Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Front Physiol ; 15: 1399037, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39092426

RESUMEN

Introduction: The mechanisms leading to the conversion of atrial fibrillation (AF) to sinus rhythm are poorly understood. This study describes the dynamic behavior of electrophysiological parameters and conduction patterns leading to spontaneous and pharmacological AF termination. Methods: Five independent groups of goats were investigated: (1) spontaneous termination of AF, and drug-induced terminations of AF by various potassium channel inhibitors: (2) AP14145, (3) PA-6, (4) XAF-1407, and (5) vernakalant. Bi-atrial contact mapping was performed during an open chest surgery and intervals with continuous and discrete atrial activity were determined. AF cycle length (AFCL), conduction velocity and path length were calculated for each interval, and the final conduction pattern preceding AF termination was evaluated. Results: AF termination was preceded by a sudden episode of discrete activity both in the presence and absence of an antiarrhythmic drug. This episode was accompanied by substantial increases in AFCL and conduction velocity, resulting in prolongation of path length. In 77% ± 4% of all terminations the conduction pattern preceding AF termination involved medial to lateral conduction along Bachmann's bundle into both atria, followed by anterior to posterior conduction. This finding suggests conduction block in the interatrial septum and/or pulmonary vein area as final step of AF termination. Conclusion: AF termination is preceded by an increased organization of fibrillatory conduction. The termination itself is a sudden process with a critical role for the interplay between spatiotemporal organization and anatomical structure.

2.
Heart Rhythm O2 ; 1(3): 206-214, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32864638

RESUMEN

BACKGROUND: Atrial-ventricular differences in voltage-gated Na+ currents might be exploited for atrial-selective antiarrhythmic drug action for the suppression of atrial fibrillation without risk of ventricular tachyarrhythmia. Eleclazine (GS-6615) is a putative antiarrhythmic drug with properties similar to the prototypical atrial-selective Na+ channel blocker ranolazine that has been shown to be safe and well tolerated in patients. OBJECTIVE: The present study investigated atrial-ventricular differences in the biophysical properties and inhibition by eleclazine of voltage-gated Na+ currents. METHODS: The fast and late components of whole-cell voltage-gated Na+ currents (respectively, I Na and I NaL) were recorded at room temperature (∼22°C) from rat isolated atrial and ventricular myocytes. RESULTS: Atrial I Na activated at command potentials ∼5.5 mV more negative and inactivated at conditioning potentials ∼7 mV more negative than ventricular I Na. There was no difference between atrial and ventricular myocytes in the eleclazine inhibition of I NaL activated by 3 nM ATX-II (IC50s ∼200 nM). Eleclazine (10 µM) inhibited I Na in atrial and ventricular myocytes in a use-dependent manner consistent with preferential activated state block. Eleclazine produced voltage-dependent instantaneous inhibition in atrial and ventricular myocytes; it caused a negative shift in voltage of half-maximal inactivation and slowed the recovery of I Na from inactivation in both cell types. CONCLUSIONS: Differences exist between rat atrial and ventricular myocytes in the biophysical properties of I Na. The more negative voltage dependence of I Na activation/inactivation in atrial myocytes underlies differences between the 2 cell types in the voltage dependence of instantaneous inhibition by eleclazine. Eleclazine warrants further investigation as an atrial-selective antiarrhythmic drug.

3.
J Cardiovasc Pharmacol ; 66(5): 412-31, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25978691

RESUMEN

There is a clear unmet medical need for new pharmacologic therapies with improved efficacy and safety for the treatment of atrial fibrillation. Considerable research efforts have been undertaken to discover and develop new safe and effective antiarrhythmic drugs that specifically target atrial K(+) channels. To realize the full value of these novel atrial-specific therapeutic drug targets, demonstration of clinical efficacy and safety is required for a new breed of atrial-selective antiarrhythmic drugs. The reward for demonstrating this in a pivotal phase III trial, on regulatory approval, will be "first-in-class" status. This article reviews the development status of new and novel K channel inhibitors currently in drug development as atrial-selective antiarrhythmics for the treatment of atrial fibrillation.


Asunto(s)
Antiarrítmicos/uso terapéutico , Fibrilación Atrial/tratamiento farmacológico , Descubrimiento de Drogas , Drogas en Investigación/uso terapéutico , Sistema de Conducción Cardíaco/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Bloqueadores de los Canales de Potasio/uso terapéutico , Canales de Potasio/efectos de los fármacos , Potenciales de Acción , Animales , Antiarrítmicos/efectos adversos , Fibrilación Atrial/diagnóstico , Fibrilación Atrial/metabolismo , Fibrilación Atrial/fisiopatología , Drogas en Investigación/efectos adversos , Sistema de Conducción Cardíaco/metabolismo , Sistema de Conducción Cardíaco/fisiopatología , Humanos , Bloqueadores de los Canales de Potasio/efectos adversos , Canales de Potasio/metabolismo , Transducción de Señal/efectos de los fármacos
4.
EMBO Mol Med ; 7(4): 394-410, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25700171

RESUMEN

Drugs targeting atrial-specific ion channels, Kv1.5 or Kir3.1/3.4, are being developed as new therapeutic strategies for atrial fibrillation. However, current preclinical studies carried out in non-cardiac cell lines or animal models may not accurately represent the physiology of a human cardiomyocyte (CM). In the current study, we tested whether human embryonic stem cell (hESC)-derived atrial CMs could predict atrial selectivity of pharmacological compounds. By modulating retinoic acid signaling during hESC differentiation, we generated atrial-like (hESC-atrial) and ventricular-like (hESC-ventricular) CMs. We found the expression of atrial-specific ion channel genes, KCNA5 (encoding Kv1.5) and KCNJ3 (encoding Kir 3.1), in hESC-atrial CMs and further demonstrated that these ion channel genes are regulated by COUP-TF transcription factors. Moreover, in response to multiple ion channel blocker, vernakalant, and Kv1.5 blocker, XEN-D0101, hESC-atrial but not hESC-ventricular CMs showed action potential (AP) prolongation due to a reduction in early repolarization. In hESC-atrial CMs, XEN-R0703, a novel Kir3.1/3.4 blocker restored the AP shortening caused by CCh. Neither CCh nor XEN-R0703 had an effect on hESC-ventricular CMs. In summary, we demonstrate that hESC-atrial CMs are a robust model for pre-clinical testing to assess atrial selectivity of novel antiarrhythmic drugs.


Asunto(s)
Fibrilación Atrial , Sistemas de Liberación de Medicamentos/métodos , Modelos Biológicos , Miocitos Cardíacos/metabolismo , Células Madre Pluripotentes/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Fibrilación Atrial/tratamiento farmacológico , Fibrilación Atrial/metabolismo , Fibrilación Atrial/patología , Evaluación Preclínica de Medicamentos/métodos , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/antagonistas & inhibidores , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/biosíntesis , Expresión Génica , Atrios Cardíacos/metabolismo , Atrios Cardíacos/patología , Humanos , Canal de Potasio Kv1.5/antagonistas & inhibidores , Canal de Potasio Kv1.5/biosíntesis , Miocitos Cardíacos/patología , Células Madre Pluripotentes/patología
5.
Circ Arrhythm Electrophysiol ; 5(6): 1184-92, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23159416

RESUMEN

BACKGROUND: Cardiac ATP-sensitive K(+) channels have been suggested to contribute to the adaptive physiological response to metabolic challenge after ß-adrenoceptor stimulation. However, an increased atrial K(+)-conductance might be expected to be proarrhythmic. We investigated the effect of ATP-sensitive K(+) channel blockade on the electrophysiological responses to ß-adrenoceptor-induced metabolic challenge in intact atria. METHODS AND RESULTS: Atrial electrograms were recorded from the left atrial epicardial surface of Langendorff-perfused rat hearts using a 5×5 electrode array. Atrial effective refractory period and conduction velocity were measured using an S(1)-S(2) protocol. The proportion of hearts in which atrial tachyarrhythmia was produced by burst-pacing was used as an index of atrial tachyarrhythmia-inducibility. Atrial nucleotide concentrations were measured by high performance liquid chromatography. Perfusion with ≥10(-9) mol/L of the ß-adrenoceptor agonist, isoproterenol (ISO), resulted in a concentration-dependent reduction of atrial effective refractory period and conduction velocity. The ISO-induced changes produced a proarrhythmic substrate such that atrial tachyarrhythmia could be induced by burst-pacing. Atrial [ATP] was significantly reduced by ISO (10(-6) mol/L). Perfusion with either of the ATP-sensitive K(+) channel blockers, glibenclamide (10(-5) mol/L) or tolbutamide (10(-3) mol/L), in the absence of ISO had no effect on basal atrial electrophysiology. On the other hand, the proarrhythmic substrate induced by 10(-6) mol/L ISO was abolished by either of the sulfonylureas, which prevented induction of atrial tachyarrhythmia. CONCLUSIONS: Atrial ATP-sensitive K(+) channels activate in response to ß-adrenergic metabolic stress in Langendorff-perfused rat hearts, resulting in a proarrhythmic substrate.


Asunto(s)
Agonistas Adrenérgicos beta/farmacología , Gliburida/farmacología , Atrios Cardíacos/fisiopatología , Canales KATP/efectos de los fármacos , Canales KATP/fisiología , Estrés Fisiológico/efectos de los fármacos , Taquicardia/fisiopatología , Animales , Modelos Animales de Enfermedad , Técnicas Electrofisiológicas Cardíacas , Sistema de Conducción Cardíaco/fisiopatología , Isoproterenol/farmacología , Canales KATP/antagonistas & inhibidores , Masculino , Bloqueadores de los Canales de Potasio/farmacología , Ratas , Estrés Fisiológico/fisiología , Factores de Tiempo , Tolbutamida/farmacología
6.
Drug Discov Today ; 17(13-14): 654-9, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22370250

RESUMEN

Atrial fibrillation (AF) is the most common cardiac arrhythmia facing physicians, afflicting 13% of men and 11% of women over 85 years of age. Epidemiological studies estimate that there are ≥ 11 million AF sufferers in the seven major economies and that its prevalence will increase two- to threefold over the next 50 years. Current strategies for treating AF involve either sinus rhythm (SR) maintenance or heart rate control, combined with anticoagulation therapy. Although SR control is the preferred and most effective treatment of AF, none of the SR control drugs currently available are able to maintain rhythm without significant side effects. In this article we discuss some of the recent advancements in developing new antiarrhythmic drugs for AF.


Asunto(s)
Antiarrítmicos/farmacología , Fibrilación Atrial/tratamiento farmacológico , Descubrimiento de Drogas , Animales , Antiarrítmicos/administración & dosificación , Antiarrítmicos/efectos adversos , Antiarrítmicos/uso terapéutico , Fibrilación Atrial/metabolismo , Fibrilación Atrial/fisiopatología , Ensayos Clínicos como Asunto , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Canales Iónicos/metabolismo , Resultado del Tratamiento
7.
J Pharmacol Toxicol Methods ; 61(2): 178-91, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20172036

RESUMEN

INTRODUCTION: Pharmacological inhibition of cardiac potassium channels encoded by hERG (human ether-à-go-go-related gene) is associated with QT interval prolongation and torsades de pointes arrhythmia. Electrophysiological assays of hERG channel inhibition are integral to the safety testing of novel drug candidates. This study was conducted to compare, for the high affinity hERG inhibitors dofetilide and cisapride, hERG blockade between action potential (AP) and conventional (step and step-ramp) screening waveforms. Furthermore, it evaluated dynamic (pulse-by-pulse) protocol-dependence of hERG channel inhibition by these drugs. METHODS: Whole-cell patch-clamp recordings were made at 37 degrees C from hERG-expressing HEK 293 cells. Half-maximal inhibitory concentrations (IC(50) values) for I(hERG) blockade were obtained using conventional voltage clamp and action potential clamp, using previously digitised ventricular and Purkinje fibre (PF) AP waveforms. RESULTS: A more marked variation in IC(50) values with different command waveforms was observed for cisapride (ranging from 7 to 72 nM) than for dofetilide (ranging from 4 to 15 nM), with higher IC(50)s obtained with AP than step or step-ramp commands. The two drugs differed little from one another in effects on voltage-dependent activation; however, I(hERG) blockade by each drug was initially voltage-dependent, but at steady-state was only voltage-dependent for cisapride. There was comparatively little difference between the two drugs in effects on I(hERG) availability or time constants of development of inactivation. Features of time-dependence of blockade and the use of protocols employing varying rest periods in drug or commands of alternating duration highlighted a pronounced ability of cisapride, but not dofetilide, to dissociate and reassociate from hERG on a pulse-by-pulse basis. DISCUSSION: Protocols described here that demonstrated dynamic variation (drug dissociation/reassociation) in hERG channel current blockade at 37 degrees C for cisapride may have future value for investigating drug interactions with the hERG channel. Downloadable digitised ventricular and PF AP waveforms that can be used in AP clamp experiments also accompany this article.


Asunto(s)
Cisaprida/farmacología , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Fenetilaminas/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Sulfonamidas/farmacología , Potenciales de Acción/efectos de los fármacos , Axones/efectos de los fármacos , Línea Celular , Cisaprida/metabolismo , Interpretación Estadística de Datos , Electrofisiología , Canales de Potasio Éter-A-Go-Go/metabolismo , Humanos , Técnicas de Placa-Clamp , Fenetilaminas/metabolismo , Bloqueadores de los Canales de Potasio/metabolismo , Sulfonamidas/metabolismo , Temperatura
8.
J Cardiovasc Pharmacol ; 52(2): 105-20, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18670369

RESUMEN

There is a clear unmet medical need for new pharmacologic therapies for the treatment of atrial fibrillation (AF) with improved efficacy and safety. This article reviews the development of new and novel Kv1.5/ultra-rapid delayed-rectifier current (I Kur) inhibitors and presents evidence that Kv1.5 modulation provides an atrial-selective mechanism for treating AF. Academia and industry have invested heavily in Kv1.5 (>500 scientific publications and >50 patents published since 1993); however, to realize the full value of this therapeutic drug target, clinical efficacy and safety data are required for a selective Kv1.5 modulator. The reward for demonstrating clinical efficacy and safety in a pivotal Phase 3 trial, on regulatory approval, is "first in class" status.


Asunto(s)
Antiarrítmicos/farmacología , Fibrilación Atrial/metabolismo , Canal de Potasio Kv1.5/metabolismo , Bloqueadores de los Canales de Potasio/uso terapéutico , Animales , Antiarrítmicos/uso terapéutico , Fibrilación Atrial/tratamiento farmacológico , Fibrilación Atrial/fisiopatología , Bloqueadores de los Canales de Potasio/farmacología
9.
Biochem Biophys Res Commun ; 351(1): 273-80, 2006 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-17056009

RESUMEN

The phenothiazine antipsychotic agent thioridazine has been linked with prolongation of the QT interval on the electrocardiogram, ventricular arrhythmias, and sudden death. Although thioridazine is known to inhibit cardiac hERG K(+) channels there is little mechanistic information on this action. We have investigated in detail hERG K(+) channel current (I(hERG)) blockade by thioridazine and identified a key molecular determinant of blockade. Whole-cell I(hERG) measurements were made at 37 degrees C from human embryonic kidney (HEK-293) cells expressing wild-type and mutant hERG channels. Thioridazine inhibited I(hERG) tails at -40mV following a 2s depolarization to +20mV with an IC(50) value of 80nM. Comparable levels of I(hERG) inhibition were seen with physiological command waveforms (ventricular and Purkinje fibre action potentials). Thioridazine block of I(hERG) was only weakly voltage-dependent, though the time dependence of I(hERG) inhibition indicated contingency of blockade upon channel gating. The S6 helix point mutation F656A almost completely abolished, and the Y652A mutation partially attenuated, I(hERG) inhibition by thioridazine. In summary, thioridazine is one of the most potent hERG K(+) channel blockers amongst antipsychotics, exhibiting characteristics of a preferential open/activated channel blocker and binding at a high affinity site in the hERG channel pore.


Asunto(s)
Clorpromazina/administración & dosificación , Canales de Potasio Éter-A-Go-Go/fisiología , Activación del Canal Iónico/fisiología , Riñón/fisiología , Potenciales de la Membrana/fisiología , Antipsicóticos/administración & dosificación , Línea Celular , Relación Dosis-Respuesta a Droga , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/química , Humanos , Riñón/efectos de los fármacos , Potenciales de la Membrana/efectos de los fármacos , Estructura Terciaria de Proteína , Relación Estructura-Actividad
10.
FEBS Lett ; 580(8): 1999-2005, 2006 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-16542653

RESUMEN

The mechanism of human ether-à-go-go-related gene (HERG) K+ channel blockade by the antifungal agent ketoconazole was investigated using patch-clamp recording from mammalian cell lines. Ketoconazole inhibited whole-cell HERG current (IHERG) with a clinically relevant half-maximal inhibitory drug concentration (IC50) value of 1.7 microM. The voltage- and time-dependent characteristics of IHERG blockade by ketoconazole indicated dependence of block on channel gating, ruling out a significant role for closed-state channel inhibition. The S6 HERG mutations Y652A and F656A produced approximately 4-fold and approximately 21-fold increases in IC50 for IHERG blockade, respectively. Thus, ketoconazole accesses the HERG channel pore-cavity on channel gating, and the S6 residue F656 is an important determinant of ketoconazole binding.


Asunto(s)
Antifúngicos/farmacología , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Canales de Potasio Éter-A-Go-Go/química , Activación del Canal Iónico/efectos de los fármacos , Cetoconazol/farmacología , Fenilalanina/metabolismo , Relación Dosis-Respuesta a Droga , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/metabolismo , Humanos , Potenciales de la Membrana/efectos de los fármacos , Mutación/genética , Factores de Tiempo
11.
J Mol Cell Cardiol ; 40(1): 107-18, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16288909

RESUMEN

Human ether-à-go-go-related gene (HERG) encodes the alpha-subunit of channels carrying the cardiac rapid delayed K+ current (Ikr), which is a major determinant of the duration of ventricular action potentials (APs) and of the QT interval. This study investigated the effects on HERG channel current (IHERG) of clemastine, a "conventional" antihistamine that has been associated with delayed ventricular repolarization in vitro, but for which no adverse effects on the human QT interval have been reported. Whole-cell patch-clamp measurements of IHERG were made at 37 degrees C from human embryonic kidney (HEK 293) cells stably expressing HERG channels. IHERG tails at -40 mV following depolarizing pulses to +20 mV were inhibited by clemastine with an IC50 value of 12 nM; this drug concentration also produced a marked inhibition of peak IHERG elicited during an AP voltage-clamp command. Clemastine produced a reversible approximately -5 mV shift in the IHERG steady-state voltage-dependent activation curve, but voltage-dependence of inactivation was unaffected. Development of IHERG inhibition by clemastine showed strong time-dependence. The S6 point mutations Y652A and F656A greatly attenuated the inhibitory effect of clemastine. We conclude that clemastine is a high potency inhibitor of IHERG, that this action is contingent upon channel gating and that clemastine interacts with a high affinity drug-binding site in the HERG channel pore cavity. The disparity between clemastine's potent IHERG inhibition and a lack of QT-prolongation in normal clinical use underscores the need to interpret HERG IC50 data for novel compounds in the context of information from other safety assays.


Asunto(s)
Clemastina/farmacología , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Antagonistas de los Receptores Histamínicos H1/farmacología , Animales , Calcio/metabolismo , Células Cultivadas , Relación Dosis-Respuesta a Droga , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/genética , Canales de Potasio Éter-A-Go-Go/metabolismo , Cobayas , Ventrículos Cardíacos/efectos de los fármacos , Humanos , Masculino , Mutación , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Potasio/farmacología , Factores de Tiempo , Función Ventricular
12.
Biochem Biophys Res Commun ; 334(2): 441-9, 2005 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-16011830

RESUMEN

The idiopathic short QT syndrome (SQTS) is characterised by an abnormally short QT interval on the electrocardiogram and by an increased risk of arrhythmia and sudden death. One variant of the syndrome is linked to missense mutations that lead to a single amino-acid change (N588K; asparagine to lysine) in the S5-Pore linker region of the cardiac HERG K(+) channel. This study was performed in order to determine how the N588K mutation alters HERG channel current (I(HERG)) kinetics at mammalian physiological temperature. The whole-cell current-voltage (I-V) relation for wild-type (WT) I(HERG) measured from Chinese Hamster Ovary cells was maximal at approximately 0 mV and showed marked inward rectification positive to this. In contrast, N588K I(HERG) showed marked rectification only at +60 mV and at more positive voltages. The voltage dependence of activation of N588K-HERG did not differ significantly from that of WT-HERG. However, N588K I(HERG) had a significantly more positive inactivation V(0.5) (-8.14+/-0.82 mV) than did WT I(HERG) (-70.05+/-0.82 mV; P<0.001, unpaired t test; n=5 for each). Its P(Na)/P(K) ratio was also greater. The instantaneous I-V relation for N588K I(HERG) under action potential voltage clamp peaked at approximately +40 mV, compared to approximately -37 mV for WT-I(HERG). These findings underscore the importance of the S5-P linker in HERG channel function and indicate that N588K-HERG contributes increased repolarising current earlier in the ventricular action potential at physiological temperature due to a approximately +60 mV shift in voltage dependence of I(HERG) inactivation.


Asunto(s)
Arritmias Cardíacas/fisiopatología , Activación del Canal Iónico , Potenciales de la Membrana , Canales de Potasio con Entrada de Voltaje , Animales , Células CHO , Cricetinae , Cricetulus , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go , Humanos , Mutagénesis Sitio-Dirigida , Relación Estructura-Actividad , Temperatura
13.
Clin Exp Pharmacol Physiol ; 32(12): 1088-96, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16445575

RESUMEN

Sipatrigine (BW 619C89), a blocker of neuronal Na+ and Ca2+ channels that is structurally related to lamotrigine, has been shown to be neuroprotective in models of cortical ischaemia. Although associated with cardiovascular effects in animal models in vivo, there is no published information concerning the effects of sipatrigine on cardiac ion currents and action potentials (AP). The aim of the present study was to examine the effects of sipatrigine on the delayed rectifier currents (I(Kr) and I(Ks)), the inward rectifier current (I(K1)), the L-type Ca2+ current (I(Ca,L)) and the fast Na+ current (I(Na)), as well as on AP duration at 30% (APD30) and 90% (APD90) repolarization, in guinea-pig isolated ventricular myocytes. Each of the currents was inhibited by sipatrigine, demonstrating the drug to be a relatively broad-spectrum blocker of cation channels in the heart. However, sipatrigine was a comparatively more potent inhibitor of I(Kr) (IC50 = 0.85 micromol/L) and I(Ks) (IC50 = 0.92 micromol/L) than of I(K1) (IC50 = 5.3 micromol/L), I(Ca,L) (IC50 = 6.0 micromol/L) and I(Na) (IC50 = 25.5 micromol/L). Consistent with block of I(Kr), I(Ks) and I(K1), sipatrigine (1-30 micromol/L) produced a concentration-dependent prolongation of APD90. Although lower concentrations of sipatrigine (< or = 3 micromol/L) caused APD(30) prolongation, higher concentrations (> or = 10 micromol/L) shortened APD30, consistent with an involvement of I(Ca,L) blockade. The contrasting effects of sipatrigine on APD30 and APD90 at higher concentrations resulted in a marked concentration-dependent triangulation of the AP. 5. The results of the present study demonstrate that sipatrigine, at concentrations previously shown to be neuroprotective in vitro, modulates cardiac K+, Ca2+ and Na+ currents and repolarization of the cardiac ventricular action potential.


Asunto(s)
Potenciales de Acción/efectos de los fármacos , Corazón/efectos de los fármacos , Canales Iónicos/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Piperazinas/farmacología , Pirimidinas/farmacología , Animales , Bloqueadores de los Canales de Calcio/farmacología , Separación Celular , Electrofisiología , Cobayas , Ventrículos Cardíacos/efectos de los fármacos , Técnicas In Vitro , Masculino , Miocitos Cardíacos/efectos de los fármacos , Bloqueadores de los Canales de Potasio/farmacología , Bloqueadores de los Canales de Sodio/farmacología
14.
Biochem Biophys Res Commun ; 325(3): 883-91, 2004 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-15541373

RESUMEN

Pharmacological inhibition of human-ether-a-go-go-related gene (HERG) K(+) channels by structurally and therapeutically diverse drugs is associated with the 'acquired' form of long QT syndrome and with potentially lethal cardiac arrhythmias. Two aromatic amino-acid residues (Y652 and F656) on the inner (S6) helices are considered to be key constituents of a high affinity drug binding site within the HERG channel pore cavity. Using wild-type (WT) and mutant HERG channels expressed in mammalian cell lines, we have investigated HERG channel current (I(HERG)) blockade at 37+/-1 degrees C by dronedarone (DRONED), a non-iodinated analogue of the Class III antiarrhythmic agent amiodarone (AMIOD). Under our conditions WT I(HERG) tails, measured at -40 mV following activating pulses to +30 mV, were blocked with IC(50) values of approximately 59 and 70 nM for DRONED and AMIOD, respectively. I(HERG) inhibition by DRONED was contingent upon channel gating, with block developing rapidly on membrane depolarization, but with no preference for activated over inactivated channels. High external [K(+)] (94 mM) reduced the potency of I(HERG) inhibition by both DRONED and AMIOD. Strikingly, mutagenesis to alanine of the S6 residue F656 (F656A) failed to eliminate blockade by both DRONED and AMIOD, whilst Y652A had comparatively little effect on DRONED but some effect on AMIOD. These findings demonstrate that high affinity drug blockade of I(HERG) can occur without a strong dependence on the Y652 and F656 aromatic amino-acid residues.


Asunto(s)
Amiodarona/análogos & derivados , Amiodarona/farmacología , Activación del Canal Iónico/efectos de los fármacos , Activación del Canal Iónico/fisiología , Riñón/efectos de los fármacos , Riñón/fisiología , Canales de Potasio con Entrada de Voltaje/efectos de los fármacos , Canales de Potasio con Entrada de Voltaje/fisiología , Sustitución de Aminoácidos , Antiarrítmicos/farmacología , Línea Celular , Relación Dosis-Respuesta a Droga , Dronedarona , Resistencia a Medicamentos/fisiología , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go , Humanos , Riñón/embriología , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Mutagénesis Sitio-Dirigida , Proteínas Recombinantes/metabolismo , Relación Estructura-Actividad
15.
Mol Pharmacol ; 66(5): 1201-12, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15308760

RESUMEN

The molecular determinants of high-affinity human ether-a-go-go-related gene (HERG) potassium channel blockade by methanesulfonanilides include two aromatic residues (Phe656 and Tyr652) on the inner helices (S6) and residues on the pore helices that face into the inner cavity, but determinants for lower-affinity HERG blockers may be different. In this study, alanine-substituted HERG channel mutants of inner cavity residues were expressed in Xenopus laevis oocytes and were used to characterize the HERG channel binding site of the antiarrhythmic propafenone. Propafenone's blockade of HERG was strongly dependent on residue Phe656 but was insensitive or weakly sensitive to mutation of Tyr652, Thr623, Ser624, Val625, Gly648, or Val659 and did not require functional inactivation. Homology models of HERG based on KcsA and MthK crystal structures, representing the closed and open forms of the channel, respectively, suggest propafenone is trapped in the inner cavity and is unable to interact exclusively with Phe656 in the closed state (whereas exclusive interactions between propafenone and Phe656 are found in the open-channel model). These findings are supported by very slow recovery of wild-type HERG channels from block at -120 mV, but extremely rapid recovery of D540K channels that reopen at this potential. The experiments and modeling suggest that the open-state propafenone binding-site may be formed by the Phe656 residues alone. The binding site for propafenone (which may involve pi-stacking interactions with two or more Phe656 side-chains) is either perturbed or becomes less accessible because of closed-channel gating. This provides further evidence for the existence of gating-induced changes in the spatial location of Phe656 side chains.


Asunto(s)
Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , Propafenona/farmacología , Secuencia de Aminoácidos , Animales , Antiarrítmicos/farmacología , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Oocitos , Canales de Potasio con Entrada de Voltaje/química , Canales de Potasio con Entrada de Voltaje/genética , Estructura Secundaria de Proteína/genética , Homología de Secuencia de Aminoácido , Transfección , Xenopus laevis
16.
J Mol Cell Cardiol ; 36(5): 701-5, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15135665

RESUMEN

Lidoflazine is an antianginal calcium channel blocker that carries a significant risk of QT interval prolongation and ventricular arrhythmia. We investigated whether or not lidoflazine inhibits current through the rapid delayed rectifier K(+) channel alpha subunit (encoded by HERG - human ether-a-go-go-related gene), since this channel has been widely linked to drug-induced QT-prolongation. Lidoflazine inhibited potently HERG current (I(HERG)) recorded from HEK 293 cells stably expressing wild-type HERG (IC(50) of approximately 16 nM). It was approximately 13-fold more potent against HERG than was verapamil under similar conditions. On membrane depolarization, I(HERG) inhibition developed gradually, ruling out closed-channel state dependent inhibition. The effect of command voltage on the drug's action suggested that lidoflazine preferentially inhibits activated/open HERG channels. The S6 mutation Y652A largely eliminated the inhibitory action of lidoflazine, whilst the F656A mutation also reduced blocking potency. We conclude: first, that lidoflazine produces high affinity blockade of the alpha subunit of the HERG channel by binding to aromatic amino acid residues within the channel pore and, second, that this is likely to represent the molecular mechanism of QT interval prolongation by this drug.


Asunto(s)
Lidoflazina/metabolismo , Lidoflazina/farmacología , Bloqueadores de los Canales de Potasio/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio con Entrada de Voltaje/antagonistas & inhibidores , Canales de Potasio con Entrada de Voltaje/metabolismo , Línea Celular , Canal de Potasio ERG1 , Electrofisiología , Canales de Potasio Éter-A-Go-Go , Humanos , Lidoflazina/química , Mutación/genética , Fenilalanina/genética , Fenilalanina/metabolismo , Bloqueadores de los Canales de Potasio/química , Canales de Potasio con Entrada de Voltaje/genética , Tirosina/genética , Tirosina/metabolismo
17.
Biochem Biophys Res Commun ; 318(2): 556-61, 2004 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-15120636

RESUMEN

The acquired form of the long-QT syndrome (LQTS) is a major safety consideration for the development and subsequent use of both cardiac and non-cardiac drugs; it is usually associated with pharmacological inhibition of cardiac HERG-encoded potassium channels. Clomiphene is an anti-estrogen agent used extensively in the treatment of infertility and is not associated with a risk of QT interval prolongation, in contrast to a structurally related compound tamoxifen. We describe here a potent inhibitory effect (IC(50) = 0.18 microM) of clomiphene on HERG ionic current (I(HERG)) recorded from a mammalian cell line expressing HERG channels. Inhibition of I(HERG) by clomiphene showed voltage-dependence and developed quickly following membrane depolarisation, indicating contingency of block on HERG channel gating. At 100 nM, clomiphene and the related anti-estrogen tamoxifen produced similar levels of I(HERG) blockade (p > 0.05). Experiments on guinea-pig isolated perfused hearts revealed that, despite its inhibitory action on I(HERG), clomiphene produced no significant effect at 1 microM on uncorrected QT interval (p > 0.1) nor on rate-corrected QT interval (QT(c); p > 0.1 for QT(c) determined using Van de Water's formula). The disparity between clomiphene's potent I(HERG) inhibition and its lack of effect on the QT interval underscores the notion that I(HERG) pharmacology may best be used alongside other screening methods when investigating the QT-prolonging tendency and related cardiotoxicity of non-cardiac drugs.


Asunto(s)
Clomifeno/farmacología , Antagonistas de Estrógenos/farmacología , Corazón/efectos de los fármacos , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio/efectos de los fármacos , Animales , Línea Celular , Relación Dosis-Respuesta a Droga , Electrocardiografía , Electrofisiología , Cobayas , Corazón/fisiología , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Síndrome de QT Prolongado , Masculino , Miocardio/metabolismo , Técnicas de Placa-Clamp , Canales de Potasio/metabolismo , Tamoxifeno/farmacología
18.
FEBS Lett ; 547(1-3): 20-6, 2003 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-12860380

RESUMEN

The scorpion toxin peptide BeKm-1 was synthesised by fluorenylmethoxycarbonyl solid phase chemistry and folded by air oxidation. The peptide's effects on heterologous human ether-a-go-go-related gene potassium current (I(HERG)) in HEK293 cells were assessed using 'whole-cell' patch clamp. Blockade of I(HERG) by BeKm-1 was concentration-dependent, temperature-dependent, and rapid in onset and reversibility. Blockade also exhibited inverse voltage dependence, inverse dependence on duration of depolarisation, and reverse use- and frequency-dependence. Blockade by BeKm-1 and recombinant ergtoxin, another scorpion toxin known to block HERG, differed in their recovery from HERG current inactivation elicited by strong depolarisation and in their ability to block HERG when the channels were already activated. We conclude that synthetic BeKm-1 toxin blocks HERG preferentially through a closed (resting) state channel blockade mechanism, although some open channel blockade also occurs.


Asunto(s)
Proteínas de Transporte de Catión , Proteínas de Unión al ADN , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio con Entrada de Voltaje , Canales de Potasio/fisiología , Venenos de Escorpión/farmacología , Transactivadores , Línea Celular , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go , Humanos , Cinética , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Potasio/síntesis química , Canales de Potasio/efectos de los fármacos , Canales de Potasio/genética , Pliegue de Proteína , Proteínas Recombinantes/antagonistas & inhibidores , Venenos de Escorpión/síntesis química , Venenos de Escorpión/química , Termodinámica , Regulador Transcripcional ERG , Transfección
19.
Br J Pharmacol ; 139(5): 887-98, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12839862

RESUMEN

1. Pharmacological blockade of the Human ether-a-go-go related gene (HERG) potassium channel is commonly linked with acquired long QT syndrome and associated proarrhythmia. The objectives of this study were (i) to identify and characterise any inhibitory action on HERG of the selective-serotonin re-uptake inhibitor fluvoxamine, (ii) to then determine whether fluvoxamine shared the consensus molecular determinants of HERG blockade of those drugs so far tested. 2. Heterologous HERG potassium current (I(HERG)) was measured at 37 degrees C, using the whole-cell patch-clamp technique, from a mammalian cell line (Human embryonic kidney 293) expressing HERG channels. I(HERG) tails, following repolarisation from +20 to -40 mV, were blocked by fluvoxamine with an IC(50) of 3.8 micro M. 3. Blockade of wild-type HERG was of extremely rapid onset (within 10 ms) and showed voltage dependence, with fluvoxamine also inducing a leftward shift in voltage-dependent activation of I(HERG). Characteristics of block were consistent with a component of closed channel (or extremely rapidly developing open channel) blockade and dependence on open and inactivated channel states. The attenuated-inactivation mutation S631A partially reduced the blocking effect of fluvoxamine. 4. The S6 mutations, Y652A and F656A, and the pore helix mutant S631A only partially attenuated blockade by fluvoxamine at concentrations causing profound blockade of wild-type HERG. 5. All HERG-blocking pharmaceuticals studied to date have been shown to block F656 mutant channels with over 100-fold reduced potency compared to their blockade of the wild-type channel. Fluvoxamine is therefore quite distinct in this regard from previously studied agents.


Asunto(s)
Sustitución de Aminoácidos/genética , Proteínas de Transporte de Catión , Proteínas de Unión al ADN , Fluvoxamina/farmacología , Mutación , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio con Entrada de Voltaje , Canales de Potasio/genética , Transactivadores , Línea Celular , Relación Dosis-Respuesta a Droga , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go , Fluvoxamina/química , Humanos , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Fenilalanina/genética , Bloqueadores de los Canales de Potasio/química , Canales de Potasio/metabolismo , Regulador Transcripcional ERG , Tirosina/genética
20.
Biochem Biophys Res Commun ; 306(2): 388-93, 2003 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-12804575

RESUMEN

Class Ia antiarrhythmic drugs, including procainamide (PROC), are associated with cardiac sodium channel blockade, delayed ventricular repolarisation and with a risk of ventricular pro-arrhythmia. The HERG K(+) channel is frequently linked to drug-induced pro-arrhythmia. Therefore, in this study, interactions between PROC and HERG K(+) channels were investigated, with particular reference to potency and mechanism of drug action. Whole-cell patch-clamp recordings of HERG current (I(HERG)) were made at 37 degrees C from human embryonic kidney (HEK 293) cells stably expressing the HERG channel. Following activating pulses to +20 mV, I(HERG) tails were inhibited by PROC with an IC(50) value of approximately 139 microM. I(HERG) blockade was found to be both time- and voltage-dependent, demonstrating contingency upon HERG channel gating. However, I(HERG) inhibition by PROC was relieved by depolarisation to a highly positive membrane potential (+80 mV) that favoured HERG channel inactivation. These data suggest that PROC inhibits the HERG K(+) channel by a primarily 'open' or 'activated' channel state blocking mechanism and that avidity of drug-binding is decreased by extensive I(HERG) inactivation. The potency of I(HERG) blockade by PROC is much lower than for other Class Ia agents that have been studied previously under analogous conditions (quinidine and disopyramide), although the blocking mechanism appears similar. Thus, differences between the chemical structure of PROC and other Class Ia antiarrhythmic drugs may help provide insight into chemical determinants of blocking potency for agents that bind to open/activated HERG channels.


Asunto(s)
Proteínas de Transporte de Catión , Proteínas de Unión al ADN , Canales de Potasio con Entrada de Voltaje , Canales de Potasio/química , Procainamida/farmacología , Transactivadores , Antiarrítmicos/farmacología , Línea Celular , Membrana Celular/metabolismo , Disopiramida/farmacología , Canal de Potasio ERG1 , Electrofisiología , Canales de Potasio Éter-A-Go-Go , Humanos , Concentración 50 Inhibidora , Síndrome de QT Prolongado/metabolismo , Potenciales de la Membrana , Técnicas de Placa-Clamp , Canales de Potasio/metabolismo , Quinidina/farmacología , Factores de Tiempo , Regulador Transcripcional ERG
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...