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










Base de datos
Intervalo de año de publicación
1.
Am J Physiol Heart Circ Physiol ; 319(2): H251-H261, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32559136

RESUMEN

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.


Asunto(s)
Antiarrítmicos/farmacología , Arritmias Cardíacas/prevención & control , Canal de Potasio ERG1/agonistas , Canales de Potasio Éter-A-Go-Go/agonistas , Frecuencia Cardíaca/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Piperidinas/farmacología , Quinolinas/farmacología , Proteínas de Pez Cebra/agonistas , Potenciales de Acción , Animales , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/fisiopatología , Modelos Animales de Enfermedad , Canal de Potasio ERG1/genética , Canal de Potasio ERG1/metabolismo , Canales de Potasio Éter-A-Go-Go/metabolismo , Cinética , Miocitos Cardíacos/metabolismo , Oocitos , Periodo Refractario Electrofisiológico , Transducción de Señal , Xenopus laevis , Pez Cebra , Proteínas de Pez Cebra/metabolismo
2.
Drug Discov Today ; 25(2): 344-366, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31756511

RESUMEN

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.


Asunto(s)
Canales de Potasio Éter-A-Go-Go , Moduladores del Transporte de Membrana , Animales , Diseño de Fármacos , Canales de Potasio Éter-A-Go-Go/agonistas , Canales de Potasio Éter-A-Go-Go/química , Canales de Potasio Éter-A-Go-Go/fisiología , Humanos , Moduladores del Transporte de Membrana/química , Moduladores del Transporte de Membrana/farmacología , Relación Estructura-Actividad
3.
Br J Pharmacol ; 175(4): 606-617, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29181850

RESUMEN

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.


Asunto(s)
Fármacos Cardiovasculares/farmacología , Drogas en Investigación/farmacología , Canales de Potasio Éter-A-Go-Go/fisiología , Frecuencia Cardíaca/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Fármacos Cardiovasculares/uso terapéutico , Evaluación Preclínica de Medicamentos/métodos , Drogas en Investigación/uso terapéutico , Canales de Potasio Éter-A-Go-Go/agonistas , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Frecuencia Cardíaca/fisiología , Humanos , Síndrome de QT Prolongado/tratamiento farmacológico , Síndrome de QT Prolongado/fisiopatología , Estudios Retrospectivos , Torsades de Pointes/tratamiento farmacológico , Torsades de Pointes/fisiopatología
4.
Eur J Pharmacol ; 818: 306-327, 2018 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-29050968

RESUMEN

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.


Asunto(s)
Compuestos de Anilina/química , Compuestos de Anilina/farmacología , Descubrimiento de Drogas , Fenómenos Electrofisiológicos/efectos de los fármacos , Canales de Potasio Éter-A-Go-Go/agonistas , Tetrazoles/química , Tetrazoles/farmacología , Compuestos de Anilina/metabolismo , Animales , Células CHO , Cricetinae , Cricetulus , Relación Dosis-Respuesta a Droga , Canales de Potasio Éter-A-Go-Go/química , Canales de Potasio Éter-A-Go-Go/metabolismo , Humanos , Activación del Canal Iónico/efectos de los fármacos , Cinética , Simulación del Acoplamiento Molecular , Conformación Proteica , Tetrazoles/metabolismo
5.
PLoS One ; 9(9): e105553, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25191697

RESUMEN

One of the main culprits in modern drug discovery is apparent cardiotoxicity of many lead-candidates via inadvertent pharmacologic blockade of K+, Ca2+ and Na+ currents. Many drugs inadvertently block hERG1 leading to an acquired form of the Long QT syndrome and potentially lethal polymorphic ventricular tachycardia. An emerging strategy is to rely on interventions with a drug that may proactively activate hERG1 channels reducing cardiovascular risks. Small molecules-activators have a great potential for co-therapies where the risk of hERG-related QT prolongation is significant and rehabilitation of the drug is impractical. Although a number of hERG1 activators have been identified in the last decade, their binding sites, functional moieties responsible for channel activation and thus mechanism of action, have yet to be established. Here, we present a proof-of-principle study that combines de-novo drug design, molecular modeling, chemical synthesis with whole cell electrophysiology and Action Potential (AP) recordings in fetal mouse ventricular myocytes to establish basic chemical principles required for efficient activator of hERG1 channel. In order to minimize the likelihood that these molecules would also block the hERG1 channel they were computationally engineered to minimize interactions with known intra-cavitary drug binding sites. The combination of experimental and theoretical studies led to identification of functional elements (functional groups, flexibility) underlying efficiency of hERG1 activators targeting binding pocket located in the S4-S5 linker, as well as identified potential side-effects in this promising line of drugs, which was associated with multi-channel targeting of the developed drugs.


Asunto(s)
Diseño de Fármacos , Canales de Potasio Éter-A-Go-Go/química , Modelos Moleculares , Potenciales de Acción/efectos de los fármacos , Sitios de Unión , Cresoles/síntesis química , Cresoles/química , Cresoles/farmacología , Bases de Datos Farmacéuticas , Relación Dosis-Respuesta a Droga , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/agonistas , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Canales de Potasio Éter-A-Go-Go/metabolismo , Humanos , Ligandos , Conformación Molecular , Simulación del Acoplamiento Molecular , Compuestos de Fenilurea/síntesis química , Compuestos de Fenilurea/química , Compuestos de Fenilurea/farmacología , Unión Proteica , Bibliotecas de Moléculas Pequeñas
6.
J Pharmacol Exp Ther ; 351(3): 596-604, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25232191

RESUMEN

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.


Asunto(s)
Cresoles/metabolismo , Cresoles/farmacología , Epilepsia/metabolismo , Canales de Potasio Éter-A-Go-Go/metabolismo , Canal de Potasio KCNQ2/metabolismo , Compuestos de Fenilurea/metabolismo , Compuestos de Fenilurea/farmacología , Animales , Células CHO , Cricetinae , Cricetulus , Relación Dosis-Respuesta a Droga , Canales de Potasio Éter-A-Go-Go/agonistas , Humanos , Canal de Potasio KCNQ2/agonistas , Estructura Secundaria de Proteína
7.
Curr Opin Pharmacol ; 15: 22-7, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24721650

RESUMEN

Type 1 human ether-a-go-go-related gene (hERG1) potassium channels are a key determinant of normal repolarization of cardiac action potentials. Loss of function mutations in hERG1 channels cause inherited long QT syndrome and increased risk of cardiac arrhythmia and sudden death. Many common medications that block hERG1 channels as an unintended side effect also increase arrhythmic risk. Routine preclinical screening for hERG1 block led to the discovery of agonists that shorten action potential duration and QT interval. Agonists have the potential to be used as pharmacotherapy for long QT syndrome, but can also be proarrhythmic. Recent studies have elucidated multiple mechanisms of action for these compounds and the structural basis for their binding to the pore domain of the hERG1 channel.


Asunto(s)
Arritmias Cardíacas/metabolismo , Canales de Potasio Éter-A-Go-Go/agonistas , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Arritmias Cardíacas/inducido químicamente , Arritmias Cardíacas/tratamiento farmacológico , Humanos , Activación del Canal Iónico/efectos de los fármacos
8.
Mol Pharmacol ; 85(5): 769-76, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24586056

RESUMEN

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.


Asunto(s)
Clozapina/farmacocinética , Canales de Potasio Éter-A-Go-Go/agonistas , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Bloqueadores de los Canales de Potasio/farmacocinética , Animales , Células CHO , Células Cultivadas , Clozapina/metabolismo , Cricetinae , Cricetulus , Interacciones Farmacológicas/fisiología , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/metabolismo , Cinética , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Bloqueadores de los Canales de Potasio/metabolismo
9.
Cell Death Dis ; 4: e652, 2013 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-23744352

RESUMEN

Traditionally the hERG1 potassium channel has been known to have a fundamental role in membrane excitability of several mammalian cells including cardiac myocytes. hERG1 has recently been found to be expressed in non-excitable cancer cells of different histogenesis, but the role of this channel in cancer biology is unknown. Results form recent studies on the effect hERG1 inhibition in some breast cancer cells are controversial as it can lead to apoptosis or protect against cell death. Nevertheless, these data suggest that the hERG1 channel could have an important role in cancer biology. Here we report the effects of hyperstimulation of hERG1 channel in human mammary gland adenocarcinoma-derived cells. Application of the hERG1 activator, the diphenylurea derivative NS1643, inhibits cell proliferation irreversibly. This event is accompanied by a preferential arrest of the cell cycle in G0/G1 phase without the occurrence of apoptotic events. Consequently, cells responded to NS1643 by developing a senescence-like phenotype associated with increased protein levels of the tumor suppressors p21 and p16(INK4a) and by a positive ß-galactosidase assay. These data suggest that prolonged stimulation of the hERG1 potassium channel may activate a senescence program and offers a compelling opportunity to develop a potential antiproliferative cancer therapy.


Asunto(s)
Proliferación Celular , Senescencia Celular , Canales de Potasio Éter-A-Go-Go/metabolismo , Animales , Neoplasias de la Mama , Células CHO , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Cresoles/farmacología , Cricetulus , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/agonistas , Femenino , Humanos , Compuestos de Fenilurea/farmacología
10.
Eur Biophys J ; 41(11): 949-58, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22936309

RESUMEN

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.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/fisiología , Membrana Dobles de Lípidos/metabolismo , Liposomas/metabolismo , Animales , Membrana Celular/metabolismo , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/agonistas , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Células HEK293 , Humanos , Potenciales de la Membrana , Técnicas de Placa-Clamp , Potasio/metabolismo , Bloqueadores de los Canales de Potasio/farmacología , Células Sf9 , Spodoptera , Veratridina/farmacología
11.
Antimicrob Agents Chemother ; 56(9): 4891-9, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22777050

RESUMEN

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.


Asunto(s)
Antivirales/farmacología , Glicósidos Cardíacos/farmacología , Citomegalovirus/efectos de los fármacos , Canales de Potasio Éter-A-Go-Go , Proteínas Virales/metabolismo , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Citomegalovirus/crecimiento & desarrollo , Digitoxina/farmacología , Digoxina/farmacología , Canales de Potasio Éter-A-Go-Go/agonistas , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Canales de Potasio Éter-A-Go-Go/genética , Fibroblastos/efectos de los fármacos , Fibroblastos/virología , Expresión Génica , Genes Reporteros , Humanos , Ratones , FN-kappa B/antagonistas & inhibidores , FN-kappa B/genética , FN-kappa B/metabolismo , Ouabaína/farmacología , Activación Transcripcional , Proteínas Virales/genética , Replicación Viral/efectos de los fármacos
12.
Biochem Biophys Res Commun ; 415(1): 141-6, 2011 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-22020101

RESUMEN

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.


Asunto(s)
Antiarrítmicos/farmacología , Canales de Potasio Éter-A-Go-Go/agonistas , Amiodarona/farmacología , Animales , Canal de Potasio ERG1 , Humanos , Hidantoínas , Imidazolidinas/farmacología , Potenciales de la Membrana/efectos de los fármacos , Piperazinas/farmacología , Pirimidinonas/farmacología , Xenopus laevis
13.
Curr Med Chem ; 18(24): 3607-21, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21774764

RESUMEN

The delayed rectifier potassium current (I(K)) is the major outward current responsible for ventricular repolarization in cardiac tissues. Based on kinetic properties and drug sensitivity it is composed of a slow (I(Ks)) and a rapid (I(Kr)) component, the latter is mediated by hERG channels. Suppression of IKr is the common mechanism of action of all class III antiarrhythmics, causing prolongation of the refractory period. However, lengthening of repolarization - either by a pathological factor or due to a pharmacological intervention - threatens with an increased risk of EAD generation and the concomitant sudden cardiac death. Therefore, a new potential anti-arrhythmic strategy, based on augmentation of the repolarization reserve, has been emerged. Recently a new class of compounds has been introduced as activators of the hERG channel. In this article we systematically review the chemical structures found to enhance IKr. Since the majority of previous experiments were performed in expression systems or in rodent cardiac preparations (neither is relevant to the human heart), in the second part of this article we present some results obtained with NS1643, the best examined hERG activator, in canine ventricular cardiomyocytes. This preparation is believed to have electrophysiological parameters most resembling those of human. NS1643 shortened the duration of canine ventricular action potential and was shown to interact with several transmembrane ion currents, including I(Ca), I(Kr), I(Ks), and I(to). However, the action potential shortening effect of NS1643 is likely related to inhibition of ICa, in addition to the enhancement of IKr. Although the multiple ion channel activity of NS1643 may carry proarrhythmic risk, the rationale of antiarrhythmic strategy based on I(Kr) activation is not questioned.


Asunto(s)
Antiarrítmicos/farmacología , Canales de Potasio Éter-A-Go-Go/metabolismo , Miocitos Cardíacos/fisiología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Canales de Calcio/química , Canales de Calcio/metabolismo , Cresoles/farmacología , Canales de Potasio Éter-A-Go-Go/agonistas , Humanos , Miocitos Cardíacos/efectos de los fármacos , Compuestos de Fenilurea/farmacología , Canales de Potasio de Rectificación Interna/agonistas , Canales de Potasio de Rectificación Interna/metabolismo
14.
J Cardiovasc Pharmacol ; 57(2): 223-30, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21135701

RESUMEN

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.


Asunto(s)
Animales Modificados Genéticamente/genética , Cresoles/farmacología , Canales de Potasio Éter-A-Go-Go/metabolismo , Síndrome de QT Prolongado/genética , Síndrome de QT Prolongado/metabolismo , Compuestos de Fenilurea/farmacología , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Estudios Cruzados , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/agonistas , Femenino , Conejos , Distribución Aleatoria
15.
Br J Pharmacol ; 161(3): 614-28, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20880400

RESUMEN

BACKGROUND AND PURPOSE: K(v)11.1 channels are involved in regulating cellular excitability in various tissues including brain, heart and smooth muscle. In these tissues, at least two isoforms, K(v)11.1a and K(v)11.1b, with different kinetics, are expressed. K(v)11.1 activators are potential therapeutic agents, but their effects have only been tested on the K(v)11.1a isoform. In this study, the effects of two different K(v)11.1 activators, NS1643 and RPR260243, were characterized on K(v)11.1a and K(v)11.1b channels. EXPERIMENTAL APPROACH: K(v)11.1a and K(v)11.1b channels were expressed in Xenopus laevis oocytes, and currents were measured using two-electrode voltage clamp. I/V curves and channel kinetics were measured before and after application of 30 µM NS1643 or 10 µM RPR260243. KEY RESULTS: NS1643 increased steady-state currents through Kv11.1b several fold more than through K(v)11.1a channels, without affecting EC(50) values. NS1643 increased activation rates and decreased rates of inactivation, recovery from inactivation and deactivation for both channels. Except for activation, where effect of NS1643 was comparable, relative changes were greater for Kv11.1b than for K(v)11.1a. RPR260243 increased steady-state currents only through Kv11.1a channels, but slowed the process of deactivation for both channels primarily by decreasing time constant of slow deactivation. This effect was greater on K(v)11.1b than on K(v)11.1a. Effects of both compounds on heteromeric K(v)11.1a/K(v)11.1b channels were similar to those on K(v)11.1a. CONCLUSIONS AND IMPLICATIONS: Both NS1643 and RPR260243 displayed differential effects on K(v)11.1a and K(v)11.1b channels, the effects being relatively more pronounced on K(v)11.1b channels. This affirms the importance of testing the effect of K(v)11.1 activators on different channel isoforms.


Asunto(s)
Cresoles/farmacología , Canales de Potasio Éter-A-Go-Go/agonistas , Compuestos de Fenilurea/farmacología , Piperidinas/farmacología , Isoformas de Proteínas/agonistas , Quinolinas/farmacología , Animales , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/genética , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/fisiología , Oocitos/efectos de los fármacos , Oocitos/fisiología , Técnicas de Placa-Clamp , Transfección , Xenopus laevis
16.
Curr Med Chem ; 17(30): 3514-32, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20738248

RESUMEN

Blockade of the human ether-a-go-go related gene 1 (hERG1) channel has been associated with an increased duration of ventricular repolarization, causing prolongation of the time interval between Q and T waves (long QT syndrome, or LQTS). LQTS may result in serious cardiovascular disorders such as tachyarrhythmia and sudden cardiac death. Diverse types of organic compounds bind to the wide intracellular cavity in the pore domain of hERG channels, leading to a full or partial blockade of ion current through the pore. The drug-induced blockade of the hERG-related component of the potassium current is thought to be a major reason for drug-induced arrhythmias in humans. Identification of specific interactions governing the high-affinity blockade of cardiac potassium (K-) channels is crucial both for the prevention of unintended ion channel block and for the design of ion channel modulators. A plethora of ligand- and receptor-based models of K-channels have been created to address these challenges. In this paper, we review the current state of knowledge regarding the structure-function relationship of hERG and discuss progress in the use of molecular modeling for developing both blockers and activators of hERG.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/química , Bloqueadores de los Canales de Potasio/química , Canales de Potasio Éter-A-Go-Go/agonistas , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Humanos , Modelos Químicos , Modelos Moleculares , Bloqueadores de los Canales de Potasio/farmacología , Estructura Terciaria de Proteína
18.
J Cardiovasc Electrophysiol ; 21(8): 923-9, 2010 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-20163495

RESUMEN

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.


Asunto(s)
Arritmias Cardíacas/tratamiento farmacológico , Canales de Potasio Éter-A-Go-Go/agonistas , Sistema de Conducción Cardíaco/efectos de los fármacos , Compuestos de Fenilurea/farmacología , Potasio/metabolismo , Tetrazoles/farmacología , Potenciales de Acción , Animales , Antiarrítmicos/efectos adversos , Antiarrítmicos/farmacología , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/fisiopatología , Estimulación Cardíaca Artificial , Canales de Potasio Éter-A-Go-Go/metabolismo , Flecainida/farmacología , Cobayas , Sistema de Conducción Cardíaco/metabolismo , Sistema de Conducción Cardíaco/fisiopatología , Técnicas In Vitro , Masculino , Perfusión , Compuestos de Fenilurea/efectos adversos , Piperidinas/farmacología , Bloqueadores de los Canales de Potasio/farmacología , Piridinas/farmacología , Bloqueadores de los Canales de Sodio/farmacología , Tetrazoles/efectos adversos , Factores de Tiempo , Imagen de Colorante Sensible al Voltaje
19.
Mol Pharmacol ; 77(1): 58-68, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19805508

RESUMEN

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.


Asunto(s)
Canales de Potasio Éter-A-Go-Go/agonistas , Bloqueadores de los Canales de Potasio/antagonistas & inhibidores , Potenciales de Acción/efectos de los fármacos , Animales , Células Cultivadas , Canales de Potasio Éter-A-Go-Go/antagonistas & inhibidores , Cobayas , Humanos , Miocitos Cardíacos/citología , Técnicas de Placa-Clamp , Piperidinas/farmacología , Piridinas/farmacología
20.
Proc Natl Acad Sci U S A ; 106(47): 20075-80, 2009 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-19892732

RESUMEN

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.


Asunto(s)
Canales de Potasio Éter-A-Go-Go , Activación del Canal Iónico , Estructura Secundaria de Proteína , ortoaminobenzoatos/química , Animales , Clorobencenos , Canal de Potasio ERG1 , Canales de Potasio Éter-A-Go-Go/agonistas , Canales de Potasio Éter-A-Go-Go/química , Canales de Potasio Éter-A-Go-Go/metabolismo , Humanos , Modelos Moleculares , Mutagénesis , Oocitos/citología , Oocitos/fisiología , Técnicas de Placa-Clamp , Potasio/metabolismo , Bloqueadores de los Canales de Potasio/química , Estructura Terciaria de Proteína , Xenopus laevis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...