Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Arterioscler Thromb Vasc Biol ; 34(7): 1522-30, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24855057

RESUMO

OBJECTIVE: Voltage-dependent K(+) (Kv) channels from the Kv7 family are expressed in blood vessels and contribute to cardiovascular physiology. Although Kv7 channel blockers trigger muscle contractions, Kv7 activators act as vasorelaxants. Kv7.1 and Kv7.5 are expressed in many vessels. Kv7.1 is under intense investigation because Kv7.1 blockers fail to modulate smooth muscle reactivity. In this study, we analyzed whether Kv7.1 and Kv7.5 may form functional heterotetrameric channels increasing the channel diversity in vascular smooth muscles. APPROACH AND RESULTS: Kv7.1 and Kv7.5 currents elicited in arterial myocytes, oocyte, and mammalian expression systems suggest the formation of heterotetrameric complexes. Kv7.1/Kv7.5 heteromers, exhibiting different pharmacological characteristics, participate in the arterial tone. Kv7.1/Kv7.5 associations were confirmed by coimmunoprecipitation, fluorescence resonance energy transfer, and fluorescence recovery after photobleaching experiments. Kv7.1/Kv7.5 heterotetramers were highly retained at the endoplasmic reticulum. Studies in HEK-293 cells, heart, brain, and smooth and skeletal muscles demonstrated that the predominant presence of Kv7.5 stimulates release of Kv7.1/Kv7.5 oligomers out of lipid raft microdomains. Electrophysiological studies supported that KCNE1 and KCNE3 regulatory subunits further increased the channel diversity. Finally, the analysis of rat isolated myocytes and human blood vessels demonstrated that Kv7.1 and Kv7.5 exhibited a differential expression, which may lead to channel diversity. CONCLUSIONS: Kv7.1 and Kv7.5 form heterotetrameric channels increasing the diversity of structures which fine-tune blood vessel reactivity. Because the lipid raft localization of ion channels is crucial for cardiovascular physiology, Kv7.1/Kv7.5 heteromers provide efficient spatial and temporal regulation of smooth muscle function. Our results shed light on the debate about the contribution of Kv7 channels to vasoconstriction and hypertension.


Assuntos
Canais de Potássio KCNQ/metabolismo , Canal de Potássio KCNQ1/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Potássio/metabolismo , Animais , Células COS , Chlorocebus aethiops , Células HEK293 , Humanos , Canais de Potássio KCNQ/química , Canais de Potássio KCNQ/efeitos dos fármacos , Canais de Potássio KCNQ/genética , Canal de Potássio KCNQ1/química , Canal de Potássio KCNQ1/efeitos dos fármacos , Canal de Potássio KCNQ1/genética , Microdomínios da Membrana/metabolismo , Potenciais da Membrana , Músculo Liso Vascular/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Estrutura Quaternária de Proteína , Ratos , Transfecção , Xenopus
2.
Circ Res ; 110(2): 211-9, 2012 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-22095730

RESUMO

RATIONALE: The mutation A341V in the S6 transmembrane segment of KCNQ1, the α-subunit of the slowly activating delayed-rectifier K(+) (I(Ks)) channel, predisposes to a severe long-QT1 syndrome with sympathetic-triggered ventricular tachyarrhythmias and sudden cardiac death. OBJECTIVE: Several genetic risk modifiers have been identified in A341V patients, but the molecular mechanisms underlying the pronounced repolarization phenotype, particularly during ß-adrenergic receptor stimulation, remain unclear. We aimed to elucidate these mechanisms and provide new insights into control of cAMP-dependent modulation of I(Ks). METHODS AND RESULTS: We characterized the effects of A341V on the I(Ks) macromolecular channel complex in transfected Chinese hamster ovary cells and found a dominant-negative suppression of cAMP-dependent Yotiao-mediated I(Ks) upregulation on top of a dominant-negative reduction in basal current. Phosphomimetic substitution of the N-terminal position S27 with aspartic acid rescued this loss of upregulation. Western blot analysis showed reduced phosphorylation of KCNQ1 at S27, even for heterozygous A341V, suggesting that phosphorylation defects in some (mutant) KCNQ1 subunits can completely suppress I(Ks) upregulation. Functional analyses of heterozygous KCNQ1 WT:G589D and heterozygous KCNQ1 WT:S27A, a phosphorylation-inert substitution, also showed such suppression. Immunoprecipitation of Yotiao with KCNQ1-A341V (in the presence of KCNE1) was not different from wild-type. CONCLUSIONS: Our results indicate the involvement of the KCNQ1-S6 region at/or around A341 in cAMP-dependent stimulation of I(Ks), a process that is under strong dominant-negative control, suggesting that tetrameric KCNQ1 phosphorylation is required. Specific long-QT1 mutations, including heterozygous A341V, disable this regulation.


Assuntos
AMP Cíclico/metabolismo , Genes Dominantes , Canal de Potássio KCNQ1/genética , Canal de Potássio KCNQ1/metabolismo , Mutação , Miócitos Cardíacos/metabolismo , Síndrome de Romano-Ward/genética , Síndrome de Romano-Ward/metabolismo , Agonistas Adrenérgicos beta/farmacologia , Alanina , Animais , Ácido Aspártico , Western Blotting , Células CHO , Simulação por Computador , Cricetinae , Cricetulus , Cães , Predisposição Genética para Doença , Heterozigoto , Humanos , Imunoprecipitação , Canal de Potássio KCNQ1/efeitos dos fármacos , Potenciais da Membrana , Modelos Cardiovasculares , Mutagênese Sítio-Dirigida , Miócitos Cardíacos/efeitos dos fármacos , Fenótipo , Fosforilação , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Processamento de Proteína Pós-Traducional , Síndrome de Romano-Ward/fisiopatologia , Fatores de Tempo , Transfecção
3.
Toxicology ; 505: 153830, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38754619

RESUMO

The use of tyrosine kinase inhibitors (TKIs) has resulted in significant occurrence of arrhythmias. However, the precise mechanism of the proarrhythmic effect is not fully understood. In this study, we found that nilotinib (NIL), vandetanib (VAN), and mobocertinib (MOB) induced the development of "cellrhythmia" (arrhythmia-like events) in a concentration-dependent manner in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Continuous administration of NIL, VAN, or MOB in animals significantly prolonged the action potential durations (APD) and increased susceptibility to arrhythmias. Using phosphoproteomic analysis, we identified proteins with altered phosphorylation levels after treatment with 3 µM NIL, VAN, and MOB for 1.5 h. Using these identified proteins as substrates, we performed kinase-substrate enrichment analysis to identify the kinases driving the changes in phosphorylation levels of these proteins. MAPK and WNK were both inhibited by NIL, VAN, and MOB. A selective inhibitor of WNK1, WNK-IN-11, induced concentration- and time-dependent cellrhythmias and prolonged field potential duration (FPD) in hiPSC-CMs in vitro; furthermore, administration in guinea pigs confirmed that WNK-IN-11 prolonged ventricular repolarization and increased susceptibility to arrhythmias. Fingding indicated that WNK1 inhibition had an in vivo and in vitro arrhythmogenic phenotype similar to TKIs. Additionally,three of TKIs reduced hERG and KCNQ1 expression at protein level, not at transcription level. Similarly, the knockdown of WNK1 decreased hERG and KCNQ1 protein expression in hiPSC-CMs. Collectively, our data suggest that the proarrhythmic effects of NIL, VAN, and MOB occur through a kinase inhibition mechanism. NIL, VAN, and MOB inhibit WNK1 kinase, leading to a decrease in hERG and KCNQ1 protein expression, thereby prolonging action potential repolarization and consequently cause arrhythmias.


Assuntos
Potenciais de Ação , Arritmias Cardíacas , Miócitos Cardíacos , Piperidinas , Proteômica , Pirimidinas , Quinazolinas , Humanos , Arritmias Cardíacas/induzido quimicamente , Animais , Proteômica/métodos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Piperidinas/farmacologia , Piperidinas/toxicidade , Pirimidinas/toxicidade , Pirimidinas/farmacologia , Quinazolinas/toxicidade , Quinazolinas/farmacologia , Potenciais de Ação/efeitos dos fármacos , Inibidores de Proteínas Quinases/toxicidade , Inibidores de Proteínas Quinases/farmacologia , Fosforilação , Canal de Potássio ERG1/metabolismo , Canal de Potássio ERG1/antagonistas & inibidores , Canal de Potássio ERG1/genética , Cobaias , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Células-Tronco Pluripotentes Induzidas/metabolismo , Masculino , Canal de Potássio KCNQ1/metabolismo , Canal de Potássio KCNQ1/genética , Canal de Potássio KCNQ1/efeitos dos fármacos , Fosfoproteínas/metabolismo , Relação Dose-Resposta a Droga
4.
Am J Physiol Gastrointest Liver Physiol ; 304(2): G157-66, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23154976

RESUMO

Gastric acid secretion by the H(+)-K(+)-ATPase at the apical surface of activated parietal cells requires luminal K(+) provided by the KCNQ1/KCNE2 K(+) channel. However, little is known about the trafficking and relative spatial distribution of KCNQ1 and H(+)-K(+)-ATPase in resting and activated parietal cells and the capacity of KCNQ1 to control acid secretion. Here we show that inhibition of KCNQ1 activity quickly curtails gastric acid secretion in vivo, even when the H(+)-K(+)-ATPase is permanently anchored in the apical membrane, demonstrating a key role of the K(+) channel in controlling acid secretion. Three-dimensional imaging analysis of isolated mouse gastric units revealed that the majority of KCNQ1 resides in an intracytoplasmic, Rab11-positive compartment in resting parietal cells, distinct from H(+)-K(+)-ATPase-enriched tubulovesicles. Upon activation, there was a significant redistribution of H(+)-K(+)-ATPase and KCNQ1 from intracytoplasmic compartments to the apical secretory canaliculi. Significantly, high Förster resonance energy transfer was detected between H(+)-K(+)-ATPase and KCNQ1 in activated, but not resting, parietal cells. These findings demonstrate that H(+)-K(+)-ATPase and KCNQ1 reside in independent intracytoplasmic membrane compartments, or membrane domains, and upon activation of parietal cells, both membrane proteins are transported, possibly via Rab11-positive recycling endosomes, to apical membranes, where the two molecules are closely physically opposed. In addition, these studies indicate that acid secretion is regulated by independent trafficking of KCNQ1 and H(+)-K(+)-ATPase.


Assuntos
Ácido Gástrico/metabolismo , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Canal de Potássio KCNQ1/metabolismo , Células Parietais Gástricas/enzimologia , Animais , Membrana Celular/enzimologia , Cromanos/farmacologia , Citoplasma/enzimologia , Endossomos/enzimologia , Transferência Ressonante de Energia de Fluorescência , Histamina/metabolismo , Canal de Potássio KCNQ1/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência , Células Parietais Gástricas/efeitos dos fármacos , Células Parietais Gástricas/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Transporte Proteico , Sulfonamidas/farmacologia , Fatores de Tempo , Proteínas rab de Ligação ao GTP/metabolismo
5.
J Appl Toxicol ; 33(8): 723-39, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22334483

RESUMO

The excitable cell membranes contain ion channels that allow the ions passage through the specific pores via a passive process. Assessment of the inhibition of the IKr (hERG) current is considered to be the main target during the drug development process, although there are other ionic currents for which drug-triggered modification can either potentiate or mask hERG channel blockade. Information describing the results of in vitro studies investigating the chemical-IKs current interactions has been developed in the current study. Based on the publicly available data sources, 145 records were collected. The final list of publications consists of 64 positions and refers to 106 different molecules connected with IKs current inhibition, with at least one IC50 value measured. Ultimately, 98 of the IC50 values expressed as absolute values were gathered. For 36 records the IC50 was expressed as a relative value. For the 11 remaining records, the inhibition was not clearly expressed. Based on the collected data the predictive models for the IC50 estimation were developed with the use of various algorithms. The extended Quantitative Structure-Activity Relationships (QSAR) methodology was applied and the in vitro research settings were included as independent variables, apart from the physico-chemical descriptors calculated with the use of the Marvin Calculator Plugins. The root mean squared error and normalized root mean squared error values for the best model (an expert system based on two independent artificial neural networks) were 0.86 and 14.04%, respectively. The model was further built into the ToxComp system, the ToxIVIVE tool specialized for cardiotoxicity assessment of drugs.


Assuntos
Canal de Potássio KCNQ1/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/farmacologia , Potássio/metabolismo , Animais , Linhagem Celular , Cricetinae , Células HEK293 , Humanos , Concentração Inibidora 50 , Canal de Potássio KCNQ1/metabolismo , Modelos Biológicos , Relação Quantitativa Estrutura-Atividade , Xenopus
6.
Am J Physiol Renal Physiol ; 299(6): F1308-19, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20861072

RESUMO

The KCNQ1 K(+) channel plays a key role in the regulation of several physiological functions, including cardiac excitability, cardiovascular tone, and body electrolyte homeostasis. The metabolic sensor AMP-activated protein kinase (AMPK) has been shown to regulate a growing number of ion transport proteins. To determine whether AMPK regulates KCNQ1, we studied the effects of AMPK activation on KCNQ1 currents in Xenopus laevis oocytes and collecting duct epithelial cells. AMPK activation decreased KCNQ1 currents and channel surface expression in X. laevis oocytes, but AMPK did not phosphorylate KCNQ1 in vitro, suggesting an indirect regulatory mechanism. As it has been recently shown that the ubiquitin-protein ligase Nedd4-2 inhibits KCNQ1 plasma membrane expression and that AMPK regulates epithelial Na(+) channels via Nedd4-2, we examined the role of Nedd4-2 in the AMPK-dependent regulation of KCNQ1. Channel inhibition by AMPK was blocked in oocytes coexpressing either a dominant-negative or constitutively active Nedd4-2 mutant, or a Nedd4-2 interaction-deficient KCNQ1 mutant, suggesting that Nedd4-2 participates in the regulation of KCNQ1 by AMPK. KCNQ1 is expressed at the basolateral membrane in mouse polarized kidney cortical collecting duct (mpkCCD(c14)) cells and in rat kidney. Treatment with the AMPK activators AICAR (2 mM) or metformin (1 mM) reduced basolateral KCNQ1 currents in apically permeabilized polarized mpkCCD(c14) cells. Moreover, AICAR treatment of rat kidney slices ex vivo induced AMPK activation and intracellular redistribution of KCNQ1 from the basolateral membrane in collecting duct principal cells. AICAR treatment also induced increased ubiquitination of KCNQ1 immunoprecipitated from kidney slice homogenates. These results indicate that AMPK inhibits KCNQ1 activity by promoting Nedd4-2-dependent channel ubiquitination and retrieval from the plasma membrane.


Assuntos
Complexos Endossomais de Distribuição Requeridos para Transporte/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Quinases Ativadas por AMP , Aminoimidazol Carboxamida/análogos & derivados , Animais , Células Epiteliais/efeitos dos fármacos , Células HEK293 , Humanos , Canal de Potássio KCNQ1/efeitos dos fármacos , Canal de Potássio KCNQ1/metabolismo , Camundongos , Ubiquitina-Proteína Ligases Nedd4 , Ratos , Ribonucleotídeos , Proteínas de Xenopus , Xenopus laevis
7.
Elife ; 92020 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-32207683

RESUMO

The cardiac ventricular action potential depends on several voltage-gated ion channels, including NaV, CaV, and KV channels. Mutations in these channels can cause Long QT Syndrome (LQTS) which increases the risk for ventricular fibrillation and sudden cardiac death. Polyunsaturated fatty acids (PUFAs) have emerged as potential therapeutics for LQTS because they are modulators of voltage-gated ion channels. Here we demonstrate that PUFA analogues vary in their selectivity for human voltage-gated ion channels involved in the ventricular action potential. The effects of specific PUFA analogues range from selective for a specific ion channel to broadly modulating cardiac ion channels from all three families (NaV, CaV, and KV). In addition, a PUFA analogue selective for the cardiac IKs channel (Kv7.1/KCNE1) is effective in shortening the cardiac action potential in human-induced pluripotent stem cell-derived cardiomyocytes. Our data suggest that PUFA analogues could potentially be developed as therapeutics for LQTS and cardiac arrhythmia.


Assuntos
Canais de Cálcio Tipo L/efeitos dos fármacos , Ácidos Graxos Insaturados/farmacologia , Canal de Potássio KCNQ1/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.5/efeitos dos fármacos , Canais de Potássio de Abertura Dependente da Tensão da Membrana/efeitos dos fármacos , Proteínas de Xenopus/efeitos dos fármacos , Potenciais de Ação/efeitos dos fármacos , Animais , Antiarrítmicos/farmacologia , Canais de Cálcio Tipo L/fisiologia , Células-Tronco Pluripotentes Induzidas/citologia , Canal de Potássio KCNQ1/fisiologia , Síndrome do QT Longo/tratamento farmacológico , Miócitos Cardíacos/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.5/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/fisiologia , Proteínas de Xenopus/fisiologia , Xenopus laevis
8.
Eur J Pharmacol ; 886: 173542, 2020 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-32910945

RESUMO

Cannabidiol (CBD) is a non-psychoactive component of Cannabis which has recently received regulatory consideration for the treatment of intractable forms of epilepsy such as the Dravet and the Lennox-Gastaut syndromes. The mechanisms of the antiepileptic effects of CBD are unclear, but several pre-clinical studies suggest the involvement of ion channels. Therefore, we have evaluated the effects of CBD on seven major cardiac currents shaping the human ventricular action potential and on Purkinje fibers isolated from rabbit hearts to assess the in vitro cardiac safety profile of CBD. We found that CBD inhibits with comparable micromolar potencies the peak and late components of the NaV1.5 sodium current, the CaV1.2 mediated L-type calcium current, as well as all the repolarizing potassium currents examined except Kir2.1. The most sensitive channels were KV7.1 and the least sensitive were KV11.1 (hERG), which underly the slow (IKs) and rapid (IKr) components, respectively, of the cardiac delayed-rectifier current. In the Purkinje fibers, CBD decreased the action potential (AP) duration more potently at half-maximal than at near complete repolarization, and slightly decreased the AP amplitude and its maximal upstroke velocity. CBD had no significant effects on the membrane resting potential except at the highest concentration tested under fast pacing rate. These data show that CBD impacts cardiac electrophysiology and suggest that caution should be exercised when prescribing CBD to carriers of cardiac channelopathies or in conjunction with other drugs known to affect heart rhythm or contractility.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Canabidiol/farmacologia , Ventrículos do Coração/efeitos dos fármacos , Coração/efeitos dos fármacos , Canais Iônicos/efeitos dos fármacos , Animais , Canabidiol/toxicidade , Canalopatias/complicações , Humanos , Técnicas In Vitro , Canal de Potássio KCNQ1/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.5/efeitos dos fármacos , Técnicas de Patch-Clamp , Ramos Subendocárdicos/efeitos dos fármacos , Coelhos
9.
Pflugers Arch ; 457(5): 1111-20, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18709386

RESUMO

This study represents an extensive characterisation of the expression and functional impact of KCNQ and KCNE accessory subunits in a murine uterus using a combination of quantitative reverse transcription polymerase chain reaction, Western blot analysis, patch clamp electrophysiology and isometric tension recording. The use of uterine tissue throughout the oestrous cycle provided a physiological model with which to assess hormonal regulation of these genes. Messenger ribonucleic acid for all KCNQ genes were detected throughout the oestrous cycle with the KCNQ1 message predominant. KCNE isoforms were detected at each stage of the cycle. KCNE4 was the most abundant (p < 0.0001), and KCNQ1, KCNQ5 and KCNE1 were up-regulated in metestrous (p < 0.0001). The K(v)7 channel inhibitor XE991 reduced outward K(+) currents and significantly increased spontaneous myometrial contractions (p < 0.05), whereas retigabine (K(v)7 activator) significantly relaxed uterine tissues (p < 0.001). These data are the first to characterise KCNQ and KCNE gene expression in a cell type outside of neurons and the cardiovascular system.


Assuntos
Ciclo Estral/fisiologia , Canal de Potássio KCNQ1/fisiologia , Miométrio/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/fisiologia , Animais , Antracenos/farmacologia , Carbamatos/farmacologia , Cromanos/farmacologia , Feminino , Canal de Potássio KCNQ1/biossíntese , Canal de Potássio KCNQ1/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Fenilenodiaminas/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/biossíntese , Sulfonamidas/farmacologia , Contração Uterina/efeitos dos fármacos
10.
J Urol ; 182(1): 330-6, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19450820

RESUMO

PURPOSE: The presence of novel KCNQ currents was investigated in guinea pig bladder interstitial cells of Cajal and their contribution to the maintenance of the resting membrane potential was assessed. MATERIALS AND METHODS: Enzymatically dispersed interstitial cells of Cajal were patch clamped with K(+) filled pipettes in voltage clamp and current clamp modes. Pharmacological modulators of KCNQ channels were tested on membrane currents and the resting membrane potential. RESULTS: Cells were stepped from -60 to 40 mV to evoke voltage dependent currents using a modified K(+) pipette solution containing ethylene glycol tetraacetic acid (5 mM) and adenosine triphosphate (3 mM) to eliminate large conductance Ca activated K channel and K(adenosine triphosphate) currents. Application of the KCNQ blockers XE991, linopirdine (Tocris Bioscience, Ellisville, Missouri) and chromanol 293B (Sigma) decreased the outward current in concentration dependent fashion. The current-voltage relationship of XE991 sensitive current revealed a voltage dependent, outwardly rectifying current that activated positive to -60 mV and showed little inactivation. The KCNQ openers flupirtine and meclofenamic acid (Sigma) increased outward currents across the voltage range. In current clamp mode XE991 or chromanol 293B decreased interstitial cell of Cajal resting membrane potential and elicited the firing of spontaneous transient depolarizations in otherwise quiescent cells. Flupirtine or meclofenamic acid hyperpolarized interstitial cells of Cajal and inhibited any spontaneous electrical activity. CONCLUSIONS: This study provides electrophysiological evidence that bladder interstitial cells of Cajal have KCNQ currents with a role in the regulation of interstitial cell of Cajal resting membrane potential and excitability. These novel findings provide key information on the ion channels present in bladder interstitial cells of Cajal and they may indicate relevant targets for the development of new therapies for bladder instability.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Canal de Potássio KCNQ1/metabolismo , Músculo Liso/fisiologia , Miócitos de Músculo Liso/metabolismo , Bloqueadores dos Canais de Potássio/farmacologia , Bexiga Urinária/citologia , Bexiga Urinária/fisiologia , Animais , Células Cultivadas , Eletrofisiologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/fisiologia , Feminino , Cobaias , Canal de Potássio KCNQ1/efeitos dos fármacos , Masculino , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Modelos Animais , Contração Muscular/efeitos dos fármacos , Contração Muscular/fisiologia , Miócitos de Músculo Liso/efeitos dos fármacos , Probabilidade , Distribuição Aleatória , Sensibilidade e Especificidade
11.
Curr Opin Pharmacol ; 8(1): 65-74, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18061539

RESUMO

The Kv7 potassium channel family encompasses five members (from Kv7.1 to Kv7.5) having distinct expression pattern and functional role. Although Kv7.1 is prevalently expressed in the cardiac muscle, Kv7.2, Kv7.3, Kv7.4, and Kv7.5 are expressed in neural tissue. Mutations in Kv7.2 and/or Kv7.3 genes are responsible for an autosomal-dominant epilepsy of the newborn defined as benign familial neonatal seizures (BFNS), whereas defects in the Kv7.4 gene have been found in families affected by a rare form of nonsyndromic autosomal-dominant hearing loss (DFNA2). Compounds acting as direct activators of neuronal channels formed by Kv7 subunits have been approved for clinical use as analgesics or are in advanced stages of clinical evaluation as anticonvulsants; in addition to these indications, solid preclinical studies reveal their potential usefulness in other diseases characterized by neuronal hyperexcitability. In the present work, we will summarize the available evidence providing proof-of-principles that neuronal Kv7 channels are highly attractive pharmacological targets, review the molecular basis of their peculiar pharmacological sensitivity, introduce some newly synthesized I(KM) openers showing improved pharmacokinetic or pharmacodynamic properties compared to older congeners, and discuss the potential novel therapeutic application of neuronal Kv7 channels in diseases additional to epilepsy.


Assuntos
Canais de Potássio KCNQ/efeitos dos fármacos , Canal de Potássio KCNQ1/efeitos dos fármacos , Canal de Potássio KCNQ2/efeitos dos fármacos , Canal de Potássio KCNQ3/efeitos dos fármacos , Aminopiridinas/farmacologia , Aminopiridinas/uso terapêutico , Animais , Sítios de Ligação , Carbamatos/farmacologia , Carbamatos/uso terapêutico , Epilepsia Neonatal Benigna/tratamento farmacológico , Perda Auditiva/tratamento farmacológico , Perda Auditiva/genética , Humanos , Canais de Potássio KCNQ/genética , Canais de Potássio KCNQ/fisiologia , Canal de Potássio KCNQ1/genética , Canal de Potássio KCNQ1/fisiologia , Canal de Potássio KCNQ2/genética , Canal de Potássio KCNQ2/fisiologia , Canal de Potássio KCNQ3/genética , Canal de Potássio KCNQ3/fisiologia , Fenilenodiaminas/farmacologia , Fenilenodiaminas/uso terapêutico
12.
Thyroid ; 29(7): 934-945, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31084419

RESUMO

Background: Hypothyroidism, the most common endocrine disease, induces cardiac electrical remodeling that creates a substrate for ventricular arrhythmias. Recent studies report that high thyrotropin (TSH) levels are related to cardiac electrical abnormalities and increased mortality rates. The aim of the present work was to investigate the direct effects of TSH on the heart and its possible causative role in the increased incidence of arrhythmia in hypothyroidism. Methods: A new rat model of central hypothyroidism (low TSH levels) was created and characterized together with the classical propylthiouracil-induced primary hypothyroidism model (high TSH levels). Electrocardiograms were recorded in vivo, and ionic currents were recorded from isolated ventricular myocytes in vitro by the patch-clamp technique. Protein and mRNA were measured by Western blot and quantitative reverse transcription polymerase chain reaction in rat and human cardiac myocytes. Adult human action potentials were simulated in silico to incorporate the experimentally observed changes. Results: Both primary and central hypothyroidism models increased the L-type Ca2+ current (ICa-L) and decreased the ultra-rapid delayed rectifier K+ current (IKur) densities. However, only primary but not central hypothyroidism showed electrocardiographic repolarization abnormalities and increased ventricular arrhythmia incidence during caffeine/dobutamine challenge. These changes were paralleled by a decrease in the density of the transient outward K+ current (Ito) in cardiomyocytes from animals with primary but not central hypothyroidism. In vitro treatment with TSH for 24 hours enhanced isoproterenol-induced spontaneous activity in control ventricular cells and diminished Ito density in cardiomyocytes from control and central but not primary hypothyroidism animals. In human myocytes, TSH decreased the expression of KCND3 and KCNQ1, Ito, and the delayed rectifier K+ current (IKs) encoding proteins in a protein kinase A-dependent way. Transposing the changes produced by hypothyroidism and TSH to a computer model of human ventricular action potential resulted in enhanced occurrence of early afterdepolarizations and arrhythmia mostly in primary hypothyroidism, especially under ß-adrenergic stimulation. Conclusions: The results suggest that suppression of repolarizing K+ currents by TSH underlies most of the electrical remodeling observed in hypothyroidism. This work demonstrates that the activation of the TSH-receptor/protein kinase A pathway in the heart is responsible for the cardiac electrical remodeling and arrhythmia generation seen in hypothyroidism.


Assuntos
Arritmias Cardíacas/metabolismo , Remodelamento Atrial/fisiologia , Hipotireoidismo/metabolismo , Miócitos Cardíacos/metabolismo , Tireotropina/metabolismo , Potenciais de Ação , Animais , Antitireóideos/toxicidade , Arritmias Cardíacas/etiologia , Arritmias Cardíacas/fisiopatologia , Bexaroteno/toxicidade , Cálcio/metabolismo , Simulação por Computador , Modelos Animais de Doenças , Suscetibilidade a Doenças , Eletrocardiografia , Humanos , Hipotireoidismo/complicações , Hipotireoidismo/fisiopatologia , Isoproterenol/farmacologia , Canal de Potássio KCNQ1/efeitos dos fármacos , Canal de Potássio KCNQ1/genética , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Miócitos Cardíacos/efeitos dos fármacos , Técnicas de Patch-Clamp , Propiltiouracila/toxicidade , RNA Mensageiro/metabolismo , Ratos , Canais de Potássio Shal/efeitos dos fármacos , Canais de Potássio Shal/genética , Tireotropina/farmacologia
13.
Mol Pharmacol ; 74(5): 1171-9, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18684841

RESUMO

Potassium channels play an important role in electrical signaling of excitable cells such as neurons, cardiac myocytes, and vascular smooth muscle cells (VSMCs). In particular, the KCNQ (Kv7) family of voltage-activated K(+) channels functions to stabilize negative resting membrane potentials and thereby opposes electrical excitability. Of the five known members of the mammalian Kv7 family, Kv7.1 was originally recognized for its role in cardiac myocytes, where it contributes to repolarization of the cardiac action potential. Kv7.2 to Kv7.5 were first discovered in neurons, in which they play a well characterized role in neurotransmitter-stimulated action potential firing. Over the past 5 years, important new roles for Kv7 channels have been identified. Kv7 channels have been found to be expressed in VSMCs from several vascular beds where they contribute to the regulation of vascular tone. There is evidence that Kv7.5 channels in VSMCs are targeted by the hormone vasopressin to mediate its physiological vasoconstrictor actions and evidence that neuronal Kv7 channels in the baroreceptors of the aortic arch adjust the sensitivity of the mechanosensitive neurons to changes in arterial blood pressure. These newly identified physiological roles for Kv7 channels in the cardiovascular system warrant increased attention because pharmacological modulators of this family of channels are being used clinically to treat a variety of neurological disorders. This raises questions about the cardiovascular side effects associated with existing therapies, but there is also obvious potential to capitalize on the established and evolving pharmacology of these channels to develop new therapies for cardiovascular diseases.


Assuntos
Sistema Cardiovascular/metabolismo , Canal de Potássio KCNQ1/fisiologia , Potenciais de Ação/efeitos dos fármacos , Humanos , Canal de Potássio KCNQ1/efeitos dos fármacos , Canal de Potássio KCNQ1/metabolismo
14.
J Pharmacol Exp Ther ; 327(1): 168-77, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18591221

RESUMO

The large-conductance voltage-gated and calcium-dependent K(+) (BK) channels are widely distributed and play important physiological roles. Commonly used BK channel inhibitors are peptide toxins that are isolated from scorpion venoms. A high-affinity, nonpeptide, synthesized BK channel blocker with selectivity against other ion channels has not been reported. We prepared several compounds from a published patent application (Doherty et al., 2004) and identified 1-[1-hexyl-6-(methyloxy)-1H-indazol-3-yl]-2-methyl-1-propanone (HMIMP) as a potent and selective BK channel blocker. The patch-clamp technique was used for characterizing the activity of HMIMP on recombinant human BK channels (alpha subunit, alpha+beta1 and alpha+beta4 subunits). HMIMP blocked all of these channels with an IC(50) of approximately 2 nM. The inhibitory effect of HMIMP was not voltage-dependent, nor did it require opening of BK channels. HMIMP also potently blocked BK channels in freshly isolated detrusor smooth muscle cells and vagal neurons. HMIMP (10 nM) reduced the open probability significantly without affecting single BK-channel current in inside-out patches. HMIMP did not change the time constant of open states but increased the time constants of the closed states. More importantly, HMIMP was highly selective for the BK channel. HMIMP had no effect on human Na(V)1.5 (1 microM), Ca(V)3.2, L-type Ca(2+), human ether-a-go-go-related gene potassium channel, KCNQ1+minK, transient outward K(+) or voltage-dependent K(+) channels (100 nM). HMIMP did not change the action potentials of ventricular myocytes, confirming its lack of effect on cardiac ion channels. In summary, HMIMP is a highly potent and selective BK channel blocker, which can serve as an important tool in the pharmacological study of the BK channel.


Assuntos
Indazóis/farmacologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/antagonistas & inibidores , Bloqueadores dos Canais de Potássio/farmacologia , Potenciais de Ação/efeitos dos fármacos , Animais , Células CHO , Canais de Cálcio/efeitos dos fármacos , Cricetinae , Cricetulus , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/efeitos dos fármacos , Cobaias , Humanos , Indóis/farmacologia , Canal de Potássio KCNQ1/efeitos dos fármacos , Canais de Potássio Ativados por Cálcio de Condutância Alta/química , Canais de Potássio Ativados por Cálcio de Condutância Alta/fisiologia , Peptídeos/farmacologia , Coelhos , Canais de Sódio/efeitos dos fármacos
15.
Eur J Pharmacol ; 590(1-3): 317-21, 2008 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-18573250

RESUMO

Tanshinone IIA, one of the main active components from Chinese herb Danshen, is widely used to treat cardiovascular diseases including arrhythmia in Asian countries especially in China. However, the mechanisms underlying its anti-arrythmia effects are not clear. In this study we investigate the effects of tanshinone IIA on human KCNQ1/KCNE1 potassium channels (I(Ks)), human ether-a-go-go-related gene potassium channels (hERG), Kv1.5 potassium channels, inward rectifier potassium channels (I(K1)) expressed in HEK 293 cells using patch clamp technique. Tanshinone IIA potently and reversibly enhanced the amplitude of I(Ks) in a concentration dependent manner with an EC(50) of 64.5 microM, accelerated the activation rate of I(Ks) channels, decelerated their deactivation and shifted the voltage dependence of I(Ks) activation to negative direction. Isoproteronol, a stimulator of beta-adrenergic receptor, at 1 microM and sodium nitroprusside (SNP), a NO donor, at 1 mM, had no significant effects on the enhancement of I(Ks) by 30 microM tanshinone IIA. N-[2-(p-bromocinnamylamino)ethyl]-5-isoquinolinesulfonamide (H89), a selective protein kinase A inhibitor, at 0.1 microM and 1H-(1,2,4)oxadiazolo(4,3-a)quinoxalin-1-one (ODQ), a selective nitric oxide-sensitive guanylyl cyclase inhibitor, at 10 microM, also had no significant effects on the enhancement of I(Ks) by 30 microM tanshinone IIA. Tanshinone IIA did not affect expressed hERG channels, Kv1.5 channels and I(K1) channels. These results indicate that tanshinone IIA directly and specifically activate human cardiac KCNQ1/KCNE1 potassium channels (I(Ks)) in HEK 293 cell through affecting the channels' kinetics.


Assuntos
Medicamentos de Ervas Chinesas/farmacologia , Coração/efeitos dos fármacos , Canal de Potássio KCNQ1/efeitos dos fármacos , Fenantrenos/farmacologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/efeitos dos fármacos , Abietanos , Células Cultivadas , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Relação Dose-Resposta a Droga , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/efeitos dos fármacos , Guanilato Ciclase/antagonistas & inibidores , Humanos , Canal de Potássio Kv1.5/efeitos dos fármacos , Doadores de Óxido Nítrico/farmacologia , Oxidiazóis/farmacologia , Quinoxalinas/farmacologia
16.
Biochim Biophys Acta ; 1758(10): 1641-52, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16836976

RESUMO

Exposure of cardiac myocytes to hyposmotic solution stimulates slowly-activating delayed-rectifying K(+) current (I(Ks)) via unknown mechanisms. In the present study, I(Ks) was measured in guinea-pig ventricular myocytes that were pretreated with modulators of cell signaling processes, and then exposed to hyposmotic solution. Pretreatment with compounds that (i) inhibit serine/threonine kinase activity (10-100 microM H89; 200 microM H8; 50 microM H7; 1 microM bisindolylmaleimide I; 10 microM LY294002; 50 microM PD98059), (ii) stimulate serine/threonine kinase activity (1-5 microM forskolin; 0.1 microM phorbol-12-myristate-13-acetate; 10 microM acetylcholine; 0.1 microM angiotensin II; 20 microM ATP), (iii) suppress G-protein activation (10 mM GDPbetaS), or (iv) disrupt the cytoskeleton (10 microM cytochalasin D), had little effect on the stimulation of I(Ks) by hyposmotic solution. In marked contrast, pretreatment with tyrosine kinase inhibitor tyrphostin A25 (20 microM) strongly attenuated both the hyposmotic stimulation of I(Ks) in myocytes and the hyposmotic stimulation of current in BHK cells co-expressing Ks channel subunits KCNQ1 and KCNE1. Since attenuation of hyposmotic stimulation was not observed in myocytes and cells pretreated with inactive tyrphostin A1, we conclude that TK has an important role in the response of cardiac Ks channels to hyposmotic solution.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Proteínas de Ligação ao GTP/fisiologia , Miócitos Cardíacos/efeitos dos fármacos , Pressão Osmótica , Canais de Potássio/efeitos dos fármacos , Proteína Quinase C/fisiologia , Proteínas Tirosina Quinases/fisiologia , 1-(5-Isoquinolinasulfonil)-2-Metilpiperazina/farmacologia , Animais , Linhagem Celular , Cromanos/farmacologia , Colforsina/farmacologia , Cricetinae , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Citocalasina D/farmacologia , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/fisiologia , Proteínas de Ligação ao GTP/antagonistas & inibidores , Cobaias , Indóis/farmacologia , Isoquinolinas/farmacologia , Canal de Potássio KCNQ1/efeitos dos fármacos , Canal de Potássio KCNQ1/fisiologia , Maleimidas/farmacologia , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Canais de Potássio/fisiologia , Proteína Quinase C/antagonistas & inibidores , Proteínas Tirosina Quinases/antagonistas & inibidores , Sulfonamidas/farmacologia , Acetato de Tetradecanoilforbol/farmacologia , Tirfostinas/farmacologia
17.
Heart Rhythm ; 4(5): 611-8, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17467630

RESUMO

BACKGROUND: We recently reported that an S140G mutation in human KCNQ1, an alpha subunit of potassium channels, was involved in the pathogenesis of familial atrial fibrillation (AF), but it is not clear whether the mutation is associated with other cardiac arrhythmias. OBJECTIVE: The purpose of this study was to further explore the association of the KCNQ1 S140G mutation with cardiac arrhythmias. METHODS: We produced a transgenic mouse model with myocardium-specific expression of the human KCNQ1 S140G mutation under the control of an alpha-cardiac myosin heavy chain promoter by standard transgenic procedure and evaluated the relationship between the KCNQ1 mutation and its phenotypes in a human family. RESULTS: Four lines of transgenic mice were established with a high level of human KCNQ1 S140G expression in the heart. Frequent episodes of first-, second-, advanced-, or third-degree atrioventricular block (AVB) occurred in at least 65% of transgenic descendants from the four lines. However, none of the five wild-type transgenic lines presented with AVBs. HMR1556, a KCNQ1-specific blocker, can terminate the AVBs. With the exception of at most three AF individuals, at least 13 AF patients were found to show obviously slow ventricular response, which may be one manifestation of AVBs. Interestingly, AF was not detected in these transgenic mice. CONCLUSION: The results suggest that human KCNQ1 S140G is also likely to be a causative mutation responsible for AVBs. The transgenic mouse model is a potential tool to explore mechanisms of AVBs.


Assuntos
Bloqueio Cardíaco/genética , Canal de Potássio KCNQ1/genética , Mutação , Animais , Fibrilação Atrial/genética , Fibrilação Atrial/fisiopatologia , Nó Atrioventricular/efeitos dos fármacos , Nó Atrioventricular/fisiopatologia , China , Cromanos/farmacologia , Eletrocardiografia , Feminino , Predisposição Genética para Doença , Glicina , Bloqueio Cardíaco/fisiopatologia , Frequência Cardíaca/efeitos dos fármacos , Frequência Cardíaca/genética , Ventrículos do Coração/efeitos dos fármacos , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/genética , Canal de Potássio KCNQ1/efeitos dos fármacos , Masculino , Camundongos , Camundongos Transgênicos , Linhagem , Fenótipo , Projetos de Pesquisa , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Serina , Índice de Gravidade de Doença , Sulfonamidas/farmacologia
18.
Assay Drug Dev Technol ; 5(5): 617-27, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17939752

RESUMO

A cardiac safety concern for QT prolongation and potential for pro-arrhythmia exists due to inhibition of the cardiac slowly activating delayed rectifier potassium current, I(Ks). Selective inhibitors of I Ks have been shown to prolong the QT interval in animal models. On the other hand, I Ks has been considered as a target for anti-arrhythmic therapy due to certain biophysical and pharmacological properties and its expression pattern in the heart. Consequently, we have developed a method utilizing a human embryonic kidney (HEK)-293 cell line expressing KCNQ1/KCNE1 (genes that encode for the I Ks channel) as a model for screening of new compounds for I Ks activity. This study was designed (1) to establish and optimize the experimental conditions for measurement of I Ks using PatchXpress() 7000A (Molecular Devices Corporation, Sunnyvale, CA) and (2) to test the effects of I Ks inhibitors and compare the 50% inhibitory concentration (IC50) values determined with PatchXpress versus conventional patch clamp in order to validate the PatchXpress approach for higher-throughput I Ks screening. Biophysical properties of HEK/I Ks recorded with PatchXpress were similar to those recorded with conventional patch-clamp and reported in the literature. The IC50 values for I Ks block determined with PatchXpress correlated well with conventional patch-clamp values from HEK-293 cells as well as from native cardiac myocytes for the majority of compounds tested. Electrophysiological recording of I Ks expressed in HEK-293 cells with the PatchXpress is of acceptable quality for screening purposes. This approach can be utilized for functional prescreening of development compounds for I Ks inhibition either for optimizing lead anti-arrhythmic or other therapeutic candidates or to exclude compounds with the potential to prolong QT.


Assuntos
Fármacos Cardiovasculares/farmacologia , Avaliação Pré-Clínica de Medicamentos/instrumentação , Canal de Potássio KCNQ1/efeitos dos fármacos , Técnicas de Patch-Clamp/instrumentação , Canais de Potássio de Abertura Dependente da Tensão da Membrana/efeitos dos fármacos , Animais , Células CHO , Linhagem Celular , Química Farmacêutica , Cricetinae , Cricetulus , Interpretação Estatística de Dados , Eletrofisiologia , Canais de Potássio Éter-A-Go-Go/efeitos dos fármacos , Cobaias , Humanos , Técnicas In Vitro , Miócitos Cardíacos/efeitos dos fármacos
19.
Neuropharmacology ; 51(6): 1068-77, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16904708

RESUMO

The family of Kv7 (KCNQ) potassium channels consists of five members. Kv7.2 and 3 are the primary molecular correlates of the M-current, but also Kv7.4 and Kv7.5 display M-current characteristics. M-channel modulators include blockers (e.g., linopirdine) for cognition enhancement and openers (e.g., retigabine) for treatment of epilepsy and neuropathic pain. We investigated the effect of a Bristol-Myers Squibb compound (S)-N-[1-(3-morpholin-4-yl-phenyl)-ethyl]-3-phenyl-acrylamide [(S)-1] on cloned human Kv7.1-5 potassium channels expressed in Xenopus laevis oocytes. Using two-electrode voltage-clamp recordings we found that (S)-1 blocks Kv7.1 and Kv7.1/KCNE1 currents. In contrast, (S)-1 produced a hyperpolarizing shift of the activation curve for Kv7.2, Kv7.2/Kv7.3, Kv7.4 and Kv7.5. Further, the compound enhanced the maximal current amplitude at all potentials for Kv7.4 and Kv7.5 whereas the combined activation/block of Kv7.2 and Kv7.2/3 was strongly voltage-dependent. The tryptophan residue 242 in S5, known to be crucial for the effect of retigabine, was also shown to be critical for the enhancing effect of (S)-1 and BMS204352. Furthermore, no additive effect on Kv7.4 current amplitude was observed when both retigabine and (S)-1 or BMS204352 were applied simultaneously. In conclusion, (S)-1 differentially affects the Kv7 channel subtypes and is dependent on a single tryptophan for the current enhancing effect in Kv7.4.


Assuntos
Acrilamidas/farmacologia , Canais de Potássio KCNQ/efeitos dos fármacos , Morfolinas/farmacologia , Neurônios/metabolismo , Acrilamidas/metabolismo , Algoritmos , Animais , Sítios de Ligação/efeitos dos fármacos , DNA Complementar/biossíntese , DNA Complementar/genética , Eletrofisiologia , Humanos , Canais de Potássio KCNQ/genética , Canal de Potássio KCNQ1/efeitos dos fármacos , Canal de Potássio KCNQ2/efeitos dos fármacos , Cinética , Morfolinas/metabolismo , Neurônios/efeitos dos fármacos , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Técnicas de Patch-Clamp , Mutação Puntual/efeitos dos fármacos , Triptofano/efeitos dos fármacos , Triptofano/metabolismo , Xenopus laevis
20.
Naunyn Schmiedebergs Arch Pharmacol ; 389(10): 1133-7, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27530870

RESUMO

Loperamide is a µ-opioid receptor agonist commonly used to treat diarrhea and often available as an over-the-counter medication. Recently, numerous reports of QRS widening accompanied by dramatic QT interval prolongation, torsades de pointe arrhythmia, and death have been reported in opioid abusers consuming large amounts of the drug to produce euphoria or prevent opiate withdrawal. The present study was undertaken to determine the mechanisms of this cardiotoxicity. Using whole-cell patch clamp electrophysiology, we tested loperamide on the cloned human cardiac sodium channel (Nav1.5) and the two main repolarizing cardiac K(+) channels cloned from the human heart: KvLQT1/minK and the human ether-a-go-go-related gene (hERG) channel. Loperamide inhibited Nav1.5 with IC50 values of 297 and 239 nM at holding potentials of -90 and -70 mV, respectively. Loperamide was weakly active on KvLQT1/minK producing 17 and 65 % inhibition at concentrations of 1 and 10 µM, respectively. Conversely, loperamide was found to be a very high affinity inhibitor of the hERG channel with an IC50 value of 89 nM at room temperature and 33 nM when measured at physiological temperature. The QRS and QT interval prolongation and the attending arrhythmias, produced by loperamide, derive from high affinity inhibition of Nav1.5 and especially hERG. Since the drug has been widely available and safely used as directed for many years, we believe that the potent inhibition loperamide possesses for cardiac ion channels has only been uncovered because of the excessive misuse of the drug as a consequence of the recent opioid abuse epidemic.


Assuntos
Antidiarreicos/toxicidade , Síndrome do QT Longo/induzido quimicamente , Loperamida/toxicidade , Miócitos Cardíacos/efeitos dos fármacos , Canal de Sódio Disparado por Voltagem NAV1.5/efeitos dos fármacos , Transtornos Relacionados ao Uso de Opioides/complicações , Torsades de Pointes/etiologia , Bloqueadores do Canal de Sódio Disparado por Voltagem/toxicidade , Potenciais de Ação , Cardiotoxicidade , Relação Dose-Resposta a Droga , Canal de Potássio ERG1/efeitos dos fármacos , Canal de Potássio ERG1/genética , Canal de Potássio ERG1/metabolismo , Células HEK293 , Humanos , Canal de Potássio KCNQ1/efeitos dos fármacos , Canal de Potássio KCNQ1/genética , Canal de Potássio KCNQ1/metabolismo , Síndrome do QT Longo/metabolismo , Síndrome do QT Longo/fisiopatologia , Miócitos Cardíacos/metabolismo , Canal de Sódio Disparado por Voltagem NAV1.5/genética , Canal de Sódio Disparado por Voltagem NAV1.5/metabolismo , Transtornos Relacionados ao Uso de Opioides/fisiopatologia , Técnicas de Patch-Clamp , Fatores de Risco , Fatores de Tempo , Torsades de Pointes/metabolismo , Torsades de Pointes/fisiopatologia , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA