Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Exp Clin Endocrinol Diabetes ; 129(12): 899-907, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32559789

RESUMO

BACKGROUND: Human glucagon-like peptide-1 analogue, Liraglutide, has shown cardioprotective effects in animal and clinical studies of type 2 diabetes mellitus. This study was conducted to assess the effect of Liraglutide on diabetes-induced myocardial electrical remodeling. MATERIALS AND METHODS: A rat model of type 2 diabetes mellitus was induced by high-fat diet and low dose Streptozotocin (35 mg/kg). Diabetic rats were randomized into 4 subgroups (n=6-7): diabetic-untreated, diabetics treated with Liraglutide, diabetics treated with Ramipril, and diabetics treated with Metformin in addition to a control group. Changes in serum glucose, insulin, lipid profile and revised quantitative insulin sensitivity check index (QUICKI index) were assessed. QT and QTc intervals were measured and the degree of cardiac interstitial and perivascular fibrosis was examined. The expression of myocardial Ito channel α subunits, gap junction protein; Kv 4.2/4.3 and connexin 43 (Cx43) respectively, were assessed by western blotting and immunohistochemistry. RESULTS: Similar to Ramipril, both Liraglutide and Metformin effectively inhibited the diabetes-induced myocardial hypertrophy and fibrosis. However, Liraglutide treatment significantly improved Kv 4.2/4.3 and Cx43 expression/distribution and prevented diabetes-related QTc interval prolongation. CONCLUSIONS: We have shown that pathological alterations in myocardial Cx43 expression and distribution, in addition to reduced Ito channel expression, may underlie the QTc interval prolongation in high-fat diet/STZ rat model of type 2 diabetes mellitus. The beneficial effects of Liraglutide, as those of Ramipril, on cardiac electrophysiology could be at least attributed to its direct ability to normalize expression and distribution of Cx43 and Ito channels in the diabetic rat heart.


Assuntos
Conexina 43/efeitos dos fármacos , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Tipo 2/tratamento farmacológico , Cardiomiopatias Diabéticas/tratamento farmacológico , Hipoglicemiantes/farmacologia , Liraglutida/farmacologia , Miocárdio/metabolismo , Canais de Potássio Shal/efeitos dos fármacos , Remodelação Ventricular/efeitos dos fármacos , Animais , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Cardiomiopatias Diabéticas/metabolismo , Distribuição Aleatória , Ratos , Canais de Potássio Shal/metabolismo
2.
FASEB J ; 34(1): 807-821, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31914636

RESUMO

Kv4 pore-forming subunits co-assemble with ß-subunits including KChIP2 and DPP6 and the resultant complexes conduct cardiac transient outward K+ current (Ito). Compound NS5806 has been shown to potentate Ito in canine cardiomyocytes; however, its effects on Ito in other species yet to be determined. We found that NS5806 inhibited native Ito in a concentration-dependent manner (0.1~30 µM) in both mouse ventricular cardiomyocytes and human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs), but potentiated Ito in the canine cardiomyocytes. In HEK293 cells co-transfected with cloned Kv4.3 (or Kv4.2) and ß-subunit KChIP2, NS5806 significantly increased the peak current amplitude and slowed the inactivation. In contrast, NS5806 suppressed the current and accelerated inactivation of the channels when cells were co-transfected with Kv4.3 (or Kv4.2), KChIP2 and another ß-subunit, DPP6-L (long isoform). Western blot analysis showed that DPP6-L was dominantly expressed in both mouse ventricular myocardium and hiPSC-CMs, while it was almost undetectable in canine ventricular myocardium. In addition, low level of DPP6-S expression was found in canine heart, whereas levels of KChIP2 expression were comparable among all three species. siRNA knockdown of DPP6 antagonized the Ito inhibition by NS5806 in hiPSC-CMs. Molecular docking simulation suggested that DPP6-L may associate with KChIP2 subunits. Mutations of putative KChIP2-interacting residues of DPP6-L reversed the inhibitory effect of NS5806 into potentiation of the current. We conclude that a pharmacological modulator can elicit opposite regulatory effects on Kv4 channel complex among different species, depending on the presence of distinct ß-subunits. These findings provide novel insight into the molecular design and regulation of cardiac Ito. Since Ito is a potential therapeutic target for treatment of multiple cardiovascular diseases, our data will facilitate the development of new therapeutic Ito modulators.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Compostos de Fenilureia/farmacologia , Canais de Potássio Shal/efeitos dos fármacos , Tetrazóis/farmacologia , Potenciais de Ação/fisiologia , Animais , Cricetulus/metabolismo , Células HEK293 , Humanos , Masculino , Camundongos Endogâmicos C57BL , Simulação de Acoplamento Molecular/métodos , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo
3.
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
4.
Psychopharmacology (Berl) ; 232(11): 1995-2006, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25510858

RESUMO

RATIONALE: Chronic alcohol-induced cognitive impairments and maladaptive plasticity of glutamatergic synapses are well-documented. However, it is unknown if prolonged alcohol exposure affects dendritic signaling that may underlie hippocampal dysfunction in alcoholics. Back-propagation of action potentials (bAPs) into apical dendrites of hippocampal neurons provides distance-dependent signals that modulate dendritic and synaptic plasticity. The amplitude of bAPs decreases with distance from the soma that is thought to reflect an increase in the density of Kv4.2 channels toward distal dendrites. OBJECTIVE: The aim of this study was to quantify changes in hippocampal Kv4.2 channel function and expression using electrophysiology, Ca(2+) imaging, and western blot analyses in a well-characterized in vitro model of chronic alcohol exposure. RESULTS: Chronic alcohol exposure significantly decreased expression of Kv4.2 channels and KChIP3 in hippocampus. This reduction was associated with an attenuation of macroscopic A-type K(+) currents in CA1 neurons. Chronic alcohol exposure increased bAP-evoked Ca(2+) transients in the distal apical dendrites of CA1 pyramidal neurons. The enhanced bAP-evoked Ca(2+) transients induced by chronic alcohol exposure were not related to synaptic targeting of N-methyl-D-aspartate (NMDA) receptors or morphological adaptations in apical dendritic arborization. CONCLUSIONS: These data suggest that chronic alcohol-induced decreases in Kv4.2 channel function possibly mediated by a downregulation of KChIP3 drive the elevated bAP-associated Ca(2+) transients in distal apical dendrites. Alcohol-induced enhancement of bAPs may affect metaplasticity and signal integration in apical dendrites of hippocampal neurons leading to alterations in hippocampal function.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Alcoolismo/fisiopatologia , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Dendritos/efeitos dos fármacos , Dendritos/fisiologia , Hipocampo/efeitos dos fármacos , Hipocampo/fisiopatologia , Proteínas Interatuantes com Canais de Kv/efeitos dos fármacos , Proteínas Interatuantes com Canais de Kv/fisiologia , Plasticidade Neuronal/efeitos dos fármacos , Plasticidade Neuronal/fisiologia , Canais de Potássio Shal/efeitos dos fármacos , Canais de Potássio Shal/fisiologia , Animais , Cálcio/metabolismo , Feminino , Células Piramidais/metabolismo , Ratos , Ratos Sprague-Dawley , Sinapses/metabolismo
5.
J Neurosci ; 34(28): 9182-9, 2014 Jul 09.
Artigo em Inglês | MEDLINE | ID: mdl-25009251

RESUMO

SNX-482, a peptide toxin isolated from tarantula venom, has become widely used as an inhibitor of Cav2.3 voltage-gated calcium channels. Unexpectedly, we found that SNX-482 dramatically reduced the A-type potassium current in acutely dissociated dopamine neurons from mouse substantia nigra pars compacta. The inhibition persisted when calcium was replaced by cobalt, showing that it was not secondary to a reduction of calcium influx. Currents from cloned Kv4.3 channels expressed in HEK-293 cells were inhibited by SNX-482 with an IC50 of <3 nM, revealing substantially greater potency than for SNX-482 inhibition of Cav2.3 channels (IC50 20-60 nM). At sub-saturating concentrations, SNX-482 produced a depolarizing shift in the voltage dependence of activation of Kv4.3 channels and slowed activation kinetics. Similar effects were seen on gating of cloned Kv4.2 channels, but the inhibition was less pronounced and required higher toxin concentrations. These results reveal SNX-482 as the most potent inhibitor of Kv4.3 channels yet identified. Because of the effects on both Kv4.3 and Kv4.2 channels, caution is needed when interpreting the effects of SNX-482 on cells and circuits where these channels are present.


Assuntos
Neurônios Dopaminérgicos/efeitos dos fármacos , Ativação do Canal Iônico/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio Shal/efeitos dos fármacos , Venenos de Aranha/farmacologia , Animais , Células Cultivadas , Neurônios Dopaminérgicos/fisiologia , Feminino , Células HEK293 , Humanos , Concentração Inibidora 50 , Ativação do Canal Iônico/fisiologia , Masculino , Potenciais da Membrana/fisiologia , Camundongos , Técnicas de Patch-Clamp , Potássio/metabolismo , Canais de Potássio Shal/fisiologia
6.
Circ Res ; 112(10): 1310-22, 2013 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-23532596

RESUMO

RATIONALE: A chromosomal haplotype producing cardiac overexpression of dipeptidyl peptidase-like protein-6 (DPP6) causes familial idiopathic ventricular fibrillation. The molecular basis of transient outward current (I(to)) in Purkinje fibers (PFs) is poorly understood. We hypothesized that DPP6 contributes to PF I(to) and that its overexpression might specifically alter PF I(to) properties and repolarization. OBJECTIVE: To assess the potential role of DPP6 in PF I(to). METHODS AND RESULTS: Clinical data in 5 idiopathic ventricular fibrillation patients suggested arrhythmia origin in the PF-conducting system. PF and ventricular muscle I(to) had similar density, but PF I(to) differed from ventricular muscle in having tetraethylammonium sensitivity and slower recovery. DPP6 overexpression significantly increased, whereas DPP6 knockdown reduced, I(to) density and tetraethylammonium sensitivity in canine PF but not in ventricular muscle cells. The K(+)-channel interacting ß-subunit K(+)-channel interacting protein type-2, essential for normal expression of I(to) in ventricular muscle, was weakly expressed in human PFs, whereas DPP6 and frequenin (neuronal calcium sensor-1) were enriched. Heterologous expression of Kv4.3 in Chinese hamster ovary cells produced small I(to); I(to) amplitude was greatly enhanced by coexpression with K(+)-channel interacting protein type-2 or DPP6. Coexpression of DPP6 with Kv4.3 and K(+)-channel interacting protein type-2 failed to alter I(to) compared with Kv4.3/K(+)-channel interacting protein type-2 alone, but DPP6 expression with Kv4.3 and neuronal calcium sensor-1 (to mimic PF I(to) composition) greatly enhanced I(to) compared with Kv4.3/neuronal calcium sensor-1 and recapitulated characteristic PF kinetic/pharmacological properties. A mathematical model of cardiac PF action potentials showed that I(to) enhancement can greatly accelerate PF repolarization. CONCLUSIONS: These results point to a previously unknown central role of DPP6 in PF I(to), with DPP6 gain of function selectively enhancing PF current, and suggest that a DPP6-mediated PF early-repolarization syndrome might be a novel molecular paradigm for some forms of idiopathic ventricular fibrillation.


Assuntos
Dipeptidil Peptidases e Tripeptidil Peptidases/fisiologia , Proteínas Interatuantes com Canais de Kv/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Canais de Potássio/fisiologia , Ramos Subendocárdicos/fisiologia , Canais de Potássio Shal/fisiologia , Fibrilação Ventricular/fisiopatologia , Adulto , Animais , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Dipeptidil Peptidases e Tripeptidil Peptidases/genética , Modelos Animais de Doenças , Cães , Feminino , Técnicas de Silenciamento de Genes , Ventrículos do Coração/patologia , Ventrículos do Coração/fisiopatologia , Humanos , Técnicas In Vitro , Proteínas Interatuantes com Canais de Kv/efeitos dos fármacos , Proteínas Interatuantes com Canais de Kv/genética , Masculino , Pessoa de Meia-Idade , Modelos Teóricos , Proteínas do Tecido Nervoso/genética , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/genética , Ramos Subendocárdicos/patologia , Canais de Potássio Shal/efeitos dos fármacos , Canais de Potássio Shal/genética , Tetraetilamônio/farmacologia , Transfecção
7.
Gut ; 62(10): 1466-74, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22833396

RESUMO

OBJECTIVE: Increased faecal butyrate levels have been reported in irritable bowel syndrome. Rectal instillation of sodium butyrate (NaB) increases visceral sensitivity in rats by an unknown mechanism. We seek to examine the signal transduction pathways responsible for the enhanced neuronal excitability in the dorsal root ganglion (DRG) following NaB enemas and demonstrate that this is responsible for the colonic hypersensitivity reported in this animal model. DESIGN: Colorectal distention (CRD) studies were performed in rats treated with NaB rectal instillation with/without intrathecal or intravenous administration of mitogen-activated protein (MAP) kinase kinase inhibitor U0126. Western blot analysis and immunocytochemistry studies elucidated intracellular signalling pathways that modulate IA. Patch-clamp recordings were performed on isolated DRG neurons treated with NaB, with/without U0126. RESULTS: Visceromotor responses (VMR) were markedly enhanced in NaB-treated rats. Western blot analysis of DRG neurons from NaB-treated rats showed a 2.2-fold increase in phosphorylated ERK1/2 (pEKR1/2) and 1.9-fold increase in phosphorylated voltage-gated potassium channel subunit 4.2 (pKv4.2). Intrathecal or intravenous administration of U0126 reduced VMR to CRD in NaB-treated rats and prevented increases in pERK1/2 and pKv4.2. Patch-clamp recordings of isolated DRG neurons showed that NaB caused a reduction in IA to 48.9%±1.4% of control and an increase in neuronal excitability, accompanied by a twofold increase in pERK1/2 and pKv4.2. Concurrent U0126 administration prevented these changes. CONCLUSIONS: Visceral hypersensitivity induced by colonic NaB treatment is mediated by activation of the MAP kinase-ERK1/2 pathway, which phosphorylates Kv4.2. This results in a reduction in IA and an enhancement of DRG neuronal excitability.


Assuntos
Butiratos/toxicidade , Gânglios Espinais/efeitos dos fármacos , Síndrome do Intestino Irritável/induzido quimicamente , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Animais , Butadienos/farmacologia , Células Cultivadas , Colo/efeitos dos fármacos , Colo/inervação , Dilatação , Enema , Ativação Enzimática/efeitos dos fármacos , Gânglios Espinais/enzimologia , Síndrome do Intestino Irritável/enzimologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Masculino , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Nitrilas/farmacologia , Técnicas de Patch-Clamp , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Ratos , Ratos Sprague-Dawley , Canais de Potássio Shal/efeitos dos fármacos , Canais de Potássio Shal/metabolismo , Dor Visceral/induzido quimicamente , Dor Visceral/enzimologia
8.
Neuropharmacology ; 63(8): 1389-403, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22964468

RESUMO

We examined the effects of the sulfonylurea compound NS5806 on neuronal A-type channel function. Using whole-cell patch-clamp we studied the effects of NS5806 on the somatodendritic A-type current (I(SA)) in cultured hippocampal neurons and the currents mediated by Kv4.2 channels coexpressed with different auxiliary ß-subunits, including both Kv channel interacting proteins (KChIPs) and dipeptidyl aminopeptidase-related proteins (DPPs), in HEK 293 cells. The amplitude of the I(SA) component in hippocampal neurons was reduced in the presence of 20 µM NS5806. I(SA) decay kinetics were slowed and the recovery kinetics accelerated, but the voltage dependence of steady-state inactivation was shifted to more negative potentials by NS5806. The peak amplitudes of currents mediated by ternary Kv4.2 channel complexes, associated with DPP6-S (short splice-variant) and either KChIP2, KChIP3 or KChIP4, were potentiated and their macroscopic inactivation slowed by NS5806, whereas the currents mediated by binary Kv4.2 channels, associated only with DPP6-S, were suppressed, and the NS5806-mediated slowing of macroscopic inactivation was less pronounced. Neither potentiation nor suppression and no effect on current decay kinetics in the presence of NS5806 were observed for Kv4.2 channels associated with KChIP3 and the N-type inactivation-conferring DPP6a splice-variant. For all recombinant channel complexes, NS5806 slowed the recovery from inactivation and shifted the voltage dependence of steady-state inactivation to more negative potentials. Our results demonstrate the activity of NS5806 on native I(SA) and possible molecular correlates in the form of recombinant Kv4.2 channels complexed with different KChIPs and DPPs, and they shed some light on the mechanism of NS5806 action.


Assuntos
Hipocampo/metabolismo , Compostos de Fenilureia/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio Shal/efeitos dos fármacos , Tetrazóis/farmacologia , Animais , Células Cultivadas , Interpretação Estatística de Dados , Dipeptidil Peptidases e Tripeptidil Peptidases/efeitos dos fármacos , Dipeptidil Peptidases e Tripeptidil Peptidases/metabolismo , Dipeptidil Peptidases e Tripeptidil Peptidases/fisiologia , Células HEK293 , Hipocampo/efeitos dos fármacos , Humanos , Técnicas In Vitro , Cinética , Proteínas Interatuantes com Canais de Kv/fisiologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Técnicas de Patch-Clamp , Ratos , Ratos Wistar
9.
Circ Res ; 108(5): 537-43, 2011 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-21252158

RESUMO

RATIONALE: The fast transient outward K(+) current (I(to,f)) plays a critical role in early repolarization of the heart. I(to,f) is consistently downregulated in cardiac disease. Despite its importance, the regulation of I(to,f) in disease remains poorly understood. OBJECTIVE: Because the transcription factor nuclear factor (NF)-κB is activated in cardiac hypertrophy and disease, we studied the role of NF-κB in mediating I(to,f) reductions induced by hypertrophy. METHODS AND RESULTS: Culturing neonatal rat ventricular myocytes in the presence of phenylephrine (PE) plus propranolol (Pro), to selectively activate α(1)-adrenergic receptors, caused reductions in I(to,f), as well as KChIP2 and Kv4.3 expression, while increasing Kv4.2 expression. Inhibition of NF-κB, via overexpression of a phosphorylation-deficient mutant of IκBα (IκBαSA) prevented PE/Pro-induced reductions in I(to,f) and KChIP2 mRNA, without affecting Kv4.2 or Kv4.3 expression, suggesting NF-κB mediates the I(to,f) reductions by repressing KChIP2. Indeed, overexpression of the NF-κB activator IκB kinase-ß also decreased KChIP2 expression and I(to,f) (despite increasing Kv4.2), whereas IκBαSA overexpression elevated KChIP2 and decreased Kv4.2 levels. In addition, the classic NF-κB activator tumor necrosis factor α also induced NF-κB-dependent reductions of KChIP2 and I(to,f). Finally, inhibition of calcineurin did not prevent PE/Pro-induced reductions in KChIP2. CONCLUSIONS: NF-κB regulates KChIP2 and Kv4.2 expression. The reductions in I(to,f) observed following α-adrenergic receptor stimulation or tumor necrosis factor α application require NF-κB-dependent decreases in KChIP2 expression.


Assuntos
Regulação para Baixo/fisiologia , Proteínas Interatuantes com Canais de Kv/metabolismo , Miócitos Cardíacos/metabolismo , NF-kappa B/fisiologia , Canais de Potássio/metabolismo , Animais , Células Cultivadas , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Hipertrofia Ventricular Esquerda/metabolismo , Hipertrofia Ventricular Esquerda/patologia , Miócitos Cardíacos/patologia , Fenilefrina/farmacologia , Propranolol/farmacologia , Ratos , Ratos Sprague-Dawley , Canais de Potássio Shal/efeitos dos fármacos , Canais de Potássio Shal/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
10.
Br J Pharmacol ; 160(8): 2028-44, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20649599

RESUMO

BACKGROUND AND PURPOSE: The compound NS5806 increases the transient outward current (I(to)) in canine ventricular cardiomyocytes and slows current decay. In human and canine ventricle, I(to) is thought to be mediated by K(V)4.3 and various ancillary proteins, yet, the exact subunit composition of I(to) channels is still debated. Here we characterize the effect of NS5806 on heterologously expressed putative I(to) channel subunits and other potassium channels. EXPERIMENTAL APPROACH: Cloned K(V)4 channels were co-expressed with KChIP2, DPP6, DPP10, KCNE2, KCNE3 and K(V)1.4 in Xenopus laevis oocytes or CHO-K1 cells. KEY RESULTS: NS5806 increased K(V)4.3/KChIP2 peak current amplitudes with an EC(50) of 5.3 +/- 1.5microM and significantly slowed current decay. KCNE2, KCNE3, DPP6 and DPP10 modulated K(V)4.3 currents and the response to NS5806, but current decay was slowed only in complexes containing KChIP2. The effect of NS5806 on K(V)4.2 was similar to that on K(V)4.3, and current decay was only slowed in presence of KChIP2. However, for K(V)4.1, the slowing of current decay by NS5806 was independent of KChIP2. K(V)1.4 was strongly inhibited by 10 microM NS5806 and K(V)1.5 was inhibited to a smaller extent. Effects of NS5806 on kinetics of currents generated by K(V)4.3/KChIP2/DPP6 with K(V)1.4 in oocytes could reproduce those on cardiac I(to) in canine ventricular myocytes. K(V)7.1, K(V)11.1 and K(ir)2 currents were unaffected by NS5806. CONCLUSION AND IMPLICATIONS: NS5806 modulated K(V)4 channel gating depending on the presence of KChIP2, suggesting that NS5806 can potentially be used to address the molecular composition as well as the physiological role of cardiac I(to).


Assuntos
Ativação do Canal Iônico/efeitos dos fármacos , Proteínas Interatuantes com Canais de Kv/metabolismo , Compostos de Fenilureia/farmacologia , Potássio/metabolismo , Canais de Potássio Shal/efeitos dos fármacos , Tetrazóis/farmacologia , Animais , Células CHO , Clonagem Molecular , Cricetinae , Cricetulus , Dipeptidil Peptidases e Tripeptidil Peptidases/metabolismo , Humanos , Cinética , Proteínas Interatuantes com Canais de Kv/genética , Canal de Potássio Kv1.4/metabolismo , Potenciais da Membrana , Proteínas do Tecido Nervoso/metabolismo , Canais de Potássio/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Canais de Potássio Shal/genética , Canais de Potássio Shal/metabolismo , Transfecção , Xenopus laevis
11.
Naunyn Schmiedebergs Arch Pharmacol ; 382(1): 63-71, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20499050

RESUMO

It was recently suggested that the antiarrhythmic effect of propranolol, a ss-adrenoceptor antagonist, on ischemic myocardium includes restoration of I(K1) current and Cx43 conductance; however, little is known whether effects on the transient outward current I(to) contribute. A model of myocardial infarction (MI) by ligating the left anterior descending coronary artery was established. Propranolol was given 1 h or daily for 3 months, whole-cell patch-clamp techniques were used to measure I(to). Kv4.2 and PKA levels were analyzed by Western blot and cAMP level was determined by radioimmunoassay. The results showed that propranolol decreased the incidence of arrhythmias induced by acute ischemia and mortality in 3 month MI rats. Propranolol restored the diminished I(to) density and Kv4.2 protein in MI hearts. In addition, neonatal cardiomyocyte pretreatment with propranolol or administrated after hypoxia can resume I(to) density. cAMP/PKA was enhanced in acute MI, the reason of decreased Kv4.2 expression. Treatment with propranolol prevented the increased cAMP/PKA in 1 h MI, whereas propranolol had little effect on decreased cAMP/PKA in 3 months MI. This study demonstrated that both short- and long-term propranolol administrations protect cardiomyocytes against arrhythmias and mortality caused by cardiac ischemia; the involvement of cAMP/PKA signal pathway in the regulation of propranolol on I(to) acted differently along with the ischemic progression.


Assuntos
Antagonistas Adrenérgicos beta/farmacologia , Propranolol/farmacologia , Canais de Potássio Shal/efeitos dos fármacos , Antagonistas Adrenérgicos beta/administração & dosagem , Animais , Western Blotting , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Modelos Animais de Doenças , Esquema de Medicação , Masculino , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/fisiopatologia , Isquemia Miocárdica/tratamento farmacológico , Isquemia Miocárdica/fisiopatologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Técnicas de Patch-Clamp , Propranolol/administração & dosagem , Ratos , Ratos Wistar , Canais de Potássio Shal/metabolismo , Fatores de Tempo
12.
Pflugers Arch ; 460(1): 87-97, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20354865

RESUMO

DPP10 is a transmembrane glycosylated protein belonging to the family of dipeptidyl aminopeptidase-like proteins (DPPLs). DPPLs are auxiliary subunits involved in the regulation of voltage-gated Kv4 channels, key determinants of cardiac and neuronal excitability. Although it is known that DPPLs are needed to generate native-like currents in heterologous expression systems, the molecular basis of this involvement are still poorly defined. In this study, we investigated the functional relevance of DPP10 glycosylation in modulating Kv4.3 channel activities. Using transfected Chinese hamster ovary (CHO) cells to reconstitute Kv4 complex, we show that the pharmacological inhibition of DPP10 glycosylation by tunicamycin and neuraminidase affects transient outward potassium current (I (to)) kinetics. Tunicamycin completely blocked DPP10 glycosylation and reduced DPP10 cell surface expression. The accelerating effects of DPP10 on Kv4.3 current kinetics, i.e. on inactivation and recovery from inactivation, were abolished. Neuraminidase produced different effects on current kinetics than tunicamycin, i.e., shifted the voltage dependence to more negative potentials. The effects of tunicamycin on the native I (to) currents of human atrial myocytes expressing DPP10 were similar to those of the KV4.3/KChIP2/DPP10 complex in CHO cells. Our results suggest that N-linked glycosylation of DPP10 plays an important role in modulating Kv4 channel activities.


Assuntos
Membrana Celular/metabolismo , Dipeptidil Peptidases e Tripeptidil Peptidases/metabolismo , Ativação do Canal Iônico , Proteínas Interatuantes com Canais de Kv/metabolismo , Potássio/metabolismo , Processamento de Proteína Pós-Traducional , Canais de Potássio Shal/metabolismo , Animais , Células CHO , Membrana Celular/efeitos dos fármacos , Cricetinae , Cricetulus , Dipeptidil Peptidases e Tripeptidil Peptidases/genética , Glicosilação , Átrios do Coração/metabolismo , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Cinética , Proteínas Interatuantes com Canais de Kv/genética , Potenciais da Membrana , Miócitos Cardíacos/metabolismo , Neuraminidase/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Transporte Proteico , Canais de Potássio Shal/efeitos dos fármacos , Canais de Potássio Shal/genética , Transfecção , Tunicamicina/farmacologia
13.
Neuropharmacology ; 58(1): 268-76, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19576909

RESUMO

The tachykinin endecapeptide substance P (SP) has been demonstrated to exert a functional role in neurodegenerative disorders, including Alzheimer's disease (AD). Aim of the present study was to evaluate the SP neuroprotective potential against apoptosis induced by the neurotoxic beta-amyloid peptide (A beta) in cultured rat cerebellar granule cells (CGCs). We found that SP protects CGCs against both A beta(25-35)- and A beta(1-42)-induced apoptotic CGCs death as revealed by live/dead cell assay, Hoechst staining and caspase(s)-induced PARP-1 cleavage, through an Akt-dependent mechanism. Since in CGCs the fast inactivating or A-type K(+) current (I(KA)) was potentiated by A beta treatment through up-regulation of Kv4 subunits, we investigated whether I(KA) and the related potassium channel subunits could be involved in the SP anti-apoptotic activity. Patch-clamp experiments showed that the A beta-induced increase of I(KA) current amplitude was reversed by SP treatment. In addition, as revealed by Western blot analysis and immunofluorescence studies, SP prevented the up-regulation of Kv4.2 and Kv4.3 channel subunits expression. These results indicate that SP plays a role in the regulation of voltage-gated potassium channels in A beta-mediated neuronal death and may represent a new approach in the understanding and treatment of AD.


Assuntos
Peptídeos beta-Amiloides/farmacologia , Cerebelo/citologia , Neurônios/efeitos dos fármacos , Canais de Potássio Shal/metabolismo , Substância P/farmacologia , Animais , Animais Recém-Nascidos , Biofísica , Caspase 3/metabolismo , Células Cultivadas , Estimulação Elétrica , Ativação do Canal Iônico/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Proteína Oncogênica v-akt/metabolismo , Técnicas de Patch-Clamp/métodos , Fragmentos de Peptídeos/farmacologia , Fosforilação/efeitos dos fármacos , Ratos , Ratos Wistar , Canais de Potássio Shal/efeitos dos fármacos
14.
Eur J Neurosci ; 29(10): 1943-50, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19453640

RESUMO

Arachidonic acid (AA) is a free fatty acid membrane-permeable second messenger that is liberated from cell membranes via receptor- and Ca(2+)-dependent events. We have shown previously that extremely low [AA](i) (1 pm) inhibits the postsynaptic voltage-gated K(+) current (I(A)) in hippocampal neurons. This inhibition is blocked by some antioxidants. The somatodendritic I(A) is mediated by Kv4.2 gene products, whereas presynaptic I(A) is mediated by Kv1.4 channel subunits. To address the interaction of AA with these alpha-subunits we studied the modulation of A-currents in human embryonic kidney 293 cells transfected with either Kv1.4 or Kv4.2 rat cDNA, using whole-cell voltage-clamp recording. For both currents 1 pm [AA](i) inhibited the conductance by > 50%. In addition, AA shifted the voltage dependence of inactivation by -9 (Kv1.4) and +6 mV (Kv4.2), respectively. Intracellular co-application of Trolox C (10 microm), an antioxidant vitamin E derivative, only slowed the effects of AA on amplitude. Notably, Trolox C shifted the voltage dependence of activation of Kv1.4-mediated I(A) by -32 mV. Extracellular Trolox for > 15 min inhibited the AA effects on I(A) amplitudes as well as the effect of intracellular Trolox on the voltage dependence of activation of Kv1.4-mediated I(A). Extracellular Trolox further shifted the voltage dependence of activation for Kv4.2 by +33 mV. In conclusion, the inhibition of maximal amplitude of Kv4.2 channels by AA can explain the inhibition of somatodendritic I(A) in hippocampal neurons, whereas the negative shift in the voltage dependence of inactivation apparently depends on other neuronal channel subunits. Both AA and Trolox potently modulate Kv1.4 and Kv4.2 channel alpha-subunits, thereby presumably tuning presynaptic transmitter release and postsynaptic somatodendritic excitability in synaptic transmission and plasticity.


Assuntos
Ácido Araquidônico/farmacologia , Canal de Potássio Kv1.4/efeitos dos fármacos , Canais de Potássio Shal/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos , Animais , Antioxidantes/farmacologia , Encéfalo/fisiologia , Células Cultivadas , Cromanos/farmacologia , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/fisiologia , Canal de Potássio Kv1.4/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Plasticidade Neuronal/efeitos dos fármacos , Plasticidade Neuronal/fisiologia , Técnicas de Patch-Clamp , Terminações Pré-Sinápticas/efeitos dos fármacos , Terminações Pré-Sinápticas/metabolismo , Ratos , Canais de Potássio Shal/metabolismo , Transmissão Sináptica/fisiologia , Transfecção
15.
Epilepsia ; 50(7): 1741-51, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19453702

RESUMO

PURPOSE: Kv4.2 subunits contribute to the pore-forming region of channels that express a transient, A-type K(+) current (A-current) in hippocampal CA1 pyramidal cell dendrites. Here, the A-current plays an important role in signal processing and synaptic integration. Kv4.2 knockout mice show a near elimination of the A-current in area CA1 dendrites, producing increased excitability in this region. In these studies, we evaluated young adult Kv4.2 knockout mice for spontaneous seizures and the response to convulsant stimulation in the whole animal in vivo and in hippocampal slices in vitro. METHODS: Electroencephalogram electrode-implanted Kv4.2 knockout and wild-type mice were observed for spontaneous behavioral and electrographic seizures. The latency to seizure and status epilepticus onset in Kv4.2 knockout and wild-type mice was assessed following intraperitoneal injection of kainate. Extracellular field potential recordings were performed in hippocampal slices from Kv4.2 knockout and wild-type mice following the bath application of bicuculline. RESULTS: No spontaneous behavioral or electrographic seizures were observed in Kv4.2 knockout mice. Following kainate, Kv4.2 knockout mice demonstrated a decreased seizure and status epilepticus latency as well as increased mortality compared to wild-type littermates. The background strain modified the seizure susceptibility phenotype in Kv4.2 knockout mice. In response to bicuculline, slices from Kv4.2 knockout mice exhibited an increase in epileptiform bursting in area CA1 as compared to wild-type littermates. DISCUSSION: These studies show that loss of Kv4.2 channels is associated with enhanced susceptibility to convulsant stimulation, supporting the concept that Kv4.2 deficiency may contribute to aberrant network excitability and regulate seizure threshold.


Assuntos
Convulsivantes/farmacologia , Hipocampo/efeitos dos fármacos , Hipocampo/fisiopatologia , Canais de Potássio Shal/fisiologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Western Blotting , Canalopatias/fisiopatologia , Dendritos/efeitos dos fármacos , Dendritos/fisiologia , Estimulação Elétrica , Eletroencefalografia/estatística & dados numéricos , Hipocampo/fisiologia , Camundongos , Camundongos Knockout , Fenótipo , Células Piramidais/efeitos dos fármacos , Células Piramidais/fisiologia , Convulsões/induzido quimicamente , Convulsões/etiologia , Convulsões/fisiopatologia , Canais de Potássio Shal/efeitos dos fármacos , Canais de Potássio Shal/genética , Estado Epiléptico/fisiopatologia
16.
Neuron ; 60(4): 657-71, 2008 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-19038222

RESUMO

The transient, A-type K+ current (IA) controls the excitability of CA1 pyramidal neuron dendrites by regulating the back-propagation of action potentials and by shaping synaptic input. Dendritic A-type K+ channels are targeted for modulation during long-term potentiation (LTP) and we have recently shown that activity-dependent internalization of the A-type channel subunit Kv4.2 enhances synaptic currents. However, the effect of changes in IA on the ability to induce subsequent synaptic plasticity (metaplasticity) has not been investigated. Here, we show that altering functional Kv4.2 expression level leads to a rapid, bidirectional remodeling of CA1 synapses. Neurons exhibiting enhanced IA showed a decrease in relative synaptic NR2B/NR2A subunit composition and did not exhibit LTP. Conversely, reducing IA by expression of a Kv4.2 dominant-negative or through genomic knockout of Kv4.2 led to an increased fraction of synaptic NR2B/NR2A and enhanced LTP. Bidirectional synaptic remodeling was mimicked in experiments manipulating intracellular Ca2+ and dependent on spontaneous activation of NMDA receptors and CaMKII activity. Our data suggest that A-type K+ channels are an integral part of a synaptic complex that regulates Ca2+ signaling through spontaneous NMDAR activation to control synaptic NMDAR expression and plasticity.


Assuntos
Hipocampo/metabolismo , Células Piramidais/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Canais de Potássio Shal/metabolismo , Sinapses/metabolismo , Transmissão Sináptica/fisiologia , Animais , Cálcio/metabolismo , Sinalização do Cálcio/genética , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/genética , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina/metabolismo , Hipocampo/citologia , Potenciação de Longa Duração/genética , Potenciais da Membrana/genética , Camundongos , Camundongos Knockout , Mutação/genética , Técnicas de Cultura de Órgãos , Subunidades Proteicas/efeitos dos fármacos , Subunidades Proteicas/metabolismo , Células Piramidais/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptores de N-Metil-D-Aspartato/efeitos dos fármacos , Receptores de N-Metil-D-Aspartato/genética , Canais de Potássio Shal/efeitos dos fármacos , Canais de Potássio Shal/genética , Sinapses/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos
17.
Auton Neurosci ; 144(1-2): 22-9, 2008 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-18818126

RESUMO

Nerve sprouting in healed myocardial infarction has been associated with increased incidences of ventricular tachyarrhythmia and sudden cardiac death. However, the underlying electrophysiological mechanisms are unclear. To investigate the linkage between nerve sprouting and potassium channel function, we developed a rat model of cardiac sympathetic nerve sprouting by chronic subcutaneous injection of 4-methylcatechol, a potent stimulator of nerve growth factor (NGF) synthesis. Cardiac sympathetic nerves were visualized by immunohistochemical staining. Myocardial necrotic injury was created by focal cold shock across intact diaphragm to mimic infarction. Transient outward current (I(to)) and inward rectifier current (I(K1)) of cardiomyocytes were recorded with the whole-cell patch clamp technique. We found that chronic 4-MC administration 1) increased cardiac NGF level and the density of cardiac sympathetic innervation; 2) decreased the expressions of Kv4.2, Kv channel-interacting protein 2 (KChIP2), Kir2.1, and the current densities of I(to) and I(K1); 3) reduced the phosphorylation of extracellular signal-regulated kinase 1/2 (pERK1/2); and 4) decreased heart rate variability and increased the susceptibility to ventricular fibrillation. Myocardial necrotic injury exerted similar effects as 4-methylcatechol, and 4-methylcatechol plus myocardial necrotic injury intensified the cardiac effects of 4-methylcatechol alone and decreased the phosphoralation of cAMP response element-binding protein (CREB). We conclude that nerve sprouting suppressed the expressions and functions of myocardial I(to) and I(K1) channels and increased the susceptibility to ventricular fibrillation. These effects are associated with decreased phosphorylation of ERK and CREB and reduced expression of KChIP2.


Assuntos
Infarto do Miocárdio/complicações , Regeneração Nervosa/fisiologia , Canais de Potássio/metabolismo , Fibras Simpáticas Pós-Ganglionares/fisiopatologia , Taquicardia Ventricular/fisiopatologia , Fibrilação Ventricular/fisiopatologia , Animais , Catecóis/farmacologia , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/efeitos dos fármacos , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Modelos Animais de Doenças , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Cones de Crescimento/efeitos dos fármacos , Cones de Crescimento/metabolismo , Cones de Crescimento/ultraestrutura , Proteínas Interatuantes com Canais de Kv/efeitos dos fármacos , Proteínas Interatuantes com Canais de Kv/metabolismo , Masculino , Fator de Crescimento Neural/agonistas , Regeneração Nervosa/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Fosforilação/efeitos dos fármacos , Canais de Potássio/efeitos dos fármacos , Canais de Potássio Corretores do Fluxo de Internalização/efeitos dos fármacos , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Ratos , Ratos Wistar , Canais de Potássio Shal/efeitos dos fármacos , Canais de Potássio Shal/metabolismo , Fibras Simpáticas Pós-Ganglionares/efeitos dos fármacos , Fibras Simpáticas Pós-Ganglionares/metabolismo , Taquicardia Ventricular/etiologia , Taquicardia Ventricular/metabolismo , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/fisiologia , Fibrilação Ventricular/etiologia , Fibrilação Ventricular/metabolismo
18.
Br J Pharmacol ; 154(4): 774-86, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18536731

RESUMO

BACKGROUND AND PURPOSE: The human cardiac transient outward potassium current (Ito) is believed to be composed of the pore-forming Kv4.3 alpha-subunit, coassembled with modulatory beta-subunits as KChIP2, MiRP1 and DPP6 proteins. beta-Subunits can alter the pharmacological response of Ito; therefore, we analysed the effects of flecainide on Kv4.3/KChIP2 channels coassembled with MiRP1 and/or DPP6 beta-subunits. EXPERIMENTAL APPROACH: Currents were recorded in Chinese hamster ovary cells stably expressing K(V)4.3/KChIP2 channels, and transiently transfected with either MiRP1, DPP6 or both, using the whole-cell patch-clamp technique. KEY RESULTS: In control conditions, Kv4.3/KChIP2/MiRP1 channels exhibited the slowest activation and inactivation kinetics and showed an 'overshoot' in the time course of recovery from inactivation. The midpoint values (Vh) of the activation and inactivation curves for Kv4.3/KChIP2/DPP6 and Kv4.3/KChIP2/MiRP1/DPP6 channels were approximately 10 mV more negative than Vh values for Kv4.3/KChIP2 and Kv4.3/KChIP2/MiRP1 channels. Flecainide (0.1-100 microM) produced a similar concentration-dependent blockade of total integrated current flow (IC50 approximately 10 microM) in all the channel complexes. However, the IC50 values for peak current amplitude and inactivated channel block were significantly different. Flecainide shifted the Vh values of both the activation and inactivation curves to more negative potentials and apparently accelerated inactivation kinetics in all channels. Moreover, flecainide slowed recovery from inactivation in all the channel complexes and suppressed the 'overshoot' in Kv4.3/KChIP2/MiRP1 channels. CONCLUSIONS AND IMPLICATIONS: Flecainide directly binds to the Kv4.3 alpha-subunit when the channels are in the open and inactivated state and the presence of the beta-subunits modulates the blockade by altering the gating function.


Assuntos
Antiarrítmicos/farmacologia , Flecainida/farmacologia , Subunidades Proteicas , Canais de Potássio Shal/efeitos dos fármacos , Animais , Antiarrítmicos/administração & dosagem , Células CHO , Cricetinae , Cricetulus , Dipeptidil Peptidases e Tripeptidil Peptidases , Relação Dose-Resposta a Droga , Feminino , Flecainida/administração & dosagem , Humanos , Concentração Inibidora 50 , Proteínas Interatuantes com Canais de Kv/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Técnicas de Patch-Clamp , Peptídeo Hidrolases/metabolismo , Canais de Potássio/metabolismo , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Canais de Potássio Shal/metabolismo , Transfecção
19.
J Gen Physiol ; 131(5): 455-71, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18411327

RESUMO

Shal-type (Kv4) channels are expressed in a large variety of tissues, where they contribute to transient voltage-dependent K+ currents. Kv4 are the molecular correlate of the A-type current of neurons (I(SA)), the fast component of I(TO) current in the heart, and also of the oxygen-sensitive K+ current (K(O2)) in rabbit carotid body (CB) chemoreceptor cells. The enormous degree of variability in the physiological properties of Kv4-mediated currents can be attributable to the complexity of their regulation together with the large number of ancillary subunits and scaffolding proteins that associate with Kv4 proteins to modify their trafficking and their kinetic properties. Among those, KChIPs and DPPX proteins have been demonstrated to be integral components of I(SA) and I(TO) currents, as their coexpression with Kv4 subunits recapitulates the kinetics of native currents. Here, we explore the presence and functional contribution of DPPX to K(O2) currents in rabbit CB chemoreceptor cells by using DPPX functional knockdown with siRNA. Additionally, we investigate if the presence of DPPX endows Kv4 channels with new pharmacological properties, as we have observed anomalous tetraethylammonium (TEA) sensitivity in the native K(O2) currents. DPPX association with Kv4 channels induced an increased TEA sensitivity both in heterologous expression systems and in CB chemoreceptor cells. Moreover, TEA application to Kv4-DPPX heteromultimers leads to marked kinetic effects that could be explained by an augmented closed-state inactivation. Our data suggest that DPPX proteins are integral components of K(O2) currents, and that their association with Kv4 subunits modulate the pharmacological profile of the heteromultimers.


Assuntos
Dipeptidil Peptidases e Tripeptidil Peptidases/metabolismo , Ativação do Canal Iônico/efeitos dos fármacos , Canais de Potássio Shal/efeitos dos fármacos , Canais de Potássio Shal/metabolismo , Tetraetilamônio/farmacologia , Animais , Corpo Carotídeo/metabolismo , Células Cultivadas , Células Quimiorreceptoras/fisiologia , Dipeptidil Peptidases e Tripeptidil Peptidases/genética , Expressão Gênica , Inativação Gênica , Humanos , Ativação do Canal Iônico/genética , Transporte de Íons/efeitos dos fármacos , Cinética , Complexos Multiproteicos/efeitos dos fármacos , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Oxigênio/farmacologia , Técnicas de Patch-Clamp , Bloqueadores dos Canais de Potássio/farmacologia , Subunidades Proteicas/genética , Subunidades Proteicas/metabolismo , Coelhos , Canais de Potássio Shal/genética
20.
J Neurochem ; 106(1): 182-92, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18363830

RESUMO

Somatodendritic voltage-dependent K(+) currents (Kv4.2) channels mediate transient A-type K(+) currents and play critical roles in controlling neuronal excitability. Accumulating evidence has indicated that Kv4.2 channels are key regulatory components of the signaling pathways that lead to synaptic plasticity. In contrast to the extensive studies of glutamate-induced AMPA [(+/-) alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid hydrate] receptors redistribution, less is known about the regulation of Kv4.2 by glutamate. In this study, we report that brief treatment with glutamate rapidly reduced total Kv4.2 levels in cultured hippocampal neurons. The glutamate effect was mimicked by NMDA, but not by AMPA. The effect of glutamate on Kv4.2 was dramatically attenuated by pre-treatment of NMDA receptors antagonist MK-801 [(5S,10R)-(+)-5-methyl-10,11-dihydro-5H-dibenzo[a,d]cyclohepten-5,10-imine hydrogen maleate] or removal of extracellular Ca(2+). Immunocytochemical analysis showed a loss of Kv4.2 clusters on the neuronal soma and dendrites following glutamate treatment, which was also dependent on the activation of NMDA receptors and the influx of Ca(2+). Furthermore, whole-cell patch-clamp recordings revealed that glutamate caused a hyperpolarized shift in the inactivation curve of A-type K(+) currents, while the activation curve remained unchanged. These results demonstrate a glutamate-induced alteration of Kv4.2 channels in cultured hippocampal neurons, which might be involved in activity-dependent changes of neuronal excitability and synaptic plasticity.


Assuntos
Membrana Celular/metabolismo , Ácido Glutâmico/metabolismo , Hipocampo/metabolismo , Neurônios/metabolismo , Canais de Potássio Shal/metabolismo , Animais , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Membrana Celular/efeitos dos fármacos , Células Cultivadas , Dendritos/efeitos dos fármacos , Dendritos/metabolismo , Dendritos/ultraestrutura , Agonistas de Aminoácidos Excitatórios/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Ácido Glutâmico/farmacologia , Hipocampo/citologia , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Plasticidade Neuronal/efeitos dos fármacos , Plasticidade Neuronal/fisiologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Técnicas de Patch-Clamp , Ratos , Ratos Wistar , Receptores de N-Metil-D-Aspartato/agonistas , Receptores de N-Metil-D-Aspartato/metabolismo , Canais de Potássio Shal/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Sinapses/metabolismo , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...