Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Membr Biol ; 207(2): 107-17, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16477531

RESUMO

Voltage- and/or conformation-dependent association and dissociation of local anesthetic-class drugs from a putative receptor site in domain IV S6 of the sodium channel and slow conformation transitions of the drug-associated channel have been proposed as mechanisms of use- and frequency-dependent reduction in sodium current. To distinguish these possibilities, we have explored the reactivity to covalent modification by thiols and block of the mutations F1760C and F1760A at the putative receptor site of the cardiac sodium channel expressed as stable cell lines in HEK-293 cells. Both mutations decreased steady-state fast inactivation, shifting V1/2h from -86 +/- 1.3 mV (WT) to -72.3 +/- 1.4 mV (F1760C) and -67.7 +/- 1 mV (F1760A). In the absence of drug, the F1760C mutant channel displayed use-dependent current reduction during pulse-train stimulation, and faster onset of slow inactivation. This mutant also retained some sensitivity to lidocaine. In contrast, the F1760A mutant showed no use-dependent current reduction or sensitivity to lidocaine. The covalent-modifying agent MTS-ET enhanced use-dependent current reduction of the F1760C mutant channel only. The use-dependent reduction in current of the covalently modified channel completely recovered with rest. Lidocaine produced no additional block during exposure to MTS-ET-treated cells (MTS-ET 43 +/- 2.7%: MTS-ET lidocaine 47 +/- 4.5%), implying interaction at a common binding site. The data suggest that use-dependent binding at the F1760 site results in enhanced slow inactivation rather than alteration of drug association and dissociation from that site and may be a general mechanism of action of sodium-channel blocking agents.


Assuntos
Inativação Gênica , Proteínas Musculares/fisiologia , Fenilalanina/fisiologia , Canais de Sódio/fisiologia , Antiarrítmicos/farmacologia , Inativação Gênica/efeitos dos fármacos , Humanos , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5 , Técnicas de Patch-Clamp , Fenilalanina/genética , Fenilalanina/metabolismo , Canais de Sódio/genética , Canais de Sódio/metabolismo
2.
Cardiovasc Res ; 57(4): 1062-71, 2003 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-12650884

RESUMO

OBJECTIVE: The fate of an impulse arising from stimulation is determined by the ability of the wave front to recruit sufficient Na channels from adjacent cells. Previous numerical studies of mutant Na channels revealed both the velocity of a conditioning wave and the recruiting capacity of the front as determinants of the vulnerable period (VP), an interval within which excitation results in unidirectional conduction. Drugs that block excitatory Na channels in a voltage dependent manner, such as antiarrhythmics, abused substances and antidepressants, slow the restoration of Na conductance trailing an action potential and are associated with proarrhythmia and sudden cardiac death. We hypothesized that drug-induced slowing of Na conductance recovery would flatten the Na conductance restoration gradient thereby reducing the recruiting capacity of a front, extending the VP and increasing the probability of unidirectional propagation. METHODS: In a cable of ventricular cells, we explored the sensitivity of the VP to voltage-dependent blockade. While varying the unbinding time constant from 100 ms to 5 s, we measured the Na conductance restoration gradient, the liminal length, the refractory period (RP) and the VP. RESULTS: Reducing the rate of drug unbinding flattened the restoration gradient, diminished the recruiting capacity of a premature impulse and extended the liminal length, RP and the VP. The VP was linearly dependent on the drug unbinding time constant. Rapidly unbinding drugs (time constant <1 s) reduced the liminal length below that of a quiescent cable. CONCLUSIONS: Slowing the unbinding rate of voltage-dependent drug block of Na channels extended the RP and the VP. Drugs with unbinding time constants greater than 1 s dramatically increased the probability of unidirectional propagation, reflecting increases in both the RP and the VP. This study provides a new mechanism linking Na channel function, compromised by voltage-dependent Na channel drug block, with proarrhythmic conditions that can lead to sudden cardiac death following premature stimulation.


Assuntos
Antiarrítmicos/farmacologia , Simulação por Computador , Sistema de Condução Cardíaco/efeitos dos fármacos , Modelos Cardiovasculares , Canais de Sódio/fisiologia , Potenciais de Ação/efeitos dos fármacos , Humanos , Período Refratário Eletrofisiológico , Bloqueadores dos Canais de Sódio/farmacologia
3.
Am J Med ; 110(4): 296-305, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11239848

RESUMO

The sodium channel is an integral membrane protein that plays a central role in conduction of the cardiac impulse in working cardiac myocytes and cells of the His-Purkinje system. The channel has two fundamental properties, ion conduction and gating. Specific domains of the channel protein control each of these functions. Ion conduction describes the mechanisms of the selective movement of sodium ion across the pore in the cell membrane. The selectivity of the channel for sodium ions is at least 10 times greater than that for other monovalent cations; the channel does not normally conduct divalent cations. Gating describes the opening and closing of the sodium channel pore. Sodium channels open transiently during membrane depolarization and close by a process termed inactivation. The cardiac sodium channel protein is a multimeric complex consisting of an alpha and an auxiliary beta-subunit. The genes encoding the sodium channel have been cloned and sequenced. The alpha subunit gene, SCN5A is sufficient to express a functional channel. However, beta subunit co-expression increases the level of channel expression and alters the voltage dependence of inactivation. Mutations of the sodium channel may result in incomplete inactivation during maintained depolarization, a decrease in the level of channel expression or acceleration of inactivation. The resulting clinical phenotypes include long QT syndrome, type III (LQT III), Brugada syndrome, and heart block. LQT III and Brugada syndromes have a high case fatality rate and are best treated with an implantable defibrillator.


Assuntos
Arritmias Cardíacas/metabolismo , Sistema de Condução Cardíaco/metabolismo , Mutação , Canais de Sódio , Arritmias Cardíacas/genética , Arritmias Cardíacas/fisiopatologia , Eletrocardiografia , Bloqueio Cardíaco/metabolismo , Sistema de Condução Cardíaco/fisiopatologia , Humanos , Síndrome do QT Longo/metabolismo , Canais de Sódio/genética , Taquicardia Ventricular/metabolismo
5.
Biophys J ; 79(6): 3019-35, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11106609

RESUMO

The role of inactivation as a central mechanism in blockade of the cardiac Na(+) channel by antiarrhythmic drugs remains uncertain. We have used whole-cell and single channel recordings to examine the block of wild-type and inactivation-deficient mutant cardiac Na(+) channels, IFM/QQQ, stably expressed in HEK-293 cells. We studied the open-channel blockers disopyramide and flecainide, and the lidocaine derivative RAD-243. All three drugs blocked the wild-type Na(+) channel in a use-dependent manner. There was no use-dependent block of IFM/QQQ mutant channels with trains of 20 40-ms pulses at 150-ms interpulse intervals during disopyramide exposure. Flecainide and RAD-243 retained their use-dependent blocking action and accelerated macroscopic current relaxation. All three drugs reduced the mean open time of single channels and increased the probability of their failure to open. From the abbreviation of the mean open times, we estimated association rates of approximately 10(6)/M/s for the three drugs. Reducing the burst duration contributed to the acceleration of macroscopic current relaxation during exposure to flecainide and RAD-243. The qualitative differences in use-dependent block appear to be the result of differences in drug dissociation rate. The inactivation gate may play a trapping role during exposure to some sodium channel blocking drugs.


Assuntos
Antiarrítmicos/farmacologia , Disopiramida/farmacologia , Flecainida/farmacologia , Coração/fisiologia , Lidocaína/análogos & derivados , Canais de Sódio/química , Canais de Sódio/fisiologia , Substituição de Aminoácidos , Linhagem Celular , Clonagem Molecular , Humanos , Lidocaína/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Modelos Biológicos , Miocárdio/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Bloqueadores dos Canais de Sódio , Transfecção
6.
Circ Res ; 86(4): 441-7, 2000 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-10700449

RESUMO

The cytoskeleton of the cardiomyocyte has been shown to modulate ion channel function. Cytoskeletal disruption in vitro alters Na(+) channel kinetics, producing a late Na(+) current that can prolong repolarization. This study describes the properties of the cardiac Na(+) channel and cardiac repolarization in neonatal mice lacking ankyrin(B), a cytoskeletal "adaptor" protein. Using whole-cell voltage clamp techniques, I(Na) density was lower in ankyrin(B)(-/-) ventricular myocytes than in wild-type (WT) myocytes (-307+/-26 versus -444+/-39 pA/pF, P<0.01). Ankyrin(B)(-/-) myocytes exhibited a hyperpolarizing shift in activation and inactivation kinetics compared with WT. Slower recovery from inactivation contributed to the negative shift in steady-state inactivation in ankyrin(B)(-/-). Single Na(+) channel mean open time was longer in ankyrin(B)(-/-) versus WT at test potentials (V(t)) of -40 mV (1.0+/-0.1 versus 0. 61+/-0.04 ms, P<0.05) and -50 mV (0.8+/-0.1 versus 0.39+/-0.05 ms, P<0.05). Ankyrin(B)(-/-) exhibited late single-channel openings at V(t) -40 and -50 mV, which were not seen in WT. Late I(Na) contributed to longer action potential durations measured at 90% repolarization (APD(90)) at 1 Hz stimulation in ankyrin(B)(-/-) compared with WT (354+/-26 versus 274+/-22 ms, P<0.05). From ECG recordings of neonatal mice, heart rates were slower in ankyrin(B)(-/-) than in WT (380+/-14 versus 434+/-13 bpm, P<0.01). Although the QT interval was similar in ankyrin(B)(-/-) and WT at physiological heart rates, QT-interval prolongation in response to heart rate deceleration was greater in ankyrin(B)(-/-). In conclusion, Na(+) channels in ankyrin(B)(-/-) display reduced I(Na) density and abnormal kinetics at the whole-cell and single-channel level that contribute to prolonged APD(90) and abnormal QT-rate adaptation.


Assuntos
Anquirinas/fisiologia , Frequência Cardíaca , Síndrome do QT Longo/fisiopatologia , Miocárdio/metabolismo , Canais de Sódio/metabolismo , Potenciais de Ação , Adaptação Fisiológica , Animais , Animais Recém-Nascidos/fisiologia , Anquirinas/deficiência , Anquirinas/genética , Eletrocardiografia , Camundongos , Camundongos Knockout/genética , Miocárdio/patologia , Técnicas de Patch-Clamp , Valores de Referência
7.
Cardiovasc Res ; 42(2): 490-502, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10533584

RESUMO

OBJECTIVE: Prior studies of the modulation of the Na+ current by sympathetic stimulation have yielded controversial results. Separation of the Na+ and Ca2+ currents poses a problem in myocyte preparations. The gating of cloned Na+ channels is different in oocytes compared with mammalian expression systems. We have examined the sympathetic modulation of the alpha-subunit of the wild-type human cardiac Na+ channel (hH1) and the long QT-associated mutant, delta KPQ, expressed in human embryonic kidney cells. METHODS: Stable cell lines of hH1 and delta KPQ were established in human embryonic kidney cells. Whole-cell and single-channel currents were measured with the patch-clamp technique. Sympathetic stimulation was effected by exposure to isoproterenol or 8-bromo-cAMP. Na+ channel activation and inactivation were determined using standard voltage clamp protocols. Ca2+:Na+ permeability ratio was determined under bi-ionic conditions. RESULTS: We observed a qualitatively different effect of sympathetic stimulation on the cardiac Na+ current from that reported in frog oocytes: activation and inactivation kinetics were shifted to more negative potentials. This shift was similar for both hH1 and delta KPQ. [delta V0.5 for inactivation: 8.3 +/- 1.7 mV, p < 0.001 (hH1); 6.8 +/- 0.9 mV, p < 0.001 (delta KPQ)]. Increased rate of closed-state inactivation contributed to the shifting of the inactivation-voltage relationship. Open-state inactivation was not affected as mean open times were unchanged. Reversal potential measurement in hH1 suggested a low Ca2+:Na+ permeability ratio of 0.017, uninfluenced by sympathetic stimulation. In delta KPQ, the size of the persistent relative to the peak current was increased with 8-bromo-cAMP from 3.0 +/- 0.7% to 4.3 +/- 0.6% (p = 0.056). CONCLUSIONS: Sympathetic stimulation exerts multiple effects on the gating of hH1. Similar effects are also seen in delta KPQ which may increase arrhythmia susceptibility in long QT syndrome by modifying the Na+ channel contribution to the action potential.


Assuntos
Agonistas Adrenérgicos beta/farmacologia , Ativação do Canal Iônico/efeitos dos fármacos , Isoproterenol/farmacologia , Síndrome do QT Longo/metabolismo , Miocárdio/metabolismo , Canais de Sódio/efeitos dos fármacos , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Canais de Cálcio/efeitos dos fármacos , Linhagem Celular , Humanos , Mutação , Técnicas de Patch-Clamp , Sódio/metabolismo , Canais de Sódio/genética , Estimulação Química
8.
Am J Cardiol ; 82(8A): 43N-49N, 1998 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-9809900

RESUMO

Drugs remain the mainstay of treatment of patients with atrial fibrillation (AF). An understanding of the mechanisms of AF and of atrioventricular (AV) conduction provides a basis for the understanding of the mechanisms of antiarrhythmic drug action. Although ectopic activity from a focus may initiate AF, re-entry is the usual mechanism of maintenance. In its classical form, reentry takes the form of circus movements around fixed anatomic structures. However, leading circle and anisotropic variants of reentry may arise as a result of functional variations of refractoriness or anistropic conduction. Electrical remodeling during AF favors its persistence. Reentry may be prevented by prolongation of the refractory period. Class III antiarrhythmic drugs prolong refractoriness by blockade of outward potassium currents. Class I drugs prolong refractoriness by delaying the recovery of of the sodium current. Many class I drugs also have potassium channel-blocking action. In AF the rate of conduction of rapid impulses to the ventricle is controlled by conduction over the AV node. Blockade of the L-type calcium channels, activation of the muscarinic and adenosine A1 receptors, or beta-adrenergic blockade will slow conduction over the AV node. The adverse cardiovascular effects of drugs used to treat AF can be predicted on the basis of their mechanisms of action. The current focus of drug development is on specific potassium channel blockers.


Assuntos
Antiarrítmicos/farmacologia , Fibrilação Atrial/tratamento farmacológico , Fibrilação Atrial/fisiopatologia , Animais , Antiarrítmicos/classificação , Função Atrial/fisiologia , Eletrofisiologia , Sistema de Condução Cardíaco/efeitos dos fármacos , Sistema de Condução Cardíaco/fisiopatologia , Humanos , Canais Iônicos/efeitos dos fármacos
9.
Am J Physiol ; 274(5): H1643-54, 1998 05.
Artigo em Inglês | MEDLINE | ID: mdl-9612375

RESUMO

Several aspects of the effect of the KPQ deletion mutation on Na+ channel gating remain unresolved. We have analyzed the kinetics of the early and late currents by recording whole cell and single-channel currents in a human embryonic kidney (HEK) cell line (HEK293) expressing wild-type and KPQ deletion mutation in cardiac Na+ channels. The rate of inactivation increased three- to fivefold between -40 and -80 mV in the mutant channel. The rate of recovery from inactivation was increased twofold. Two modes of gating accounted for the late current: 1) isolated brief openings with open times that were weakly voltage dependent and the same as the initial transient and 2) bursts of opening with highly voltage-dependent prolonged open times. Latency to first opening was accelerated, suggesting an acceleration of the rate of activation. The delta KPQ mutation has multiple effects on activation and inactivation. The aggregate effects may account for the increased susceptibility to arrhythmias.


Assuntos
Coração/fisiologia , Ativação do Canal Iônico/genética , Canais de Sódio/fisiologia , Animais , Linhagem Celular , Eletrofisiologia , Predisposição Genética para Doença , Humanos , Síndrome do QT Longo/genética , Síndrome do QT Longo/fisiopatologia , Deleção de Sequência
14.
Cardiovasc Drugs Ther ; 11(2): 149-67, 1997 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9140692

RESUMO

The goal of developing an antiarrhythmic agent effective against malignant ventricular arrhythmias while maintaining a low side-effect profile remains elusive. The class III drugs amiodarone and sotalol are the best available agents. However, both drugs possess properties outside the realm of a pure class III effect, and their use is limited by a variety of dose-related side effects. There are several drugs with more selective class III properties currently in development. This review provides an overview of the optimal characteristics of an effective theoretical class III drug and a summary of the properties of a number of class III drugs under active investigation. An ideal class III antiarrhythmic agent for a reentrant arrhythmia should provide use-dependent prolongation of the action potential duration with slow onset and rapid offset kinetics. This drug would prolong the effective refractory period of cardiac tissue selectively at the rapid heart rates achieved during ventricular tachycardia or fibrillation with a delayed onset of action, and a rapid resolution of its effects on resumption of physiologic heart rates. With little effect on the refractory period at normal or slow heart rates, the ability to induce torsade de pointes would be lessened. In contrast to these ideal properties, most currently available and investigational agents have a reverse use-dependent effect on the action potential duration, producing more effects on the refractory period at slower heart rates. This property results in part from preferential block of the rapidly activating component of the delayed rectifier potassium channel (IKr), with little or no effect on the slowly activating component (IKs). The development of a drug with favorable blocking kinetics that selectively blocks IKs may results in lower proarrhythmic events while still maintaining effective antiarrhythmic properties.


Assuntos
Antiarrítmicos/farmacologia , Arritmias Cardíacas/tratamento farmacológico , Animais , Antiarrítmicos/efeitos adversos , Antiarrítmicos/uso terapêutico , Arritmias Cardíacas/induzido quimicamente , Arritmias Cardíacas/fisiopatologia , Humanos
15.
Pacing Clin Electrophysiol ; 20(2 Pt 2): 432-44, 1997 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9058847

RESUMO

Over the past decade, the strategies for arrhythmia management have been in transition. Physical methods to treat arrhythmias, such as ICDs and RF ablation, have undergone considerable refinement and wider application. Ischemic heart disease and congestive heart failure have been identified as clinical situations in which antiarrhythmic drugs have a significant proarrhythmic potential. However, drugs retain an important role in arrhythmia management. Strategies to mitigate the structural and functional changes that occur in hypertrophy, ischemia, and infarction have not been thoroughly explored. Membrane ion channels and receptors are the targets for the action of currently available drugs. The cloning and sequencing of these ion channels and receptors should improve the efficacy and specificity of drug design. Cardiac Na+, Ca2+, K+, and nonspecific cation channels have a clearly defined role in the generation of the normal action potential. Their specific roles in the various clinical arrhythmias is less certain. There are sufficient data to associate specific ionic channels with normal and abnormal automaticity and with reentry occurring in specific regions of the heart. A rational choice of antiarrthymic drugs can be made when an arrhythmogenic mechanism and the putative underlying membrane currents can be identified based on the clinical characteristics of the arrhythmia. For a majority of clinical arrhythmias, this ideal has not been achieved. When a particular drug is used to treat an arrhythmia, the full complement of its actions will depend on which multiple ion channels or receptors are blocked and the kinetics of drug interaction with these sites.


Assuntos
Antiarrítmicos/metabolismo , Antiarrítmicos/farmacologia , Arritmias Cardíacas/tratamento farmacológico , Arritmias Cardíacas/fisiopatologia , Ativação do Canal Iônico/efeitos dos fármacos , Canais Iônicos/efeitos dos fármacos , Arritmias Cardíacas/etiologia , Humanos , Ativação do Canal Iônico/fisiologia , Canais Iônicos/metabolismo , Canais Iônicos/fisiologia , Miocárdio/patologia
16.
J Clin Invest ; 99(2): 288-96, 1997 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-9005997

RESUMO

Our laboratory has been testing the hypothesis that genetic modulation of the beta-adrenergic signaling cascade can enhance cardiac function. We have previously shown that transgenic mice with cardiac overexpression of either the human beta2-adrenergic receptor (beta2AR) or an inhibitor of the beta-adrenergic receptor kinase (betaARK), an enzyme that phosphorylates and uncouples agonist-bound receptors, have increased myocardial inotropy. We now have created recombinant adenoviruses encoding either the beta2AR (Adeno-beta2AR) or a peptide betaARK inhibitor (consisting of the carboxyl terminus of betaARK1, Adeno-betaARKct) and tested their ability to potentiate beta-adrenergic signaling in cultured adult rabbit ventricular myocytes. As assessed by radioligand binding, Adeno-beta2AR infection led to approximately 20-fold overexpression of beta-adrenergic receptors. Protein immunoblots demonstrated the presence of the Adeno-betaARKct transgene. Both transgenes significantly increased isoproterenol-stimulated cAMP as compared to myocytes infected with an adenovirus encoding beta-galactosidase (Adeno-betaGal) but did not affect the sarcolemmal adenylyl cyclase response to Forskolin or NaF. beta-Adrenergic agonist-induced desensitization was significantly inhibited in Adeno-betaARKct-infected myocytes (16+/-2%) as compared to Adeno-betaGal-infected myocytes (37+/-1%, P < 0.001). We conclude that recombinant adenoviral gene transfer of the beta2AR or an inhibitor of betaARK-mediated desensitization can potentiate beta-adrenergic signaling.


Assuntos
Adenoviridae/genética , Técnicas de Transferência de Genes , Ventrículos do Coração/metabolismo , Receptores Adrenérgicos beta 2/genética , Receptores Adrenérgicos beta 2/metabolismo , Adenilil Ciclases/análise , Agonistas Adrenérgicos beta/farmacologia , Animais , Sobrevivência Celular , Células Cultivadas , AMP Cíclico/metabolismo , Vetores Genéticos , Ventrículos do Coração/citologia , Humanos , Isoproterenol/farmacologia , Masculino , Coelhos , Sarcolema/enzimologia , Transdução de Sinais , Transgenes
17.
Mol Pharmacol ; 50(6): 1643-50, 1996 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8967988

RESUMO

Inactivation has been implicated as an important determinant of the block of Na+ channel by local anesthetic-class drugs. This proposition has been difficult to examine because agents used to modify inactivation change other channel properties and both inactivated and blocked channels do not conduct. We used site-directed mutagenesis of Phe1304 to glutamine in the linker between the third and fourth domains of the mu-1 Na+ channel to slow inactivation. Wild-type and mutant channels were expressed in frog oocytes. Macropatch and single-channel currents were recorded in cell-attached membrane patches. The F1304Q mutation increased mean open time (1.7 fold at -20 mV) and reduced the probability that the channel would fail to open. Closed times were best fit by a double-exponential function, suggesting that the inactivated state transitions were no longer absorbing. In wild-type channels, 100 microM disopyramide decreased mean open time from 1.64 +/- 0.08 to 0.34 +/- 0.04 msec. Total open time per trial was decreased 2-fold. There also was a marked increase in the fraction of null sweeps. In the inactivation-deficient mutant channel, mean and total open times were also reduced. These data indicate that even when inactivation is slowed by a localized specific mutation, open-channel block by disopyramide persists. Inactivation may not be a necessary requirement for open-channel block.


Assuntos
Bloqueadores dos Canais de Sódio , Clonagem Molecular , Disopiramida/farmacologia , Glutamina/genética , Ativação do Canal Iônico/genética , Cinética , Mutagênese Sítio-Dirigida , Fenilalanina/genética , Canais de Sódio/genética
18.
J Cardiovasc Electrophysiol ; 7(4): 353-64, 1996 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8777484

RESUMO

Propafenone is a sodium channel blocking antiarrhythmic drug. It also has beta-adrenergic, potassium channel, and weak calcium channel blocking activity. The drug is metabolized in the liver with rates dependent on the debrisoquin phenotype. The saturable metabolism results in nonlinear pharmacokinetics. The metabolites retain sodium channel blocking activity but little beta-adrenergic blocking activity. Both controlled and noncontrolled studies have documented its efficacy in a variety of supraventricular arrhythmias. Intravenous propafenone is effective in converting atrial fibrillation to normal sinus rhythm. Chronic oral administration decreases the frequency of recurrence of atrial fibrillation and paroxysmal supraventricular tachycardia. The drug is particularly effective in the Wolff-Parkinson-White syndrome. The drug may produce SA block in patients with underlying sinus node dysfunction. Propafenone has comparatively few noncardiac side effects. It is a useful primary drug or an alternative to more commonly used drugs used for the treatment of supraventricular arrhythmias.


Assuntos
Antiarrítmicos/uso terapêutico , Arritmias Cardíacas/tratamento farmacológico , Propafenona/uso terapêutico , Disfunção Ventricular/tratamento farmacológico , Animais , Antiarrítmicos/farmacocinética , Antiarrítmicos/farmacologia , Humanos , Propafenona/farmacocinética , Propafenona/farmacologia , Taquicardia Supraventricular/tratamento farmacológico
19.
Am J Physiol ; 270(1 Pt 2): H386-97, 1996 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8769775

RESUMO

Recent voltage-clamp studies of isolated myocytes have demonstrated widespread occurrence of a transient outward current (I(to)) carried by potassium ions. In the canine ventricle, this current is well developed in epicardial cells but not in endocardial cells. The resultant spatial dispersion of refractoriness is potentially proarrhythmic and may be amplified by channel blockade. The inactivation and recovery time constants of this channel are in excess of several hundred milliseconds, and consequently channel availability is frequency dependent at physiological stimulation rates. When the time constants associated with transitions between different channel conformations are rapid relative to drug binding kinetics, the interactions between drugs and an ion channel can be approximated by a sequence of first-order reactions, in which binding occurs in pulses in response to pulse train stimulation (pulse chemistry). When channel conformation transition time constants do not meet this constraint, analytical characterizations of the drug-channel interaction must then be modified to reflect the channel time-dependent properties. Here we report that the rate and steady-state amount of frequency-dependent inactivation of I(to) are consistent with a generalization of the channel blockade model: channel availability is reduced in a pulsatile exponential pattern as the stimulation frequency is increased, and the rate of reduction is a linear function of the pulse train depolarizing and recovery intervals. I(to) was reduced in the presence of quinidine. After accounting for the use-dependent availability of I(to) channels, we found little evidence of an additional use-dependent component of block after exposure to quinidine, suggesting that quinidine reacts with both open and closed I(to) channels as though the binding site is continuously accessible. The model provides a useful tool for assessing drug-channel interactions when the reaction cannot be continuously monitored.


Assuntos
Coração/fisiologia , Modelos Cardiovasculares , Modelos Químicos , Canais de Potássio/fisiologia , Animais , Condutividade Elétrica , Estimulação Elétrica , Cinética , Bloqueadores dos Canais de Potássio , Quinidina/farmacologia , Coelhos , Fatores de Tempo
20.
Pacing Clin Electrophysiol ; 18(9 Pt 1): 1686-704, 1995 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-7491312

RESUMO

Antiarrhythmic drugs have relative specificity for blocking each of the major classes of ion channels that control the action potential. The kinetics of block is determined by the state of the channel. Those channel states occupied at depolarized potentials generally have greater affinity for the blocking drugs. The kinetics of the drug-channel interaction is important in determining the blocking profile observed clinically. The increased mortality resulting from drug treatment in CAST and several atrial fibrillation trials has resulted in a shift in antiarrhythmic drug development from the Na+ channel blocking (Class I) drugs to the K+ channel blocking (Class III) drugs. While both Classes of drugs have a proarrhythmic potential, this may be less for the Class III agents. Their lack of negative inotropy also make them more attractive. It is important that the potential advantages of these agents be evaluated in controlled clinical trials. In several laboratories, the techniques of molecular biology and biophysics are being combined to determine the block site of available drugs. This information will aid in the future development of agents with greater specificity, and hopefully greater efficacy and safety than those currently in clinical use.


Assuntos
Antiarrítmicos/farmacologia , Coração/fisiologia , Canais Iônicos/antagonistas & inibidores , Potenciais de Ação/efeitos dos fármacos , Antiarrítmicos/classificação , Bloqueadores dos Canais de Cálcio/farmacologia , Ensaios Clínicos Controlados como Assunto , Desenho de Fármacos , Eletrofisiologia , Coração/efeitos dos fármacos , Humanos , Canais Iônicos/efeitos dos fármacos , Bloqueadores dos Canais de Potássio , Canais de Potássio/efeitos dos fármacos , Bloqueadores dos Canais de Sódio , Canais de Sódio/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...