Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Circ Res ; 117(7): 612-621, 2015 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-26224794

RESUMO

RATIONALE: In the working heart, coronary blood flow is linked to the production of metabolites, which modulate tone of smooth muscle in a redox-dependent manner. Voltage-gated potassium channels (Kv), which play a role in controlling membrane potential in vascular smooth muscle, have certain members that are redox-sensitive. OBJECTIVE: To determine the role of redox-sensitive Kv1.5 channels in coronary metabolic flow regulation. METHODS AND RESULTS: In mice (wild-type [WT], Kv1.5 null [Kv1.5(-/-)], and Kv1.5(-/-) and WT with inducible, smooth muscle-specific expression of Kv1.5 channels), we measured mean arterial pressure, myocardial blood flow, myocardial tissue oxygen tension, and ejection fraction before and after inducing cardiac stress with norepinephrine. Cardiac work was estimated as the product of mean arterial pressure and heart rate. Isolated arteries were studied to establish whether genetic alterations modified vascular reactivity. Despite higher levels of cardiac work in the Kv1.5(-/-) mice (versus WT mice at baseline and all doses of norepinephrine), myocardial blood flow was lower in Kv1.5(-/-) mice than in WT mice. At high levels of cardiac work, tissue oxygen tension dropped significantly along with ejection fraction. Expression of Kv1.5 channels in smooth muscle in the null background rescued this phenotype of impaired metabolic dilation. In isolated vessels from Kv1.5(-/-) mice, relaxation to H2O2 was impaired, but responses to adenosine and acetylcholine were normal compared with those from WT mice. CONCLUSIONS: Kv1.5 channels in vascular smooth muscle play a critical role in coupling myocardial blood flow to cardiac metabolism. Absence of these channels disassociates metabolism from flow, resulting in cardiac pump dysfunction and tissue hypoxia.


Assuntos
Circulação Coronária/fisiologia , Vasos Coronários/metabolismo , Canal de Potássio Kv1.5/fisiologia , Músculo Liso Vascular/metabolismo , Vasodilatação/fisiologia , Animais , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos
2.
Biochem Pharmacol ; 84(4): 498-506, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22583923

RESUMO

The natural flavones and polymethylflavone have been reported to have cardiovascular protective effects. In the present study, we determined whether quecertin, apigenin and their methylated compounds (3,7,3',4'-tetramethylquecertin, 3,5,7,3',4'-pentamethylquecertin, 7,4'-dimethylapigenin, and 5,7,4'-trimethylapigenin) would block the atrial specific potassium channel hKv1.5 using a whole-cell patch voltage-clamp technique. We found that only trimethylapigenin showed a strong inhibitory effect on hKv1.5 channel current. This compound suppressed hKv1.5 current in HEK 293 cell line (IC50=6.4 µM), and the ultra-rapid delayed rectify K⁺ current I(Kur) in human atrial myocytes (IC50=8.0 µM) by binding to the open channels and showed a use- and frequency-dependent manner. In addition, trimethylapigenin decreased transient outward potassium current (I(to)) in human atrial myocytes, inhibited acetylcholine-activated K⁺ current (IC50=6.8µM) in rat atrial myocytes. Interestingly, trimethylapigenin had a weak inhibition of hERG channel current. Our results indicate that trimethyapigenin significantly inhibits the atrial potassium currents hKv1.5/I(Kur) and I(KACh), which suggests that trimethylapigenin may be a potential candidate for anti-atrial fibrillation.


Assuntos
Apigenina/farmacologia , Miocárdio/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/fisiologia , Animais , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Feminino , Células HEK293 , Átrios do Coração/citologia , Humanos , Canal de Potássio Kv1.5/antagonistas & inibidores , Canal de Potássio Kv1.5/fisiologia , Masculino , Miócitos Cardíacos/fisiologia , Técnicas de Patch-Clamp , Quercetina/análogos & derivados , Quercetina/farmacologia , Ratos , Ratos Sprague-Dawley
3.
Cancer Invest ; 30(3): 203-8, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22360360

RESUMO

Because Kv1.3 and Kv1.5 K(+) channels are remodeled during tumorigenesis and participate in skeletal muscle proliferation, we analyzed their expression in human skeletal muscle sarcomas. Aggressive alveolar rhabdomyosarcoma (ARMS) and embryonal rhabdomyosarcoma (ERMS) were studied. Kv1.5 expression was moderate in adult muscle and low in ERMS, whereas it was notable in ARMS and embryonic samples. Kv1.3 expression showed no major differences between RMS and healthy samples. We found a correlation of Kv1.3 and Kv1.5 expression with the tumor malignancy.


Assuntos
Canal de Potássio Kv1.3/análise , Canal de Potássio Kv1.5/análise , Músculo Esquelético/patologia , Rabdomiossarcoma/metabolismo , Adolescente , Adulto , Idoso , Criança , Pré-Escolar , Feminino , Fase G1 , Humanos , Imuno-Histoquímica , Canal de Potássio Kv1.3/fisiologia , Canal de Potássio Kv1.5/fisiologia , Masculino , Pessoa de Meia-Idade , Rabdomiossarcoma/patologia
4.
Eur J Pharmacol ; 649(1-3): 14-22, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-20826138

RESUMO

Repolarization of cardiac action potentials is regulated by several types of K(+) currents. The present study examined the presence and functional significance of rapid delayed rectifier (I(Kr)) in left and right atrial myocytes of mouse heart, using whole-cell patch-clamp method. The functional role of ultrarapid delayed rectifier (I(Kur)) in the repolarization was also examined by blocking with 4-aminopyridine (50 µM). The presence of I(Kr) was detected in left and right atrial myocytes as an E-4031 (5 µM)-sensitive current that exhibited relatively rapid activation during depolarization and half activation voltage of -17.5 and -17.4 mV for left and right atrial myocytes, respectively. The current density of I(Kr) was similar between left and right atria. The prolongation of action potential measured at 50% repolarization evoked by 4-aminopyridine was significantly larger in left than in right atrium, which appears to be consistent with the larger amplitude of I(Kur) in left atrium. On the other hand, the prolongation of action potential measured at 90% repolarization caused by E-4031 was significantly larger in right than in left atrium. The longer action potential of right atrium, which may result at least partly from smaller amplitude of I(Kur), is likely to enhance the functional significance of I(Kr) in repolarization process of right atrium, despite of similar magnitude of I(Kr) in left and right atria. Our data thus identifies I(Kr) in mouse atria and indicates the presence of functional interaction between I(Kr) and I(Kur) that potentially contributes to repolarization heterogeneity in left and right atria of mouse heart.


Assuntos
Potenciais de Ação/efeitos dos fármacos , Função Atrial/efeitos dos fármacos , Canais de Potássio Éter-A-Go-Go/fisiologia , Canal de Potássio Kv1.5/fisiologia , Aminopiridinas/farmacologia , Animais , Células Cultivadas , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Átrios do Coração/citologia , Átrios do Coração/efeitos dos fármacos , Cinética , Canal de Potássio Kv1.5/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/fisiologia , Especificidade de Órgãos , Técnicas de Patch-Clamp , Piperidinas/farmacologia , Bloqueadores dos Canais de Potássio/farmacologia , Piridinas/farmacologia
5.
J Ethnopharmacol ; 131(1): 187-95, 2010 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-20600762

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Aconitum has been widely used to treat various diseases in China for a long time. However, improper use of this drug results in severe intoxication. Aconitine (ACO), a diterpenoid alkaloid from aconitum, mainly contributes to cardio-toxic effects of aconitum and has also been commonly known to induce arrhythmias in animal models. However, its pro-arrhythmic mechanisms are not clear. AIM OF THE STUDY: The effects of ACO on HERG and Kv1.5 channels were investigated. MATERIALS AND METHODS: HERG and Kv1.5 channels were expressed in Xenopus laevis oocytes, and the resulting currents were recorded using a two-microelectrode voltage clamp technique. RESULTS: In HERG channels, ACO exhibited a blockade in a voltage- and time-dependent manner. The blockade was enhanced by further activation of currents, which were consistent with an open-channel blockade. In Kv1.5 channels, ACO produced a voltage-, time-, and frequency-dependent inhibition. The blockade was enhanced by higher rates of stimulation, consistent with preferential binding of the drug to the open state. In addition, ACO blocked Kv1.5 and HERG channels in a concentration-dependent manner with an IC(50) of 0.796+/-0.123 and 1.801+/-0.332 microM, respectively. CONCLUSIONS: ACO blocks HERG and Kv1.5 potassium channels in the open state. Blockade of potassium channels, particular the HERG channel, may be one of the important mechanisms of how ACO induces arrhythmias.


Assuntos
Aconitina/farmacologia , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Canal de Potássio Kv1.5/antagonistas & inibidores , Bloqueadores dos Canais de Potássio/farmacologia , Animais , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/fisiologia , Feminino , Humanos , Canal de Potássio Kv1.5/fisiologia , Xenopus laevis
6.
Can J Cardiol ; 24(1): 41-4, 2008 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18209767

RESUMO

BACKGROUND: Postoperative atrial fibrillation (AF) is a frequent complication following cardiac surgery. Risk factors leading to the development of postoperative AF are not well known and may be influenced by mutations of specific channels involved in atrial repolarization. Recently, the authors have identified three single nucleotide polymorphisms (SNPs) (R87Q, A251T and P307S) in the voltage-gated potassium channel hKv1.5 in a French-Canadian population. Two of these, R87Q and P307S, modified the gating process and the expression level of the hKv1.5 channel. OBJECTIVES: Considering that these SNPs may accelerate atrial repolarization, it was hypothesized that they may predispose patients to postoperative AF. METHODS: The authors tested the presence of SNPs in the hKv1.5 channel among 185 patients undergoing coronary artery bypass graft surgery. RESULTS: In the postoperative period, 96 patients (52%) developed a new onset of AF. A higher prevalence of SNPs was found among patients who developed postoperative AF than in the population without this postoperative arrhythmia (6.25% versus 3.37%; P=0.42). Respective allelic frequencies for R87Q and P307S were 0.52% and 1.56% in the postoperative AF group versus 0% and 0.56% in the non-AF group. Families of the carrier patients were also screened, and several members were found who carried the SNPs but did not have AF. The A251T SNP is not likely to be responsible for AF because it does not modify hKv1.5 channel functions. CONCLUSIONS: A genetic background that may be involved in the occurrence of postoperative AF was identified. Therefore, R87Q and P307S polymorphisms in hKv1.5, possibly in combination with other risk factors, may influence the development of postoperative AF.


Assuntos
Fibrilação Atrial/genética , Ponte de Artéria Coronária , Canal de Potássio Kv1.5/genética , Polimorfismo de Nucleotídeo Único , Fibrilação Atrial/epidemiologia , Fibrilação Atrial/fisiopatologia , Canadá/epidemiologia , Feminino , Humanos , Canal de Potássio Kv1.5/fisiologia , Masculino , Pessoa de Meia-Idade , Mutação , Linhagem , Projetos Piloto , Complicações Pós-Operatórias/epidemiologia , Prevalência , Fatores de Risco , População Branca/genética
7.
Am J Physiol Heart Circ Physiol ; 294(2): H570-8, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18083891

RESUMO

Pulmonary arterial hypertension (PAH) is a lethal syndrome characterized by vascular obstruction and right ventricular failure. Although the fundamental cause remains elusive, many predisposing and disease-modifying abnormalities occur, including endothelial injury/dysfunction, bone morphogenetic protein receptor-2 gene mutations, decreased expression of the O(2)-sensitive K(+) channel (Kv1.5), transcription factor activation [hypoxia-inducible factor-1alpha (HIF-1alpha) and nuclear factor-activating T cells], de novo expression of survivin, and increased expression/activity of both serotonin transporters and platelet-derived growth factor receptors. Together, these abnormalities create a cancerlike, proliferative, apoptosis-resistant phenotype in pulmonary artery smooth muscle cells (PASMCs). A possible unifying mechanism for PAH comes from studies of fawn-hooded rats, which manifest spontaneous PAH and impaired O(2) sensing. PASMC mitochondria normally produce reactive O(2) species (ROS) in proportion to P(O2). Superoxide dismutase 2 (SOD2) converts intramitochondrial superoxide to diffusible H(2)O(2), which serves as a redox-signaling molecule, regulating pulmonary vascular tone and structure through effects on Kv1.5 and transcription factors. O(2) sensing is mediated by this mitochondria-ROS-HIF-1alpha-Kv1.5 pathway. In PAH and cancer, mitochondrial metabolism and redox signaling are reversibly disordered, creating a pseudohypoxic redox state characterized by normoxic decreases in ROS, a shift from oxidative to glycolytic metabolism and HIF-1alpha activation. Three newly recognized mitochondrial abnormalities disrupt the mitochondria-ROS-HIF-1alpha-Kv1.5 pathway: 1) mitochondrial pyruvate dehydrogenase kinase activation, 2) SOD2 deficiency, and 3) fragmentation and/or hyperpolarization of the mitochondrial reticulum. The pyruvate dehydrogenase kinase inhibitor, dichloroacetate, corrects the mitochondrial abnormalities in experimental models of PAH and human cancer, causing a regression of both diseases. Mitochondrial abnormalities that disturb the ROS-HIF-1alpha-Kv1.5 O(2)-sensing pathway contribute to the pathogenesis of PAH and cancer and constitute promising therapeutic targets.


Assuntos
Hipertensão Pulmonar/genética , Hipertensão Pulmonar/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Canal de Potássio Kv1.5/fisiologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Mitocôndrias/metabolismo , Consumo de Oxigênio/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/fisiologia , Animais , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Canal de Potássio Kv1.5/genética , Oxirredução , Ratos , Superóxido Dismutase/genética , Superóxido Dismutase/fisiologia
8.
Biochem Pharmacol ; 74(11): 1596-607, 2007 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-17826747

RESUMO

BAPTA-AM is a well-known membrane permeable Ca(2+) chelator. The present study found that BAPTA-AM rapidly and reversibly suppressed human ether a-go-go-related gene (hERG or Kv11.1) K(+) current, human Kv1.3 and human Kv1.5 channel currents stably expressed in HEK 293 cells, and the effects were not related to Ca(2+) chelation. The externally applied BAPTA-AM inhibited hERG channels in a concentration-dependent manner (IC(50): 1.3 microM). Blockade of hERG channels was dependent on channel opening, and tonic block was minimal. Steady-state activation V(0.5) of hERG channels was negatively shifted by 8.5 mV (from -3.7+/-2.8 of control to -12.2+/-3.1 mV, P<0.01), while inactivation V(0.5) was negatively shifted by 6.1 mV (from -37.9+/-2.0 mV of control to -44.0+/-1.6 mV, P<0.05) with application of 3 microM BAPTA-AM. The S6 mutant Y652A and the pore helix mutant S631A significantly attenuated blockade by BAPTA-AM at 10 microM causing profound blockade of wild-type hERG channels. In addition, BAPTA-AM inhibited hKv1.3 and hKv1.5 channels in a concentration-dependent manner (IC(50): 1.45 and 1.23 microM, respectively), and the blockade of these two types of channels was also dependent on channel opening. Moreover, EGTA-AM was found to be an open channel blocker of hERG, hKv1.3, hKv1.5 channels, though its efficacy is weaker than that of BAPTA-AM. These results indicate that the membrane permeable Ca(2+) chelator BAPTA-AM (also EGTA-AM) exerts an open channel blocking effect on hERG, hKv1.3 and hKv1.5 channels.


Assuntos
Permeabilidade da Membrana Celular/efeitos dos fármacos , Ácido Egtázico/análogos & derivados , Canais de Potássio Éter-A-Go-Go/fisiologia , Cálcio/metabolismo , Linhagem Celular , Quelantes/farmacologia , Relação Dose-Resposta a Droga , Ácido Egtázico/farmacologia , Canais de Potássio Éter-A-Go-Go/genética , Humanos , Canal de Potássio Kv1.3/genética , Canal de Potássio Kv1.3/fisiologia , Canal de Potássio Kv1.5/genética , Canal de Potássio Kv1.5/fisiologia , Potenciais da Membrana/efeitos dos fármacos , Mutagênese Sítio-Dirigida , Porinas/genética , Porinas/fisiologia , Transfecção
9.
Am J Physiol Cell Physiol ; 292(3): C1041-52, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16956964

RESUMO

In Kv1.5, protonation of histidine 463 in the S5-P linker (turret) increases the rate of depolarization-induced inactivation and decreases the peak current amplitude. In this study, we examined how amino acid substitutions that altered the physico-chemical properties of the side chain at position 463 affected slow inactivation and then used the substituted cysteine accessibility method (SCAM) to probe the turret region (E456-P468) to determine whether residue 463 was unique in its ability to modulate the macroscopic current. Substitutions at position 463 of small, neutral (H463G and H463A) or large, charged (H463R, H463K, and H463E) side groups accelerated inactivation and induced a dependency of the current amplitude on the external potassium concentration. When cysteine substitutions were made in the distal turret (T462C-P468C), modification with either the positively charged [2-(trimethylammonium)ethyl] methanethiosulfonate bromide (MTSET) or negatively charged sodium (2-sulfonatoethyl) methanethiosulfonate reagent irreversibly inhibited current. This inhibition could be antagonized either by the R487V mutation (homologous to T449V in Shaker) or by raising the external potassium concentration, suggesting that current inhibition by MTS reagents resulted from an enhancement of inactivation. These results imply that protonation of residue 463 does not modulate inactivation solely by an electrostatic interaction with residues near the pore mouth, as proposed by others, and that residue 463 is part of a group of residues within the Kv1.5 turret that can modulate P/C-type inactivation.


Assuntos
Ativação do Canal Iônico/fisiologia , Rim/fisiologia , Canal de Potássio Kv1.5/química , Canal de Potássio Kv1.5/fisiologia , Potenciais da Membrana/fisiologia , Substituição de Aminoácidos , Linhagem Celular , Cisteína/química , Cisteína/metabolismo , Humanos , Mutagênese Sítio-Dirigida , Porosidade , Conformação Proteica , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA