Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Arch Biochem Biophys ; 634: 47-56, 2017 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-28778458

RESUMO

Controlling target gene expression is a vital step in the procedure of gene therapy upon acute lung injury (ALI). Excessive activation of nuclear factor-kappa B (NF-κB) has been the key point of the inflammation overwhelming process in onset of ALI. We designed and tested a variety of plasmid named pHSP70/IκBαm which conditionally carries a mutant inhibitor of kappa B (IκB) transgene to regulate the activity of NF-κB signaling pathway in its response to an inflammatory stimulus that causes acute lung injury. Results recorded along our experiments showed that pHSP70/IκBαm was able to control mutant IκB expression in RAW264.7 cells with reference to the level of inflammatory response induced by LPS, thereby inhibiting NF-κB activation and downstream inflammatory cytokine expression. Vivo experiments revealed that construction naming pHSP70/IκBαm reduced LPS-induced lung injury and the secretion of inflammatory factors from lungs, hearts, and livers of sample mice in a LPS dose-dependent manner. In conclusion, the promoter heat shocking protein 70(HSP70) regulatory sequence of the construction was shown to drive mutant IκB expression so that its levels were positively associated with the dose of LPS used to induce acute lung injury. NF-κB activation and the downstream expression of inflammatory factors were therefore down-regulated in along an efficient path and ameliorating the damage as a consequence of LPS-induced acute lung injury.


Assuntos
Lesão Pulmonar Aguda/imunologia , Lesão Pulmonar Aguda/prevenção & controle , Terapia de Alvo Molecular , NF-kappa B/imunologia , Plasmídeos/administração & dosagem , Lesão Pulmonar Aguda/genética , Animais , Citocinas/imunologia , Desenho de Fármacos , Regulação da Expressão Gênica/genética , Regulação da Expressão Gênica/imunologia , Marcação de Genes , Proteínas de Choque Térmico HSP70/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , NF-kappa B/genética , Plasmídeos/genética , Regiões Promotoras Genéticas/genética , Células RAW 264.7 , Sequências Reguladoras de Ácido Ribonucleico/genética , Resultado do Tratamento
2.
Am J Respir Cell Mol Biol ; 49(6): 1057-63, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23855826

RESUMO

Leptin is reported to be involved in acute lung injury (ALI). However, the role and underlying mechanisms of leptin in ALI remain unclear. The aim of this study was to determine whether leptin deficiency promoted the development of ALI. LPS or oleic acid (OA) were administered to wild-type and leptin deficient (ob/ob) mice to induce ALI. Leptin level, survival rate, and lung injury were examined. Results showed that leptin levels were predominantly increased in the lung, but also in the heart, liver, kidney, and adipose tissue after LPS adminiatration. Compared with wild-type mice, LPS- or OA-induced lung injury was worse and the survival rate was lower in ob/ob mice. Moreover, leptin deficiency promoted the release of proinflammatory cytokines. Exogenous administration of leptin reduced lethality in ob/ob mice and ameliorated lung injury partly through inhibiting the activation of NF-κB, p38, and ERK pathways. These results indicated that leptin deficiency contributed to the development of lung injury by enhancing inflammatory response, and a high level of leptin improved survival and protected against ALI.


Assuntos
Lesão Pulmonar Aguda/etiologia , Lesão Pulmonar Aguda/prevenção & controle , Leptina/fisiologia , Lipopolissacarídeos/toxicidade , Ácido Oleico/toxicidade , Lesão Pulmonar Aguda/fisiopatologia , Animais , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Mediadores da Inflamação/metabolismo , Leptina/deficiência , Leptina/genética , Pulmão/efeitos dos fármacos , Pulmão/metabolismo , Pulmão/patologia , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Obesos , NF-kappa B/metabolismo , Regulação para Cima/efeitos dos fármacos
3.
Biochim Biophys Acta ; 1823(2): 282-9, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22061963

RESUMO

Human ether á-go-go gene potassium channels (hEAG1 or Kv10.1) are expressed in brain and various human cancers and play a role in neuronal excitement and tumor progression. However, the functional regulation of hEAG channels by signal transduction is not fully understood. The present study was therefore designed to investigate whether hEAG1 channels are regulated by protein tyrosine kinases (PTKs) in HEK 293 cells stably expressing hEAG1 gene using whole-cell patch voltage-clamp, immunoprecipitation, Western blot, and mutagenesis approaches. We found that the selective epidermal growth factor receptor (EGFR) kinase inhibitor AG556 (10 µM), but not the platelet growth factor receptor (PDGFR) kinase inhibitor AG1295 (10 µM) or the Src-family inhibitor PP2 (10 µM), can inhibit hEAG1 current, and the inhibitory effect can be reversed by the protein tyrosine phosphatase (PTP) inhibitor orthovanadate. Immunoprecipitation and Western blot analysis revealed that tyrosine phosphorylation level of hEAG1 channels was reduced by AG556, and the reduction was significantly countered by orthovanadate. The hEAG1 mutants Y90A, Y344A and Y485A, but not Y376A and Y479A, exhibited reduced response to AG556. Interestingly, the inhibition effect of AG556 was lost in triple mutant hEAG1 channels at Y90, Y344, and Y485 with alanine. These results demonstrate for the first time that hEAG1 channel activity is regulated by EGFR kinase at the tyrosine residues Tyr90, Try344, and Try485. This effect is likely involved in regulating neuronal activity and/or tumor growth.


Assuntos
Receptores ErbB/metabolismo , Canais de Potássio Éter-A-Go-Go/metabolismo , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , Canais de Potássio Éter-A-Go-Go/genética , Células HEK293 , Humanos , Técnicas de Patch-Clamp , Interferência de RNA , Transdução de Sinais/fisiologia , Tirosina/metabolismo , Tirfostinas/metabolismo , Vanadatos/metabolismo
4.
Microvasc Res ; 83(2): 205-12, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22005047

RESUMO

BACKGROUND: Hypoxic pulmonary vasoconstriction may lead to pulmonary hypertension, but the underlying mechanisms of persistent vasoconstriction are still unclear. There is evidence that pulmonary inflammation contributes to the abnormalities of function in the pulmonary artery (PA) following chronic hypoxia exposure. Macrophage migration inhibitory factor (MIF) is an important pro-inflammatory cytokine, and we found that expression of MIF was increased in the smooth muscle of PA from hypoxic pulmonary hypertensive rats. Therefore, the aim of the study was to investigate the role of MIF in modulating vasoreactivity of isolated PA rings. METHODS: Sprague-Dawley rats were challenged by intermittent chronic hypoxia exposure for 4 weeks to establish hypoxic pulmonary hypertension models. Subsequently, immunohistochemistry and western blot assay were used to examine the MIF expression in pulmonary artery. Moreover, isometric force displacement was measured in isolated intrapulmonary artery. RESULTS: In the isolated PA, our results showed that MIF mediated the enhanced pulmonary arterial vasoconstriction in response to chronic hypoxia, and the delayed hypoxic constriction in a biphasic pattern of constriction occurs in response to acute hypoxia. We also present the finding that MIF had no effect on force on its own, but concentration-dependently potentiated constrictions pre-evoked by phenylephrine under normoxic condition. The potentiation was independent of the endothelium. MIF-induced potentiation of phenylephrine-evoked constriction was partially inhibited by PKC inhibitor chelerythrine, p38 inhibitor SB 203580, ERK1/2 inhibitor U0126, respectively. CONCLUSIONS: Our results suggested that MIF enhanced vasoconstriction of pulmonary artery elicited by agonist through PKC, p38 and ERK1/2 signal pathways, which may contributes to hypoxic pulmonary vasoconstriction.


Assuntos
Hipertensão Pulmonar/etiologia , Hipóxia/complicações , Oxirredutases Intramoleculares/metabolismo , Fatores Inibidores da Migração de Macrófagos/metabolismo , Artéria Pulmonar/metabolismo , Artéria Pulmonar/fisiopatologia , Vasoconstrição , Animais , Western Blotting , Células Cultivadas , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Hipertensão Pulmonar Primária Familiar , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/fisiopatologia , Hipóxia/metabolismo , Hipóxia/fisiopatologia , Imuno-Histoquímica , Masculino , Proteína Quinase 1 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/antagonistas & inibidores , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Cadeias Leves de Miosina/metabolismo , Fosforilação , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Artéria Pulmonar/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Vasoconstrição/efeitos dos fármacos , Vasoconstritores/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
5.
Biochim Biophys Acta ; 1798(5): 995-1001, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20085748

RESUMO

The aim of the present study was to investigate whether/how the recombinant human cardiac I(Ks) could be regulated by epidermal growth factor receptor kinase in HEK 293 cells stably expressing hKCNQ1/hKCNE1 genes using the approaches of perforated patch clamp technique, immunoprecipitation and Western blot analysis. It was found that the broad spectrum isoflavone tyrosine kinase inhibitor genistein and the selective epidermal growth factor receptor kinase inhibitor tyrphostin AG556 suppressed the recombinant I(Ks), and their inhibition was countered by the protein tyrosine phosphatase inhibitor orthovanadate. The Src-family kinase inhibitor PP2 reduced the current, but the effect was not antagonized by orthovanadate. Immunoprecipitation and Western blot analysis revealed that tyrosine phosphorylation level of hKCNQ1 protein was decreased by genistein or AG556, but not by PP2. These results provide the novel information that epidermal growth factor receptor kinase, but not Src-family kinases, regulates the recombinant cardiac I(Ks) stably expressed in HEK 293 cells via phosphorylating KCNQ1 protein of the channel.


Assuntos
Receptores ErbB/metabolismo , Canal de Potássio KCNQ1/metabolismo , Proteínas Recombinantes/metabolismo , Animais , Linhagem Celular , Eletrofisiologia , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , Genisteína/metabolismo , Humanos , Ativação do Canal Iônico/fisiologia , Canal de Potássio KCNQ1/genética , Fosforilação , Inibidores de Proteínas Quinases/metabolismo , Proteínas Recombinantes/genética , Tirosina/metabolismo , Tirfostinas/metabolismo , Vanadatos/metabolismo , Quinases da Família src/genética , Quinases da Família src/metabolismo
6.
Pulm Pharmacol Ther ; 24(2): 240-6, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21199676

RESUMO

Bicyclol is synthesized based on schisandrin, which is one of the main active components of Chinese herb Fructus Schisandrae. The purpose of this study is to investigate whether bicyclol has a beneficial effect on lipopolysaccharide (LPS)-induced acute lung injury (ALI) in mice. Bicyclol was given to mice by gavage for three times. ALI was induced by vena caudalis injection of LPS. The last dose of bicyclol was administrated 1 h before LPS given. Mice in each group were sacrificed at different time point after LPS administration. As revealed by survival study, pretreatment with high doses of bicyclol reduced the mortality of mice from ALI. Bicyclol pretreatment significantly improved LPS-induced lung pathological changes, inhibited myeloperoxidase (MPO) activity, and reduced lung/body and lung wet/dry weight ratios. Bicyclol also inhibited the release of TNF-α, IL-1ß and HMGB1, whereas simultaneously increased the expression of IL-10. Furthermore, the phosphorylation level of NF-κB p65 was markedly decreased by bicyclol. Taken together, our study showed that bicyclol improves survival rate and attenuates LPS-induced ALI. The protective mechanism may be due to the inhibition of NF-κB activation and regulation of cytokine secretion.


Assuntos
Lesão Pulmonar Aguda/prevenção & controle , Compostos de Bifenilo/farmacologia , NF-kappa B/metabolismo , Lesão Pulmonar Aguda/mortalidade , Animais , Compostos de Bifenilo/administração & dosagem , Citocinas/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Lipopolissacarídeos/toxicidade , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Peroxidase/metabolismo , Fosforilação/efeitos dos fármacos , Fatores de Tempo , Fator de Transcrição RelA/metabolismo
7.
Respir Res ; 11: 182, 2010 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-21182801

RESUMO

BACKGROUND: Pulmonary vascular structure remodeling (PVSR) is a hallmark of pulmonary hypertension. P27(kip1), one of critical cyclin-dependent kinase inhibitors, has been shown to mediate anti-proliferation effects on various vascular cells. Beta-estradiol (ß-E2) has numerous biological protective effects including attenuation of hypoxic pulmonary hypertension (HPH). In the present study, we employed ß-E2 to investigate the roles of p27(kip1) and its closely-related kinase (Skp-2) in the progression of PVSR and HPH. METHODS: Sprague-Dawley rats treated with or without ß-E2 were challenged by intermittent chronic hypoxia exposure for 4 weeks to establish hypoxic pulmonary hypertension models, which resemble moderate severity of hypoxia-induced PH in humans. Subsequently, hemodynamic and pulmonary pathomorphology data were gathered. Additionally, pulmonary artery smooth muscle cells (PASMCs) were cultured to determine the anti-proliferation effect of ß-E2 under hypoxia exposure. Western blotting or reverse transcriptional polymerase chain reaction (RT-PCR) were adopted to test p27(kip1), Skp-2 and Akt-P changes in rat lung tissue and cultured PASMCs. RESULTS: Chronic hypoxia significantly increased right ventricular systolic pressures (RVSP), weight of right ventricle/left ventricle plus septum (RV/LV+S) ratio, medial width of pulmonary arterioles, accompanied with decreased expression of p27(kip1) in rats. Whereas, ß-E2 treatment repressed the elevation of RVSP, RV/LV+S, attenuated the PVSR of pulmonary arterioles induced by chronic hypoxia, and stabilized the expression of p27(kip1). Study also showed that ß-E2 application suppressed the proliferation of PASMCs and elevated the expression of p27(kip1) under hypoxia exposure. In addition, experiments both in vivo and in vitro consistently indicated an escalation of Skp-2 and phosphorylated Akt under hypoxia condition. Besides, all these changes were alleviated in the presence of ß-E2. CONCLUSIONS: Our results suggest that ß-E2 can effectively attenuate PVSR and HPH. The underlying mechanism may partially be through the increased p27(kip1) by inhibiting Skp-2 through Akt signal pathway. Therefore, targeting up-regulation of p27(kip1) or down-regulation of Skp-2 might provide new strategies for treatment of HPH.


Assuntos
Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Estradiol/administração & dosagem , Hipertensão Pulmonar/tratamento farmacológico , Hipertensão Pulmonar/metabolismo , Hipóxia/tratamento farmacológico , Hipóxia/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Relação Dose-Resposta a Droga , Expressão Gênica/efeitos dos fármacos , Hipertensão Pulmonar/complicações , Hipóxia/complicações , Masculino , Ratos , Ratos Sprague-Dawley , Resultado do Tratamento
8.
Chin J Physiol ; 53(1): 36-44, 2010 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-21789883

RESUMO

Chronic hypoxia results in pulmonary hypertension. To investigate the role of Na+/H+ exchange in this process, we determined the effect of amiloride, a Na+/H+ exchange inhibitor, on hypoxic pulmonary hypertension and pulmonary arterial smooth muscle cell proliferation, both in vivo and in vitro. Sprague-Dawley rats were placed either in a hypobaric, hypoxic chamber (10.5% 02) or under normal 21% O2 atmosphere for 8 h each day for 3 weeks. Rats under hypoxic conditions received 1, 3, or 10 mg/kg/d amiloride or the vehicle alone. Hematologic indices, including red blood cells, hemoglobin, hematocrit and mean corpuscular hemoglobin increased in hypoxic rats, but these changes were prevented by treatment with amiloride. In the hypoxic rats, the right ventricular systolic pressure and right ventricular hypertension index (weight ratio of right ventricular to left and septum together) were increased by 88% and 129%, respectively. Arteriolar wall thickness and area in the hypoxia-treated animals increased 3- and 2-fold, respectively, over normoxic controls; the increase in each of these indices was attenuated by amiloride in a dose-dependent manner. In cultured pulmonary arterial smooth muscle cells, hypoxia greatly increased cellular proliferation, and this similarly showed a dose-dependent attenuation in the presence of amiloride. Amiloride did not affect blood pressure in vivo or cause cell damage in vitro. These data suggest that the Na+/H+ exchange inhibitor amiloride may represent an effective adjunctive therapy in pulmonary hypertension induced by chronic hypoxia.


Assuntos
Amilorida/farmacologia , Proliferação de Células/efeitos dos fármacos , Hipertensão Pulmonar/etiologia , Hipóxia/complicações , Músculo Liso Vascular/patologia , Artéria Pulmonar/patologia , Trocadores de Sódio-Hidrogênio/antagonistas & inibidores , Animais , Pressão Sanguínea/efeitos dos fármacos , Células Cultivadas , Contagem de Eritrócitos , Hematócrito , Hemoglobinas/metabolismo , Concentração de Íons de Hidrogênio , Hipertensão Pulmonar/sangue , Hipertensão Pulmonar/patologia , Masculino , Modelos Animais , Músculo Liso Vascular/efeitos dos fármacos , Artéria Pulmonar/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Bloqueadores dos Canais de Sódio/farmacologia , Trocadores de Sódio-Hidrogênio/efeitos dos fármacos , Trocadores de Sódio-Hidrogênio/metabolismo
9.
Mol Ther Methods Clin Dev ; 17: 975-985, 2020 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-32426413

RESUMO

Hypoxia-induced pulmonary vascular constriction and structure remodeling are the main causes of hypoxic pulmonary hypertension. In the present study, an adeno-associated virus vector, containing Tie2 promoter and hypoxia response elements, was designed and named HTSFcAng(1-7). Its targeting, hypoxic inducibility, and vascular relaxation were examined in vitro, and its therapeutic effects on hypobaric hypoxia-induced pulmonary hypertension were examined in rats. Transfection of HTSFcAng(1-7) specifically increased the expression of angiotensin-(1-7) in endothelial cells in normoxia. Hypoxia increased the expression of angiotensin-(1-7) in HTSFcAng(1-7)-transfected endothelial cells. The condition medium from HTSFcAng(1-7)-transfected endothelial cells inhibited the hypoxia-induced proliferation of pulmonary artery smooth muscle cells, relaxed the pulmonary artery rings, totally inhibited hypoxia-induced early contraction, enhanced maximum relaxation, and reversed phase II constriction to sustained relaxation. In hypoxic pulmonary hypertension rats, treatment with HTSFcAng(1-7) by nasal drip adeno-associated virus significantly reversed hypoxia-induced hemodynamic changes and pulmonary artery-wall remodeling, accompanied by the concomitant overexpression of angiotensin-(1-7), mainly in the endothelial cells in the lung. Therefore, hypoxia-inducible overexpression of angiotensin-(1-7) in pulmonary endothelial cells may be a potential strategy for the gene therapy of hypoxic pulmonary hypertension.

10.
Clin Exp Pharmacol Physiol ; 36(10): e57-64, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19515065

RESUMO

1. Reactive oxygen species (ROS) cause vascular complications and impair vasodilation in diabetes mellitus. Large-conductance Ca(2+)-activated potassium channels (BK(Ca)) modulate vascular tone and play an important negative feedback role in vasoconstriction. In the present study, we tested the hypothesis that ROS regulate the function of BK(Ca) in diabetic cerebral artery smooth muscle cells. 2. Diabetes was induced in male BALB/c mice by injection of streptozotocin (STZ; 180 mg/kg, i.p., dissolved in sterile saline). Control and diabetic mice were treated with 12.7 micromol/L rotenone, an inhibitor of the mitochondrial electron transport chain complex I, or placebo every other day for 5 weeks. The whole-cell patch clamp-technique and functional vasomotor methods were used to record BK(Ca) currents and myogenic tone of cerebral artery smooth muscle cells. 3. In the diabetic group, there was a significant decrease in spontaneous transient outward currents in cerebral artery smooth muscle cells compared with control. Although the currents were only moderately increased in rotenone-treated diabetic mice, they remained significantly lower than in the control group. Furthermore, the macroscopic BK(Ca) currents that were decreased in diabetic mice were partially recovered in rotenone-treated diabetic mice (P < 0.05 vs untreated diabetic group). 4. The posterior cerebral artery from diabetic mice had a significantly higher myogenic tone than the control group, but this impaired contraction was partially reversed in the rotenone-treated diabetic group (P < 0.05 vs untreated diabetic group). 5. The H(2)O(2) concentration was significantly increased in cerebral arteries from diabetic mice compared with control. This increase in H(2)O(2) was significantly blunted by rotenone treatment. 6. In conclusion, rotenone partially reverses the decreased macroscopic BK(Ca) currents in STZ-induced Type 1 diabetic mice and this reversal of BK(Ca) currents may be related to the inhibitory effects of rotenone on H(2)O(2) production. Reactive oxygen species, particularly H(2)O(2), are important regulators of BK(Ca) channels and myogenic tone in diabetic cerebral artery.


Assuntos
Artérias Cerebrais/efeitos dos fármacos , Diabetes Mellitus Experimental/fisiopatologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/efeitos dos fármacos , Músculo Liso Vascular/efeitos dos fármacos , Rotenona/farmacologia , Estreptozocina , Animais , Sinalização do Cálcio/efeitos dos fármacos , Artérias Cerebrais/metabolismo , Artérias Cerebrais/patologia , Artérias Cerebrais/fisiopatologia , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Avaliação Pré-Clínica de Medicamentos , Peróxido de Hidrogênio/metabolismo , Peróxido de Hidrogênio/farmacologia , Inseticidas/farmacologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Alta/fisiologia , Masculino , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patologia , Músculo Liso Vascular/fisiopatologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/patologia , Miócitos de Músculo Liso/fisiologia , Técnicas de Patch-Clamp , Vasoconstrição/efeitos dos fármacos
11.
Aviat Space Environ Med ; 80(8): 698-702, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19653571

RESUMO

INTRODUCTION: Exposure to high +Gz acceleration forces on a centrifuge or in an aircraft can severely decrease cerebral blood perfusion and cause rapid G-induced loss of consciousness. However, milder acceleration may gradually reduce cerebral blood flow and affect cognitive function in subtler ways. This study used lower body negative pressure (LBNP) to mimic +Gz circulatory effects in order to study cerebral hemodynamics and brain function. METHODS: Subjects were 15 healthy men, 19-21 yr of age. They were exposed to LBNP at two levels for 5 min each separated by a 10-min recovery period. The conditions were low (LO), -4.00 kPa (-30 mmHg) and high (HI), -6.67 kPa (-50 mmHg).Variables measured before, during, and after LBNP included cerebral blood flow velocity (CBFV) in the middle cerebral artery, blood oxygen saturation (SaO2), heart rate (HR), blood pressure, P300 of event-related EEG potentials, reaction time, and tracking error. RESULTS: LO significantly reduced CBFV at 4 and 5 min, increased HR, and decreased the amplitude of P300, but none of the other variables changed from baseline. In contrast, HI produced significant changes in most variables: CBFV decreased at 2 min and then fell further at 4 and 5 min, HR increased, and SaO2 decreased. Significant neurocognitive changes included increased latency and reduced amplitude of P300, slower reaction time, and greater tracking error. CONCLUSION: The higher level of LBNP used here reduced cerebral perfusion sufficiently to impair neurocognitive function. This model may be useful for further studies of these and other variables under closely controlled conditions.


Assuntos
Circulação Cerebrovascular/fisiologia , Gravitação , Hemodinâmica/fisiologia , Pressão Negativa da Região Corporal Inferior/efeitos adversos , Artéria Cerebral Média/fisiopatologia , Medicina Aeroespacial , Velocidade do Fluxo Sanguíneo , Humanos , Masculino , Tempo de Reação , Ultrassonografia Doppler Transcraniana , Adulto Jovem
12.
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
13.
J Am Heart Assoc ; 6(6)2017 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-28645933

RESUMO

BACKGROUND: Pulmonary arterial remodeling, a main characteristic of hypoxic pulmonary hypertension, can gradually reverse once oxygen has been restored. Previous studies documented that apoptosis increased markedly during the reversal of remodeled pulmonary arteries, but the types of cells and mechanisms related to the apoptosis have remained elusive. This study aimed to determine whether pulmonary artery smooth muscle cell (PASMC)-specific apoptosis was involved in the reoxygenation-induced reversal of hypoxic pulmonary arterial remodeling and elucidate the underlying mechanism. METHODS AND RESULTS: Hypoxic pulmonary hypertension was induced in adult male Sprague-Dawley rats (n=6/group) by chronic hypobaric hypoxia. and the hypoxic pulmonary hypertension rats were then transferred to a normoxia condition. During reoxygenation, hypoxia-induced pulmonary arterial remodeling gradually reversed. The reversal of remodeled pulmonary arteries was associated with increased H2O2 and with changes in lung expression of cleaved caspase3/PARP, Bax, and Bcl-2, consistent with increased apoptosis. The PASMC apoptosis, in particular, increased remarkably during this reversal. In vitro, reoxygenation induced the apoptosis of cultured rat primary PASMCs accompanied by increased mitochondrial reactive oxygen species, mitochondrial dysfunction, and the release of cytochrome C from mitochondria to cytoplasm. Clearance of reactive oxygen species alleviated mitochondrial dysfunction as well as the release of cytochrome C and, finally, decreased PASMC apoptosis. CONCLUSIONS: Reoxygenation-induced apoptosis of PASMCs is implicated in the reversal of hypoxic pulmonary arterial remodeling, which may be attributed to the mitochondrial reactive oxygen species-mediated mitochondrial dysfunction.


Assuntos
Apoptose , Hipertensão Pulmonar/etiologia , Hipóxia/complicações , Mitocôndrias Musculares/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Oxigênio/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Remodelação Vascular , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Hipertensão Pulmonar/metabolismo , Hipertensão Pulmonar/patologia , Hipertensão Pulmonar/fisiopatologia , Hipóxia/metabolismo , Hipóxia/patologia , Hipóxia/fisiopatologia , Masculino , Mitocôndrias Musculares/patologia , Músculo Liso Vascular/patologia , Músculo Liso Vascular/fisiopatologia , Miócitos de Músculo Liso/patologia , Artéria Pulmonar/metabolismo , Artéria Pulmonar/patologia , Artéria Pulmonar/fisiopatologia , Ratos Sprague-Dawley , Transdução de Sinais , Fatores de Tempo
14.
Br J Pharmacol ; 147(3): 289-97, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16341233

RESUMO

The antifungal antibiotic clotrimazole (CLT) shows therapeutic effects on cancer, sickle cell disease, malaria, etc. by inhibiting membrane intermediate-conductance Ca2+ -activated K+ channels (IKCa). However, it is unclear whether this drug would affect human cardiac K+ currents. The present study was therefore designed to investigate the effects of CLT on transient outward K+ current (Ito1), and ultra-rapid delayed rectifier K+ current (IKur) in isolated human atrial myocytes, and cloned hERG channel current (IhERG) and recombinant human cardiac KCNQ1/KCNE1 channel current (IKs) expressed in HEK 293 cells. It was found that CLT inhibited Ito1 with an IC50 of 29.5 microM, accelerated Ito1 inactivation, and decreased recovery of Ito1 from inactivation. In addition, CLT inhibited human atrial I(Kur) in a concentration-dependent manner (IC50 = 7.6 microM). CLT substantially suppressed IhERG (IC50 = 3.6 microM), and negatively shifted the activation conductance of IhERG. Moreover, CLT inhibited IKs (IC50 = 15.1 microM), and positively shifted the activation conductance of the current. These results indicate that the antifungal antibiotic CLT substantially inhibits human cardiac repolarization K+ currents including Ito1, IKur, IhERG, and IKs. However, caution is recommended when correlating the observed in vitro effects on cardiac ion currents to the clinical relevance.


Assuntos
Antifúngicos/farmacologia , Clotrimazol/farmacologia , Coração/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio/efeitos dos fármacos , Adulto , Idoso , Células Cultivadas , Canais de Potássio de Retificação Tardia/antagonistas & inibidores , Canal de Potássio ERG1 , Canais de Potássio Éter-A-Go-Go/antagonistas & inibidores , Humanos , Canal de Potássio KCNQ1/antagonistas & inibidores , Pessoa de Meia-Idade , Canais de Potássio/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/antagonistas & inibidores
15.
Br J Pharmacol ; 173(10): 1569-79, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26914282

RESUMO

BACKGROUND AND PURPOSE: Berberine, a small molecule derived from Coptidis rhizome, has been found to be potent at lowering blood glucose and regulating lipid metabolism. Recent clinical studies have shown that berberine reduces blood pressure and increases systemic insulin sensitivity in patients with metabolic syndrome; however, the underlying mechanism is still unclear. Here, we investigated the mechanism by which berberine improves vascular insulin sensitivity in diabetic rats. EXPERIMENTAL APPROACH: Diabetes was induced in male Sprague­Dawley rats by feeding a high-fat diet and administration of a low dose of streptozotocin. These diabetic rats were treated with berberine (200 mg·kg(−1)·day(−1), gavage) for 4 weeks. Vascular dilation was determined in isolated mesenteric artery rings. Effects of berberine on insulin signalling were also studied in human artery endothelial cells cultured in high glucose (25 mmol·L(−1)) and palmitate (500 µmol·L(−1)). KEY RESULTS: Berberine treatment for 4 weeks significantly restored the impaired ACh- and insulin-induced vasodilatation of mesenteric arteries from diabetic rats. In isolated mesenteric artery rings, berberine (2.5­10 µmol·L(−1)) elicited dose-dependent vasodilatation and significantly enhanced insulin-induced vasodilatation. Mechanistically, berberine up-regulated phosphorylation of the insulin receptor and its downstream signalling molecules AMPK, Akt and eNOS, and increased cell viability and autophagy in cultured endothelial cells. Moreover, down-regulating insulin receptors with specific siRNA significantly attenuated berberine-induced phosphorylation of AMPK. CONCLUSIONS AND IMPLICATIONS: Berberine improves diabetic vascular insulin sensitivity and mesenteric vasodilatation by up-regulating insulin receptor-mediated signalling in diabetic rats. These findings suggest berberine has potential as a preventive or adjunctive treatment of diabetic vascular complications. LINKED ARTICLES: This article is part of a themed section on Chinese Innovation in Cardiovascular Drug Discovery. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2015.172.issue-23.


Assuntos
Berberina/farmacologia , Resistência à Insulina , Artérias Mesentéricas/efeitos dos fármacos , Receptor de Insulina/fisiologia , Transdução de Sinais/fisiologia , Regulação para Cima , Acetilcolina/farmacologia , Animais , Glicemia/metabolismo , Linhagem Celular , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/enzimologia , Endotélio Vascular/metabolismo , Humanos , Insulina/farmacologia , Masculino , Artérias Mesentéricas/metabolismo , RNA Interferente Pequeno/genética , Ratos , Ratos Sprague-Dawley , Receptor de Insulina/genética , Vasodilatação/efeitos dos fármacos
16.
Hypertension ; 65(2): 414-20, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25421979

RESUMO

Activation of pulmonary adventitial fibroblasts plays a key role in the pulmonary vascular remodeling in hypoxic pulmonary hypertension. Previous studies showed that miRNAs participated in the regulation of fibroblast activation. This study explored the role of miR-29 in the activation of pulmonary adventitial fibroblasts and the therapeutic potential in hypoxic pulmonary hypertension. We found that hypoxia-induced pulmonary adventitial fibroblasts activation was accompanied with a drastic decrease of miR-29a-3p expression. Knockdown of hypoxia-inducible factor-1 α or Smad3 reversed the hypoxia-induced decrease of miR-29-3p in cultured pulmonary adventitial fibroblasts. In vitro, miR-29a-3p mimic inhibited the hypoxia-induced proliferation, migration, and secretion of pulmonary adventitial fibroblasts, suppressed the hypoxia-induced expression of α-smooth muscle actin and extracellular matrix collagen in pulmonary adventitial fibroblasts; however, miR-29a-3p inhibitor mimicked the effect of hypoxia on the activation of pulmonary adventitial fibroblasts. Further studies revealed that preventative or therapeutic administration of miR-29a-3p significantly decreased pulmonary artery pressure and right ventricle hypertrophy index and ameliorated pulmonary vascular remodeling in hypoxic pulmonary hypertension rats. These findings suggest that miR-29a-3p regulates the activation and phenotype of pulmonary adventitial fibroblasts in hypoxia and has preventative and therapeutic potential in hypoxic pulmonary hypertension.


Assuntos
Fibroblastos/efeitos dos fármacos , Vetores Genéticos/uso terapêutico , Hipertensão Pulmonar/prevenção & controle , Hipertrofia Ventricular Direita/prevenção & controle , Hipóxia/complicações , Pulmão/patologia , MicroRNAs/uso terapêutico , Remodelação Vascular/efeitos dos fármacos , Túnica Adventícia/patologia , Animais , Sequência de Bases , Células Cultivadas , Dependovirus/genética , Regulação para Baixo , Proteínas da Matriz Extracelular/biossíntese , Proteínas da Matriz Extracelular/genética , Fibroblastos/fisiologia , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Terapia Genética , Hipertensão Pulmonar/tratamento farmacológico , Hipertensão Pulmonar/etiologia , Hipertensão Pulmonar/fisiopatologia , Hipertrofia Ventricular Direita/etiologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Masculino , MicroRNAs/antagonistas & inibidores , MicroRNAs/biossíntese , MicroRNAs/genética , MicroRNAs/fisiologia , Dados de Sequência Molecular , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Proteína Smad3/antagonistas & inibidores , Proteína Smad3/fisiologia , Transfecção , Remodelação Vascular/fisiologia
17.
Sheng Li Xue Bao ; 55(2): 187-90, 2003 Apr 25.
Artigo em Zh | MEDLINE | ID: mdl-12715109

RESUMO

The purpose of this study was to investigate the vasorelaxing effect of vasonatrin peptide (VNP) on human intramammary artery (HIMA).The vasorelaxing effect of VNP on HIMA was measured by means of perfusion in vitro. The effects of HS-142-1, TEA, 8-Br-cGMP and methylene blue (MB) were also observed. It was found that VNP caused a concentration-dependent relaxation in HIMA which was independent of the endothelium. 8-Br-cGMP (0.1-1000 micromol/L) also caused a concentration-dependent relaxation in HIMA. The vasorelaxing effect of VNP disappeared in the presence of HS-142-1 (20 micromol/L), an antagonist of the natriuretic peptide guanylate cyclase (GC) receptor. MB (10 micromol/L), an inhibitor of GC, not only blocked completely the relaxation of HIMA, but also enhanced the vascular contraction induced by norepinephrine. TEA (1 mmol/L), an antagonist of calcium activated potassium channels (K(Ca)), reduced but not completely blocked the vasorelaxing effect of VNP. These findings suggest that VNP can relax HIMA, which is independent of the endothelium. This effect is possibly achieved by the binding of VNP with the natriuretic peptide GC receptors in the smooth muscle cells (SMCs), leading to an increase in intracellular cGMP level. Moreover, the vasorelaxing effect of VNP is associated with K(Ca).


Assuntos
Fator Natriurético Atrial/farmacologia , Artéria Torácica Interna/fisiologia , Vasodilatação/fisiologia , Idoso , Relação Dose-Resposta a Droga , Humanos , Técnicas In Vitro , Artéria Torácica Interna/efeitos dos fármacos , Pessoa de Meia-Idade , Canais de Potássio Cálcio-Ativados/metabolismo , Receptores Acoplados a Guanilato Ciclase/metabolismo , Vasodilatação/efeitos dos fármacos
18.
PLoS One ; 8(2): e56774, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23437233

RESUMO

We previously showed that tanshinone IIA ameliorated the hypoxia-induced pulmonary hypertension (HPH) partially by attenuating pulmonary artery remodeling. The hypoxia-induced proliferation of pulmonary artery smooth muscle cells (PASMCs) is one of the major causes for pulmonary arterial remodeling, therefore the present study was performed to explore the effects and underlying mechanism of tanshinone IIA on the hypoxia-induced PASMCs proliferation. PASMCs were isolated from male Sprague-Dawley rats and cultured in normoxic (21%) or hypoxic (3%) condition. Cell proliferation was measured with 3 - (4, 5 - dimethylthiazal - 2 - yl) - 2, 5 - diphenyltetrazoliumbromide assay and cell counting. Cell cycle was measured with flow cytometry. The expression of of p27, Skp-2 and the phosphorylation of Akt were measured using western blot and/or RT-PCR respectively. The results showed that tanshinone IIA significantly inhibited the hypoxia-induced PASMCs proliferation in a concentration-dependent manner and arrested the cells in G1/G0-phase. Tanshinone IIA reversed the hypoxia-induced reduction of p27 protein, a cyclin-dependent kinase inhibitor, in PASMCs by slowing down its degradation. Knockdown of p27 with specific siRNA abolished the anti-proliferation of tanshinone IIA. Moreover, tanshinone IIA inhibited the hypoxia-induced increase of S-phase kinase-associated protein 2 (Skp2) and the phosphorylation of Akt, both of which are involved in the degradation of p27 protein. In vivo tanshinone IIA significantly upregulated the hypoxia-induced p27 protein reduction and downregulated the hypoxia-induced Skp2 increase in pulmonary arteries in HPH rats. Therefore, we propose that the inhibition of tanshinone IIA on hypoxia-induce PASMCs proliferation may be due to arresting the cells in G1/G0-phase by slowing down the hypoxia-induced degradation of p27 via Akt/Skp2-associated pathway. The novel information partially explained the anti-remodeling property of tanshinone IIA on pulmonary artery in HPH.


Assuntos
Abietanos/farmacologia , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/metabolismo , Proteínas Quinases Associadas a Fase S/metabolismo , Animais , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Hipóxia Celular , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p27/genética , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/metabolismo , Masculino , Modelos Biológicos , Fosforilação/efeitos dos fármacos , Proteólise , Interferência de RNA , Ratos , Transdução de Sinais/efeitos dos fármacos
19.
Oncol Rep ; 30(4): 1753-61, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23863999

RESUMO

Chemokines and chemokine receptor 4 (CXCR4) play an important role in metastasis. CXCR4 is also expressed in the human osteosarcoma cell line 9607-F5M2 (F5M2), which has a high tumorigenic ability and potential for spontaneous pulmonary metastasis. Mesenchymal stem cells (MSCs) contribute to the formation of the tumor stroma and promote metastasis. However, mechanisms underlying the promotion of osteosarcoma growth and pulmonary metastasis by MSCs are still elusive. Our study co-injected the human MSCs and F5M2 cells into the caudal vein of nude mice. The total number of tumor nodules per lung was significantly increased in the F5M2+MSC group compared to the other groups (control, F5M2 cells alone and MSCs alone) at week six. Moreover, a high number of Dil-labeled MSCs was present also at the osteosarcoma metastasis sites in the lung. Using Transwell assays, we found that F5M2 cells migrate towards MSCs, while the CXCR4 inhibitor AMD3100 decreased the migration potential of F5M2 cells towards MSCs. Furthermore, upon treatment with F5M2-conditioned medium, MSCs expressed and secreted higher levels of VEGF as determined by immunohistochemistry, western blotting and ELISA, respectively. Importantly, co-cultured with F5M2 cells, MSCs expressed and secreted higher VEGF levels, while AMD3100 dramatically decreased the VEGF secretion by MSCs. However, CXCR4 expression on F5M2 cells was not significantly increased in the co-culture system. Additionally, VEGF increased the proliferation of both MSCs and F5M2 cells. These findings suggest that CXCR4-mediated osteosarcoma growth and pulmonary metastasis are promoted by MSCs through VEGF.


Assuntos
Neoplasias Pulmonares/secundário , Células-Tronco Mesenquimais/metabolismo , Osteossarcoma/metabolismo , Receptores CXCR4/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Benzilaminas , Neoplasias Ósseas/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Técnicas de Cocultura , Meios de Cultivo Condicionados/metabolismo , Ciclamos , Compostos Heterocíclicos/farmacologia , Xenoenxertos , Humanos , Neoplasias Pulmonares/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Transplante de Neoplasias , Receptores CXCR4/antagonistas & inibidores
20.
Adv Pharmacol ; 59: 93-134, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20933200

RESUMO

Cardiac K(+) channels are cardiomyocyte membrane proteins that regulate K(+) ion flow across the cell membrane on the electrochemical gradient and determine the resting membrane potential and the cardiac action potential morphology and duration. Several K(+) channels have been well studied in the human heart. They include the transient outward K(+) current I(to1), the ultra-rapidly activating delayed rectifier current I(Kur), the rapidly and slowly activating delayed rectifier currents I(Kr) and I(Ks), the inward rectifier K(+) current I(K1), and ligand-gated K(+) channels, including adenosine-5'-triphosphate (ATP)-sensitive K(+) current (I(KATP)) and acetylcholine-activated current (I(KACh)). Regional differences of K(+) channel expression contribute to the variable morphologies and durations of cardiac action potentials from sinus node and atrial to ventricular myocytes, and different ventricular layers from endocardium and midmyocardium to epicardium. They also show different responses to endogenous regulators and/or pharmacological agents. K(+) channels are well-known targets for developing novel anti-arrhythmic drugs that can effectively prevent/inhibit cardiac arrhythmias. Especially, atrial-specific K(+) channel currents (I(Kur) and I(KACh)) are the targets for developing atrial-selective anti-atrial fibrillation drugs, which has been greatly progressed in recent years. This chapter concentrates on recent advances in intracellular signaling regulation and pharmacology of cardiac K(+) channels under physiological and pathophysiological conditions.


Assuntos
Antiarrítmicos , Arritmias Cardíacas/tratamento farmacológico , Ativação do Canal Iônico , Canais de Potássio , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/fisiologia , Animais , Antiarrítmicos/química , Antiarrítmicos/farmacologia , Antiarrítmicos/uso terapêutico , Arritmias Cardíacas/metabolismo , Arritmias Cardíacas/fisiopatologia , Canais de Potássio de Retificação Tardia/efeitos dos fármacos , Canais de Potássio de Retificação Tardia/fisiologia , Descoberta de Drogas , Átrios do Coração/efeitos dos fármacos , Átrios do Coração/metabolismo , Ventrículos do Coração/efeitos dos fármacos , Ventrículos do Coração/metabolismo , Humanos , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/fisiologia , Canais KATP/fisiologia , Especificidade de Órgãos , Bloqueadores dos Canais de Potássio/química , Bloqueadores dos Canais de Potássio/farmacologia , Bloqueadores dos Canais de Potássio/uso terapêutico , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização/efeitos dos fármacos , Canais de Potássio Corretores do Fluxo de Internalização/fisiologia , Canais de Potássio de Domínios Poros em Tandem/efeitos dos fármacos , Canais de Potássio de Domínios Poros em Tandem/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/efeitos dos fármacos , Canais de Potássio de Abertura Dependente da Tensão da Membrana/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA