left ventricle of 2-3week old canine neonate hearts were studied using patch-clamp techniques. RESULTS: Neonate cells were ~6-fold smaller than those of adults (28.8±8.8 vs. 176±6.7pF). IK1 was larger in neonate myocytes and displayed a substantial inward component and an outward component with negative slope conductance, peaking at -60mV (4.13 pA/pF). IKr tail currents (at -40mV), were small (<20pA). IKs could not be detected, even after exposure to isoproterenol (100nM). Ito was also absent in the neonate, consistent with the absence of a phase 1 in the action potential. Peak INa, late INa and ICa were smaller in the neonate compared with adults. KCND3, KCNIP2 and KCNQ1 mRNA expression was half, while KCNH2 was equal and KCNJ2 was greater in the neonate when compared with adults. CONCLUSIONS: Two major repolarizing K(+) currents (IKs and Ito) present in adult ventricular cells are absent in the 2week old neonate. Peak and late INa are significantly smaller in the neonate. Our results suggest that the absence of these two currents in the neonate heart may increase the susceptibility to arrhythmias under certain long QT conditions.


Assuntos
Canais Iônicos/genética , Canais Iônicos/metabolismo , Função Ventricular/fisiologia , Potenciais de Ação , Animais , Animais Recém-Nascidos , Antiarrítmicos/farmacologia , Cálcio/metabolismo , Cães , Feminino , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/metabolismo , Humanos , Masculino , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Técnicas de Patch-Clamp , Piperidinas/farmacologia , Potássio/metabolismo , Canais de Potássio/fisiologia , Piridinas/farmacologia , Sódio/metabolismo , Função Ventricular/efeitos dos fármacos
3.
J Cardiovasc Electrophysiol ; 23(4): 423-32, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22034916

RESUMO

INTRODUCTION: Tricyclic antidepressants are known to induce cardiac arrhythmias at therapeutic or supratherapeutic doses. The tricyclic antidepressant, amitriptyline, is reported to induce ST segment elevation in the right precordial electrocardiogram (ECG) leads, thus unmasking Brugada syndrome (BrS). The mechanism by which antidepressants induce the BrS phenotype and associated sudden death is not well established. METHODS AND RESULTS: Action potentials (AP) were simultaneously recorded from epicardial and endocardial sites of isolated coronary-perfused canine right ventricular wedge preparations, together with a transmural pseudo-ECG. Amitriptyline alone (0.2 µM-1 mM) failed to induce a BrS phenotype. NS5806 (8 µM), a transient outward potassium channel current (I(to) ) agonist, was used to produce an outward shift of current mimicking a genetic predisposition to BrS. In the presence of NS5806, a therapeutic concentration of amitriptyline (0.2 µM) accentuated the epicardial AP notch leading to ST-segment elevation of the ECG. All-or-none repolarization at some epicardial sites but not others gave rise to phase-2-reentry and polymorphic ventricular tachycardia (VT) in 6 of 9 preparations. Isoproterenol (100 nM) or quinidine (10 µM) reversed the effects of amitriptyline aborting phase 2 reentry and VT (4/4). Using voltage-clamp techniques applied to isolated canine ventricular myocytes, 0.2 µM amitriptyline was shown to produce use-dependent inhibition of sodium channel current (I(Na) ), without significantly affecting I(to) (n = 5). CONCLUSIONS: Our data suggest that amitriptyline-induced inhibition of I(Na) unmasks the Brugada ECG phenotype and facilitates development of an arrhythmogenic substrate only in the setting of a genetic predisposition by creating repolarization heterogeneities that give rise to phase 2 reentry and VT.


Assuntos
Amitriptilina/efeitos adversos , Antidepressivos Tricíclicos/efeitos adversos , Síndrome de Brugada/induzido quimicamente , Sistema de Condução Cardíaco/efeitos dos fármacos , Bloqueadores dos Canais de Sódio/efeitos adversos , Potenciais de Ação , Agonistas Adrenérgicos beta/farmacologia , Animais , Antiarrítmicos/farmacologia , Síndrome de Brugada/genética , Síndrome de Brugada/metabolismo , Síndrome de Brugada/fisiopatologia , Síndrome de Brugada/prevenção & controle , Linhagem Celular , Cães , Eletrocardiografia , Feminino , Predisposição Genética para Doença , Genótipo , Sistema de Condução Cardíaco/metabolismo , Sistema de Condução Cardíaco/fisiopatologia , Humanos , Técnicas In Vitro , Masculino , Canal de Sódio Disparado por Voltagem NAV1.5 , Técnicas de Patch-Clamp , Perfusão , Fenótipo , Bloqueadores dos Canais de Potássio/efeitos adversos , Canais de Sódio/efeitos dos fármacos , Canais de Sódio/genética , Canais de Sódio/metabolismo , Taquicardia Ventricular/induzido quimicamente , Taquicardia Ventricular/metabolismo , Taquicardia Ventricular/fisiopatologia , Fatores de Tempo , Transfecção , Fibrilação Ventricular/induzido quimicamente , Fibrilação Ventricular/metabolismo , Fibrilação Ventricular/fisiopatologia
4.
Am J Physiol Heart Circ Physiol ; 301(4): H1615-24, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21821780

RESUMO

Block of Na(+) channel conductance by ranolazine displays marked atrial selectivity that is an order of magnitude higher that of other class I antiarrhythmic drugs. Here, we present a Markovian model of the Na(+) channel gating, which includes activation-inactivation coupling, aimed at elucidating the mechanisms underlying this potent atrial selectivity of ranolazine. The model incorporates experimentally observed differences between atrial and ventricular Na(+) channel gating, including a more negative position of the steady-state inactivation curve in atrial versus ventricular cells. The model assumes that ranolazine requires a hydrophilic access pathway to the channel binding site, which is modulated by both activation and inactivation gates of the channel. Kinetic rate constants were obtained using guarded receptor analysis of the use-dependent block of the fast Na(+) current (I(Na)). The model successfully reproduces all experimentally observed phenomena, including the shift of channel availability, the sensitivity of block to holding or diastolic potential, and the preferential block of slow versus fast I(Na.) Using atrial and ventricular action potential-shaped voltage pulses, the model confirms significantly greater use-dependent block of peak I(Na) in atrial versus ventricular cells. The model highlights the importance of action potential prolongation and of a steeper voltage dependence of the time constant of unbinding of ranolazine from the atrial Na(+) channel in the development of use-dependent I(Na) block. Our model predictions indicate that differences in channel gating properties as well as action potential morphology between atrial and ventricular cells contribute equally to the atrial selectivity of ranolazine. The model indicates that the steep voltage dependence of ranolazine interaction with the Na(+) channel at negative potentials underlies the mechanism of the predominant block of I(Na) in atrial cells by ranolazine.


Assuntos
Acetanilidas/farmacologia , Coração/efeitos dos fármacos , Piperazinas/farmacologia , Bloqueadores dos Canais de Sódio , Animais , Simulação por Computador , Relação Dose-Resposta a Droga , Átrios do Coração/efeitos dos fármacos , Ventrículos do Coração/efeitos dos fármacos , Cinética , Cadeias de Markov , Modelos Biológicos , Modelos Estatísticos , Miócitos Cardíacos/efeitos dos fármacos , Ranolazina , Canais de Sódio/efeitos dos fármacos , Canais de Sódio/metabolismo
5.
Am J Physiol Heart Circ Physiol ; 301(4): H1606-14, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21821778

RESUMO

Atrial-selective inhibition of cardiac Na(+) channel current (I(Na)) and I(Na)-dependent parameters has been shown to contribute to the safe and effective management of atrial fibrillation. The present study examined the basis for the atrial-selective actions of ranolazine. Whole cell I(Na) was recorded at 15°C in canine atrial and ventricular myocytes and in human embryonic kidney (HEK)-293 cells expressing SCN5A. Tonic block was negligible at holding potentials from -140 to -100 mV, suggesting minimal drug interactions with the closed state. Trains of 40 pulses were elicited over a range of holding potentials to determine use-dependent block. Guarded receptor formalism was used to analyze the development of block during pulse trains. Use-dependent block by ranolazine increased at more depolarized holding potentials, consistent with an interaction of the drug with either preopen or inactivated states, but was unaffected by longer pulse durations between 5 and 200 ms, suggesting a weak interaction with the inactivated state. Block was significantly increased at shorter diastolic intervals between 20 and 200 ms. Responses in atrial and ventricular myocytes and in HEK-293 cells displayed a similar pattern. Ranolazine is an open state blocker that unbinds from closed Na(+) channels unusually fast but is trapped in the inactivated state. Kinetic rates of ranolazine interactions with different states of atrial and ventricular Na(+) channels were similar. Our data suggest that the atrial selectivity of ranolazine is due to a more negative steady-state inactivation curve, less negative resting membrane potential, and shorter diastolic intervals in atrial cells compared with ventricular cells at rapid rates.


Assuntos
Acetanilidas/farmacologia , Piperazinas/farmacologia , Bloqueadores dos Canais de Sódio , Acetanilidas/metabolismo , Algoritmos , Animais , Sítios de Ligação/efeitos dos fármacos , Interpretação Estatística de Dados , Cães , Feminino , Células HEK293 , Frequência Cardíaca/efeitos dos fármacos , Humanos , Técnicas In Vitro , Cinética , Modelos Lineares , Masculino , Miócitos Cardíacos/efeitos dos fármacos , Técnicas de Patch-Clamp , Piperazinas/metabolismo , Ranolazina , Canais de Sódio/metabolismo
6.
J Cardiovasc Pharmacol ; 56(1): 80-90, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20386458

RESUMO

Recent development of drugs for the treatment of atrial fibrillation (AF) has focused on atrial selective agents. We examined the atrioventricular differences in sodium channel block of the antiarrhythmic agent AZD1305 in atria and ventricles of anesthetized dogs in vivo, canine isolated arterially perfused preparations in vitro, and isolated myocytes using whole-cell patch-clamp techniques. AZD1305 did not change heart rate or blood pressure in vivo but prolonged action potential duration and increased effective refractory period, diastolic threshold of excitation, and conduction time preferentially in atria both in vitro and in vivo. AZD1305 reduced the maximum rate of rise of the action potential upstroke (V(max)) predominantly in atria (-51% +/- 10% in atria vs. -31% +/- 23% in ventricles; 3 microM; cycle length = 500 milliseconds). Fast sodium current (I(Na)) was blocked by AZD1305 to a greater degree in atrial versus ventricular myocytes (particularly tonic inhibition). In coronary-perfused right atria, AZD1305 very effectively prevented induction of persistent acetylcholine-mediated AF and, in a different set of atria, terminated persistent AF (in 5 of 5 and 7 of 8 atria, respectively). In conclusion, AZD1305 exerts atrial predominant sodium channel-blocking effects in vitro and in vivo and effectively suppresses AF.


Assuntos
Antiarrítmicos/farmacologia , Fibrilação Atrial/tratamento farmacológico , Compostos Azabicíclicos/farmacologia , Carbamatos/farmacologia , Acetilcolina , Potenciais de Ação/efeitos dos fármacos , Animais , Fibrilação Atrial/fisiopatologia , Modelos Animais de Doenças , Cães , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Feminino , Átrios do Coração/efeitos dos fármacos , Átrios do Coração/metabolismo , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/metabolismo , Masculino , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Sódio/farmacologia
7.
Circulation ; 116(13): 1449-57, 2007 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-17785620

RESUMO

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.


Assuntos
Acetanilidas/farmacologia , Antiarrítmicos/farmacologia , Átrios do Coração/efeitos dos fármacos , Piperazinas/farmacologia , Bloqueadores dos Canais de Sódio/farmacologia , Potenciais de Ação/efeitos dos fármacos , Animais , Fibrilação Atrial/tratamento farmacológico , Cardiotônicos/farmacologia , Cães , Avaliação Pré-Clínica de Medicamentos , Ventrículos do Coração/efeitos dos fármacos , Canais Iônicos/efeitos dos fármacos , Lidocaína/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Especificidade de Órgãos , Técnicas de Patch-Clamp , Ramos Subendocárdicos/efeitos dos fármacos , Ranolazina
8.
Ann N Y Acad Sci ; 1123: 105-12, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18375582

RESUMO

UNLABELLED: Antiarrhythmic drug therapy remains the principal approach for suppression of atrial fibrillation (AF) and flutter (AFl) and prevention of their recurrence. Among the current strategies for suppression of AF/AFl is the development of antiarrhythmic agents that preferentially affect atrial, rather than ventricular electrical parameters. Inhibition of the ultrarapid delayed rectifier potassium current (IKur), present in the atria, but not in the ventricles, is an example of an atrial-selective approach. Our recent study examined the hypothesis that sodium channel characteristics differ between atrial and ventricular cells and that atrial-selective sodium channel block is another effective strategy for the management of AF. We have demonstrated very significant differences in the inactivation characteristics of atrial versus ventricular sodium channels and a striking atrial selectivity for the action of ranolazine, an inactivated-state sodium channel blocker, to produce use-dependent block of the sodium channels, leading to depression of excitability, development of post-repolarization refractoriness (PRR), and suppression of AF. Lidocaine and chronic amiodarone, both predominantly inactivated-state sodium channel blockers, also produced a preferential depression of sodium channel-dependent parameters (VMax conduction velocity, diastolic threshold of excitation, and PRR) in the atria. Propafenone, a predominantly open-state sodium channel blocker, produced similar changes of electrophysiological parameters, which were was not atrial-selective. The ability of ranolazine, chronic amiodarone, and propafenone to prolong the atrial action potential potentiated their ability to suppress AF in coronary-perfused canine atrial preparations. IN CONCLUSION: Our data demonstrate important differences in the inactivation characteristics of atrial versus ventricular sodium channels and a striking atrial selectivity for the action of agents like ranolazine to produce use-dependent block of sodium channels leading to suppression of AF. Our findings suggest that atrial-selective sodium channel block may be a valuable strategy to combat AF.


Assuntos
Antiarrítmicos/uso terapêutico , Fibrilação Atrial/prevenção & controle , Átrios do Coração/fisiopatologia , Bloqueadores dos Canais de Sódio/uso terapêutico , Função Ventricular , Acetanilidas/uso terapêutico , Amiodarona/uso terapêutico , Animais , Fibrilação Atrial/fisiopatologia , Cães , Eletrofisiologia , Humanos , Lidocaína/uso terapêutico , Piperazinas/uso terapêutico , Propafenona/uso terapêutico , Ranolazina
9.
Heart Rhythm ; 4(5): 638-47, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17467634

RESUMO

BACKGROUND: Timothy syndrome is a multisystem disorder associated with QT interval prolongation and ventricular cardiac arrhythmias. The syndrome has been linked to mutations in Ca(V)1.2 resulting in gain of function of the L-type calcium current (I(Ca,L)). Ranolazine is an antianginal agent shown to exert an antiarrhythmic effect in experimental models of long QT syndrome. OBJECTIVE: The purpose of this study was to develop and characterize an experimental model of Timothy syndrome by using BayK8644 to mimic the gain of function of I(Ca,L) and to examine the effects of ranolazine. METHODS: Action potentials from epicardial and M regions and a pseudo-electrocardiogram (ECG) were simultaneously recorded from coronary-perfused left ventricular wedge preparations, before and after addition of BayK8644 (1 microM). RESULTS: BayK8644 preferentially prolonged action potential duration of the M cell, leading to prolongation of the QT interval and an increase in transmural dispersion of repolarization (from 44.3 +/- 7 ms to 86.5 +/- 25 ms). Stimulation at cycle lengths of 250-500 ms led to ST-T wave alternans due to alternation of the plateau voltage of the M cell action potential as well as development of delayed afterdepolarizations in epicardial and M cell action potentials. Ventricular extrasystoles and tachycardia (monomorphic, bidirectional, or torsades de pointes) developed spontaneously or after rapid pacing. Peak and late I(Na) were unaffected by BayK8644. Clinically relevant concentrations of ranolazine (10 microM) suppressed all actions of BayK8644. CONCLUSION: A left ventricular wedge model of long QT syndrome created by augmentation of I(Ca,L) recapitulates the ECG and arrhythmic manifestations of Timothy syndrome, which can be suppressed by ranolazine.


Assuntos
Acetanilidas/farmacologia , Antiarrítmicos/farmacologia , Eletrocardiografia , Síndrome do QT Longo/tratamento farmacológico , Síndrome do QT Longo/fisiopatologia , Miócitos Cardíacos/efeitos dos fármacos , Piperazinas/farmacologia , Éster Metílico do Ácido 3-Piridinacarboxílico, 1,4-Di-Hidro-2,6-Dimetil-5-Nitro-4-(2-(Trifluormetil)fenil)/efeitos adversos , Potenciais de Ação/efeitos dos fármacos , Análise de Variância , Animais , Agonistas dos Canais de Cálcio/efeitos adversos , Modelos Animais de Doenças , Cães , Endocárdio/citologia , Endocárdio/efeitos dos fármacos , Endocárdio/fisiopatologia , Inibidores Enzimáticos/farmacologia , Ventrículos do Coração/citologia , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/fisiopatologia , Síndrome do QT Longo/etiologia , Técnicas de Patch-Clamp , Pericárdio/citologia , Pericárdio/efeitos dos fármacos , Pericárdio/fisiopatologia , Ranolazina , Taquicardia Ventricular/induzido quimicamente , Taquicardia Ventricular/tratamento farmacológico , Taquicardia Ventricular/fisiopatologia , Torsades de Pointes/induzido quimicamente , Torsades de Pointes/tratamento farmacológico , Torsades de Pointes/fisiopatologia , Complexos Ventriculares Prematuros/induzido quimicamente , Complexos Ventriculares Prematuros/tratamento farmacológico , Complexos Ventriculares Prematuros/fisiopatologia
10.
Circulation ; 110(8): 904-10, 2004 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-15302796

RESUMO

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.


Assuntos
Antiarrítmicos/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Piperazinas/farmacologia , Acetanilidas , Potenciais de Ação , Animais , Cálcio/metabolismo , Células Cultivadas/efeitos dos fármacos , Canais de Potássio de Retificação Tardia , Cães , Avaliação Pré-Clínica de Medicamentos , Eletrocardiografia , Ventrículos do Coração , Canais Iônicos/efeitos dos fármacos , Transporte de Íons/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Técnicas de Patch-Clamp , Piperidinas/farmacologia , Potássio/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/antagonistas & inibidores , Piridinas/farmacologia , Ranolazina , Sódio/metabolismo , Trocador de Sódio e Cálcio/antagonistas & inibidores
11.
Circulation ; 106(15): 2004-11, 2002 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-12370227

RESUMO

BACKGROUND: The Brugada syndrome displays an autosomal dominant mode of transmission with low penetrance. Despite equal genetic transmission of the disease, the clinical phenotype is 8 to 10 times more prevalent in males than in females. The basis for this intriguing sex-related distinction is unknown. The present study tests the hypothesis that the disparity in expression of the Brugada phenotype is a result of a more prominent I(to)-mediated action potential notch in the right ventricular (RV) epicardium of males versus females. METHODS AND RESULTS: We studied epicardial tissue slices, arterially perfused wedge preparations, and dissociated epicardial myocytes isolated from male and female canine hearts. RV epicardium action potential phase 1 amplitude was 64.8+/-2.0% of that of phase 2 in males compared with 73.8+/-4.4% in females (P<0.05) at a cycle length of 2000 ms. I(to) density was 26% smaller and time constant for inactivation 17% smaller at +40 mV in female versus male RV epicardial cells (P<0.05). The other functional characteristics of I(to), including the voltage dependence of inactivation and time course of reactivation, were no different between the sexes. Pinacidil caused loss of action potential dome in male, but not female, RV epicardial tissue slices. Terfenadine (5 micromol/L) induced phase 2 reentry in 6 of 7 male but only 2 of 7 female arterially perfused wedge preparations. Two of 6 male and 1 of 2 female preparations developed polymorphic ventricular tachycardia/ventricular fibrillation. CONCLUSIONS: Our results suggest that the predominance of the Brugada phenotype in males is a result of the presence of a more prominent I(to) in males versus females.


Assuntos
Potenciais de Ação , Pericárdio/fisiologia , Fatores Sexuais , Potenciais de Ação/efeitos dos fármacos , Animais , Arritmias Cardíacas/diagnóstico , Arritmias Cardíacas/etiologia , Arritmias Cardíacas/fisiopatologia , Células Cultivadas , Técnicas de Cultura , Cães , Feminino , Coração/fisiologia , Cinética , Masculino , Pericárdio/efeitos dos fármacos , Fenótipo , Pinacidil/farmacologia , Síndrome , Terfenadina/farmacologia
12.
J Cardiovasc Pharmacol Ther ; 9 Suppl 1: S65-83, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15378132

RESUMO

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.


Assuntos
Angina Pectoris/tratamento farmacológico , Síndrome do QT Longo/prevenção & controle , Piperazinas/uso terapêutico , Torsades de Pointes/tratamento farmacológico , Acetanilidas , Potenciais de Ação/efeitos dos fármacos , Angina Pectoris/classificação , Angina Pectoris/fisiopatologia , Animais , Técnicas Eletrofisiológicas Cardíacas/métodos , Humanos , Canais Iônicos/classificação , Canais Iônicos/efeitos dos fármacos , Canais Iônicos/fisiologia , Síndrome do QT Longo/induzido quimicamente , Síndrome do QT Longo/fisiopatologia , Piperazinas/metabolismo , Piperazinas/farmacologia , Ranolazina , Estereoisomerismo , Torsades de Pointes/induzido quimicamente , Torsades de Pointes/fisiopatologia
13.
Circ Heart Fail ; 7(4): 627-33, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24874201

RESUMO

BACKGROUND: There is a critical need for safer and more effective pharmacological management of atrial fibrillation (AF) in the setting of heart failure (HF). METHODS AND RESULTS: This study investigates the electrophysiological, antiarrhythmic, and proarrhythmic effects of a clinically relevant concentration of ranolazine (5 µmol/L) in coronary-perfused right atrial and left ventricular preparations isolated from the hearts of HF dogs. HF was induced by ventricular tachypacing (2-6 weeks at 200-240 beats per minute; n=17). Transmembrane action potentials were recorded using standard microelectrode techniques. In atria, ranolazine slightly prolonged action potential duration but significantly depressed sodium channel current-dependent parameters causing a reduction of maximum rate of rise of the action potential upstroke, a prolongation of the effective refractory period secondary to the development of postrepolarization refractoriness, and an increase in diastolic threshold of excitation and atrial conduction time. Ranolazine did not significantly alter these parameters or promote arrhythmias in the ventricles. Ranolazine produced greater inhibition of peak sodium channel current in atrial cells isolated from HF versus normal dogs. A single premature beat reproducibly induced self-terminating AF in 10 of 17 atria. Ranolazine (5 µmol/L) suppressed induction of AF in 7 of 10 (70%) atria. In the remaining 3 atria, ranolazine reduced frequency and duration of AF. CONCLUSIONS: Our results demonstrate more potent suppression of AF by ranolazine in the setting of HF than previously demonstrated in nonfailing hearts and absence of ventricular proarrhythmia. The data suggest that ranolazine may be of benefit as an alternative to amiodarone and dofetilide in the management of AF in patients with HF.


Assuntos
Acetanilidas/administração & dosagem , Fibrilação Atrial/tratamento farmacológico , Eletrocardiografia/efeitos dos fármacos , Sistema de Condução Cardíaco/efeitos dos fármacos , Insuficiência Cardíaca/prevenção & controle , Miócitos Cardíacos/metabolismo , Piperazinas/administração & dosagem , Potenciais de Ação/efeitos dos fármacos , Animais , Fibrilação Atrial/complicações , Fibrilação Atrial/fisiopatologia , Modelos Animais de Doenças , Cães , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/administração & dosagem , Seguimentos , Átrios do Coração/metabolismo , Átrios do Coração/patologia , Átrios do Coração/fisiopatologia , Sistema de Condução Cardíaco/fisiopatologia , Insuficiência Cardíaca/etiologia , Insuficiência Cardíaca/fisiopatologia , Ventrículos do Coração/metabolismo , Ventrículos do Coração/patologia , Ventrículos do Coração/fisiopatologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Técnicas de Patch-Clamp , Ranolazina , Bloqueadores dos Canais de Sódio
14.
J Am Coll Cardiol ; 57(8): 986-93, 2011 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-21329846

RESUMO

OBJECTIVES: The purpose of this study was to determine the electrophysiologic effects of simvastatin in canine pulmonary vein (PV) sleeve preparations. BACKGROUND: Ectopic activity arising from the PV plays a prominent role in the development of atrial fibrillation. METHODS: Transmembrane action potentials were recorded from canine superfused left superior or inferior PV sleeves using standard microelectrode techniques. Acetylcholine (1 µM), isoproterenol (1 µM), high calcium ([Ca(2+)](o) = 5.4 mM), or a combination was used to induce early afterdepolarizations or delayed afterdepolarizations and triggered activity. Voltage clamp experiments were performed in the left atrium measuring fast and late sodium currents. RESULTS: Under steady-state conditions, simvastatin (10 nM, n = 9) induced a small increase in action potential duration measured at 85% repolarization and a significant decrease in action potential amplitude, take-off potential, and maximum rate of rise of action potential upstroke at the fastest rates. The V(max) decreased from 175.1 ± 34 V/s to 151.7 ± 28 V/s and from 142 ± 47 V/s to 97.4 ± 39 V/s at basic cycle lengths of 300 and 200 ms, respectively. Simvastatin (10 to 20 nM) eliminated delayed afterdepolarizations and delayed afterdepolarization-induced triggered activity in 7 of 7 PV sleeve preparations and eliminated or reduced late-phase 3 early afterdepolarizations in 6 of 6 PV sleeve preparations. Simvastatin (20 nM) did not affect late or fast sodium currents measured using voltage clamp techniques. CONCLUSIONS: Our data suggest that in addition to its upstream actions to reduce atrial structural remodeling, simvastatin exerts a direct antiarrhythmic effect by suppressing triggers responsible for the genesis of atrial fibrillation.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Veias Pulmonares/efeitos dos fármacos , Sinvastatina/farmacologia , Acetilcolina/farmacologia , Potenciais de Ação/fisiologia , Animais , Antiarrítmicos/farmacologia , Células Cultivadas , Cães , Relação Dose-Resposta a Droga , Técnicas Eletrofisiológicas Cardíacas , Isoproterenol/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Modelos Animais , Células Musculares/efeitos dos fármacos , Células Musculares/fisiologia , Veias Pulmonares/fisiologia , Distribuição Aleatória , Valores de Referência , Sensibilidade e Especificidade
15.
Ann Biomed Eng ; 37(7): 1294-309, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19353268

RESUMO

Determining the effect of a compound on I (Kr) is a standard screen for drug safety. Often the effect is described using a single IC(50) value, which is unable to capture complex effects of a drug. Using verapamil as an example, we present a method for using recordings from native myocytes at several drug doses along with qualitative features of I (Kr) from published studies of HERG current to estimate parameters in a mathematical model of the drug effect on I (Kr). I (Kr) was recorded from canine left ventricular myocytes using ruptured patch techniques. A voltage command protocol was used to record tail currents at voltages from -70 to -20 mV, following activating pulses over a wide range of voltages and pulse durations. Model equations were taken from a published I (Kr) Markov model and the drug was modeled as binding to the open state. Parameters were estimated using a combined global and local optimization algorithm based on collected data with two additional constraints on I (Kr) I-V relation and I (Kr) inactivation. The method produced models that quantitatively reproduce both the control I (Kr) kinetics and dose dependent changes in the current. In addition, the model exhibited use and rate dependence. The results suggest that: (1) the technique proposed here has the practical potential to develop data-driven models that quantitatively reproduce channel behavior in native myocytes; (2) the method can capture important drug effects that cannot be reproduced by the IC(50) method. Although the method was developed for I (Kr), the same strategy can be applied to other ion channels, once appropriate channel-specific voltage protocols and qualitative features are identified.


Assuntos
Canais de Potássio Éter-A-Go-Go/metabolismo , Ativação do Canal Iônico/fisiologia , Potenciais da Membrana/fisiologia , Modelos Cardiovasculares , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Verapamil/administração & dosagem , Animais , Antiarrítmicos/administração & dosagem , Células Cultivadas , Simulação por Computador , Cães , Relação Dose-Resposta a Droga , Canal de Potássio ERG1 , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Cinética , Potenciais da Membrana/efeitos dos fármacos , Taxa de Depuração Metabólica , Potássio/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa