Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Bioorg Chem ; 98: 103717, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32171994

RESUMO

A series of 2-aryl-2-(pyridin-2-yl)acetamides were synthesized and screened for their anticonvulsant activity in animal models of epilepsy. The compounds were broadly active in the 'classical' maximal electroshock seizure (MES) and subcutaneous Metrazol (scMET) tests as well as in the 6 Hz and kindling models of pharmacoresistant seizures. Furthermore, the compounds showed good therapeutic indices between anticonvulsant activity and motor impairment. Structure-activity relationship (SAR) trends clearly showed the highest activity resides in unsubstituted phenyl derivatives or compounds having ortho- and meta- substituents on the phenyl ring. The 2-aryl-2-(pyridin-2-yl)acetamides were derived by redesign of the cardiotoxic sodium channel blocker Disopyramide (DISO). Our results show that the compounds preserve the capability of the parent compound to inhibit voltage gated sodium currents in patch-clamp experiments; however, in contrast to DISO, a representative compound from the series 1 displays high levels of cardiac safety in a panel of in vitro and in vivo experiments.


Assuntos
Acetamidas/uso terapêutico , Anticonvulsivantes/uso terapêutico , Disopiramida/uso terapêutico , Convulsões/tratamento farmacológico , Acetamidas/administração & dosagem , Acetamidas/química , Animais , Anticonvulsivantes/administração & dosagem , Anticonvulsivantes/química , Disopiramida/administração & dosagem , Disopiramida/química , Relação Dose-Resposta a Droga , Eletrochoque , Feminino , Injeções Intraperitoneais , Injeções Subcutâneas , Masculino , Camundongos , Estrutura Molecular , Pentilenotetrazol/administração & dosagem , Ratos , Ratos Wistar , Convulsões/induzido quimicamente , Relação Estrutura-Atividade
2.
J Biol Chem ; 293(18): 7040-7057, 2018 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-29545312

RESUMO

Cardiac potassium channels encoded by human ether-à-go-go-related gene (hERG) are major targets for structurally diverse drugs associated with acquired long QT syndrome. This study characterized hERG channel inhibition by a minimally structured high-affinity hERG inhibitor, Cavalli-2, composed of three phenyl groups linked by polymethylene spacers around a central amino group, chosen to probe the spatial arrangement of side chain groups in the high-affinity drug-binding site of the hERG pore. hERG current (IhERG) recorded at physiological temperature from HEK293 cells was inhibited with an IC50 of 35.6 nm with time and voltage dependence characteristic of blockade contingent upon channel gating. Potency of Cavalli-2 action was markedly reduced for attenuated inactivation mutants located near (S620T; 54-fold) and remote from (N588K; 15-fold) the channel pore. The S6 Y652A and F656A mutations decreased inhibitory potency 17- and 75-fold, respectively, whereas T623A and S624A at the base of the selectivity filter also decreased potency (16- and 7-fold, respectively). The S5 helix F557L mutation decreased potency 10-fold, and both F557L and Y652A mutations eliminated voltage dependence of inhibition. Computational docking using the recent cryo-EM structure of an open channel hERG construct could only partially recapitulate experimental data, and the high dependence of Cavalli-2 block on Phe-656 is not readily explainable in that structure. A small clockwise rotation of the inner (S6) helix of the hERG pore from its configuration in the cryo-EM structure may be required to optimize Phe-656 side chain orientations compatible with high-affinity block.


Assuntos
Canal de Potássio ERG1/antagonistas & inibidores , Canal de Potássio ERG1/química , Bloqueadores dos Canais de Potássio/farmacologia , Alanina/química , Simulação por Computador , Microscopia Crioeletrônica , Cristalografia por Raios X , Relação Dose-Resposta a Droga , Canal de Potássio ERG1/genética , Canal de Potássio ERG1/metabolismo , Células HEK293 , Humanos , Concentração Inibidora 50 , Ativação do Canal Iônico/efeitos dos fármacos , Simulação de Acoplamento Molecular , Mutação , Técnicas de Patch-Clamp , Fenilalanina/química , Bloqueadores dos Canais de Potássio/administração & dosagem , Ligação Proteica , Conformação Proteica
3.
PLoS Comput Biol ; 13(6): e1005593, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28609477

RESUMO

Gain-of-function mutations in KCNJ2-encoded Kir2.1 channels underlie variant 3 (SQT3) of the short QT syndrome, which is associated with atrial fibrillation (AF). Using biophysically-detailed human atria computer models, this study investigated the mechanistic link between SQT3 mutations and atrial arrhythmogenesis, and potential ion channel targets for treatment of SQT3. A contemporary model of the human atrial action potential (AP) was modified to recapitulate functional changes in IK1 due to heterozygous and homozygous forms of the D172N and E299V Kir2.1 mutations. Wild-type (WT) and mutant formulations were incorporated into multi-scale homogeneous and heterogeneous tissue models. Effects of mutations on AP duration (APD), conduction velocity (CV), effective refractory period (ERP), tissue excitation threshold and their rate-dependence, as well as the wavelength of re-entry (WL) were quantified. The D172N and E299V Kir2.1 mutations produced distinct effects on IK1 and APD shortening. Both mutations decreased WL for re-entry through a reduction in ERP and CV. Stability of re-entrant excitation waves in 2D and 3D tissue models was mediated by changes to tissue excitability and dispersion of APD in mutation conditions. Combined block of IK1 and IKr was effective in terminating re-entry associated with heterozygous D172N conditions, whereas IKr block alone may be a safer alternative for the E299V mutation. Combined inhibition of IKr and IKur produced a synergistic anti-arrhythmic effect in both forms of SQT3. In conclusion, this study provides mechanistic insights into atrial proarrhythmia with SQT3 Kir2.1 mutations and highlights possible pharmacological strategies for management of SQT3-linked AF.


Assuntos
Arritmias Cardíacas/fisiopatologia , Fibrilação Atrial/fisiopatologia , Acoplamento Excitação-Contração , Sistema de Condução Cardíaco/fisiopatologia , Modelos Cardiovasculares , Canais de Potássio Corretores do Fluxo de Internalização/genética , Potenciais de Ação , Arritmias Cardíacas/complicações , Fibrilação Atrial/complicações , Simulação por Computador , Predisposição Genética para Doença/genética , Átrios do Coração/fisiopatologia , Humanos , Ativação do Canal Iônico/genética , Modelos Genéticos , Mutação/genética , Contração Miocárdica , Potássio/metabolismo
4.
J Mol Cell Cardiol ; 86: 42-53, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26159617

RESUMO

The class Ic antiarrhythmic drug flecainide inhibits KCNH2-encoded "hERG" potassium channels at clinically relevant concentrations. The aim of this study was to elucidate the underlying molecular basis of this action. Patch clamp recordings of hERG current (IhERG) were made from hERG expressing cells at 37°C. Wild-type (WT) IhERG was inhibited with an IC50 of 1.49µM and this was not significantly altered by reversing the direction of K(+) flux or raising external [K(+)]. The use of charged and uncharged flecainide analogues showed that the charged form of the drug accesses the channel from the cell interior to produce block. Promotion of WT IhERG inactivation slowed recovery from inhibition, whilst the N588K and S631A attenuated-inactivation mutants exhibited IC50 values 4-5 fold that of WT IhERG. The use of pore-helix/selectivity filter (T623A, S624A V625A) and S6 helix (G648A, Y652A, F656A) mutations showed <10-fold shifts in IC50 for all but V625A and F656A, which respectively exhibited IC50s 27-fold and 142-fold their WT controls. Docking simulations using a MthK-based homology model suggested an allosteric effect of V625A, since in low energy conformations flecainide lay too low in the pore to interact directly with that residue. On the other hand, the molecule could readily form π-π stacking interactions with aromatic residues and particularly with F656. We conclude that flecainide accesses the hERG channel from the cell interior on channel gating, binding low in the inner cavity, with the S6 F656 residue acting as a principal binding determinant.


Assuntos
Arritmias Cardíacas/tratamento farmacológico , Flecainida/administração & dosagem , Torsades de Pointes/tratamento farmacológico , Transativadores/genética , Antiarrítmicos/administração & dosagem , Arritmias Cardíacas/genética , Arritmias Cardíacas/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Humanos , Simulação de Acoplamento Molecular , Mutação , Técnicas de Patch-Clamp , Conformação Proteica , Torsades de Pointes/genética , Torsades de Pointes/patologia , Transativadores/biossíntese , Transativadores/química , Regulador Transcricional ERG
5.
J Mol Cell Cardiol ; 74: 220-30, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24877995

RESUMO

The antianginal drug ranolazine, which combines inhibitory actions on rapid and sustained sodium currents with inhibition of the hERG/IKr potassium channel, shows promise as an antiarrhythmic agent. This study investigated the structural basis of hERG block by ranolazine, with lidocaine used as a low potency, structurally similar comparator. Recordings of hERG current (IhERG) were made from cell lines expressing wild-type (WT) or mutant hERG channels. Docking simulations were performed using homology models built on MthK and KvAP templates. In conventional voltage clamp, ranolazine inhibited IhERG with an IC50 of 8.03µM; peak IhERG during ventricular action potential clamp was inhibited ~62% at 10µM. The IC50 values for ranolazine inhibition of the S620T inactivation deficient and N588K attenuated inactivation mutants were respectively ~73-fold and ~15-fold that for WT IhERG. Mutations near the bottom of the selectivity filter (V625A, S624A, T623A) exhibited IC50s between ~8 and 19-fold that for WT IhERG, whilst the Y652A and F656A S6 mutations had IC50s ~22-fold and 53-fold WT controls. Low potency lidocaine was comparatively insensitive to both pore helix and S6 mutations, but was sensitive to direction of K(+) flux and particularly to loss of inactivation, with an IC50 for S620T-hERG ~49-fold that for WT IhERG. Docking simulations indicated that the larger size of ranolazine gives it potential for a greater range of interactions with hERG pore side chains compared to lidocaine, in particular enabling interaction of its two aromatic groups with side chains of both Y652 and F656. The N588K mutation is responsible for the SQT1 variant of short QT syndrome and our data suggest that ranolazine is unlikely to be effective against IKr/hERG in SQT1 patients.


Assuntos
Acetanilidas/química , Antiarrítmicos/química , Canais de Potássio Éter-A-Go-Go/química , Piperazinas/química , Potássio/metabolismo , Acetanilidas/farmacologia , Potenciais de Ação/efeitos dos fármacos , Antiarrítmicos/farmacologia , Relação Dose-Resposta a Droga , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/genética , Canais de Potássio Éter-A-Go-Go/metabolismo , Expressão Gênica , Células HEK293 , Humanos , Transporte de Íons , Lidocaína/química , Lidocaína/farmacologia , Simulação de Acoplamento Molecular , Mutação , Técnicas de Patch-Clamp , Piperazinas/farmacologia , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Ranolazina , Relação Estrutura-Atividade , Transgenes
6.
J Pharmacol Toxicol Methods ; 123: 107293, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37468081

RESUMO

Pharmacological blockade of the IKr channel (hERG) by diverse drugs in clinical use is associated with the Long QT Syndrome that can lead to life threatening arrhythmia. Various computational tools including machine learning models (MLM) for the prediction of hERG inhibition have been developed to facilitate the throughput screening of drugs in development and optimise thus the prediction of hERG liabilities. The use of MLM relies on large libraries of training compounds for the quantitative structure-activity relationship (QSAR) modelling of hERG inhibition. The focus on inhibition omits potential effects of hERG channel agonist molecules and their associated QT shortening risk. It is instructive, therefore, to consider how known hERG agonists are handled by MLM. Here, two highly developed online computational tools for the prediction of hERG liability, Pred-hERG and HergSPred were probed for their ability to detect hERG activator drug molecules as hERG interactors. In total, 73 hERG blockers were tested with both computational tools giving overall good predictions for hERG blockers with reported IC50s below Pred-hERG and HergSPred cut-off threshold for hERG inhibition. However, for compounds with reported IC50s above this threshold such as disopyramide or sotalol discrepancies were observed. HergSPred identified all 20 hERG agonists selected as interacting with the hERG channel. Further studies are warranted to improve online MLM prediction of hERG related cardiotoxicity, by explicitly taking into account channel agonism as well as inhibition.


Assuntos
Canais de Potássio Éter-A-Go-Go , Bloqueadores dos Canais de Potássio , Humanos , Bloqueadores dos Canais de Potássio/farmacologia , Arritmias Cardíacas , Aprendizado de Máquina , Internet
7.
Pharmaceuticals (Basel) ; 16(9)2023 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-37765012

RESUMO

hERG (human Ether-à-go-go Related Gene)-encoded potassium channels underlie the cardiac rapid delayed rectifier (IKr) potassium current, which is a major target for antiarrhythmic agents and diverse non-cardiac drugs linked to the drug-induced form of long QT syndrome. E-4031 is a high potency hERG channel inhibitor from the methanesulphonanilide drug family. This study utilized a methanesulphonate-lacking E-4031 analogue, "E-4031-17", to evaluate the role of the methanesulphonamide group in E-4031 inhibition of hERG. Whole-cell patch-clamp measurements of the hERG current (IhERG) were made at physiological temperature from HEK 293 cells expressing wild-type (WT) and mutant hERG constructs. For E-4031, WT IhERG was inhibited by a half-maximal inhibitory concentration (IC50) of 15.8 nM, whilst the comparable value for E-4031-17 was 40.3 nM. Both compounds exhibited voltage- and time-dependent inhibition, but they differed in their response to successive applications of a long (10 s) depolarisation protocol, consistent with greater dissociation of E-4031-17 than the parent compound between applied commands. Voltage-dependent inactivation was left-ward voltage shifted for E-4031 but not for E-4031-17; however, inhibition by both compounds was strongly reduced by attenuated-inactivation mutations. Mutations of S6 and S5 aromatic residues (F656V, Y652A, F557L) greatly attenuated actions of both drugs. The S624A mutation also reduced IhERG inhibition by both molecules. Overall, these results demonstrate that the lack of a methanesulphonate in E-4031-17 is not an impediment to high potency inhibition of IhERG.

8.
J Mol Cell Cardiol ; 52(1): 185-95, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21989164

RESUMO

The Class Ia antiarrhythmic drug disopyramide (DISO) causes QT interval prolongation that is potentially dangerous in acquired Long QT Syndrome but beneficial in short QT syndrome, through inhibition of the hERG-encoded channels responsible for rapid delayed rectifier K(+) current (I(Kr)). In this study, alanine mutants of hERG S6 and pore helix residues and MthK-based homology modelling and ligand docking were used to investigate molecular determinants of DISO binding to hERG. Whole-cell hERG current (I(hERG)) recordings were made at 37°C from HEK-293 cells expressing WT or mutant hERG channels. WT outward I(hERG) tails were inhibited with an IC(50) of 7.3µM, whilst inward I(hERG) tails in a high [K(+)](e) of 94mM were blocked with an IC(50) of 25.7µM. The IC(50) for the Y652A mutation was ~55-fold that of WT I(hERG); this mutation also abolished a leftward shift in voltage-dependent I(hERG) activation present for WT hERG. The IC(50) for F656A I(hERG) was ~51 fold its corresponding WT control. In contrast to previously studied methanesulphonanilide hERG inhibitors, neither the G648A S6 nor the T623A and S624A pore helical mutations modified DISO IC(50). Computational docking with the hERG model showed that DISO did not exhibit a single unique binding pose; instead several low energy binding poses at the lower end of the pore cavity favoured interactions with Y652 and F656. In the WT hERG model DISO did not interact directly with residues at the base of the pore helix, consistent with the minimal effect of mutation of these residues on drug block.


Assuntos
Antiarrítmicos/farmacologia , Disopiramida/farmacologia , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Canais de Potássio Éter-A-Go-Go/genética , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/genética , Sequência de Aminoácidos , Relação Dose-Resposta a Droga , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/química , Células HEK293 , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Mutação , Conformação Proteica
9.
J Arrhythm ; 38(4): 554-569, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35936037

RESUMO

In the human heart, the rapid delayed rectifier K+ current (I Kr) contributes significantly to ventricular action potential (AP) repolarization and to set the duration of the QT interval of the surface electrocardiogram (ECG). The pore-forming (α) subunit of the I Kr channel is encoded by KCNH2 or human ether-à-go-go-related gene 1 (hERG1). Impairment of hERG function through either gene mutation (congenital) or pharmacological blockade by diverse drugs in clinical use (acquired) can cause a prolongation of the AP duration (APD) reflected onto the surface ECG as a prolonged QT interval or Long QT Syndrome (LQTS). LQTS can increase the risk of triggered activity of ventricular cardiomyocytes and associated life-threatening arrhythmia. Current treatments all focus on reducing the incidence of arrhythmia or terminating it after its onset but there is to date no prophylactic treatment for the pharmacological management of LQTS. A new class of hERG modulators (agonists) have been suggested through direct interaction with the hERG channel to shorten the action potential duration (APD) and/or increase the postrepolarisation refractoriness period (PRRP) of ventricular cardiomyocytes protecting thereby against triggered activity and associated arrhythmia. Although promising drug candidates, there remain major obstacles to their clinical development. The aim of this review is to summarize the latest advances as well as the limitations of this proposed pharmacotherapy.

10.
J Mol Cell Cardiol ; 51(5): 713-21, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21787784

RESUMO

Complete atrioventricular block (CAVB) and related ventricular bradycardia are known to induce ventricular hypertrophy and arrhythmias. Different animal models of CAVB have been established with the most common being the dog model. Related studies were mainly focused on the consequences on the main repolarizing currents in these species, i.e. IKr and IKs, with a limited time point kinetics post-AVB. In order to explore at a genomic scale the electrical remodeling induced by AVB and its chronology, we have developed a novel model of CAVB in the mouse using a radiofrequency-mediated ablation procedure. We investigated transcriptional changes in ion channels and contractile proteins in the left ventricles as a function of time (12h, 1, 2 and 5 days after CAVB), using high-throughput real-time RT-PCR. ECG in conscious and anesthetized mice, left ventricular pressure recordings and patch-clamp were used for characterization of this new mouse model. As expected, CAVB was associated with a lengthening of the QT interval. Moreover, polymorphic ventricular tachycardia was recorded in 6/9 freely-moving mice during the first 24h post-ablation. Remarkably, myocardial hypertrophy was only evident 48 h post-ablation and was associated with increased heart weight and altered expression of contractile proteins. During the first 24 hours post-CAVB, genes encoding ion channel subunits were either up-regulated (such as Nav1.5, +74%) or down-regulated (Kv4.2, -43%; KChIP2, -47%; Navß1, -31%; Cx43, -29%). Consistent with the transient alteration of Kv4.2 expression, I(to) was reduced at day 1, but restored at day 5. In conclusion, CAVB induces two waves of molecular remodeling: an early one (≤24 h) leading to arrhythmias, a later one related to hypertrophy. These results provide new molecular basis for ventricular tachycardia induced by AV block.


Assuntos
Arritmias Cardíacas/metabolismo , Bloqueio Atrioventricular/metabolismo , Ventrículos do Coração/metabolismo , Hipertrofia Ventricular Esquerda/metabolismo , Canais Iônicos/metabolismo , Miocárdio/metabolismo , Subunidades Proteicas/metabolismo , Taquicardia Ventricular/metabolismo , Potenciais de Ação/fisiologia , Animais , Arritmias Cardíacas/etiologia , Arritmias Cardíacas/fisiopatologia , Bloqueio Atrioventricular/complicações , Bloqueio Atrioventricular/fisiopatologia , Modelos Animais de Doenças , Regulação para Baixo , Eletrocardiografia , Expressão Gênica , Perfilação da Expressão Gênica , Ventrículos do Coração/fisiopatologia , Hemodinâmica , Hipertrofia Ventricular Esquerda/etiologia , Hipertrofia Ventricular Esquerda/fisiopatologia , Canais Iônicos/genética , Masculino , Camundongos , Miocárdio/patologia , Tamanho do Órgão , Subunidades Proteicas/genética , Reação em Cadeia da Polimerase em Tempo Real , Taquicardia Ventricular/etiologia , Taquicardia Ventricular/fisiopatologia , Fatores de Tempo , Regulação para Cima
12.
J Cardiovasc Electrophysiol ; 21(10): 1160-9, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20455975

RESUMO

INTRODUCTION: Potassium channels encoded by human ether-à-go-go-related gene (hERG) underlie the cardiac rapid delayed rectifier K(+) channel current (I(Kr)). Acidosis occurs in a number of pathological situations and modulates a range of ionic currents including I(Kr) . The aim of this study was to characterize effects of extracellular acidosis on hERG current (I(hERG)), with particular reference to quantifying effects on I(hERG) elicited by physiological waveforms and upon the protective role afforded by hERG against premature depolarizing stimuli. METHODS AND RESULTS: I(hERG) recordings were made from hERG-expressing Chinese Hamster Ovary cells using whole-cell patch-clamp at 37°C. I(hERG) during action potential (AP) waveforms was rapidly suppressed by reducing external pH from 7.4 to 6.3. Peak repolarizing current and steady state I(hERG) activation were shifted by ∼+6 mV; maximal I(hERG) conductance was reduced. The voltage-dependence of I(hERG) inactivation was little-altered. Fast and slow time-constants of I(hERG) deactivation were smaller across a range of voltages at pH 6.3 than at pH 7.4, and the contribution of fast deactivation increased. A modest acceleration of the time-course of recovery of I(hERG) from inactivation was observed, but time-course of activation was unaffected. The amplitude of outward I(hERG) transients elicited by premature stimuli following an AP command was significantly decreased at lower pH. Computer simulations showed that after AP repolarization a subthreshold stimulus at pH 7.4 could evoke an AP at pH 6.3. CONCLUSION: During acidosis the contribution of I(hERG) to action potential repolarization is reduced and hERG may be less effective in counteracting proarrhythmogenic depolarizing stimuli.


Assuntos
Canais de Potássio Éter-A-Go-Go/química , Canais de Potássio Éter-A-Go-Go/fisiologia , Ativação do Canal Iônico/fisiologia , Potenciais da Membrana/fisiologia , Acidose/fisiopatologia , Animais , Células CHO , Cricetinae , Cricetulus , Concentração de Íons de Hidrogênio , Técnicas de Patch-Clamp
13.
Physiol Rep ; 8(5): e14385, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32147975

RESUMO

The macrolide antibiotic erythromycin has been associated with QT interval prolongation and inhibition of the hERG-encoded channels responsible for the rapid delayed rectifier K+ current I(Kr ). It has been suggested that low concentrations of erythromycin may have a protective effect against hERG block and associated drug-induced arrhythmia by reducing the affinity of the pore-binding site for high potency hERG inhibitors. This study aimed to explore further the notion of a potentially protective effect of erythromycin. Whole-cell patch-clamp experiments were performed in which hERG-expressing mammalian (Human Embryonic Kidney; HEK) cells were preincubated with low to moderate concentrations of erythromycin (3 or 30 µM) prior to whole-cell patch clamp recordings of hERG current (IhERG ) at 37°C. In contrast to a previous report, exposure to low concentrations of erythromycin did not reduce pharmacological sensitivity of hERG to the antipsychotic thioridazine and antihistamine terfenadine. The IC50 value for IhERG tail inhibition by terfenadine was decreased by ~32-fold in the presence of 3 µM erythromycin (p < .05 vs. no preincubation). Sensitivity to thioridazine remained unchanged (p > .05 vs. no preincubation). The effects of low concentrations of erythromycin were investigated for a series of pore blocking drugs, and the results obtained were consistent with additive and/or synergistic effects. Experiments with the externally acting blocker BeKm-1 on WT hERG and a pore mutant (F656V) were used to explore the location of the binding site for erythromycin. Our data are inconsistent with the use of erythromycin for the management of drug-induced QT prolongation.


Assuntos
Antipsicóticos/administração & dosagem , Canal de Potássio ERG1/antagonistas & inibidores , Eritromicina/administração & dosagem , Antagonistas não Sedativos dos Receptores H1 da Histamina/administração & dosagem , Terfenadina/administração & dosagem , Tioridazina/administração & dosagem , Sítios de Ligação/efeitos dos fármacos , Canal de Potássio ERG1/fisiologia , Eritromicina/química , Células HEK293 , Humanos , Concentração Inibidora 50 , Macrolídeos/administração & dosagem , Macrolídeos/química , Técnicas de Patch-Clamp
14.
Front Cardiovasc Med ; 7: 85, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32432127

RESUMO

The COVID-19 pandemic is an unprecedented challenge and will require novel therapeutic strategies. Affected patients are likely to be at risk of arrhythmia due to underlying comorbidities, polypharmacy and the disease process. Importantly, a number of the medications likely to receive significant use can themselves, particularly in combination, be pro-arrhythmic. Drug-induced prolongation of the QT interval is primarily caused by inhibition of the hERG potassium channel either directly and/or by impaired channel trafficking. Concurrent use of multiple hERG-blocking drugs may have a synergistic rather than additive effect which, in addition to any pre-existing polypharmacy, critical illness or electrolyte imbalance, may significantly increase the risk of arrhythmia and Torsades de Pointes. Knowledge of these risks will allow informed decisions regarding appropriate therapeutics and monitoring to keep our patients safe.

15.
Biophys J ; 97(5): 1323-34, 2009 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-19720020

RESUMO

Studies in Shaker, a voltage-dependent potassium channel, suggest a coupling between activation and inactivation. This coupling is controversial in hERG, a fast-inactivating voltage-dependent potassium channel. To address this question, we transferred to hERG the S3-S4 linker of the voltage-independent channel, rolf, to selectively disrupt the activation process. This chimera shows an intact voltage-dependent inactivation process consistent with a weak coupling, if any, between both processes. Kinetic models suggest that the chimera presents only an open and an inactivated states, with identical transition rates as in hERG. The lower sensitivity of the chimera to BeKm-1, a hERG preferential closed-state inhibitor, also suggests that the chimera presents mainly open and inactivated conformations. This chimera allows determining the mechanism of action of hERG blockers, as exemplified by the test on ketoconazole.


Assuntos
Canais de Cátion Regulados por Nucleotídeos Cíclicos/química , Canais de Potássio Éter-A-Go-Go/química , Canais de Potássio Éter-A-Go-Go/metabolismo , Ativação do Canal Iônico , Análise de Variância , Animais , Células COS , Chlorocebus aethiops , Canais de Cátion Regulados por Nucleotídeos Cíclicos/genética , Canais de Cátion Regulados por Nucleotídeos Cíclicos/metabolismo , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/genética , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Cetoconazol/farmacologia , Cinética , Potenciais da Membrana , Modelos Biológicos , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/farmacologia , Ratos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Venenos de Escorpião/farmacologia
16.
J Mol Cell Cardiol ; 47(5): 743-7, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19285083

RESUMO

Recently identified genetic forms of short QT syndrome (SQTS) are associated with an increased risk of arrhythmia and sudden death. The SQT3 variant is associated with an amino-acid substitution (D172N) in the KCNJ2-encoded Kir2.1 K+ channel. In this study, whole-cell action potential (AP) clamp recording from transiently transfected Chinese Hamster Ovary cells at 37 degrees C showed marked augmentation of outward Kir2.1 current through D172N channels, associated with right-ward voltage-shifts of peak repolarizing current during both ventricular and atrial AP commands. Peak outward current elicited by ventricular AP commands was inhibited by chloroquine with an IC50 of 2.45 microM for wild-type (WT) Kir2.1, of 3.30 microM for D172N-Kir2.1 alone and of 3.11 microM for co-expressed WT and D172N (P>0.05 for all). These findings establish chloroquine as an effective inhibitor of SQT3 mutant Kir2.1 channels.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Antirreumáticos/farmacologia , Arritmias Cardíacas/genética , Arritmias Cardíacas/metabolismo , Cloroquina/farmacologia , Técnicas de Patch-Clamp/métodos , Canais de Potássio Corretores do Fluxo de Internalização/genética , Potenciais de Ação/genética , Animais , Células CHO , Cricetinae , Cricetulus , Eletrofisiologia , Humanos , Canais de Potássio Corretores do Fluxo de Internalização/fisiologia
17.
Pharmacol Ther ; 119(2): 118-32, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18616963

RESUMO

Drug-induced torsades de pointes (TdP) arrhythmia is a major safety concern in the process of drug design and development. The incidence of TdP tends to be low, so early pre-clinical screens rely on surrogate markers of TdP to highlight potential problems with new drugs. hERG (human ether-à-go-go-related gene, alternative nomenclature KCNH2) is responsible for channels mediating the 'rapid' delayed rectifier K+ current (IKr) which plays an important role in ventricular repolarization. Pharmacological inhibition of native IKr and of recombinant hERG channels is a shared feature of diverse drugs associated with TdP. In vitro hERG assays therefore form a key element of an integrated assessment of TdP liability, with patch-clamp electrophysiology offering a 'gold standard'. However, whilst clearly necessary, hERG assays cannot be assumed automatically to provide sufficient information, when considered in isolation, to differentiate 'safe' from 'dangerous' drugs. Other relevant factors include therapeutic plasma concentration, drug metabolism and active metabolites, severity of target condition and drug effects on other cardiac ion channels that may mitigate or exacerbate effects of hERG blockade. Increased understanding of the nature of drug-hERG channel interactions may ultimately help eliminate potential hERG blockade early in the design and development process. Currently, for promising drug candidates integration of data from hERG assays with information from other pre-clinical safety screens remains essential.


Assuntos
Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Torsades de Pointes/induzido quimicamente , Animais , Canais de Potássio de Retificação Tardia/antagonistas & inibidores , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos/métodos , Técnicas Eletrofisiológicas Cardíacas , Humanos , Concentração Inibidora 50 , Técnicas de Patch-Clamp , Preparações Farmacêuticas/administração & dosagem , Preparações Farmacêuticas/metabolismo
18.
Exp Physiol ; 94(4): 400-11, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19151075

RESUMO

The beta(3)-adrenoceptors (beta(3)-ARs) have been identified and characterized in the human heart. Specific beta(3)-AR stimulation, unlike beta(1)-AR or beta(2)-AR stimulation, decreases cardiac contractility, partly via the G(i)-NO pathway. However, the precise role of cardiac beta(3)-ARs is not yet completely understood. Indeed, under normal conditions, the beta(3)-AR response is present only to a very low degree in rats and mice. Therefore, we evaluated whether beta(3)-ARs were present and functional in rabbit ventricular cardiomyocytes, and whether the rabbit could serve as a relevant model for the study of cardiac beta(3)-ARs. We used RT-PCR and Western blot to measure the beta(3)-AR transcripts and protein levels in rabbit ventricular cardiomyocytes. We also analysed the effect of beta(3)-AR stimulation using isoproterenol in combination with nadolol or SR 58611A on cardiomyocyte shortening, Ca(2+) transient, L-type Ca(2+) current (I(Ca,L)), delayed rectifier potassium current (I(Ks)) and action potential duration (APD). For the first time, we show that beta(3)-ARs are expressed in rabbit ventricular cardiomyocytes. The mRNA and protein sequences present a high homology to those of rat and human beta(3)-ARs. Furthermore, beta(3)-AR stimulation decreases cardiomyocyte shortening, Ca(2+) transient and I(Ca,L) amplitudes, via a G(i)-NO pathway. Importantly, beta(3)-AR stimulation enhances I(Ks) amplitude and shortens the APD. Taken together, our results indicate that the rabbit provides a relevant model, easily used in laboratories, to study the roles of cardiac beta(3)-ARs in physiological conditions.


Assuntos
Sinalização do Cálcio , Contração Miocárdica , Miócitos Cardíacos/metabolismo , Receptores Adrenérgicos beta 3/metabolismo , Potenciais de Ação , Agonistas Adrenérgicos beta/farmacologia , Antagonistas Adrenérgicos beta/farmacologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Canais de Cálcio Tipo L/metabolismo , Sinalização do Cálcio/efeitos dos fármacos , Células Cultivadas , Canais de Potássio de Retificação Tardia/metabolismo , Relação Dose-Resposta a Droga , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Ventrículos do Coração/metabolismo , Humanos , Isoproterenol/farmacologia , Masculino , Modelos Animais , Dados de Sequência Molecular , Contração Miocárdica/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Nadolol/farmacologia , Óxido Nítrico/metabolismo , RNA Mensageiro/metabolismo , Coelhos , Ratos , Receptores Adrenérgicos beta 3/efeitos dos fármacos , Receptores Adrenérgicos beta 3/genética , Tetra-Hidronaftalenos/farmacologia , Fatores de Tempo
20.
Cardiovasc Res ; 79(3): 427-35, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18390900

RESUMO

AIMS: KCNQ1 (alias KvLQT1 or Kv7.1) and KCNE1 (alias IsK or minK) co-assemble to form the voltage-activated K(+) channel responsible for I(Ks)-a major repolarizing current in the human heart-and their dysfunction promotes cardiac arrhythmias. The channel is a component of larger macromolecular complexes containing known and undefined regulatory proteins. Thus, identification of proteins that modulate its biosynthesis, localization, activity, and/or degradation is of great interest from both a physiological and pathological point of view. METHODS AND RESULTS: Using a yeast two-hybrid screening, we detected a direct interaction between beta-tubulin and the KCNQ1 N-terminus. The interaction was confirmed by co-immunoprecipitation of beta-tubulin and KCNQ1 in transfected COS-7 cells and in guinea pig cardiomyocytes. Using immunocytochemistry, we also found that they co-localized in cardiomyocytes. We tested the effects of microtubule-disrupting and -stabilizing agents (colchicine and taxol, respectively) on the KCNQ1-KCNE1 channel activity in COS-7 cells by means of the permeabilized-patch configuration of the patch-clamp technique. None of these agents altered I(Ks). In addition, colchicine did not modify the current response to osmotic challenge. On the other hand, the I(Ks) response to protein kinase A (PKA)-mediated stimulation depended on microtubule polymerization in COS-7 cells and in cardiomyocytes. Strikingly, KCNQ1 channel and Yotiao phosphorylation by PKA-detected by phospho-specific antibodies-was maintained, as was the association of the two partners. CONCLUSION: We propose that the KCNQ1-KCNE1 channel directly interacts with microtubules and that this interaction plays a major role in coupling PKA-dependent phosphorylation of KCNQ1 with I(Ks) activation.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Canal de Potássio KCNQ1/metabolismo , Microtúbulos/metabolismo , Miócitos Cardíacos/enzimologia , Tubulina (Proteína)/metabolismo , Proteínas de Ancoragem à Quinase A/metabolismo , Potenciais de Ação , Animais , Células COS , Chlorocebus aethiops , Cobaias , Canal de Potássio KCNQ1/genética , Cinética , Masculino , Camundongos , Microtúbulos/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Pressão Osmótica , Fosforilação , Ligação Proteica , Estrutura Terciária de Proteína , Transfecção , Tubulina (Proteína)/genética , Moduladores de Tubulina/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA