Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 132
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Br J Clin Pharmacol ; 90(4): 1027-1035, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37990600

RESUMEN

AIMS: AP30663 is a novel compound under development for pharmacological conversion of atrial fibrillation by targeting the small conductance Ca2+ activated K+ (KCa2) channel. The aim of this extension phase 1 study was to test AP30663 at higher single doses compared to the first-in-human trial. METHODS: Sixteen healthy male volunteers were randomized into 2 cohorts: 6- and 8-mg/kg intravenous single-dose administration of AP30663 vs. placebo. Safety, pharmacokinetic and pharmacodynamic data were collected. RESULTS: AP30663 was associated with mild and transient infusion site reactions with no clustering of other adverse events but with an estimated maximum mean QTcF interval prolongation of 45.2 ms (95% confidence interval 31.5-58.9) in the 6 mg/kg dose level and 50.4 ms (95% confidence interval 36.7-64.0) with 8 mg/kg. Pharmacokinetics was dose proportional with terminal half-life of around 3 h. CONCLUSION: AP30663 in doses up to 8 mg/kg was associated with mild and transient infusion site reactions and an increase of the QTcF interval. Supporting Information support that the QTc effect may be explained by an off-target inhibition of the IKr channel.


Asunto(s)
Fibrilación Atrial , Humanos , Masculino , Fibrilación Atrial/inducido químicamente , Fibrilación Atrial/tratamiento farmacológico , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Electrocardiografía , Frecuencia Cardíaca , Reacción en el Punto de Inyección
2.
Physiol Rev ; 94(2): 609-53, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24692356

RESUMEN

About 10 distinct potassium channels in the heart are involved in shaping the action potential. Some of the K+ channels are primarily responsible for early repolarization, whereas others drive late repolarization and still others are open throughout the cardiac cycle. Three main K+ channels drive the late repolarization of the ventricle with some redundancy, and in atria this repolarization reserve is supplemented by the fairly atrial-specific KV1.5, Kir3, KCa, and K2P channels. The role of the latter two subtypes in atria is currently being clarified, and several findings indicate that they could constitute targets for new pharmacological treatment of atrial fibrillation. The interplay between the different K+ channel subtypes in both atria and ventricle is dynamic, and a significant up- and downregulation occurs in disease states such as atrial fibrillation or heart failure. The underlying posttranscriptional and posttranslational remodeling of the individual K+ channels changes their activity and significance relative to each other, and they must be viewed together to understand their role in keeping a stable heart rhythm, also under menacing conditions like attacks of reentry arrhythmia.


Asunto(s)
Arritmias Cardíacas/metabolismo , Sistema de Conducción Cardíaco/metabolismo , Miocardio/metabolismo , Canales de Potasio/metabolismo , Potasio/metabolismo , Potenciales de Acción , Animales , Antiarrítmicos/farmacología , Arritmias Cardíacas/tratamiento farmacológico , Arritmias Cardíacas/fisiopatología , Sistema de Conducción Cardíaco/efectos de los fármacos , Sistema de Conducción Cardíaco/fisiopatología , Humanos , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio/efectos de los fármacos , Periodo Refractario Electrofisiológico
3.
Proc Natl Acad Sci U S A ; 115(34): E8077-E8085, 2018 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-30076230

RESUMEN

Dravet syndrome is a catastrophic, pharmacoresistant epileptic encephalopathy. Disease onset occurs in the first year of life, followed by developmental delay with cognitive and behavioral dysfunction and substantially elevated risk of premature death. The majority of affected individuals harbor a loss-of-function mutation in one allele of SCN1A, which encodes the voltage-gated sodium channel NaV1.1. Brain NaV1.1 is primarily localized to fast-spiking inhibitory interneurons; thus the mechanism of epileptogenesis in Dravet syndrome is hypothesized to be reduced inhibitory neurotransmission leading to brain hyperexcitability. We show that selective activation of NaV1.1 by venom peptide Hm1a restores the function of inhibitory interneurons from Dravet syndrome mice without affecting the firing of excitatory neurons. Intracerebroventricular infusion of Hm1a rescues Dravet syndrome mice from seizures and premature death. This precision medicine approach, which specifically targets the molecular deficit in Dravet syndrome, presents an opportunity for treatment of this intractable epilepsy.


Asunto(s)
Epilepsias Mioclónicas/tratamiento farmacológico , Interneuronas/metabolismo , Mutación , Canal de Sodio Activado por Voltaje NAV1.1/metabolismo , Venenos de Araña/farmacología , Transmisión Sináptica/efectos de los fármacos , Animales , Células CHO , Cricetulus , Epilepsias Mioclónicas/genética , Epilepsias Mioclónicas/metabolismo , Epilepsias Mioclónicas/patología , Células HEK293 , Humanos , Interneuronas/patología , Ratones , Ratones Mutantes , Canal de Sodio Activado por Voltaje NAV1.1/genética
4.
J Cardiovasc Pharmacol ; 76(2): 164-172, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32453071

RESUMEN

The mechanisms underlying atrial-selective prolongation of effective refractory period (ERP) and suppression of atrial fibrillation (AF) by NS8593 and UCL1684, small conductance calcium-activated potassium (SK) channel blockers, are poorly defined. The purpose of the study was to confirm the effectiveness of these agents to suppress AF and to probe the underlying mechanisms. Transmembrane action potentials and pseudoelectrocardiograms were recorded from canine isolated coronary-perfused canine atrial and ventricular wedge preparations. Patch clamp techniques were used to record sodium channel current (INa) in atrial and ventricular myocytes and human embryonic kidney cells. In both atria and ventricles, NS8593 (3-10 µM) and UCL1684 (0.5 µM) did not significantly alter action potential duration, suggesting little to no SK channel inhibition. Both agents caused atrial-selective: (1) prolongation of ERP secondary to development of postrepolarization refractoriness, (2) reduction of Vmax, and (3) increase of diastolic threshold of excitation (all are sodium-mediated parameters). NS8593 and UCL1684 significantly reduced INa density in human embryonic kidney cells as well as in atrial but not in ventricular myocytes at physiologically relevant holding potentials. NS8593 caused a shift of steady-state inactivation to negative potentials in atrial but not ventricular cells. NS8593 and UCL1684 prevented induction of acetylcholine-mediated AF in 6/6 and 8/8 preparations, respectively. This anti-AF effect was associated with strong rate-dependent depression of excitability. The SK channel blockers, NS8593 and UCL1684, are effective in preventing the development of AF due to potent atrial-selective inhibition of INa, causing atrial-selective prolongation of ERP secondary to induction of postrepolarization refractoriness.


Asunto(s)
1-Naftilamina/análogos & derivados , Alcanos/farmacología , Antiarrítmicos/farmacología , Fibrilación Atrial/prevención & control , Atrios Cardíacos/efectos de los fármacos , Frecuencia Cardíaca/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Canal de Sodio Activado por Voltaje NAV1.5/efectos de los fármacos , Compuestos de Quinolinio/farmacología , Bloqueadores de los Canales de Sodio/farmacología , 1-Naftilamina/farmacología , Potenciales de Acción/efectos de los fármacos , Animales , Fibrilación Atrial/metabolismo , Fibrilación Atrial/fisiopatología , Perros , Femenino , Células HEK293 , Atrios Cardíacos/metabolismo , Atrios Cardíacos/fisiopatología , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/fisiopatología , Humanos , Masculino , Miocitos Cardíacos/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/genética , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Periodo Refractario Electrofisiológico/efectos de los fármacos , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/antagonistas & inhibidores , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/metabolismo
5.
Europace ; 21(10): 1584-1593, 2019 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-31408093

RESUMEN

AIMS: Acute myocardial infarction (AMI) is associated with intracellular Ca2+ build-up. In healthy ventricles, small conductance Ca2+-activated K+ (SK) channels are present but do not participate in repolarization. However, SK current is increased in chronic myocardial infarction and heart failure, and recently, SK channel inhibition was demonstrated to reduce arrhythmias in AMI rats. Hence, we hypothesized that SK channel inhibitors (NS8593 and AP14145) could reduce arrhythmia development during AMI in a porcine model. METHODS AND RESULTS: Twenty-seven pigs were randomized 1:1:1 to control, NS8593, or AP14145. Haemodynamic and electrophysiological parameters [electrocardiogram (ECG) and monophasic action potentials (MAP)] were continuously recorded. A balloon was placed in the mid-left anterior descending artery, blinded to treatment. Infusion lasted from 10 min before occlusion until 30 min after. Occlusion was maintained for 1 h, followed by 2 h of reperfusion. Upon occlusion, cardiac output dropped similarly in all groups, while blood pressure remained stable. Heart rate decreased in the NS8593 and AP14145 groups. QRS duration increased upon occlusion in all groups but more prominently in AP14145-treated pigs. Inhibition of SK channels did not affect QT interval. Infarct MAP duration shortened comparably in all groups. Ventricular fibrillation developed in 4/9 control-, 4/9 AP14145-, and 2/9 NS8593-treated pigs. Ventricular tachycardia was rarely observed in either group, whereas ventricular extrasystoles occurred comparably in all groups. CONCLUSION: Inhibition of SK channels was neither beneficial nor detrimental to ventricular arrhythmia development in the setting of AMI in this porcine model.


Asunto(s)
1-Naftilamina/análogos & derivados , Electrocardiografía , Frecuencia Cardíaca/efectos de los fármacos , Ventrículos Cardíacos/fisiopatología , Infarto del Miocardio/tratamiento farmacológico , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/antagonistas & inhibidores , Taquicardia Ventricular/etiología , 1-Naftilamina/farmacología , Animales , Modelos Animales de Enfermedad , Femenino , Infarto del Miocardio/complicaciones , Infarto del Miocardio/fisiopatología , Porcinos , Taquicardia Ventricular/fisiopatología
6.
Eur J Neurosci ; 46(3): 1887-1896, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28635024

RESUMEN

Nav 1.1 (SCN1A) channels primarily located in gamma-aminobutyric acid (GABA)ergic fast-spiking interneurons are pivotal for action potential generation and propagation in these neurons. Inappropriate function of fast-spiking interneurons, leading to disinhibition of pyramidal cells and network desynchronization, correlates with decreased cognitive capability. Further, reduced functionality of Nav 1.1 channels is linked to various diseases in the central nervous system. There is, at present, however no subtype selective pharmacological activators of Nav 1.1 channels available for studying pharmacological modulation of interneuron function. In the current study, we identified a small molecule Nav 1.1 activator, 3-amino-5-(4-methoxyphenyl)thiophene-2-carboxamide, named AA43279, and provided an in vitro to in vivo characterization of the compound. In HEK-293 cells expressing human Nav 1.1 channels, AA43279 increased the Nav 1.1-mediated current in a concentration-dependent manner mainly by impairing the fast inactivation kinetics of the channels. In rat hippocampal brain slices, AA43279 increased the firing activity of parvalbumin-expressing, fast-spiking GABAergic interneurons and increased the spontaneous inhibitory post-synaptic currents (sIPSCs) recorded from pyramidal neurons. When tested in vivo, AA43279 had anti-convulsive properties in the maximal electroshock seizure threshold test. AA43279 was tested for off-target effects on 72 different proteins, including Nav 1.2, Nav 1.4, Nav 1.5, Nav 1.6 and Nav 1.7 and exhibited reasonable selectivity. Taken together, AA43279 might constitute a valuable tool compound for revealing biological functions of Nav 1.1 channels.


Asunto(s)
Anticonvulsivantes/farmacología , Neuronas GABAérgicas/efectos de los fármacos , Interneuronas/efectos de los fármacos , Canal de Sodio Activado por Voltaje NAV1.1/metabolismo , Convulsiones/tratamiento farmacológico , Bloqueadores de los Canales de Sodio/farmacología , Tiofenos/farmacología , Potenciales de Acción , Animales , Anticonvulsivantes/síntesis química , Anticonvulsivantes/uso terapéutico , Región CA1 Hipocampal/citología , Región CA1 Hipocampal/metabolismo , Región CA1 Hipocampal/fisiología , Potenciales Postsinápticos Excitadores , Neuronas GABAérgicas/metabolismo , Neuronas GABAérgicas/fisiología , Células HEK293 , Humanos , Interneuronas/metabolismo , Interneuronas/fisiología , Masculino , Ratones , Ratas , Ratas Sprague-Dawley , Bloqueadores de los Canales de Sodio/síntesis química , Bloqueadores de los Canales de Sodio/uso terapéutico
7.
Pflugers Arch ; 468(11-12): 1853-1863, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27722784

RESUMEN

Dose is an important parameter in terms of both efficacy and adverse effects in pharmacological treatment of atrial fibrillation (AF). Both of the class III antiarrhythmics dofetilide and amiodarone have documented anti-AF effects. While dofetilide has dose-related ventricular side effects, amiodarone primarily has adverse non-cardiac effects. Pharmacological inhibition of small conductance Ca2+-activated K+ (SK) channels has recently been reported to be antiarrhythmic in a number of animal AF models. In a Langendorff model of acutely induced AF on guinea pig hearts, it was investigated whether a combination of the SK channel blocker N-(pyridin-2-yl)-4-(pyridin-2-yl)thiazol-2-amine (ICA) together with either dofetilide or amiodarone provided a synergistic effect. The duration of AF was reduced with otherwise subefficacious concentrations of either dofetilide or amiodarone when combined with ICA, also at a subefficacious concentration. At a concentration level effective as monotherapy, dofetilide produced a marked increase in the QT interval. This QT prolonging effect was absent when combined with ICA at non-efficacious monotherapy concentrations. The results thereby reveal that combination of subefficacious concentrations of an SK channel blocker and either dofetilide or amiodarone can maintain anti-AF properties, while the risk of ventricular arrhythmias is reduced.


Asunto(s)
Amiodarona/farmacología , Antiarrítmicos/farmacología , Fibrilación Atrial/tratamiento farmacológico , Corazón/efectos de los fármacos , Fenetilaminas/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Piridinas/farmacología , Sulfonamidas/farmacología , Tiazoles/farmacología , Amiodarona/uso terapéutico , Animales , Antiarrítmicos/uso terapéutico , Sinergismo Farmacológico , Cobayas , Frecuencia Cardíaca , Preparación de Corazón Aislado , Fenetilaminas/uso terapéutico , Piridinas/uso terapéutico , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/antagonistas & inhibidores , Sulfonamidas/uso terapéutico , Tiazoles/uso terapéutico
8.
Pflugers Arch ; 468(4): 643-54, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26729267

RESUMEN

In isolated human atrial cardiomyocytes, inhibition of K2P3.1 K(+) channels results in action potential (action potential duration (APD)) prolongation. It has therefore been postulated that K2P3.1 (KCNK3), together with K2P9.1 (KCNK9), could represent novel drug targets for the treatment of atrial fibrillation (AF). However, it is unknown whether these findings in isolated cells translate to the whole heart. The purposes of this study were to investigate the expression levels of KCNK3 and KCNK9 in human hearts and two relevant rodent models and determine the antiarrhythmic potential of K2P3.1 inhibition in isolated whole-heart preparations. By quantitative PCR, we found that KCNK3 is predominantly expressed in human atria whereas KCNK9 was not detectable in heart human tissue. No differences were found between patients in AF or sinus rhythm. The expression in guinea pig heart resembled humans whereas rats displayed a more uniform expression of KCNK3 between atria and ventricle. In voltage-clamp experiments, ML365 and A293 were found to be potent and selective inhibitors of K2P3.1, but at pH 7.4, they failed to prolong atrial APD and refractory period (effective refractory period (ERP)) in isolated perfused rat and guinea pig hearts. At pH 7.8, which augments K2P3.1 currents, pharmacological channel inhibition produced a significant prolongation of atrial ERP (11.6 %, p = 0.004) without prolonging ventricular APD but did not display a significant antiarrhythmic effect in our guinea pig AF model (3/8 hearts converted on A293 vs 0/7 hearts in time-matched controls). These results suggest that when K2P3.1 current is augmented, K2P3.1 inhibition leads to atrial-specific prolongation of ERP; however, this ERP prolongation did not translate into significant antiarrhythmic effects in our AF model.


Asunto(s)
Potenciales de Acción , Arritmias Cardíacas/metabolismo , Función Atrial , Proteínas del Tejido Nervioso/metabolismo , Canales de Potasio de Dominio Poro en Tándem/metabolismo , Protones , Periodo Refractario Electrofisiológico , Adolescente , Adulto , Animales , Arritmias Cardíacas/fisiopatología , Células Cultivadas , Femenino , Cobayas , Atrios Cardíacos/citología , Atrios Cardíacos/metabolismo , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Masculino , Persona de Mediana Edad , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteínas del Tejido Nervioso/genética , Canales de Potasio de Dominio Poro en Tándem/antagonistas & inhibidores , Canales de Potasio de Dominio Poro en Tándem/genética , Ratas , Ratas Wistar , Especificidad de la Especie , Función Ventricular
9.
Circulation ; 129(4): 430-40, 2014 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-24190961

RESUMEN

BACKGROUND: Recent evidence points to functional Ca²âº-dependent K⁺ (SK) channels in the heart that may govern atrial fibrillation (AF) risk, but the underlying mechanisms are unclear. This study addressed the role of SK channels in atrial repolarization and AF persistence in a canine AF model. METHODS AND RESULTS: Electrophysiological variables were assessed in dogs subjected to atrial remodeling by 7-day atrial tachypacing (AT-P), as well as controls. Ionic currents and single-channel properties were measured in isolated canine atrial cardiomyocytes by patch clamp. NS8593, a putative selective SK blocker, suppressed SK current with an IC50 of ≈5 µmol/L, without affecting Na⁺, Ca²âº, or other K⁺ currents. Whole-cell SK current sensitive to NS8593 was significantly larger in pulmonary vein (PV) versus left atrial (LA) cells, without a difference in SK single-channel open probability (P(o)), whereas AT-P enhanced both whole-cell SK currents and single-channel P(o). SK-current block increased action potential duration in both PV and LA cells after AT-P; but only in PV cells in absence of AT-P. SK2 expression was more abundant at both mRNA and protein levels for PV versus LA in control dogs, in both control and AT-P; AT-P upregulated only SK1 at the protein level. Intravenous administration of NS8593 (5 mg/kg) significantly prolonged atrial refractoriness and reduced AF duration without affecting the Wenckebach cycle length, left ventricular refractoriness, or blood pressure. CONCLUSIONS: SK currents play a role in canine atrial repolarization, are larger in PVs than LA, are enhanced by atrial-tachycardia remodeling, and appear to participate in promoting AF maintenance. These results are relevant to the potential mechanisms underlying the association between SK single-nucleotide polymorphisms and AF and suggest SK blockers as potentially interesting anti-AF drugs.


Asunto(s)
Fibrilación Atrial/patología , Fibrilación Atrial/fisiopatología , Fenómenos Electrofisiológicos/fisiología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/fisiología , 1-Naftilamina/análogos & derivados , 1-Naftilamina/farmacología , Animales , Modelos Animales de Enfermedad , Perros , Técnicas Electrofisiológicas Cardíacas , Atrios Cardíacos/efectos de los fármacos , Atrios Cardíacos/patología , Miocitos Cardíacos/patología , Miocitos Cardíacos/fisiología , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Potasio/farmacología , Venas Pulmonares/efectos de los fármacos , Venas Pulmonares/patología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/efectos de los fármacos
10.
J Cardiovasc Pharmacol ; 66(3): 294-9, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25978690

RESUMEN

During recent years, small conductance Ca-activated K (SK) channels have been reported to play a role in cardiac electrophysiology. SK channels seem to be expressed in atria and ventricles, but from a functional perspective, atrial activity is predominant. A general notion seems to be that cardiac SK channels are predominantly coming into play during arrhythmogenic events where intracellular concentration of Ca is increased. During ventricular fibrillation (VF), a surge of [Ca]i has the potential to bind to and open SK channels. To obtain mechanistic insight into possible roles of SK channels during VF, we conducted experiments with an SK channel pore blocker (ICA) and a negatively allosteric modulator (NS8395) in a Langendorff-perfused heart model. Both compounds increased the action potential duration, effective refractory period, and Wenckebach cycle length to comparable extents. Despite these similarities, the SK channel modulator was found to revert and prevent VF more efficiently than the SK channel pore blocker. In conclusion, either negative allosteric modulation of the SK channel with NS8593 is more favorable than pure channel block with ICA or the 2 compounds have different selectivity profiles that makes NS8593 more antiarrhythmic than ICA in a setting of VF.


Asunto(s)
1-Naftilamina/análogos & derivados , Potenciales de Acción/efectos de los fármacos , Antiarrítmicos/uso terapéutico , Bloqueadores de los Canales de Potasio/uso terapéutico , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/antagonistas & inhibidores , Fibrilación Ventricular/tratamiento farmacológico , 1-Naftilamina/administración & dosificación , 1-Naftilamina/farmacología , 1-Naftilamina/uso terapéutico , Animales , Antiarrítmicos/administración & dosificación , Antiarrítmicos/farmacología , Modelos Animales de Enfermedad , Técnicas Electrofisiológicas Cardíacas , Femenino , Cobayas , Técnicas In Vitro , Preparación de Corazón Aislado , Bloqueadores de los Canales de Potasio/administración & dosificación , Bloqueadores de los Canales de Potasio/farmacología , Fibrilación Ventricular/metabolismo , Fibrilación Ventricular/fisiopatología
11.
J Cardiovasc Pharmacol ; 66(5): 441-8, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25830485

RESUMEN

Small-conductance Ca(2+)-activated potassium (SK) channels are relative newcomers within the field of cardiac electrophysiology. In recent years, an increased focus has been given to these channels because they might constitute a relatively atrial-selective target. This review will give a general introduction to SK channels followed by their proposed function in the heart under normal and pathophysiological conditions. It is revealed how antiarrhythmic effects can be obtained by SK channel inhibition in a number of species in situations of atrial fibrillation. On the contrary, the beneficial effects of SK channel inhibition in situations of heart failure are questionable and still needs investigation. The understanding of cardiac SK channels is rapidly increasing these years, and it is hoped that this will clarify whether SK channel inhibition has potential as a new anti-atrial fibrillation principle.


Asunto(s)
Fibrilación Atrial/metabolismo , Sistema de Conducción Cardíaco/metabolismo , Frecuencia Cardíaca , Canales de Potasio Calcio-Activados/metabolismo , Animales , Antiarrítmicos/uso terapéutico , Fibrilación Atrial/tratamiento farmacológico , Fibrilación Atrial/fisiopatología , Drogas en Investigación/uso terapéutico , Sistema de Conducción Cardíaco/efectos de los fármacos , Sistema de Conducción Cardíaco/fisiopatología , Frecuencia Cardíaca/efectos de los fármacos , Humanos , Terapia Molecular Dirigida , Bloqueadores de los Canales de Potasio/uso terapéutico , Canales de Potasio Calcio-Activados/antagonistas & inhibidores , Transducción de Señal
12.
J Cardiovasc Pharmacol ; 66(2): 165-76, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25856531

RESUMEN

INTRODUCTION: SK channels have functional importance in the cardiac atrium of many species, including humans. Pharmacological blockage of SK channels has been reported to be antiarrhythmic in animal models of atrial fibrillation; however, the exact antiarrhythmic mechanism of SK channel inhibition remains unclear. OBJECTIVES: We speculated that together with a direct inhibition of repolarizing SK current, the previously observed depolarization of the atrial resting membrane potential (RMP) after SK channel inhibition reduces sodium channel availability, thereby prolonging the effective refractory period and slowing the conduction velocity (CV). We therefore aimed at elucidating these properties of SK channel inhibition and the underlying antiarrhythmic mechanisms using microelectrode action potential (AP) recordings and CV measurements in isolated rat atrium. Automated patch clamping and two-electrode voltage clamp were used to access INa and IK,ACh, respectively. RESULTS: The SK channel inhibitor N-(pyridin-2-yl)-4-(pyridin-2-yl)thiazol-2-amine (ICA) exhibited antiarrhythmic effects. ICA prevented electrically induced runs of atrial fibrillation in the isolated right atrium and induced atrial postrepolarization refractoriness and depolarized RMP. Moreover, ICA (1-10 µM) was found to slow CV; however, because of a marked prolongation of effective refractory period, the calculated wavelength was increased. Furthermore, at increased pacing frequencies, SK channel inhibition by ICA (10-30 µM) demonstrated prominent depression of other sodium channel-dependent parameters. ICA did not inhibit IK,ACh, but at concentrations above 10 µM, ICA use dependently inhibited INa. CONCLUSIONS: SK channel inhibition modulates multiple parameters of AP. It prolongs the AP duration and shifts the RMP towards more depolarized potentials through direct ISK block. This indirectly leads to sodium channel inhibition through accumulation of state dependently inactivated channels, which ultimately slows conduction and decreases excitability. However, a contribution from a direct sodium channel inhibition cannot be ruled. We here propose that the primary antiarrhythmic mechanism of SK channel inhibition is through direct potassium channel block and through indirect sodium channel inhibition.


Asunto(s)
Antiarrítmicos/farmacología , Función del Atrio Derecho/efectos de los fármacos , Bloqueadores de los Canales de Potasio/farmacología , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/antagonistas & inhibidores , Animales , Antiarrítmicos/uso terapéutico , Fibrilación Atrial/tratamiento farmacológico , Fibrilación Atrial/fisiopatología , Función del Atrio Derecho/fisiología , Células CHO , Cricetinae , Cricetulus , Femenino , Atrios Cardíacos/efectos de los fármacos , Masculino , Técnicas de Cultivo de Órganos , Bloqueadores de los Canales de Potasio/uso terapéutico , Ratas , Ratas Sprague-Dawley , Canales de Potasio de Pequeña Conductancia Activados por el Calcio/fisiología , Xenopus laevis
13.
Scand J Clin Lab Invest ; 75(8): 699-709, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26403377

RESUMEN

BACKGROUND: Congenital long QT syndrome (LQTS) is a hereditary cardiac channelopathy characterized by delayed ventricular repolarization, syncope, torsades de pointes and sudden cardiac death. Thirty-three members of five apparently 'unrelated' Danish families carry the KCNH2:c.87C> A; p.F29L founder mutation. METHODS AND RESULTS: Linkage disequilibrium mapping with microsatellites around KCNH2 enabled us to estimate the age of the founder mutation to be approximately 22 generations, corresponding to around 550 years. Neighbouring-Joining analysis disclosed one early and three later nodes. The median QTc time of the carriers was 490 ms (range: 415-589 ms) and no difference was seen between the different branches of the family. The mutation is malignant with a penetrance of 73%. Ten F29L carriers received implantable defibrillators (ICDs) (median age at implant 20 years), and of those four individuals experienced eight appropriate shocks. Patch-clamp analysis in HEK 293 cells, performed at 34°C disclosed a loss-of-function phenotype with fast deactivation, reduced steady-state inactivation current density and a positive voltage shift in inactivation. Western blotting of HEK 293 cells transfected with KCNH2:WT and KCNH2:c.87C> A revealed a reduced fraction of fully glycosylated hERG:p.F29L suggesting that this mutation results in defective trafficking. CONCLUSION: The altered channel gating kinetics in combination with defective trafficking of mutated channels is expected to result in reduced repolarizing current density and, thus, a LQTS phenotype.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/genética , Síndrome de QT Prolongado/genética , Dinamarca , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/fisiología , Femenino , Efecto Fundador , Estudios de Asociación Genética , Células HEK293 , Haplotipos , Humanos , Activación del Canal Iónico , Cinética , Masculino , Potenciales de la Membrana , Repeticiones de Microsatélite , Mutación Missense , Fenotipo , Polimorfismo de Nucleótido Simple , Transporte de Proteínas , Análisis de Secuencia de ADN
14.
Artículo en Inglés | MEDLINE | ID: mdl-21987061

RESUMEN

Atrial fibrillation (AF) is recognised as the most common sustained cardiac arrhythmia in clinical practice. Ongoing drug development is aiming at obtaining atrial specific effects in order to prevent pro-arrhythmic, devastating ventricular effects. In principle, this is possible due to a different ion channel composition in the atria and ventricles. The present text will review the aetiology of arrhythmias with focus on AF and include a description of cardiac ion channels. Channels that constitute potentially atria-selective targets will be described in details. Specific focus is addressed to the recent discovery that Ca(2+)-activated small conductance K(+) channels (SK channels) are important for the repolarisation of atrial action potentials. Finally, an overview of current pharmacological treatment of AF is included.


Asunto(s)
Antiarrítmicos/farmacología , Arritmias Cardíacas/tratamiento farmacológico , Arritmias Cardíacas/fisiopatología , Fibrilación Atrial/tratamiento farmacológico , Fibrilación Atrial/prevención & control , Fibrilación Atrial/fisiopatología , Bloqueadores de los Canales de Calcio/farmacología , Canales Iónicos/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Potenciales de Acción , Animales , Calcio/metabolismo , Electrofisiología , Corazón/fisiología , Humanos , Iones , Modelos Biológicos , Canales de Potasio/metabolismo
15.
J Pharmacol Exp Ther ; 350(3): 520-30, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24951278

RESUMEN

Large-conductance Ca(2+)-activated K(+) channels (BK, KCa1.1, MaxiK) are important regulators of urinary bladder function and may be an attractive therapeutic target in bladder disorders. In this study, we established a high-throughput fluorometric imaging plate reader-based screening assay for BK channel activators and identified a small-molecule positive modulator, NS19504 (5-[(4-bromophenyl)methyl]-1,3-thiazol-2-amine), which activated the BK channel with an EC50 value of 11.0 ± 1.4 µM. Hit validation was performed using high-throughput electrophysiology (QPatch), and further characterization was achieved in manual whole-cell and inside-out patch-clamp studies in human embryonic kidney 293 cells expressing hBK channels: NS19504 caused distinct activation from a concentration of 0.3 and 10 µM NS19504 left-shifted the voltage activation curve by 60 mV. Furthermore, whole-cell recording showed that NS19504 activated BK channels in native smooth muscle cells from guinea pig urinary bladder. In guinea pig urinary bladder strips, NS19504 (1 µM) reduced spontaneous phasic contractions, an effect that was significantly inhibited by the specific BK channel blocker iberiotoxin. In contrast, NS19504 (1 µM) only modestly inhibited nerve-evoked contractions and had no effect on contractions induced by a high K(+) concentration consistent with a K(+) channel-mediated action. Collectively, these results show that NS19504 is a positive modulator of BK channels and provide support for the role of BK channels in urinary bladder function. The pharmacologic profile of NS19504 indicates that this compound may have the potential to reduce nonvoiding contractions associated with spontaneous bladder overactivity while having a minimal effect on normal voiding.


Asunto(s)
Agonistas de los Canales de Calcio/farmacología , Canales de Potasio de Gran Conductancia Activados por el Calcio/agonistas , Contracción Muscular/efectos de los fármacos , Relajación Muscular/efectos de los fármacos , Vejiga Urinaria/efectos de los fármacos , Animales , Agonistas de los Canales de Calcio/química , Femenino , Cobayas , Células HEK293 , Humanos , Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Masculino , Contracción Muscular/fisiología , Relajación Muscular/fisiología , Técnicas de Cultivo de Órganos , Vejiga Urinaria/fisiología
16.
Circ J ; 78(5): 1136-43, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24599044

RESUMEN

BACKGROUND: Mutations in SCN5A can result in both long QT type 3 (LQT3) and Brugada syndrome (BrS), and a few mutations have been found to have an overlapping phenotype. Long QT syndrome is characterized by prolonged QT interval, and a prerequisite for a BrS diagnosis is ST elevation in the right precordial leads of the electrocardiogram. METHODS AND RESULTS: In a Danish family suffering from long QT syndrome, a novel missense mutation in SCN5A, changing a leucine residue into a glutamine residue at position 1786 (L1786Q), was found to be present in heterozygous form co-segregating with prolonged QT interval. The proband presented with an aborted cardiac arrest, and his mother died suddenly and unexpectedly at the age of 65. Flecainide treatment revealed coved ST elevation in all mutation carriers. Electrophysiological investigations of the mutant in HEK293 cells indicated a reduced peak current, a negative shift in inactivation properties and a positive shift in activation properties, compatible with BrS. Furthermore, the sustained (I(Na,late)) tetrodotoxin-sensitive sodium current was found to be drastically increased, explaining the association between the mutation and LQT syndrome. CONCLUSIONS: The L1786Q mutation is associated with a combined LQT3 and concealed BrS phenotype explained by gating characteristics of the mutated ion channel protein. Hence, sodium channel blockade should be considered in clinical evaluation of apparent LQT3 patients.


Asunto(s)
Antiarrítmicos/administración & dosificación , Síndrome de Brugada , Familia , Flecainida/administración & dosificación , Síndrome de QT Prolongado , Mutación Missense , Canal de Sodio Activado por Voltaje NAV1.5 , Sustitución de Aminoácidos , Animales , Síndrome de Brugada/tratamiento farmacológico , Síndrome de Brugada/genética , Síndrome de Brugada/metabolismo , Síndrome de Brugada/fisiopatología , Femenino , Células HEK293 , Heterocigoto , Humanos , Síndrome de QT Prolongado/dietoterapia , Síndrome de QT Prolongado/genética , Síndrome de QT Prolongado/metabolismo , Síndrome de QT Prolongado/fisiopatología , Masculino , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Canal de Sodio Activado por Voltaje NAV1.5/genética , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo
17.
Nat Med ; 30(1): 106-111, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38092897

RESUMEN

Existing antiarrhythmic drugs to treat atrial fibrillation (AF) have incomplete efficacy, contraindications and adverse effects, including proarrhythmia. AP30663, an inhibitor of the KCa2 channel, has demonstrated AF efficacy in animals; however, its efficacy in humans with AF is unknown. Here we conducted a phase 2 trial in which patients with a current episode of AF lasting for 7 days or less were randomized to receive an intravenous infusion of 3 or 5 mg kg-1 AP30663 or placebo. The trial was prematurely discontinued because of slow enrollment during the coronavirus disease 2019 pandemic. The primary endpoint of the trial was cardioversion from AF to sinus rhythm within 90 min from the start of the infusion, analyzed with Bayesian statistics. Among 59 patients randomized and included in the efficacy analyses, the primary endpoint occurred in 42% (5 of 12), 55% (12 of 22) and 0% (0 of 25) of patients treated with 3 mg kg-1 AP30663, 5 mg kg-1 AP30663 or placebo, respectively. Both doses demonstrated more than 99.9% probability of superiority over placebo, surpassing the prespecified 95% threshold. The mean time to cardioversion, a secondary endpoint, was 47 (s.d. = 23) and 41 (s.d. = 24) minutes for 3 mg kg-1 and 5 mg kg-1 AP30663, respectively. AP30663 caused a transient increase in the QTcF interval, with a maximum mean effect of 37.7 ms for the 5 mg kg-1 dose. For both dose groups, no ventricular arrhythmias occurred and adverse event rates were comparable to the placebo group. AP30663 demonstrated AF cardioversion efficacy in patients with recent-onset AF episodes. KCa2 channel inhibition may be an attractive mechanism for rhythm control of AF that should be studied further in randomized trials. ClinicalTrials.gov registration: NCT04571385 .


Asunto(s)
Fibrilación Atrial , Humanos , Fibrilación Atrial/tratamiento farmacológico , Teorema de Bayes , Resultado del Tratamiento , Antiarrítmicos/efectos adversos , Infusiones Intravenosas
18.
Am J Hum Genet ; 86(6): 872-80, 2010 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-20560207

RESUMEN

Congenital long QT syndrome (LQTS) is a hereditary disorder that leads to sudden cardiac death secondary to fatal cardiac arrhythmias. Although many genes for LQTS have been described, the etiology remains unknown in 30%-40% of cases. In the present study, a large Chinese family (four generations, 49 individuals) with autosomal-dominant LQTS was clinically evaluated. Genome-wide linkage analysis was performed by using polymorphic microsatellite markers to map the genetic locus, and positional candidate genes were screened by sequencing for mutations. The expression pattern and functional characteristics of the mutated protein were investigated by western blotting and patch-clamp electrophysiology. The genetic locus of the LQTS-associated gene was mapped to chromosome 11q23.3-24.3. A heterozygous mutation (Kir3.4-Gly387Arg) was identified in the G protein-coupled, inwardly rectifying potassium channel subunit Kir3.4, encoded by the KCNJ5 gene. The Kir3.4-Gly387Arg mutation was present in all nine affected family members and absent in 528 ethnically matched controls. Western blotting of human cardiac tissue demonstrated significant Kir3.4 expression levels in the cardiac ventricles. Heterologous expression studies with Kir3.4-Gly387Arg revealed a loss-of-function electrophysiological phenotype resulting from reduced plasma membrane expression. Our findings suggest a role for Kir3.4 in the etiology of LQTS.


Asunto(s)
Canales de Potasio Rectificados Internamente Asociados a la Proteína G/genética , Adolescente , Adulto , Anciano , Aminofilina , Atropina , Mapeo Cromosómico , Combinación de Medicamentos , Femenino , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Ligamiento Genético , Humanos , Recién Nacido , Síndrome de QT Prolongado/genética , Masculino , Persona de Mediana Edad , Mutación , Miocardio/metabolismo , Nitroglicerina , Papaverina , Linaje , Fenobarbital
19.
J Cardiovasc Pharmacol ; 62(2): 192-8, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23609329

RESUMEN

G protein-coupled inwardly rectifying K⁺ channels (GIRK) are important in the regulation of heart rate and atrial electrophysiology. GIRK channels are activated by G protein-coupled receptors, including muscarinic M2 receptors and adenosine A1 receptors. The aim of this study was to characterize and compare the electrophysiological effects of acetylcholine (ACh) and adenosine on GIRK channels in rat atria. Action potential duration at 90% repolarization (APD90), effective refractory period (ERP), and resting membrane potential (RMP) were investigated in isolated rat atria by intracellular recordings. Both the adenosine analog N6-cyclopentyladenosine (CPA) and ACh profoundly shortened APD90 and ERP and hyperpolarized the RMP. No additive or synergistic effect of CPA and ACh coapplication was observed. To antagonize GIRK channel activation, the specific inhibitor rTertiapin Q (TTQ) was applied. The coapplication of TTQ reversed the CPA and ACh-induced effects. When TTQ was applied without exogenous receptor activator, both APD90 and ERP were prolonged and RMP was depolarized, confirming a basal activity of the GIRK current. The results reveal that activation of A1 and M2 receptors has a profound and equal effect on the electrophysiology in rat atrium. This effect is to a major extent mediated through GIRK channels. Furthermore, these results support the notion that atrial GIRK currents from healthy hearts have a basal component and additional activation can be mediated via at least 2 different receptor mechanisms.


Asunto(s)
Arritmias Cardíacas/prevención & control , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/agonistas , Corazón/efectos de los fármacos , Moduladores del Transporte de Membrana/farmacología , Miocardio/metabolismo , Receptor Muscarínico M2/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Acetilcolina/metabolismo , Adenosina/metabolismo , Agonistas de Receptores Adrenérgicos alfa 1/farmacología , Animales , Arritmias Cardíacas/inducido químicamente , Arritmias Cardíacas/metabolismo , Función Atrial/efectos de los fármacos , Fenómenos Electrofisiológicos/efectos de los fármacos , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/antagonistas & inhibidores , Canales de Potasio Rectificados Internamente Asociados a la Proteína G/metabolismo , Técnicas In Vitro , Masculino , Potenciales de la Membrana/efectos de los fármacos , Bloqueadores de los Canales de Potasio/farmacología , Agonistas del Receptor Purinérgico P1/farmacología , Ratas , Ratas Sprague-Dawley , Receptor Muscarínico M2/agonistas , Periodo Refractario Electrofisiológico/efectos de los fármacos
20.
Cell Physiol Biochem ; 30(3): 642-52, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22854413

RESUMEN

Angiotensin II (AngII) is a key peptide in cardiovascular homeostasis and is a ligand for the Angiotensin II type 1 and 2 seven transmembrane receptors (AT(1)R and AT(2)R). The AT(1) receptor is a seven-transmembrane (7TM) G protein-coupled receptor (GPCR) mediating the majority of the physiological functions of AngII. The AT(1)R mediates its effects through both G protein-dependent and independent signaling, which can be separated by functionally selective agonists. In the present study we investigate the effect of AngII and the ß-arrestin biased agonist [SII]AngII on ischemia-reperfusion injury in rat hearts. Isolated hearts mounted in a Langendorff perfused rat heart preparations showed that preconditioning with [SII]AngII reduced the infarct size induced by global ischemia from 46±8.4% to 22±3.4%. In contrast, neither preconditioning with AngII nor postconditioning with AngII or [SII]AngII had a protective effect. Together these results demonstrate a cardioprotective effect of simultaneous blockade of G protein signaling and activation of G protein independent signaling through AT(1) receptors.


Asunto(s)
Receptor de Angiotensina Tipo 1/metabolismo , Daño por Reperfusión/metabolismo , Angiotensina II/farmacología , Animales , Arrestinas/farmacología , Cardiotónicos/farmacología , Proteínas de Unión al GTP/metabolismo , Frecuencia Cardíaca/efectos de los fármacos , Ventrículos Cardíacos/efectos de los fármacos , Hemodinámica/efectos de los fármacos , Técnicas In Vitro , Masculino , Presión , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/fisiopatología , beta-Arrestinas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA