Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Sheng Li Xue Bao ; 65(3): 269-75, 2013 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-23788183

RESUMO

The present study was designed to investigate the role of opioid receptors in the vasorelaxation effect of chronic intermittent hypobaric hypoxia (CIHH) in thoracic aorta rings and the underlying mechanism in rats. Adult male Sprague-Dawley (SD) rats were randomly divided into 2 groups: CIHH treatment group and control group. The rats in CIHH group were exposed to hypoxia in a hypobaric chamber (simulated 5 000 m altitude) for 28 days, 6 h per day. The rats in control group were kept in the same environment as CIHH rats except no hypoxia exposure. The relaxation of thoracic aorta rings was recorded by organ bath perfusion technique, and expression of opioid receptors was measured by Western blot. Results are shown as follows. (1) The acetylcholine (ACh)-induced endothelium-dependent relaxation of thoracic aorta in CIHH rats was increased obviously in a concentration-dependent manner compared with that in control rats (P < 0.05). (2) This enhancement of ACh-induced relaxation in CIHH rats was abolished by naloxone, a non-specific opioid receptor blocker (P < 0.05). (3) The expressions of δ, µ and κ opioid receptors in thoracic aorta of CIHH rats were up-regulated compared with those in control rats (P < 0.05). (4) The enhancement of CIHH on relaxation of thoracic aorta was reversed by glibenclamide, an ATP-sensitive potassium channel (KATP) blocker (P < 0.05). The results suggest that opioid receptors are involved in CIHH-enhanced ACh-induced vasorelaxation of thoracic aorta through KATP channel pathways.


Assuntos
Acetilcolina/farmacologia , Aorta Torácica/efeitos dos fármacos , Hipóxia/fisiopatologia , Receptores Opioides/metabolismo , Altitude , Animais , Glibureto/farmacologia , Canais KATP/antagonistas & inibidores , Masculino , Ratos , Ratos Sprague-Dawley , Vasodilatação
2.
Biochem Biophys Res Commun ; 427(1): 73-9, 2012 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-22975349

RESUMO

Cyclic adenosine 3',5'-monophosphate (cAMP), which is synthesized by adenylyl cyclase (AC) and degraded by phosphodiesterase (PDE), plays crucial roles in the regulation of multiple cellular functions and physiological processes. Prolyl hydroxylase domain (PHD) proteins, which belong to a family of dioxygenases that function as oxygen sensors through their hydroxylation activity, have been implicated in multiple signaling pathways. Here, we aimed to determine whether PHD played a role in regulating intracellular cAMP level in cardiomyocytes. Through the overexpression/knockdown of the PHD gene and the measurement of the cAMP content, we found that PHD2, but not PHD1 or PHD3, acts as a regulator of intracellular cAMP. In neonatal rat cardiomyocytes and H9c2 cells, the overexpression of PHD2 increased the intracellular cAMP level, whereas the PHD2 knockdown reduced it. There was no alteration in the AC expression or activity in cells that overexpressed or downregulated PHD2. The overexpression of PHD2 decreased both the protein expression and the activity of phosphodiesterase 4D (PDE4D), whereas the PHD2 knockdown increased the PDE4D expression and activity. Co-immunoprecipitation experiments revealed a direct binding between PHD2 and PDE4D and liquid chromatography-tandem mass spectrometry analyses identified the specific hydroxylation sites on PDE4D. In conclusion, PHD2 may directly interact with PDE4D to function as a novel regulator of the intracellular cAMP levels in cardiomyocytes.


Assuntos
AMP Cíclico/metabolismo , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/metabolismo , Miócitos Cardíacos/metabolismo , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Animais , Linhagem Celular , Regulação para Baixo , Humanos , Prolina Dioxigenases do Fator Induzível por Hipóxia , Miócitos Cardíacos/enzimologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Ratos
3.
Blood Cells Mol Dis ; 49(2): 67-73, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22595196

RESUMO

High altitude sickness (HAS) occurs among humans visiting or inhabiting high altitude environments. Genetic differences in the EPAS1 and EGLN1 genes have been found between lowland (Han) and highland (Tibetan) Chinese. Three SNPs within EPAS1 and EGLN1 were evaluated in Han and Tibetan patients with acute mountain sickness (AMS) and chronic mountain sickness (CMS). We compared 85 patients with AMS to 79 Han unaffected with mountain sickness (MS) as well as 45 CMS patients to 34 unaffected Tibetan subjects. The three SNPs studied were EPAS1 [ch2: 46441523 (hg18], EGLN1 (rs480902) and (rs516651). Direct sequencing was used to identify individual genotypes for the three SNPs. Age was found to be significantly associated with the EPAS1 SNP in the CMS patients while heart rate (HR) and oxygen saturation level of hemoglobin (SaO(2)) were found to be significantly associated with the EGLN1 (rs480902) SNP in the Han patients with AMS. The individuals with CMS were found to diverge significantly for the EPAS1 SNP compared to their Tibetan control group as measured by genetic distance (0.123) indicating positive selection of the EPAS-G allele with age and illness. The EGLN1 (rs480902) SNP had a significant correlation with hematocrit (HCT), HR and SaO(2) in AMS patients. AMS and CMS were found to be significantly associated with the EPAS1 and EGLN1 SNPs compared to their Han and Tibetan control groups, respectively, indicating these nucleotide alterations have a physiological effect for the development of high altitude sickness.


Assuntos
Doença da Altitude/genética , Povo Asiático , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Polimorfismo de Nucleotídeo Único , Pró-Colágeno-Prolina Dioxigenase/genética , Doença Aguda , Adulto , Fatores Etários , Alelos , Altitude , Doença da Altitude/etnologia , China/epidemiologia , Feminino , Genótipo , Frequência Cardíaca , Hemoglobinas/metabolismo , Humanos , Prolina Dioxigenases do Fator Induzível por Hipóxia , Masculino , Pessoa de Meia-Idade , Oxigênio/metabolismo , Análise de Sequência de DNA
4.
Exp Physiol ; 97(10): 1105-18, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22562809

RESUMO

Intermittent hypobaric hypoxia (IHH) is an effective protective strategy against myocardial ischaemia-reperfusion (I/R) injury, but the precise mechanisms are far from clear. To understand the overall effects of IHH on the myocardial proteins during I/R, we analysed functional performance and the protein expression profile in isolated hearts from normoxic rats and from rats adapted to IHH (5000 m, 4 h day(-1), 4 weeks) following I/R injury (30 min/45 min). Intermittent hypobaric hypoxia significantly improved the postischaemic recovery of left ventricular function compared with the recovery in time-matched normoxic control hearts. Two-dimensional electrophoresis with matrix-assisted laser desorption/ionization and time-of-flight mass spectrometric analysis was then used to assess protein alterations in left ventricles from normoxic and IHH groups, with or without I/R. The expressions of 16 proteins changed by over fivefold; nine of these proteins are involved in energy metabolism. Immunoblot and real-time PCR analysis confirmed the IHH-increased expressions of the ATP synthase subunit ß, mitochondrial aldehyde dehydrogenase and heat shock protein 27 in left ventricles. Furthermore, IHH significantly attenuated the reduction of myocardial ATP content, mitochondrial ATP synthase activity, membrane potential and respiratory control ratios due to I/R. In addition, inhibition of mitochondrial ATP synthase by oligomycin (1 µmol l(-1)) abolished the IHH-induced improvements in three parameters: postischaemic recovery of left ventricular function, mitochondrial membrane potential and respiratory control ratios. These results suggest that an improvement in mitochondrial energy metabolism makes an important contribution to the cardioprotection afforded by IHH against postischaemic myocardial dysfunction.


Assuntos
Hipóxia/metabolismo , Mitocôndrias Cardíacas/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Trifosfato de Adenosina/metabolismo , Aldeído Desidrogenase/metabolismo , Animais , Metabolismo Energético , Proteínas de Choque Térmico HSP27/metabolismo , Ventrículos do Coração/metabolismo , Masculino , Potencial da Membrana Mitocondrial , ATPases Mitocondriais Próton-Translocadoras/metabolismo , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Proteoma/metabolismo , Ratos , Ratos Sprague-Dawley , Função Ventricular Esquerda/fisiologia
5.
Eur J Appl Physiol ; 112(3): 1037-46, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21735218

RESUMO

Intermittent hypoxia (IH) markedly enhances cardiac tolerance against ischemia/reperfusion injury, but its mechanism and molecular basis remain unclear. For exploring the expression of mitochondrial proteins induced by IH, two-dimensional electrophoresis and Thermo Finnigan LTQ mass spectrometer (MS) were applied. After comparing the protein profiles of myocardial mitochondria between IH and normoxic hearts, 14 protein spots were found to be altered more than threefold between the two groups, 11 of which were identified by Finnigan LTQ MS. Among these 11 proteins, 9 were involved in energy metabolism, including 7 that were increased after IH. The latter were identified as aldehyde dehydrogenase, methylmalonate-semialdehyde dehydrogenase, ATP synthase ß chain, mitochondrial aconitase, malate dehydrogenase, electron transfer flavoprotein α subunit and sirtuin 5. Two other proteins, ubiquinol-cytochrome C reductase iron-sulfur subunit and aspartate aminotransferase, were decreased after IH. Biochemical tests for energy metabolism in mitochondria supported the proteomic results. IH exposure also increased the expression of a molecular chaperone-heat shock protein 60 and an antioxidant protein, peroxiredoxin 5. These findings will provide clues for understanding the mechanism of IH-induced cardiac protection and may lead to the development of interventional strategies designed to utilize the advantages of IH clinically.


Assuntos
Hipóxia/metabolismo , Proteínas Mitocondriais/análise , Miócitos Cardíacos/química , Proteômica , Animais , Eletroforese em Gel Bidimensional , Hipóxia/patologia , Masculino , Espectrometria de Massas , Mitocôndrias Cardíacas/química , Mitocôndrias Cardíacas/metabolismo , Mitocôndrias Cardíacas/patologia , Proteínas Mitocondriais/metabolismo , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Periodicidade , Proteoma/análise , Proteômica/métodos , Ratos , Ratos Sprague-Dawley
6.
Chin J Physiol ; 55(1): 62-70, 2012 Feb 29.
Artigo em Inglês | MEDLINE | ID: mdl-22242956

RESUMO

Our previous study showed that chronic intermittent hypobaric hypoxia (CIHH) could prevent decreases in systemic arterial blood pressure (SABP) during acute hypoxia. However, the mechanism was not clear. The purpose of the present study was to observe whether the carotid sinus baroreflex (CSB) was involved in the antagonizing effect of CIHH on SABP decrease induced by acute hypoxia and to explore the underlying mechanism using perfusion technique in rat isolated carotid sinus area. After 14-day and 28-day CIHH exposure, the CSB in rats was enhanced markedly, manifesting as increases in PS and RD, and decreases in TP and SP. This facilitation of CSB was partly abolished by Glibenclamide (Gli, 10 µM), a K ATP channel blocker, but was not influenced by L-NAME (100 µM), a nitric oxide synthase (NOS) inhibitor. The results of the study suggested that CIHH facilitated CSB through opening the K ATP channels in carotid sinus of anesthetic rats and might be one of mechanisms of CIHH keeping SABP homeostasis during acute hypoxia.


Assuntos
Barorreflexo , Seio Carotídeo/fisiopatologia , Hipóxia/fisiopatologia , Anestesia , Animais , Gasometria , Pressão Sanguínea , Peso Corporal , Glibureto , Hipoglicemiantes , Hipóxia/patologia , Masculino , Miocárdio/patologia , NG-Nitroarginina Metil Éster , Tamanho do Órgão , Perfusão , Ratos , Ratos Sprague-Dawley
7.
J Cell Mol Med ; 15(1): 134-40, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19863695

RESUMO

Abnormal activation of mitochondrial translocator protein (TSPO) contributes to arrhythmogenesis during cardiac metabolic compromise; however, its role in the antiarrhythmic activities of chronic hypoxia adaptation remains unclear. Our results demonstrated that 80% of normoxic rats developed ischaemic VF, whereas this condition was seldom observed in rats with 14 days of chronic intermittent hypobaric hypoxia (CIHH). TSPO stimulation or inhibition affected the arrhythmias incidence in normoxic rats, but did not change the CIHH-mediated antiarrhythmic effects. Abrupt and excessive elevation of TSPO activity was positively linked to ischaemic VF, and CIHH preserved TSPO activity during ischaemia. The preservation of TSPO activity by CIHH also contributed to the maintenance of intracellular Ca homeostasis. These results suggest that the blunt sensitivity of TSPO to ischaemic stress may be responsible for the antiarrhythmic effects by CIHH.


Assuntos
Antiarrítmicos/farmacologia , Arritmias Cardíacas/metabolismo , Proteínas de Transporte/metabolismo , Hipóxia/metabolismo , Isquemia/prevenção & controle , Mitocôndrias/metabolismo , Receptores de GABA-A/metabolismo , Animais , Arritmias Cardíacas/tratamento farmacológico , Arritmias Cardíacas/patologia , Western Blotting , Cálcio/metabolismo , Doença Crônica , Modelos Animais de Doenças , Isquemia/patologia , Masculino , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa
8.
J Cell Mol Med ; 15(12): 2712-22, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21255264

RESUMO

Adrenergic receptor (AR)-mediated signalling is modulated by oxygen levels. Prolyl hydroxylases (PHDs) are crucial for intracellular oxygen sensing and organism survival. However, it remains to be clarified whether or how PHDs are involved in the regulation of ß(2) -adrenoceptor (ß(2) -AR) signalling. Here we show that PHD2 can modulate the rate of ß(2) -AR internalization through interactions with ß-arrestin 2. PHD2 hydroxylates ß-arrestin 2 at the proline (Pro)(176), Pro(179) and Pro(181) sites, which retards the recruitment of ß-arrestin 2 to the plasma membrane and inhibits subsequent co-internalization with ß(2) -AR into the cytosol. ß(2) -AR internalization is critical to control the temporal and spatial aspects of ß(2) -AR signalling. Identifying novel regulators of ß(2) -AR internalization will enable us to develop new strategies to manipulate receptor signalling and provide potential targets for drug development in the prevention and treatment of diseases associated with ß(2) -AR signalling dysregulation.


Assuntos
Arrestinas/metabolismo , Membrana Celular/metabolismo , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Células Cultivadas , Citosol/metabolismo , Endocitose , Humanos , Prolina Dioxigenases do Fator Induzível por Hipóxia , Imunoprecipitação , Rim/citologia , Rim/metabolismo , Fosforilação , Pró-Colágeno-Prolina Dioxigenase/antagonistas & inibidores , Pró-Colágeno-Prolina Dioxigenase/genética , RNA Interferente Pequeno/genética , Transdução de Sinais , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , beta-Arrestina 2 , beta-Arrestinas
9.
Am J Physiol Heart Circ Physiol ; 301(4): H1695-705, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21821784

RESUMO

Intermittent hypobaric hypoxia (IHH) protects hearts against ischemia-reperfusion (I/R) injury, but the underlying mechanisms are far from clear. ROS are paradoxically regarded as a major cause of myocardial I/R injury and a trigger of cardioprotection. In the present study, we investigated whether the ROS generated during early reperfusion contribute to IHH-induced cardioprotection. Using isolated perfused rat hearts, we found that IHH significantly improved the postischemic recovery of left ventricular (LV) contractile function with a concurrent reduction of lactate dehydrogenase release and myocardial infarct size (20.5 ± 5.3% in IHH vs. 42.1 ± 3.8% in the normoxic control, P < 0.01) after I/R. Meanwhile, IHH enhanced the production of protein carbonyls and malondialdehyde, respective products of protein oxidation and lipid peroxidation, in the reperfused myocardium and ROS generation in reperfused cardiomyocytes. Such effects were blocked by the mitochondrial ATP-sensitive K(+) channel inhibitor 5-hydroxydecanoate. Moreover, the IHH-improved postischemic LV performance, enhanced phosphorylation of PKB (Akt), PKC-ε, and glycogen synthase kinase-3ß, as well as translocation of PKC-ε were not affected by applying H(2)O(2) (20 µmol/l) during early reperfusion but were abolished by the ROS scavengers N-(2-mercaptopropionyl)glycine (MPG) and manganese (III) tetrakis (1-methyl-4-pyridyl)porphyrin. Furthermore, IHH-reduced lactate dehydrogenase release and infarct size were reversed by MPG. Consistently, inhibition of Akt with wortmannin and PKC-ε with εV1-2 abrogated the IHH-improved postischemic LV performance. These findings suggest that IHH-induced cardioprotection depends on elevated ROS production during early reperfusion.


Assuntos
Hipóxia/fisiopatologia , Contração Miocárdica/fisiologia , Isquemia Miocárdica/fisiopatologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Reperfusão Miocárdica , Transdução de Sinais/fisiologia , Pressão do Ar , Animais , Antioxidantes/farmacologia , Sobrevivência Celular/fisiologia , Cistina/análogos & derivados , Cistina/farmacologia , Ativação Enzimática/fisiologia , Canais KATP/metabolismo , L-Lactato Desidrogenase/metabolismo , Peroxidação de Lipídeos , Masculino , Mitocôndrias Cardíacas/metabolismo , Mitocôndrias Cardíacas/fisiologia , Infarto do Miocárdio/patologia , Miócitos Cardíacos/fisiologia , Proteína Oncogênica v-akt/metabolismo , Proteína Oncogênica v-akt/fisiologia , Oxirredução , Proteína Quinase C-épsilon/metabolismo , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Recuperação de Função Fisiológica
10.
Basic Res Cardiol ; 106(3): 329-42, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21298517

RESUMO

Intermittent hypobaric hypoxia (IHH) preconditioning protects the heart against ischemic injuries. However, little is known about the therapeutic effect of IHH on myocardial infarction (MI). The aim of this study was to test whether IHH treatment influences infarct size and cardiac performance after MI. Seven days after sham operation or left anterior descending coronary artery ligation, male Sprague-Dawley rats were randomly exposed to normoxia or one 6-h period each day of IHH (5,000 m) for 14 and 28 days. Echocardiography analysis showed that IHH significantly reduced left ventricular (LV) dilation and improved cardiac performance after 14- or 28-day treatment compared with MI-normoxic groups. The improvement of LV function was further confirmed in isolated perfused MI-IHH hearts. Such protection was associated with attenuated infarct size, myocardial fibrosis, and apoptotic cardiomyocytes. IHH treatment also enhanced coronary flow and phosphorylation of heat shock protein 27 in both sham and MI groups compared with the control groups. In addition, IHH increased the capillary density and vascular endothelial growth factor expression in peri-infarcted zones compared with sham-IHH and MI-normoxic groups. Our data demonstrated for the first time that IHH treatment exerts a therapeutic effect on MI by attenuating progressive myocardial remodeling and improving myocardial contractility. IHH treatment might provide a unique and promising therapeutic approach for ischemic heart diseases.


Assuntos
Hipóxia/complicações , Hipóxia/fisiopatologia , Precondicionamento Isquêmico Miocárdico/métodos , Infarto do Miocárdio/prevenção & controle , Infarto do Miocárdio/fisiopatologia , Animais , Apoptose/fisiologia , Western Blotting , Marcação In Situ das Extremidades Cortadas , Masculino , Infarto do Miocárdio/patologia , Ratos , Ratos Sprague-Dawley , Remodelação Ventricular
11.
Sheng Li Xue Bao ; 63(3): 205-10, 2011 Jun 25.
Artigo em Zh | MEDLINE | ID: mdl-21681337

RESUMO

The present study is aimed to investigate the effect of chronic intermittent hypobaric hypoxia (CIHH) on contractile activities in isolated thoracic aorta and pulmonary artery rings and the underlying mechanism in rats. Sprague-Dawley (SD) rats were randomly divided into 4 groups: control group (CON), 14 days CIHH treatment group (CIHH14), 28 days CIHH treatment group (CIHH28) and 42 days CIHH treatment group (CIHH42). CIHH rats were exposed to hypoxia in a hypobaric chamber simulating 5 000 m altitude, 6 h daily for 14, 28 and 42 d, respectively. After artery rings were prepared from pulmonary artery and thoracic aorta, the contractile activity of the artery rings was recorded using organ bath technique. Results are shown as follows. (1) There were no significant differences of noradrenaline (NA)- and KCl-induced contractions in thoracic aorta and pulmonary artery rings among CIHH and CON rats. (2) Angiotensin Ⅱ (ANGⅡ)-induced contraction in thoracic aorta rings, not in pulmonary artery rings, of CIHH rats was decreased compared with that in CON rats. There was no significant difference of ANGⅡ-induced contraction in thoracic aorta rings among CIHH rats. (3) Inhibitory effect of CIHH on ANGⅡ-induced contraction in thoracic aorta rings was endothelium-independent, and was reversed by glibenclamide (Gli), an ATP-sensitive potassium channels (K(ATP)) blocker, and L-NAME, a NO synthase inhibitor, but not by indomethacin (Indo), a cyclooxygenase inhibitor. These results suggest that CIHH attenuates the contraction induced by ANGⅡ in thoracic aorta rings of rat, which is related to the opening of K(ATP) channel and the increased production of NO.


Assuntos
Aorta Torácica/fisiopatologia , Hipóxia/fisiopatologia , Músculo Liso Vascular/fisiopatologia , Artéria Pulmonar/fisiopatologia , Vasoconstrição/fisiologia , Angiotensina II/farmacologia , Animais , Canais KATP/metabolismo , Masculino , Contração Muscular/fisiologia , Óxido Nítrico/biossíntese , Ratos , Ratos Sprague-Dawley
12.
J Physiol ; 588(Pt 16): 2987-98, 2010 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-20547682

RESUMO

beta-Adrenoceptors (beta-ARs) play a critical role in the regulation of cardiovascular function. Intracellular oxygen homeostasis is crucial for the survival of cardiomyocytes. However, it is still unclear whether beta-AR activation can modulate intracellular oxygen. Here we used mitochondrial and cytosolic target Renilla luciferase to detect intracellular oxygen concentration. Pharmacological experiments revealed that beta2-AR activation specifically regulates intracellular oxygen in cardiomyocytes and COS7 cells. This effect was abrogated by inhibitory G protein (Gi) inhibition, endothelial nitric oxide synthase (eNOS) blockade, and NO scavenging, implicating that the beta2-AR-Gi-eNOS pathway is involved in this regulation. beta2-AR activation increased the AMP/ATP ratio, AMPK activity, ROS production and prolyl hydroxylase activity. These effects also contribute to the regulation of beta2-AR signalling, thus providing an additional layer of complexity to enforce the specificity of beta1-AR and beta2-AR signalling. Collectively, the study provides novel insight into the modulation of oxygen homeostasis, broadens the scope of beta2-AR function, and may have crucial implications for beta2-AR signalling regulation.


Assuntos
Miócitos Cardíacos/metabolismo , Oxigênio/metabolismo , Receptores Adrenérgicos beta 1/metabolismo , Receptores Adrenérgicos beta 2/metabolismo , Transdução de Sinais , Proteínas Quinases Ativadas por AMP/metabolismo , Monofosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Agonistas de Receptores Adrenérgicos beta 1/farmacologia , Agonistas de Receptores Adrenérgicos beta 2/farmacologia , Animais , Animais Recém-Nascidos , Células COS , Chlorocebus aethiops , Citosol/metabolismo , Inibidores Enzimáticos/farmacologia , Subunidades alfa Gi-Go de Proteínas de Ligação ao GTP/metabolismo , Homeostase , Luciferases de Renilla/biossíntese , Luciferases de Renilla/genética , Mitocôndrias Cardíacas/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/antagonistas & inibidores , Óxido Nítrico Sintase Tipo III/metabolismo , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Receptores Adrenérgicos beta 1/efeitos dos fármacos , Receptores Adrenérgicos beta 2/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Transfecção
13.
Am J Physiol Heart Circ Physiol ; 298(4): H1267-72, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20097768

RESUMO

Chronic intermittent hypobaric hypoxia (CIHH) has an effective cardiac protection against ischemia-reperfusion injury. However, the underlying mechanisms are not fully known. It has been shown that blockade of beta-adrenergic receptor exerts anti-arrhythmic action and improves cardiac remodeling in ischemic myocardium. Thus we determined the influence of CIHH on beta-adrenergic receptor activity in right ventricular papillary muscle of rats. We found that the action potential duration in right ventricular papillary muscle was significantly longer in CIHH rats than in control rats. Activation of beta-adrenergic receptor with dl-isoproterenol dose-dependently increased action potential duration and the contractility in right ventricular papillary muscle. In CIHH rats, the prolonged effect of dl-isoproterenol on action potential duration and the positive inotropic effect were significantly decreased compared with that in control rats. Furthermore, radioligand-binding experiments revealed that the density and affinity of beta-adrenergic receptor in right ventricular myocardium was significantly lower in CIHH rats. In addition, Western blot analysis revealed that the membrane-bound G protein G(s)alpha expression level in cardiac myocardium was significantly lower in CIHH rats than that in control rats. Collectively, these data suggest that CIHH suppresses beta-adrenergic receptor action in right ventricular papillary muscle through decreasing receptor density and affinity, as well as membrane-bound G(s)alpha. This mechanism may be involved in the cardiac protective effect of CIHH.


Assuntos
Ventrículos do Coração/metabolismo , Hipóxia/metabolismo , Músculos Papilares/metabolismo , Receptores Adrenérgicos beta/metabolismo , Potenciais de Ação/efeitos dos fármacos , Agonistas Adrenérgicos beta/farmacologia , Animais , Peso Corporal/efeitos dos fármacos , Relação Dose-Resposta a Droga , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Isoproterenol/farmacologia , Masculino , Modelos Animais , Contração Muscular/efeitos dos fármacos , Tamanho do Órgão/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley
14.
Biochem Biophys Res Commun ; 401(2): 231-7, 2010 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-20849813

RESUMO

Prolyl hydroxylases (PHDs) are dioxygenases that use oxygen as a co-substrate to hydroxylate proline residues. Three PHD isoforms (PHD1, PHD2 and PHD3) have been identified in mammalian cells. PHD3 expression is upregulated in some cardiac diseases such as cardiomyopathy, myocardial ischemia-reperfusion injury and congestive heart failure, all of which are associated with apoptosis. However, the role of PHDs in cardiomyocyte apoptosis remains unknown. Here, we have found that exposure of embryonic rat heart-derived H9c2 cells to doxorubicin (DOX) induced cell apoptosis as evaluated by caspase-3/7 activity, mitochondrial membrane potential (Δψm) and cell viability, and that this apoptosis was linked to PHD3 upregulation. PHD inhibition or PHD3 silencing substantially ameliorated DOX-induced apoptosis, but PHD1 or PHD2 knockdown did not significantly influence apoptosis. Furthermore, immunoprecipitation experiments showed that PHD3 upregulation reduced the formation of the Bax-Bcl-2 complex, inhibiting the anti-apoptotic effect of Bcl-2. Thus, PHD3 upregulation may be partially responsible for DOX-induced cardiomyocyte apoptosis via its interaction with Bcl-2. Inhibition of PHD3 is likely to be cardioprotective against apoptosis in some heart disorders.


Assuntos
Apoptose , Miócitos Cardíacos/fisiologia , Pró-Colágeno-Prolina Dioxigenase/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Animais , Linhagem Celular , Doxorrubicina/farmacologia , Humanos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-bcl-2/genética , Ratos
15.
Pharmacol Res ; 60(1): 61-7, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19427588

RESUMO

The cardioprotective effects of estrogen remain controversial in clinical practice. Previous reports have shown that cardioprotective mechanisms converge on the mitochondria, but the role of mitochondria in estrogen's actions on cardiac arrhythmias is unclear. Here, we report that stimulation or inhibition of mitochondrial benzodiazepine receptors (mBzR) affected ventricular fibrillation (VF) almost in an "all-or-none" manner in an in vitro rat heart model of ischemic VF. Low concentrations of estrogen did not provide antiarrhythmic effects; however, the combination of mBzR activator and estrogen reduced VF incidence in hearts from either gender. Such synergistic actions also enabled cardiomyocytes to resist metabolic stress-induced intracellular [Ca(2+)](i) overload. Ligand binding experiments revealed that estrogen itself did not affect mBzR activity under basal conditions but promoted its up-regulation under myocardial ischemia. Our results suggest that mBzR may be an important molecule for ischemic arrhythmia and may act as a molecular switch for estrogen's antiarrhythmic effects. This finding provides a clue for elucidating the conflicting results regarding estrogen's cardiac effects in clinical studies and also suggests potential new strategies for hormone treatment in the female population.


Assuntos
Cardiotônicos/farmacologia , Proteínas de Transporte/metabolismo , Estradiol/farmacologia , Estrogênios/farmacologia , Mitocôndrias Cardíacas/efeitos dos fármacos , Isquemia Miocárdica/complicações , Receptores de GABA-A/metabolismo , Fibrilação Ventricular/prevenção & controle , Animais , Cálcio/metabolismo , Proteínas de Transporte/biossíntese , Modelos Animais de Doenças , Feminino , Técnicas In Vitro , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias Cardíacas/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Perfusão , Ratos , Ratos Sprague-Dawley , Receptores de GABA-A/biossíntese , Fibrilação Ventricular/etiologia , Fibrilação Ventricular/metabolismo
16.
Sheng Li Xue Bao ; 61(1): 21-6, 2009 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-19224050

RESUMO

The purpose of the present study was to investigate the effect of chronic intermittent hypobaric hypoxia (CIHH) on α(1)-adrenergic receptors and the role of alpha(1)-adrenergic receptors in the protection of CIHH against ischemic injury of myocardium. Sixty-six adult male Sprague-Dawley rats were randomly divided into four groups: control group (Con), 14-day CIHH treatment group (CIHH14), 28-day CIHH treatment group (CIHH28) and 42-day CIHH treatment group (CIHH42). CIHH rats were exposed to hypoxia mimicking 5 000 m altitude (p(B)=404 mmHg, p(O(2))=84 mmHg) in a hypobaric chamber, 6 h daily for 14, 28 and 42 d, respectively. Control animals lived in the same environment as CIHH animals except hypoxia exposure. After anesthesia with sodium pentobarbital (3.0-3.5 mL/kg body weight, i.p.), papillary muscle was taken from the right ventricle of rat and perfused with modified Tyrode's solution continuously, at constant temperature (37 °C) and perfusion speed (12 mL/min). Muscle contraction was evoked by electric stimuli. Different concentrations (1x10(-7), 1x10(-6) and 1x10(-5) mol/L) of phenylephrine (PE), an alpha(1)-adrenergic receptor agonist, were applied cumulatively to investigate the effect of PE on the mechanic contraction of right ventricular papillary muscles of rats in Con, CIHH14, CIHH28 and CIHH42 groups. Also, prazosin (1x10(-6) mol/L), an α(1)-adrenergic receptor antagonist, was used to investigate the role of α(1)-adrenergic receptor in the protective effect of CIHH on papillary muscle. The results showed: (1) PE increased the maximal isometric tension (P(max)) and maximal velocity of tension development (P(dT/dt)) of muscle contraction in a dose-dependent manner (P<0.05), and the increase of the muscle contraction was much greater in CIHH28 and CIHH42 rats than that in Con rats (P<0.05). Under 1x10(-5) mol/L of PE, the increases of P(max) and P(dT/dt) over the baseline were 51.2% and 44.5% in CIHH28 group, 48.6% and 44.5% in CIHH42 group, and 28.7% and 24.5% in Con group, respectively; (2) The contraction of papillary muscle decreased during simulated ischemia, but the decrease was slighter in CIHH rats than that in Con rats (P<0.05). The decreases in P(max) and P(dT/dt) were 59.6% and 53.6% in CIHH28 group, 60.4% and 49.9% in CIHH42 group, and 74.4% and 64.7% in Con group, respectively; (3) The protective effect of CIHH on ischemic papillary muscle was abolished by prazosin (1x10(-6) mol/L). The results of the present study suggest that CIHH increases the activity of α(1)-adrenergic receptor, which is possibly one of the mechanisms for the cardioprotection of CIHH.


Assuntos
Ventrículos do Coração/fisiopatologia , Hipóxia/metabolismo , Miocárdio/metabolismo , Músculos Papilares/metabolismo , Receptores Adrenérgicos alfa 1/metabolismo , Altitude , Animais , Masculino , Contração Muscular , Ratos , Ratos Sprague-Dawley
17.
Sheng Li Xue Bao ; 60(6): 687-94, 2008 Dec 25.
Artigo em Inglês | MEDLINE | ID: mdl-19082422

RESUMO

The aim of this study is to investigate the effects of chronic intermittent hypobaric hypoxia (IHH) and chronic continuous hypobaric hypoxia (CHH) on hemodynamics under basic normoxia and acute hypoxia conditions and to find the difference of two types of chronic hypoxia. Forty adult male Sprague-Dawley (SD) rats were randomly divided into 5 groups: Control group (CON), 28 days IHH group (IHH28), 42 days IHH group (IHH42), 28 days CHH group (CHH28) and 42 days CHH group (CHH42). The rats in IHH groups were treated with intermittent hypoxia (11.1% O2) mimicking 5 000 m altitude in a hypobaric chamber for 28 or 42 d, 6 h a day, respectively. The rats in CHH groups lived in the hypobaric chamber with the same degree of hypoxia like IHH rats except half an hour in normoxia each day for feeding and cleaning. The body weight of rats was measured once a week. The parameters in hemodynamics, such as mean artery blood pressure (MAP), heart rate (HR), left ventricular systolic pressure (LVSP), maximum change rate of left ventricular pressure (+/-LVdP/dt(max)) were recorded under basic normoxia and acute hypoxia conditions through catheterization technique. The superoxide dismutase (SOD) activity and malondialdehyde (MDA) content in myocardium of rats were measured by biochemical method. The weights of whole heart, left and right ventricles were measured separately. The results showed: (1) The basic HR and MAP in CHH42 rats were lower than those in CON, IHH and CHH28 rats (P<0.05). (2) IHH showed a cardioprotection against acute hypoxia and reoxygenation injury, manifested as the result that the changes of HR, MAP, LVSP, and +/- LVdP/dt(max) were smaller than those in CON rats during acute hypoxia and reoxygenation. CHH showed a rather strong cardioprotection during acute hypoxia, manifested as the result that the decreases of HR, MAP, LVSP, and +/- LVdP/dt(max)were much smaller, but it did damage during reoxygenation, manifested as the result that the recovery of hemodynamics was the worst among three groups (P<0.05). (3) The antioxygenation of heart was increased in both IHH and CHH rats compared with that in CON rats manifested by the increased SOD activity and decreased MDA content (P<0.05, P<0.01). (4) IHH had no effect on heart weight, but CHH rats showed an obvious right ventricular hypertrophy compared with CON and IHH animals (P<0.01). The result indicates that IHH can induce a more effective cardioprotection with no much side effect, which might have a potential value for practical use.


Assuntos
Coração/fisiopatologia , Hemodinâmica , Hipóxia/metabolismo , Miocárdio/patologia , Altitude , Animais , Hipertrofia Ventricular Direita/patologia , Masculino , Malondialdeído/metabolismo , Miocárdio/metabolismo , Ratos , Ratos Sprague-Dawley , Superóxido Dismutase/metabolismo
18.
Sheng Li Xue Bao ; 59(5): 601-13, 2007 Oct 25.
Artigo em Zh | MEDLINE | ID: mdl-17940700

RESUMO

Intermittent hypoxia (IH), or periodic hypoxia is referred as exposure to hypoxia interrupted by normoxia that occurs under many physiological and pathophysiological conditions. A lot of researches showed that IH adaptation, like ischemic preconditioning (IPC) and long-term high-altitude hypoxic adaptation (LHA), had significant cardioprotective effects including increasing the tolerance of myocardium to ischemia/reperfusion injury, limiting infarction size and morphologic damage, inhibiting apoptosis of myocardial cells, enhancing recovery of cardiac function in ischemia/reperfusion, and antiarrhythmia. However, the precise mechanisms underlying the protective effects of IH against ischemia/reperfusion injury are far from clear. The potential candidates participating in the protective effects of IH include oxygen transport, energy metabolism, neurohumoral regulation, antioxidase, stress protein, adenosine, ATP-sensitive potassium channel, mitochondrion, calcium control, nitric oxide and protein kinase. The effects of IH are affected by the protocol of hypoxic exposure, age and sex of experimental animals. IH adaptation, for longer lasting time than IPC and lesser side effect than LHA, might have a practical value for using.


Assuntos
Hipóxia , Precondicionamento Isquêmico , Traumatismo por Reperfusão/prevenção & controle , Adaptação Fisiológica , Cálcio , Metabolismo Energético , Humanos , Canais KATP , Miocárdio , Miócitos Cardíacos , Canais de Potássio , Traumatismo por Reperfusão/fisiopatologia
19.
Sheng Li Xue Bao ; 59(5): 660-6, 2007 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-17940707

RESUMO

The aim of the present study was to explore the effects of two different modes of intermittent hypobaric hypoxia (IHH) on myocardial ischemia/reperfusion injury in developing rat hearts. Postnatal male sprague-Dawley rats (n=72) were divided randomly into 3 groups: intermittent hypoxia at 3 000 m (IHH3000) group, intermittent hypoxia at 5 000 m (IHH5000) group and control group. The isolated hearts were perfused in the Langendorff apparatus, undergoing 30 min of global ischemia and 60 min of reperfusion. Cardiac function, coronary flow and lactate dehydrogenase (LDH) activity were recorded at 5 min before ischemia and 1, 5, 10, 20, 30, 60 min during reperfusion, respectively. The heart weight was measured at the end of the experiment. The results showed that: (1) There was no difference in body weight gaining between IHH3000 and control groups. The gain of body weight in IHH5000 group was much lower than that in IHH3000 and control groups (P<0.01). (2) Compared with that in the control group, the recovery of cardiac function in IHH3000 group was enhanced at 60 min after ischemia/reperfusion, coronary flow was increased, and LDH activity was decreased (P<0.05), meaning a cardioprotective effect occurred. There was no significant difference in heart weight between IHH3000 and control groups. In addition, cardiac function restored better in IHH3000 group after 42 d of hypoxic exposure than that after 28 d of hypoxic exposure (P<0.05). (3) Compared with that in the control group, the recovery of cardiac function in IHH5000 group was lower, coronary flow was decreased, and LDH activity was increased (P<0.05). There was a hypertrophy in the right ventricle in IHH5000 group. All changes indicated definitely that a detrimental effect developed in IHH5000 group. The results suggest that proper IHH can protect developing rat hearts against ischemia/reperfusion injury while this effect could be affected by the modes of intermittent hypoxic exposure.


Assuntos
Hipóxia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Animais , Coração , Ventrículos do Coração , Masculino , Substâncias Protetoras , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA