Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Am J Physiol Heart Circ Physiol ; 319(2): H251-H261, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-32559136

RESUMO

Human ether-à-go-go related gene (hERG) K+ channels are important in cardiac repolarization, and their dysfunction causes prolongation of the ventricular action potential, long QT syndrome, and arrhythmia. As such, approaches to augment hERG channel function, such as activator compounds, have been of significant interest due to their marked therapeutic potential. Activator compounds that hinder channel inactivation abbreviate action potential duration (APD) but carry risk of overcorrection leading to short QT syndrome. Enhanced risk by overcorrection of the APD may be tempered by activator-induced increased refractoriness; however, investigation of the cumulative effect of hERG activator compounds on the balance of these effects in whole organ systems is lacking. Here, we have investigated the antiarrhythmic capability of a hERG activator, RPR260243, which primarily augments channel function by slowing deactivation kinetics in ex vivo zebrafish whole hearts. We show that RPR260243 abbreviates the ventricular APD, reduces triangulation, and steepens the slope of the electrical restitution curve. In addition, RPR260243 increases the post-repolarization refractory period. We provide evidence that this latter effect arises from RPR260243-induced enhancement of hERG channel-protective currents flowing early in the refractory period. Finally, the cumulative effect of RPR260243 on arrhythmogenicity in whole organ zebrafish hearts is demonstrated by the restoration of normal rhythm in hearts presenting dofetilide-induced arrhythmia. These findings in a whole organ model demonstrate the antiarrhythmic benefit of hERG activator compounds that modify both APD and refractoriness. Furthermore, our results demonstrate that targeted slowing of hERG channel deactivation and enhancement of protective currents may provide an effective antiarrhythmic approach.NEW & NOTEWORTHY hERG channel dysfunction causes long QT syndrome and arrhythmia. Activator compounds have been of significant interest due to their therapeutic potential. We used the whole organ zebrafish heart model to demonstrate the antiarrhythmic benefit of the hERG activator, RPR260243. The activator abbreviated APD and increased refractoriness, the combined effect of which rescued induced ventricular arrhythmia. Our findings show that the targeted slowing of hERG channel deactivation and enhancement of protective currents caused by the RPR260243 activator may provide an effective antiarrhythmic approach.


Assuntos
Antiarrítmicos/farmacologia , Arritmias Cardíacas/prevenção & controle , Canal de Potássio ERG1/agonistas , Canais de Potássio Éter-A-Go-Go/agonistas , Frequência Cardíaca/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Piperidinas/farmacologia , Quinolinas/farmacologia , Proteínas de Peixe-Zebra/agonistas , Potenciais de Ação , Animais , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/fisiopatologia , Modelos Animais de Doenças , Canal de Potássio ERG1/genética , Canal de Potássio ERG1/metabolismo , Canais de Potássio Éter-A-Go-Go/metabolismo , Cinética , Miócitos Cardíacos/metabolismo , Oócitos , Período Refratário Eletrofisiológico , Transdução de Sinais , Xenopus laevis , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo
2.
Mol Pharmacol ; 85(5): 769-76, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24586056

RESUMO

The Kv11.1 potassium channel is the molecular target for the majority of drugs implicated in acquired long QT syndrome, the most common cause of drug-induced sudden cardiac death, and a common reason for drug restriction or withdrawal from the market. While the IC50 for block of Kv11.1 is commonly used to estimate the risk of acquired long QT syndrome, this approach is crude, and it is widely accepted that the kinetics of drug interactions with the channel are a critical component in understanding their mechanism of action and risk profiles. In this study we report the first directly measured kinetics of block and unblock of Kv11.1 by a QT prolonging drug: the antipsychotic clozapine. Our data show that clozapine binding to Kv11.1 is complex. There are at least two kinetically distinct components to both block and unblock, while the kinetics of unblock are dependent on the dose or duration of drug application. Based on these observations, we have proposed a model incorporating kinetically distinct binding to the open and inactivated states of Kv11.1 that can describe the observed kinetic features of clozapine block and correctly predict the overall affinity and apparent nonstate-dependent interaction of clozapine with Kv11.1. Mechanistic insights into drug block of Kv11.1 gained though detailed kinetic analyses such as this have a potential role in development of drugs targeted to specific channel states to reduce unwanted side effects, as well as in the design of better high-throughput preclinical tests for assessing the proarrhythmic effects of QT prolonging drugs.


Assuntos
Clozapina/farmacocinética , Canais de Potássio Éter-A-Go-Go/agonistas , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Bloqueadores dos Canais de Potássio/farmacocinética , Animais , Células CHO , Células Cultivadas , Clozapina/metabolismo , Cricetinae , Cricetulus , Interações Medicamentosas/fisiologia , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/metabolismo , Cinética , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Bloqueadores dos Canais de Potássio/metabolismo
3.
J Pharmacol Exp Ther ; 351(3): 596-604, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25232191

RESUMO

Human ether-a-go-go-related gene (hERG) and KCNQ channels are two classes of voltage-gated potassium channels. Specific mutations have been identified that are causal for type II long QT (LQT2) syndrome, neonatal epilepsy, and benign familial neonatal convulsions. Increasing evidence from clinical studies suggests that LQT2 and epilepsy coexist in some patients. Therefore, an integral approach to investigating and treating the two diseases is likely more effective. In the current study, we found that NS1643 [1,3-bis-(2-hydroxy-5-trifluoromethyl-phenyl)-urea], a previously reported hERG activator, is also an activator of KCNQ channels. It potentiates the neuronal KCNQ2, KCNQ4, and KCNQ2/Q3 channels, but not the cardiac KCNQ1. The effects of NS1643 on the KCNQ2 channel include left shifting of voltage for reaching 50% of the maximum conductance and slowing of deactivation. Analysis of the dose-response curve of NS1643 revealed an EC50 value of 2.44 ± 0.25 µM. A hydrophobic phenylalanine (F137) located at the middle region of the voltage-sensing domain was identified as critical for NS1643 activity on KCNQ2. When testing NS1643 effects in rescuing LQT2 hERG mutants and the KCNQ2 BFNC mutants, we found it is particularly efficacious in some cases. Considering the substantial relationship between LQT2 and epilepsy, these findings reveal that NS1643 is a useful compound to elucidate the causal connection of LQT2 and epilepsy. More generally, this may provide a strategy in the development of therapeutics for LQT2 and epilepsy.


Assuntos
Cresóis/metabolismo , Cresóis/farmacologia , Epilepsia/metabolismo , Canais de Potássio Éter-A-Go-Go/metabolismo , Canal de Potássio KCNQ2/metabolismo , Compostos de Fenilureia/metabolismo , Compostos de Fenilureia/farmacologia , Animais , Células CHO , Cricetinae , Cricetulus , Relação Dose-Resposta a Droga , Canais de Potássio Éter-A-Go-Go/agonistas , Humanos , Canal de Potássio KCNQ2/agonistas , Estrutura Secundária de Proteína
4.
Antimicrob Agents Chemother ; 56(9): 4891-9, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22777050

RESUMO

Infection with human cytomegalovirus (HCMV) continues to be a major threat for pregnant women and the immunocompromised population. Although several anti-HCMV therapies are available, the development of new anti-HCMV agents is highly desired. There is growing interest in identifying compounds that might inhibit HCMV by modulating the cellular milieu. Interest in cardiac glycosides (CG), used in patients with congestive heart failure, has increased because of their established anticancer and their suggested antiviral activities. We report that the several CG--digoxin, digitoxin, and ouabain--are potent inhibitors of HCMV at nM concentrations. HCMV inhibition occurred prior to DNA replication, but following binding to its cellular receptors. The levels of immediate early, early, and late viral proteins and cellular NF-κB were significantly reduced in CG-treated cells. The activity of CG in infected cells correlated with the expression of the potassium channel gene, hERG. CMV infection upregulated hERG, whereas CG significantly downregulated its expression. Infection with mouse CMV upregulated mouse ERG (mERG), but treatment with CG did not inhibit virus replication or mERG transcription. These findings suggest that CG may inhibit HCMV by modulating human cellular targets associated with hERG and that these compounds should be studied for their antiviral activities.


Assuntos
Antivirais/farmacologia , Glicosídeos Cardíacos/farmacologia , Citomegalovirus/efeitos dos fármacos , Canais de Potássio Éter-A-Go-Go , Proteínas Virais/metabolismo , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Citomegalovirus/crescimento & desenvolvimento , Digitoxina/farmacologia , Digoxina/farmacologia , Canais de Potássio Éter-A-Go-Go/agonistas , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Canais de Potássio Éter-A-Go-Go/genética , Fibroblastos/efeitos dos fármacos , Fibroblastos/virologia , Expressão Gênica , Genes Reporter , Humanos , Camundongos , NF-kappa B/antagonistas & inibidores , NF-kappa B/genética , NF-kappa B/metabolismo , Ouabaína/farmacologia , Ativação Transcricional , Proteínas Virais/genética , Replicação Viral/efeitos dos fármacos
5.
Eur Biophys J ; 41(11): 949-58, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22936309

RESUMO

The purpose of this study was to obtain functional hERG ion channel protein for use in a non-cell-based ion channel assay. hERG was expressed in Sf9 insect cells. Attempts to solubilise the hERG protein from Sf9 insect cell membranes using non-ionic detergents (NP40 and DDM) were not successful. We therefore generated liposomes from the unpurified membrane fraction and incorporated these into porous Teflon-supported bilayer lipid membranes. Macroscopic potassium currents (1 nA) were recorded that approximated those in whole-cell patch-clamping, but the channels were bidirectional in the bilayer lipid membrane (BLM). Currents were partially inhibited by the hERG blockers E4031, verapamil, and clofilium, indicating that the protein of interest is present at high levels in the BLM relative to endogenous channels. Cell liposomes produced from Sf9 insect cell membranes expressing voltage-gated sodium channels also gave current responses that were activated by veratridine and inhibited by saxitoxin. These results demonstrate that purification of the ion channel of interest is not always necessary for liposomes used in macro-current BLM systems.


Assuntos
Canais de Potássio Éter-A-Go-Go/fisiologia , Bicamadas Lipídicas/metabolismo , Lipossomos/metabolismo , Animais , Membrana Celular/metabolismo , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/agonistas , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Células HEK293 , Humanos , Potenciais da Membrana , Técnicas de Patch-Clamp , Potássio/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Células Sf9 , Spodoptera , Veratridina/farmacologia
6.
Proc Natl Acad Sci U S A ; 106(47): 20075-80, 2009 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-19892732

RESUMO

Human ether-a-go-go-related gene 1 (hERG1) K(+) channels mediate repolarization of cardiac action potentials. Unintended block of hERG1 channels by some drugs can prolong the QT interval and induce arrhythmia. Recently, hERG1 channel agonists were discovered and, based on their mechanisms of action can be classified into two types. RPR260243 [(3R,4R)-4-[3-(6-methoxy-quinolin-4-yl)-3-oxo-propyl]-1-[3-(2,3,5 trifluorophenyl)-prop-2-ynyl]-piperidine-3-carboxylic acid], a type 1 agonist, binds to residues located near the intracellular end of S5 and S6 transmembrane segments and activates hERG1 channels by a dual mechanism of slowed deactivation and attenuated P-type inactivation. As defined here, type 2 agonists such as PD-118057 [2-(4-[2-(3,4-dichloro-phenyl)-ethyl]-phenylamino)-benzoic acid] attenuate inactivation but do not slow deactivation. At 10 muM, PD-118057 shifted the half-point for inactivation of wild-type hERG1 channels by +19 mV and increased peak outward current by 136%. Scanning mutagenesis and functional characterization of 44 mutant channels expressed in Xenopus oocytes was used to identify the major structural determinants of the binding site for PD-118057. Single mutations of residues in the pore helix (F619) or the S6 segment (L646) of hERG1 eliminated agonist activity. Mutation of a nearby residues in the S6 segment (C643, M645) enhanced drug activity, presumably by reducing steric hindrance for drug binding. Molecular modeling indicates that PD-118057 binds to a hydrophobic pocket formed by L646 of one hERG1 subunit and F619 of an adjacent subunit. We conclude that direct interaction of PD-118057 with the pore helix attenuates fast P-type inactivation and increases open probability of hERG1 channels.


Assuntos
Canais de Potássio Éter-A-Go-Go , Ativação do Canal Iônico , Estrutura Secundária de Proteína , ortoaminobenzoatos/química , Animais , Clorobenzenos , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/agonistas , Canais de Potássio Éter-A-Go-Go/química , Canais de Potássio Éter-A-Go-Go/metabolismo , Humanos , Modelos Moleculares , Mutagênese , Oócitos/citologia , Oócitos/fisiologia , Técnicas de Patch-Clamp , Potássio/metabolismo , Bloqueadores dos Canais de Potássio/química , Estrutura Terciária de Proteína , Xenopus laevis
7.
Biochem Biophys Res Commun ; 415(1): 141-6, 2011 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-22020101

RESUMO

Nifekalant and azimilide, Class III antiarrhythmic agents, block the human ether-à-go-go-related gene K(+) (hERG) channel. However, when a depolarizing membrane potential is applied, they also increase the current at low potentials by shifting its activation curve towards hyperpolarizing voltages. This phenomenon is called 'facilitation'. In this study, we tried to address the mechanism underlying the facilitation by analyzing the effects of various compounds on hERG expressed in Xenopus oocytes. Like nifekalant, amiodarone, quinidine and carvedilol, but not by dofetilide, caused the current facilitation of hERG, suggesting that the facilitation is a common effect to a subset of hERG blockers. As the concentration of each compound was increased, the total hERG current was suppressed progressively, while the current at low potentials was augmented. Activation curves of the remaining hERG current in the facilitation condition could be described as the sum of two Boltzmann functions reflecting two populations of hERG currents having different activation curves. The voltage shift in the activation curve from control was constant for each compound even at different concentrations; -31 mV in amiodarone, -27 mV in nifekalant, -17 mV in quinidine and -12 mV in carvedilol. Therefore, the facilitation is based on the appearance of hERG whose voltage-dependence for the activation is shifted towards hyperpolarizing voltages.


Assuntos
Antiarrítmicos/farmacologia , Canais de Potássio Éter-A-Go-Go/agonistas , Amiodarona/farmacologia , Animais , Canal de Potássio ERG1 , Humanos , Hidantoínas , Imidazolidinas/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Piperazinas/farmacologia , Pirimidinonas/farmacologia , Xenopus laevis
8.
J Cardiovasc Pharmacol ; 57(2): 223-30, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21135701

RESUMO

Transgenic rabbits expressing pore mutants of K(V)7.1 display a long QT syndrome 1 (LQT1) phenotype. Recently, NS1643 has been described to increase I(Kr).We hypothesized that NS1643 would shorten the action potential duration (APD(90)) in LQT1 rabbits. Transgenic LQT1 rabbits were compared with littermate control (LMC) rabbits. In vivo electrocardiogram studies in sedated animals were performed at baseline and during 45 minutes of intravenous infusion of NS1643 or vehicle in a crossover design. Ex vivo monophasic action potentials were recorded from Langendorff-perfused hearts at baseline and during 45-minute perfusion with NS1643. Left ventricular refractory periods were assessed before and after NS1643 infusion. Genotype differences in APD accommodation were also addressed. In vivo NS1643 shortened the QTc significantly in LQT1 compared with vehicle. In Langendorff experiments, NS1643 significantly shortened the APD(90) in LQT1 and LMC [32.0 ± 4.3 milliseconds (ms); 21.0 ± 5.0 ms] and left ventricular refractory periods (23.7 ± 8.3; 22.6 ± 9.9 ms). NS1643 significantly decreased dp/dt (LQT1: 49% ± 3%; LMC: 63% ± 4%) and increased the incidence of arrhythmia. The time course of APD adaptation was impaired in LQT1 rabbits and unaffected by I(Kr) augmentation. In conclusion, K(V)11.1 channel activation shortens the cardiac APD in a rabbit model of inherited LQT1, but it comes with the risk of excessive shortening of APD.


Assuntos
Animais Geneticamente Modificados/genética , Cresóis/farmacologia , Canais de Potássio Éter-A-Go-Go/metabolismo , Síndrome do QT Longo/genética , Síndrome do QT Longo/metabolismo , Compostos de Fenilureia/farmacologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Estudos Cross-Over , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/agonistas , Feminino , Coelhos , Distribuição Aleatória
9.
Mol Pharmacol ; 77(1): 58-68, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19805508

RESUMO

Human ether-à-go-go-related gene (hERG) potassium channel activity helps shape the cardiac action potential and influences its duration. In this study, we report the discovery of 3-nitro-N-(4-phenoxyphenyl) benzamide (ICA-105574), a potent and efficacious hERG channel activator with a unique mechanism of action. In whole-cell patch-clamp studies of recombinant hERG channels, ICA-105574 steeply potentiated current amplitudes more than 10-fold with an EC(50) value of 0.5 +/- 0.1 microM and a Hill slope (n(H)) of 3.3 +/- 0.2. The effect on hERG channels was confirmed because the known hERG channel blockers, N-[4-[[1-[2-(6-methyl-2-pyridinyl)ethyl]-4-piperidinyl]carbonyl]phenyl]methanesulfonamide, 2HCl (E-4031) and BeKm-1, potently blocked the stimulatory effects of ICA-105574. The primary mechanism by which ICA-105574 potentiates hERG channel activity is by removing hERG channel inactivation, because ICA-105574 (2 microM) shifts the midpoint of the voltage-dependence of inactivation by >180 mV from -86 to +96 mV. In addition to the effects on inactivation, greater concentrations of ICA-105574 (3 microM) produced comparatively small hyperpolarizing shifts (up to 11 mV) in the voltage-dependence of channel activation and a 2-fold slowing of channel deactivation. In isolated guinea pig ventricular cardiac myocytes, ICA-105574 induced a concentration-dependent shortening of action potential duration (>70%, 3 microM) that could be prevented by preincubation with E-4031. In conclusion, we identified a novel agent that can prevent the inactivation of hERG potassium channels. This compound may provide a useful tool to further understand the mechanism by which hERG channels inactivate and affect cardiac function in addition to the role of hERG channels in other cell systems.


Assuntos
Canais de Potássio Éter-A-Go-Go/agonistas , Bloqueadores dos Canais de Potássio/antagonistas & inibidores , Potenciais de Ação/efeitos dos fármacos , Animais , Células Cultivadas , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Cobaias , Humanos , Miócitos Cardíacos/citologia , Técnicas de Patch-Clamp , Piperidinas/farmacologia , Piridinas/farmacologia
10.
J Cardiovasc Electrophysiol ; 21(8): 923-9, 2010 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-20163495

RESUMO

INTRODUCTION: The hERG (Kv11.1) potassium channel underlies cardiac I(Kr) and is important for cardiac repolarization. Recently, hERG agonists have emerged as potential antiarrhythmic drugs. As modulation of outward potassium currents has been suggested to modulate cardiac conduction, we tested the hypothesis that pharmacological activation of I(Kr) results in impaired cardiac conduction. METHODS AND RESULTS: Cardiac conduction was assessed in Langendorff-perfused guinea pig hearts. Application of the hERG agonist NS3623 (10 microM) prolonged the QRS rate dependently. A significant prolongation (16 +/- 6%) was observed at short basic cycle length (BCL 90 ms) but not at longer cycle lengths (BCL 250 ms). The effect could be reversed by the I(Kr) blocker E4031 (1 microM). While partial I(Na) inhibition with flecainide (1 microM) alone prolonged the QRS (34 +/- 3%, BCL 250 ms), the QRS was further prolonged by 19 +/- 2% when NS3623 was added in the presence of flecainide. These data suggest that the effect of NS3623 was dependent on sodium channel availability. Surprisingly, in the presence of the voltage sensitive dye di-4-ANEPPS a similar potentiation of the effect of NS3623 was observed. With di-4-ANEPPS, NS3623 prolonged the QRS significantly (26 +/- 4%, BCL 250 ms) compared to control with a corresponding decrease in conduction velocity. CONCLUSION: Pharmacological activation of I(Kr) by the hERG agonist NS3623 impairs cardiac conduction. The effect is dependent on sodium channel availability. These findings suggest a role for I(Kr) in modulating cardiac conduction and may have implications for the use of hERG agonists as antiarrhythmic drugs.


Assuntos
Arritmias Cardíacas/tratamento farmacológico , Canais de Potássio Éter-A-Go-Go/agonistas , Sistema de Condução Cardíaco/efeitos dos fármacos , Compostos de Fenilureia/farmacologia , Potássio/metabolismo , Tetrazóis/farmacologia , Potenciais de Ação , Animais , Antiarrítmicos/efeitos adversos , Antiarrítmicos/farmacologia , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/fisiopatologia , Estimulação Cardíaca Artificial , Canais de Potássio Éter-A-Go-Go/metabolismo , Flecainida/farmacologia , Cobaias , Sistema de Condução Cardíaco/metabolismo , Sistema de Condução Cardíaco/fisiopatologia , Técnicas In Vitro , Masculino , Perfusão , Compostos de Fenilureia/efeitos adversos , Piperidinas/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Piridinas/farmacologia , Bloqueadores dos Canais de Sódio/farmacologia , Tetrazóis/efeitos adversos , Fatores de Tempo , Imagens com Corantes Sensíveis à Voltagem
11.
Drug Discov Today ; 25(2): 344-366, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31756511

RESUMO

hERG is best known as a primary anti-target, the inhibition of which is responsible for serious side effects. A renewed interest in hERG as a desired target, especially in oncology, was sparked because of its role in cellular proliferation and apoptosis. In this study, we survey the most recent advances regarding hERG by focusing on SAR in the attempt to elucidate, at a molecular level, off-target and on-target actions of potential hERG binders, which are highly promiscuous and largely varying in structure. Understanding the rationale behind hERG interactions and the molecular determinants of hERG activity is a real challenge and comprehension of this is of the utmost importance to prioritize compounds in early stages of drug discovery and to minimize cardiotoxicity attrition in preclinical and clinical studies.


Assuntos
Canais de Potássio Éter-A-Go-Go , Moduladores de Transporte de Membrana , Animais , Desenho de Fármacos , Canais de Potássio Éter-A-Go-Go/agonistas , Canais de Potássio Éter-A-Go-Go/química , Canais de Potássio Éter-A-Go-Go/fisiologia , Humanos , Moduladores de Transporte de Membrana/química , Moduladores de Transporte de Membrana/farmacologia , Relação Estrutura-Atividade
12.
J Physiol ; 587(Pt 10): 2313-26, 2009 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-19332483

RESUMO

There is a growing appreciation that ion channels encoded by the ether-à-go-go-related gene family have a functional impact in smooth muscle in addition to their accepted role in cardiac myocytes and neurones. This study aimed to assess the expression of ERG1-3 (KCNH1-3) genes in the murine myometrium (smooth muscle layer of the uterus) and determine the functional impact of the ion channels encoded by these genes in pregnant and non-pregnant animals. Quantitative RT-PCR did not detect message for ERG2 and 3 in whole myometrial tissue extracts. In contrast, message for two isoforms of mERG1 were readily detected with mERG1a more abundant than mERG1b. In isometric tension studies of non-pregnant myometrium, the ERG channel blockers dofetilide (1 microM), E4031 (1 microM) and Be-KM1 (100 nM) increased spontaneous contractility and ERG activators (PD118057 and NS1643) inhibited spontaneous contractility. In contrast, neither ERG blockade nor activation had any effect on the inherent contractility in myometrium from late pregnant (19 days gestation) animals. Moreover, dofetilide-sensitive K(+) currents with distinctive 'hooked' kinetics were considerably smaller in uterine myocytes from late pregnant compared to non-pregnant animals. Expression of mERG1 isoforms did not alter throughout gestation or upon delivery, but the expression of genes encoding auxillary subunits (KCNE) were up-regulated considerably. This study provides the first evidence for a regulation of ERG-encoded K(+) channels as a precursor to late pregnancy physiological activity.


Assuntos
Canais de Potássio Éter-A-Go-Go/fisiologia , Trabalho de Parto/fisiologia , Miométrio/fisiologia , 4-Aminopiridina/farmacologia , Animais , Clorobenzenos , Cresóis/farmacologia , Canal de Potássio ERG1 , Estimulação Elétrica , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Fenômenos Eletrofisiológicos/fisiologia , Canais de Potássio Éter-A-Go-Go/agonistas , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Feminino , Expressão Gênica/genética , Camundongos , Camundongos Endogâmicos BALB C , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/fisiologia , Miométrio/efeitos dos fármacos , Ocitocina/farmacologia , Técnicas de Patch-Clamp , Compostos de Fenilureia/farmacologia , Pinacidil/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Gravidez , Contração Uterina/efeitos dos fármacos , Contração Uterina/fisiologia , ortoaminobenzoatos/farmacologia
13.
Biochem Biophys Res Commun ; 381(2): 159-64, 2009 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-19232322

RESUMO

The zebrafish is increasingly recognized as an animal model for the analysis of hERG-related diseases. However, functional properties of the zebrafish orthologue of hERG have not been analyzed yet. We heterologously expressed cloned ERG channels in Xenopus oocytes and analyzed biophysical properties using the voltage clamp technique. zERG channels conduct rapidly activating and inactivating potassium currents. However, compared to hERG, the half-maximal activation voltage of zERG current is shifted towards more positive potentials and the half maximal steady-state inactivation voltage is shifted towards more negative potentials. zERG channel activation is delayed and channel deactivation is accelerated significantly. However, time course of zERG conducted current under action potential clamp is highly similar to the human orthologue. In summary, we show that ERG channels in zebrafish exhibit biophysical properties similar to the human orthologue. Considering the conserved channel function, the zebrafish represents a valuable model to investigate human ERG channel related diseases.


Assuntos
Canais de Potássio Éter-A-Go-Go/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Peixe-Zebra/fisiologia , Animais , Canais de Potássio Éter-A-Go-Go/agonistas , Canais de Potássio Éter-A-Go-Go/genética , Humanos , Potenciais da Membrana , Oócitos , Xenopus , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/agonistas , Proteínas de Peixe-Zebra/genética
14.
Prog Biophys Mol Biol ; 98(2-3): 347-62, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19351523

RESUMO

The cardiac action potential is the result of an orchestrated function of a number of different ion channels. Action potential repolarisation in humans relies on three potassium current components named I(Kr), I(Ks) and I(K1) with party overlapping functions. The ion channel alpha-subunits conducting these currents are hERG1 (Kv11.1), KCNQ1 (Kv7.1) and Kir2.1. Loss-of-function in any of these currents can result in long QT syndrome. Long QT is a pro-arrhythmic disease with increased risk of developing lethal ventricular arrhythmias such as Torsade de Pointes and ventricular fibrillation. In addition to congenital long QT, acquired long QT can also constitute a safety risk. Especially unintended inhibition of the hERG1 channel constitutes a major concern in the development of new drugs. Based on this knowledge is has been speculated whether activation of the hERG1 channel could be anti-arrhythmic and thereby constitute a new principle in treatment of cardiac arrhythmogenic disorders. The first hERG1 channel agonist was reported in 2005 and a limited number of such compounds are now available. In the present text we review results obtained by hERG1 channel activation in a number of cardiac relevant settings from in vitro to in vivo. It is demonstrated how the principle of hERG1 channel activation under certain circumstances can constitute a new anti-arrhythmogenic principle. Finally, important conceptual differences between the short QT syndrome and the hERG1 channel activation, are evaluated.


Assuntos
Antiarrítmicos/farmacologia , Arritmias Cardíacas/tratamento farmacológico , Canais de Potássio Éter-A-Go-Go/agonistas , Potenciais de Ação , Arritmias Cardíacas/etiologia , Arritmias Cardíacas/metabolismo , Fenômenos Biofísicos , Humanos , Ativação do Canal Iônico , Canais KATP/agonistas
15.
Mol Pharmacol ; 74(6): 1696-704, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18809671

RESUMO

1,3-Bis-[2-hydroxy-5-(trifluoromethyl)phenyl]urea (NS1643) is reported to be an activator of human ether-à-go-go-related gene current. However, it remains unknown whether it has any effects on other types of ion channels. The effects of NS1643 on ion currents and membrane potential were investigated in this study. NS1643 stimulated Ca(2+)-activated K(+) current [I(K(Ca))] in a concentration-dependent manner with an EC(50) value of 1.8 microM in pituitary tumor (GH(3)) cells. In inside-out recordings, this compound applied to the intracellular side of the detached channels stimulated large-conductance Ca(2+)-activated K(+) (BK(Ca)) channels with no change in single-channel conductance. It shifted the activation curve of BK(Ca) channels to less depolarized voltages without altering the gating charge of the channels. NS1643-stimulated channel activity depended on intracellular Ca(2+), and mean closed time during exposure to NS1643 was reduced. NS1643 (3 microM) had little or no effect on peak amplitude of ether-à-go-go-related gene-mediated K(+) current evoked by membrane hyperpolarization, although it increased the amplitude of late-sustained components of K(+) inward current, which was suppressed by paxilline but not by azimilide. NS1643 (3 microM) had no effect on L-type Ca(2+) current. This compound reduced repetitive firing of action potentials, and further application of paxilline attenuated its decrease in firing rate. In addition, NS1643 enhanced BK(Ca)-channel activity in human embryonic kidney 293T cells expressing alpha-hSlo. In summary, we clearly show that NS1643 interacts directly with the BK(Ca) channel to increase the amplitude of I(K(Ca)) in pituitary tumor (GH(3)) cells. The alpha-subunit of the channel may be a target for the action of this small compound.


Assuntos
Cresóis/farmacologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/agonistas , Compostos de Fenilureia/farmacologia , Potenciais de Ação , Animais , Cálcio/metabolismo , Linhagem Celular Tumoral , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/agonistas , Humanos , Ativação do Canal Iônico , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/genética , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/fisiologia , Técnicas de Patch-Clamp , Neoplasias Hipofisárias , Ratos
16.
Mol Pharmacol ; 73(6): 1709-21, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18372399

RESUMO

We studied the mechanisms and sites of activator actions of 2-[2-(3,4-dichloro-phenyl)-2,3-dihydro-1H-isoindol-5-ylamino]-nicotinic acid [PD307243 (PD)] and 1,3-bis-(2-hydroxy-5-trifluoromethyl-phenyl)-urea [NS1643 (NS)] on the human ether-a-go-go-related gene (hERG) channel expressed in oocytes and COS-7 cells. PD and NS affected hERG in a concentration-dependent manner, reaching a maximal increase in current amplitude by 100% and > or = 300% (1-s test pulse to 0 mV), with apparent K(d) values of 3 and 20 microM, respectively. Both drugs slowed hERG inactivation. NS additionally shifted the activation curve in the negative direction, accelerated activation, and slowed deactivation. Kinetic model simulations suggested that the activator effects of PD and NS could be largely accounted for by their effects on the hERG gating kinetics. Both drugs worked from outside the cell membrane but their binding sites seemed to be distinctly different. Perturbing the conformation of outer vestibule/external pore entrance (by cysteine substitution at high-impact positions or cysteine side chain modification at intermediate-impact positions) prevented the activator effect of NS but not that of PD. Furthermore, the peptide toxin BeKm-1, which bound to the outer mouth of the hERG channel, suppressed NS effect but potentiated PD effect. We propose that NS is a "gating-modifier": it binds to the outer vestibule/pore entrance of hERG and increases current amplitudes by promoting channel activation while retarding inactivation. The activator effect of PD was prevented by external quaternary ammonium cations or dofetilide, which approached the hERG selectivity filter from opposite sides of the membrane and depleted K(+) ions in the selectivity filter. We suggest that PD may work as a "pore-modifier" of the hERG channel.


Assuntos
Cresóis/metabolismo , Canais de Potássio Éter-A-Go-Go/agonistas , Canais de Potássio Éter-A-Go-Go/metabolismo , Isoindóis/metabolismo , Ácidos Nicotínicos/metabolismo , Compostos de Fenilureia/metabolismo , Animais , Sítios de Ligação/efeitos dos fármacos , Sítios de Ligação/fisiologia , Células COS , Chlorocebus aethiops , Cresóis/farmacologia , Feminino , Cobaias , Humanos , Isoindóis/farmacologia , Masculino , Ácidos Nicotínicos/farmacologia , Compostos de Fenilureia/farmacologia , Xenopus
17.
J Cardiovasc Pharmacol ; 52(1): 35-41, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18594476

RESUMO

The long QT syndrome is characterized by a prolongation of the QT interval measured on the surface electrocardiogram. Prolonging the QT interval increases the risk of dangerous ventricular fibrillations, eventually leading to sudden cardiac death. Pharmacologically induced QT interval prolongations are most often caused by antagonizing effects on the repolarizing cardiac current called IKr. In humans IKr is mediated by the human ether-a-go-go related gene (hERG) potassium channel. We recently presented NS3623, a compound that selectively activates this channel. The present study was dedicated to examining the in vivo effects of NS3623. Injection of 30 mg/kg NS3623 shortened the corrected QT interval by 25 +/- 4% in anaesthetized guinea pigs. Accordingly, 50 mg/kg of NS3623 shortened the QT interval by 30 +/- 6% in conscious guinea pigs. Finally, pharmacologically induced QT prolongation by a hERG channel antagonist (0.15 mg/kg E-4031) could be reverted by injection of NS3623 (50 mg/kg) in conscious guinea pigs. In conclusion, the present in vivo study demonstrates that injection of the hERG channel agonist NS3623 results in shortening of the QTc interval as well as reversal of a pharmacologically induced QT prolongation in both anaesthetized and conscious guinea pigs.


Assuntos
Antiarrítmicos/farmacologia , Canais de Potássio de Retificação Tardia/agonistas , Canais de Potássio Éter-A-Go-Go/agonistas , Frequência Cardíaca/efeitos dos fármacos , Compostos de Fenilureia/farmacologia , Tetrazóis/farmacologia , Animais , Estado de Consciência , Canais de Potássio de Retificação Tardia/antagonistas & inibidores , Relação Dose-Resposta a Droga , Eletrocardiografia , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Feminino , Cobaias , Frequência Cardíaca/fisiologia , Humanos , Síndrome do QT Longo/induzido quimicamente , Síndrome do QT Longo/fisiopatologia , Piperidinas/efeitos adversos , Piperidinas/farmacologia , Piridinas/efeitos adversos , Piridinas/farmacologia
18.
Br J Pharmacol ; 175(4): 606-617, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29181850

RESUMO

BACKGROUND AND PURPOSE: Translation of non-clinical markers of delayed ventricular repolarization to clinical prolongation of the QT interval corrected for heart rate (QTc) (a biomarker for torsades de pointes proarrhythmia) remains an issue in drug discovery and regulatory evaluations. We retrospectively analysed 150 drug applications in a US Food and Drug Administration database to determine the utility of established non-clinical in vitro IKr current human ether-à-go-go-related gene (hERG), action potential duration (APD) and in vivo (QTc) repolarization assays to detect and predict clinical QTc prolongation. EXPERIMENTAL APPROACH: The predictive performance of three non-clinical assays was compared with clinical thorough QT study outcomes based on free clinical plasma drug concentrations using sensitivity and specificity, receiver operating characteristic (ROC) curves, positive (PPVs) and negative predictive values (NPVs) and likelihood ratios (LRs). KEY RESULTS: Non-clinical assays demonstrated robust specificity (high true negative rate) but poor sensitivity (low true positive rate) for clinical QTc prolongation at low-intermediate (1×-30×) clinical exposure multiples. The QTc assay provided the most robust PPVs and NPVs (ability to predict clinical QTc prolongation). ROC curves (overall test accuracy) and LRs (ability to influence post-test probabilities) demonstrated overall marginal performance for hERG and QTc assays (best at 30× exposures), while the APD assay demonstrated minimal value. CONCLUSIONS AND IMPLICATIONS: The predictive value of hERG, APD and QTc assays varies, with drug concentrations strongly affecting translational performance. While useful in guiding preclinical candidates without clinical QT prolongation, hERG and QTc repolarization assays provide greater value compared with the APD assay.


Assuntos
Fármacos Cardiovasculares/farmacologia , Drogas em Investigação/farmacologia , Canais de Potássio Éter-A-Go-Go/fisiologia , Frequência Cardíaca/efeitos dos fármacos , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Fármacos Cardiovasculares/uso terapêutico , Avaliação Pré-Clínica de Medicamentos/métodos , Drogas em Investigação/uso terapêutico , Canais de Potássio Éter-A-Go-Go/agonistas , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Frequência Cardíaca/fisiologia , Humanos , Síndrome do QT Longo/tratamento farmacológico , Síndrome do QT Longo/fisiopatologia , Estudos Retrospectivos , Torsades de Pointes/tratamento farmacológico , Torsades de Pointes/fisiopatologia
19.
Eur J Pharmacol ; 818: 306-327, 2018 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-29050968

RESUMO

Despite the importance of the hERG channel in drug discovery and the sizable number of antagonist molecules discovered, only a few hERG agonists have been discovered. Here we report a novel hERG agonist; SKF-32802 and a structural analog of the agonist NS3623, SB-335573. These were discovered through a similarity search of published hERG agonists. SKF-32802 incorporates an amide linker rather than NS3623's urea, resulting in a compound with a different mechanism of action. We find that both compounds decrease the time constant of open channel kinetics, increase the amplitude of the envelope of tails assay, mildly increased the amplitude of the IV curve, bind the hERG channel in either open or closed states, increase the plateau of the voltage dependence of activation and modulate the effects of the hERG antagonist, quinidine. Neither compound affects inactivation nor deactivation kinetics, a property unique among hERG agonists. Additionally, SKF-32802 induces a leftward shift in the voltage dependence of activation. Our structural models show that both compounds make strong bridging interactions with multiple channel subunits and are stabilized by internal hydrogen bonding similar to NS3623, PD-307243 and RPR26024. While SB-335573 binds in a nearly identical fashion as NS3623, SKF-32802 makes an additional hydrogen bond with neighboring threonine 623. In summary, SB-335573 is a type 4 agonist which increases open channel probability while SKF-32802 is a type 3 agonist which induces a leftward shift in the voltage dependence of activation.


Assuntos
Compostos de Anilina/química , Compostos de Anilina/farmacologia , Descoberta de Drogas , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Canais de Potássio Éter-A-Go-Go/agonistas , Tetrazóis/química , Tetrazóis/farmacologia , Compostos de Anilina/metabolismo , Animais , Células CHO , Cricetinae , Cricetulus , Relação Dose-Resposta a Droga , Canais de Potássio Éter-A-Go-Go/química , Canais de Potássio Éter-A-Go-Go/metabolismo , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Cinética , Simulação de Acoplamento Molecular , Conformação Proteica , Tetrazóis/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA