Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Eur J Pharmacol ; 971: 176556, 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38574840

RESUMO

AIMS: Endothelial-mesenchymal transition (EndMT) is a crucial pathological process contributing to cardiac fibrosis. Bradykinin has been found to protect the heart against fibrosis. Whether bradykinin regulates EndMT has not been determined. MATERIALS AND METHODS: Rats were subjected to ligation of the left anterior descending coronary artery for 1 h and subsequent reperfusion to induce cardiac ischemia-reperfusion (IR) injury. Bradykinin (0.5 µg/h) was infused by an osmotic pump implanted subcutaneously at the onset of reperfusion. Fourteen days later, the functional, histological, and molecular analyses were performed to investigate the changes in cardiac fibrosis and EndMT. Human coronary artery endothelial cells were utilized to determine the molecular mechanisms in vitro. RESULTS: Bradykinin treatment improved cardiac function and decreased fibrosis following cardiac IR injury, accompanied by ameliorated EndMT and increased nitric oxide (NO) production. In vitro experiments found that bradykinin mitigated transforming growth factor ß1 (TGFß1)-induced EndMT. Significantly, the bradykinin B2 receptor antagonist or endothelial nitric oxide synthase inhibitor abolished the effects of bradykinin on EndMT inhibition, indicating that the bradykinin B2 receptor and NO might mediate the effects of bradykinin on EndMT inhibition. CONCLUSION: Bradykinin plays an essential role in the process of cardiac fibrosis. Bradykinin preserves the cellular signature of endothelial cells, preventing them from EndMT following cardiac IR injury, possibly mediated by bradykinin B2 receptor activation and NO production.


Assuntos
Cardiomiopatias , Traumatismo por Reperfusão , Humanos , Ratos , Animais , Células Endoteliais , Bradicinina/farmacologia , Bradicinina/metabolismo , Transição Endotélio-Mesênquima , Cardiomiopatias/metabolismo , Receptores da Bradicinina/metabolismo , Óxido Nítrico/metabolismo , Traumatismo por Reperfusão/metabolismo , Fibrose , Transição Epitelial-Mesenquimal
2.
Life Sci ; 329: 121973, 2023 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-37482211

RESUMO

AIMS: Tissue kallikrein-related peptidase8 (KLK8) has been found to mitigate acute myocardial ischemia-reperfusion (IR) injury. However, the effect of KLK8 on cardiac remodeling in response to IR injury has not been determined. MATERIALS AND METHODS: KLK8 overexpressing transgenic rat (KLK8-TG) was used as the animal model. IR injury was induced by ligating the left anterior descending coronary artery for 1 h and subsequent reperfusion. The functional and morphological changes of the heart were examined 14 days after the injury. Neonatal rat cardiac fibroblasts (CFs) were used to investigate the molecular mechanisms in vitro. KEY FINDINGS: KLK8 overexpression enhanced cardiac diastolic dysfunction, fibrosis, and hypertrophy after IR injury, indicating that KLK8 accentuated cardiac remodeling in response to IR injury. Moreover, KLK8 overexpression increased epidermal growth factor (EGF) release and promoted the phosphorylation of EGF receptor (EGFR) and ERK1/2 in the heart after IR injury. It was interesting to find that both EGFR antagonist (AG 1478) and MEK inhibitor (PD98059) attenuated the KLK8-induced proliferation and activation of CFs in vitro, indicating that EGFR signaling might mediate the pro-fibrotic action of KLK8. SIGNIFICANCE: KLK8 plays a crucial role in cardiac remodeling after myocardial infarction. KLK8 accentuates cardiac fibrosis after IR injury, possibly mediated by EGFR signaling in CFs.


Assuntos
Traumatismo por Reperfusão Miocárdica , Ratos , Animais , Traumatismo por Reperfusão Miocárdica/metabolismo , Calicreínas Teciduais/genética , Calicreínas Teciduais/metabolismo , Calicreínas Teciduais/farmacologia , Remodelação Ventricular , Receptores ErbB/metabolismo , Fibrose , Fibroblastos/metabolismo , Miocárdio/metabolismo
3.
Eur J Pharmacol ; 911: 174493, 2021 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-34506777

RESUMO

BACKGROUND: Dexmedetomidine (DEX) has been reported to protect the heart against ischemia reperfusion (I/R) injury. However, the exact mechanisms are still not fully understood. METHODS AND RESULTS: A rat cardiac I/R injury model was induced by ligation of the left anterior descending coronary artery for 1 h and subsequent reperfusion for 2 h, and DEX was administered intravenously 30 min before ischemia. We confirmed that DEX treatment mitigated cardiac I/R injury. Interestingly, we found that DEX regulated the expression of bradykinin (BK) receptors (B1R and B2R) in rat hearts during I/R injury and enhanced the protective action of BK administered during reperfusion. Moreover, in vitro hypoxia reoxygenation (H/R) injury was induced in neonatal rat cardiomyocytes (CMs), and DEX was administered 1 h before hypoxia. The in vitro findings were consistent with the in vivo experiments. We found that an α2-adrenoceptor (α2-AR) antagonist (yohimbine) completely aborted DEX-induced B1R and B2R regulation; an adenylyl cyclase (AC) agonist (forskolin) blocked B1R downregulation, while a phosphatidylinositol 3-kinase (PI3K) inhibitor (LY294002) blocked B2R upregulation. The above findings indicated that DEX interacted with α2-AR in cardiomyocytes, inhibited B1R expression via suppression of AC, and stimulated B2R expression via activation of PI3K. CONCLUSIONS: DEX regulates BK receptor expression and potentiates the protection of BK in cardiac I/R injury, which suggests that modulating endogenous cardioprotective factors may play an important role in DEX-induced cardioprotection.


Assuntos
Dexmedetomidina
4.
Int J Biol Macromol ; 140: 1126-1133, 2019 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-31449861

RESUMO

Tissue kallikrein-related peptidases (KLKs) play important roles in acute cardiac injury and cardiac remodeling. However, the exact cardiac actions of KLK8 have not been determined. Transgenic rat overexpressing KLK8 was established to examine the role of KLK8 in the heart. Cardiac injury was induced by ischemia/reperfusion (I/R) and examined by infarct size measurement and TUNEL staining. The molecular mechanisms were investigated in cultured neonatal rat cardiomyocytes (CMs). Western blot analysis was used to determine the protein levels. KLK8 protein level was significantly increased in the cardiac ischemic risk area. KLK8 overexpression mitigated I/R-induced cardiac injury, as evidenced by decreased infarct size and apoptosis in cardiac ischemic risk area in vivo. Via in vitro studies, it was found that KLK8 overexpression attenuated the Hypoxia/Reoxygenation (H/R) injury in CMs; both B2R and PAR2 antagonist significantly attenuated KLK8-induced protective actions under H/R injury. Moreover, KLK8 overexpressed CMs showed significant higher phosphorylation levels of Akt, ERK1/2 and PKA under H/R stimulation; B2R antagonist attenuated the phosphorylation levels of Akt and ERK1/2, while PAR2 antagonist attenuated the phosphorylation levels of PKA and ERK1/2. KLK8 protects the heart against I/R-induced cardiac injury, which may represent a new therapeutic target in cardiac medicine.


Assuntos
Cardiotônicos/metabolismo , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Serina Endopeptidases/metabolismo , Animais , Bradicinina/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Feminino , Traumatismo por Reperfusão Miocárdica/patologia , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos Sprague-Dawley , Receptor B2 da Bradicinina/metabolismo , Receptor PAR-2/metabolismo , Transdução de Sinais , Regulação para Cima
5.
Sheng Li Xue Bao ; 69(5): 571-578, 2017 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-29063105

RESUMO

Myocardial infarction (MI) is the leading cause of morbidity and mortality worldwide. The regeneration capacity of the adult mammalian heart is very limited, so that the lost cells are replaced by fibrotic scar. This is followed by remodeling of the surrounding myocardium, which includes cardiac hypertrophy and fibrosis, and makes the ventricular wall thicken and stiffen. This adverse cardiac remodeling leads to impaired cardiac function and eventually leads to heart failure. Extensive studies have revealed that microRNAs (miRNAs) play an essential role in cardiovascular diseases. microRNA-22 (miR-22) is one of the most abundant miRNA in the heart. Many studies have demonstrated that miR-22 plays critical roles in MI and subsequent cardiac remodeling. In this review, we summarized the recent research progresses, including the regulatory effects of miR-22 in oxidative stress, cardiac apoptosis, autophagy, hypertrophy, fibrosis and regeneration.


Assuntos
MicroRNAs/fisiologia , Infarto do Miocárdio/etiologia , Animais , Autofagia , Cardiomegalia/etiologia , Humanos , Estresse Oxidativo , Remodelação Ventricular
6.
Free Radic Biol Med ; 96: 406-17, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27174562

RESUMO

Mitochondrial oxidative damage is critically involved in cardiac ischemia reperfusion (I/R) injury. MicroRNA-22 (miR-22) has been predicted to potentially target sirtuin-1 (Sirt1) and peroxisome proliferator-activated receptor-γ coactivator-1α (PGC1α), both of which are known to provide protection against mitochondrial oxidative injury. The present study aims to investigate whether miR-22 is involved in the regulation of cardiac I/R injury by regulation of mitochondrial function. We found that miR-22 level was significantly increased in rat hearts subjected to I/R injury, as compared with the sham group. Intra-myocardial injection of 20 ug miR-22 inhibitor reduced I/R injury as evidenced by significant decreases in cardiac infarct size, serum lactate dehydrogenase (LDH) and creatine kinase (CK) levels and the number of apoptotic cardiomyocytes. H9c2 cardiomyocytes exposed to hypoxia/reoxygenation (H/R) insult exhibited an increase in miR-22 expression, which was blocked by reactive oxygen species (ROS) scavenger and p53 inhibitor. In addition, miR-22 inhibitor attenuated, whereas miR-22 mimic aggravated H/R-induced injury in H9c2 cardiomyocytes. MiR-22 inhibitor per se had no significant effect on cardiac mitochondrial function. Mitochondria from rat receiving miR-22 inhibitor 48h before ischemia were found to have a significantly less mitochondrial superoxide production and greater mitochondrial membrane potential and ATP production as compared with rat receiving miR control. In H9c2 cardiomyocyte, it was found that miR-22 mimic aggravated, whilst miR-22 inhibitor significantly attenuated H/R-induced mitochondrial damage. By using real time PCR, western blot and dual-luciferase reporter gene analyses, we identified Sirt1 and PGC1α as miR-22 targets in cardiomyocytes. It was found that silencing of Sirt1 abolished the protective effect of miR-22 inhibitor against H/R-induced mitochondrial dysfunction and cell injury in cardiomyocytes. Taken together, our findings reveal a novel molecular mechanism for cardiac mitochondrial dysfunction during myocardial I/R injury at the miRNA level and demonstrate the therapeutic potential of miR-22 inhibition for acute myocardial I/R injury by maintaining cardiac mitochondrial function.


Assuntos
MicroRNAs/genética , Traumatismo por Reperfusão Miocárdica/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Sirtuína 1/genética , Trifosfato de Adenosina/biossíntese , Animais , Creatina Quinase/sangue , Regulação da Expressão Gênica/genética , Humanos , L-Lactato Desidrogenase/sangue , Potencial da Membrana Mitocondrial/genética , MicroRNAs/antagonistas & inibidores , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Traumatismo por Reperfusão Miocárdica/sangue , Traumatismo por Reperfusão Miocárdica/patologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Estresse Oxidativo/genética , Ratos , Superóxidos/metabolismo
8.
Sci Rep ; 7: 20024, 2016 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-26823023

RESUMO

The tissue kallikrein-related peptidase family (KLK) is a group of trypsin- and chymotrypsin-like serine proteases that share a similar homology to parent tissue kallikrein (KLK1). KLK1 is identified in heart and has anti-hypertrophic effects. However, whether other KLK family members play a role in regulating cardiac function remains unknown. In the present study, we demonstrated for the first time that KLK8 was expressed in myocardium. KLK8 expression was upregulated in left ventricle of cardiac hypertrophy models. Both intra-cardiac adenovirus-mediated and transgenic-mediated KLK8 overexpression led to cardiac hypertrophy in vivo. In primary neonatal rat cardiomyocytes, KLK8 knockdown inhibited phenylephrine (PE)-induced cardiomyocyte hypertrophy, whereas KLK8 overexpression promoted cardiomyocyte hypertrophy via a serine protease activity-dependent but kinin receptor-independent pathway. KLK8 overexpression increased epidermal growth factor (EGF) production, which was blocked by the inhibitors of serine protease. EGF receptor (EGFR) antagonist and EGFR knockdown reversed the hypertrophy induced by KLK8 overexpression. KLK8-induced cardiomyocyte hypertrophy was also significantly decreased by blocking the protease-activated receptor 1 (PAR1) or PAR2 pathway. Our data suggest that KLK8 may promote cardiomyocyte hypertrophy through EGF signaling- and PARs-dependent but a kinin receptor-independent pathway. It is implied that different KLK family members can subtly regulate cardiac function and remodeling.


Assuntos
Cardiomegalia/genética , Ventrículos do Coração/metabolismo , Miocárdio/metabolismo , Serina Endopeptidases/genética , Animais , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Fator de Crescimento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Regulação da Expressão Gênica , Ventrículos do Coração/patologia , Humanos , Calicreínas/genética , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Ratos , Receptor PAR-1/metabolismo , Receptor PAR-2/metabolismo , Serina Endopeptidases/biossíntese , Transdução de Sinais , Ativação Transcricional
9.
Gynecol Endocrinol ; 31(9): 742-6, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26182188

RESUMO

Exercise training has been looked on as a non-pharmacologic approach to treating ovariectomy (OVX)-induced dysfunctions. In this study, we investigated whether chronic exercise impacts on expression of urocortins (UCNs) and corticotropin-releasing hormone receptor type 2 (CRHR2) in myocardium of OVX rats. Bilateral OVX or sham-operation was performed under anesthesia. Both groups were then divided into two subgroups, with or without treadmill training for 8 weeks. It was found that OVX as well as exercise did not affect the mRNA levels of UCN, UCN2 and UCN3 in myocardium. OVX caused down-regulation of CRHR2 in myocardium. Exercise training reversed the OVX-induced reduction of CRHR2, but had no influence on CRHR2 level in sham rats. OVX resulted in a decrease in estrogen receptor α (ERα) expression in myocardium, which was restored by exercise. Moreover, exercise training also reversed OVX-induced down-regulation of specific protein-1 (Sp-1) expression in myocardium. CRHR2 expression level correlated with Sp-1 and ERα level in myocardium. These results indicate that exercise training can restore the CRHR2 level in myocardium of OVX rats, which is associated with ERα and Sp-1 expression.


Assuntos
Receptor alfa de Estrogênio/genética , Miocárdio/metabolismo , Ovariectomia , Condicionamento Físico Animal , RNA Mensageiro/metabolismo , Receptores de Hormônio Liberador da Corticotropina/genética , Fator de Transcrição Sp1/genética , Urocortinas/genética , Animais , Western Blotting , Hormônio Liberador da Corticotropina/genética , Hormônio Liberador da Corticotropina/metabolismo , Regulação para Baixo , Receptor alfa de Estrogênio/metabolismo , Feminino , Regulação da Expressão Gênica , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição Sp1/metabolismo , Urocortinas/metabolismo
11.
Can J Cardiol ; 30(6): 687-95, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24882544

RESUMO

BACKGROUND: Urocortin-1 (UCN1) exerts protective effects on hypoxia/reoxygenation injury in the heart. Serum- and glucocorticoid- responsive kinase-1 (SGK1), a serine-threonine kinase, has been shown to be crucial for cardiomyocyte survival. The purpose of the present study was to investigate whether SGK1 is involved in UCN1-induced cardioprotection. METHODS: Cardiomyocytes were obtained from neonatal rats and used as a model to investigate UCN1 regulation of SGK1. Specific small interfering RNA targeting SGK1 was used to knock down SGK1 expression. The messenger RNA (mRNA) level of SGK1 was measured using quantitative real time reverse transcription polymerase chain reaction, and the protein levels of SGK1 and phosphorylated SGK1 were determined using Western blot analysis. RESULTS: SGK1 knockdown attenuated the protective effects of UCN1 against hypoxia/reoxygenation injury in cardiomyocytes. Treatment of cardiomyocytes with UCN1 stimulated SGK1 mRNA and protein expression and time-dependently increased phosphorylated SGK1 level. These effects were completely reversed with corticotrophin-releasing hormone receptor type 2 antagonist. Adenylate cyclase and protein kinase A inhibitors abolished the stimulatory effect of UCN1 on SGK1 expression. SGK1 phosphorylation induced by UCN1 was blocked by phosphorinositide-3-kinase inhibitor. CONCLUSIONS: SGK1 is involved in the cardioprotective effects of UCN1 in cardiomyocytes. UCN1 stimulates SGK1 phosphorylation via the phosphorinositide-3-kinase signalling pathway and it induces SGK1 expression via the adenylate cyclase/protein kinase A pathway.


Assuntos
Hipóxia/prevenção & controle , Proteínas Imediatamente Precoces/fisiologia , Miócitos Cardíacos/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Traumatismo por Reperfusão/prevenção & controle , Urocortinas/farmacologia , Animais , Animais Recém-Nascidos , Western Blotting , Proteínas Quinases Dependentes de Cálcio-Calmodulina/antagonistas & inibidores , Células Cultivadas , Cromonas/farmacologia , Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Técnicas de Silenciamento de Genes , Hipóxia/metabolismo , Proteínas Imediatamente Precoces/genética , Isoquinolinas/farmacologia , Modelos Animais , Morfolinas/farmacologia , Fragmentos de Peptídeos/farmacologia , Peptídeos Cíclicos/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Hormônio Liberador da Corticotropina/antagonistas & inibidores , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sulfonamidas/farmacologia
13.
Mol Cell Endocrinol ; 382(1): 17-25, 2014 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-24035863

RESUMO

Estrogens have been suggested to exert cardioprotection through maintaining endogenous cardioprotective mechanisms. In the present study, we investigated whether estrogens protect cardiomyocytes against hypoxia/reoxygenation (H/R) via modulating urocortins (UCNs) and their receptor corticotrophin-releasing hormone receptor type 2 (CRHR2). We found that 17ß-estradiol (E2) enhanced UCN cardioprotection against H/R and increased CRHR2 expression in neonatal rat cardiomyocytes. E2 protected cardiomyocytes against H/R, which was impaired by CRHR2 antagonist or knockdown of CRHR2. Estrogen receptor α (ERα) antagonist treatment or ERα knockdown could abolish E2-induced CRHR2 up-regulation. Moreover, knockdown of Sp1 also attenuated E2-induced CRHR2 up-regulation. Ovariectomy resulted in down-regulation of CRHR2 and Sp-1 in myocardium of mice, which was restored by E2 or ERα agonist treatment. These results suggest that estrogens act on ERα to up-regulate CRHR2 expression in cardiomyocytes, thereby enhancing cardioprotection of UCNs against H/R.


Assuntos
Cardiotônicos/farmacologia , Estradiol/farmacologia , Miócitos Cardíacos/metabolismo , Oxigênio/farmacologia , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Regulação para Cima/efeitos dos fármacos , Animais , Hipóxia Celular/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Receptor alfa de Estrogênio/agonistas , Receptor alfa de Estrogênio/metabolismo , Feminino , Técnicas de Silenciamento de Genes , Camundongos , Miocárdio/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Ovariectomia , Ratos , Ratos Sprague-Dawley , Receptores de Hormônio Liberador da Corticotropina/antagonistas & inibidores , Fator de Transcrição Sp1/metabolismo , Urocortinas/farmacologia
14.
Int J Cardiol ; 168(5): 4755-60, 2013 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-23962778

RESUMO

BACKGROUND: The mechanism underlying estrogen cardioprotection remains largely unknown. Urocortin (UCN), a member of corticotropin-releasing hormone (CRH) family, is one of endogenous cardioprotective factors. The goal of present study is to investigate whether estrogens regulate UCN and its receptor CRH receptor type 2 (CRHR2) in female rat heart. METHODS: 17ß-estradiol (E2) was subcutaneously administrated to ovariectomized (OVX) rats for eight weeks. UCN was administrated before simulated myocardial ischemia/reperfusion (I/R). Cell damage was assessed by measurement of infarct size, activity of serum creatine kinase (CK) and lactate dehydrogenase (LDH) and percentage of TUNEL staining in myocardium. The mRNA and protein levels of UCN and CRHR2 were determined in sham operated and OVX rats with or without E2 replacement. DNA methylation frequency of CRHR2 gene promoter was determined by bisulfite-sequencing. RESULTS: UCN administration reduced infarct size, LDH and CK level and percentage of TUNEL staining upon I/R injury. The cardioprotective effects of UCN were abrogated in OVX rats and E2 replacement restored UCN-induced cardioprotection.CRHR2 mRNA and protein expression were down-regulated more than 40% in OVX rats, both of which were restored by E2 replacement. UCN mRNA and protein levels were not affected by ovariectomy and E2 replacement. Hypermethylation in CRHR2 promoter was found in OVX rats, and two of the methylated CpG sites were seated at cis-acting elements. Hypermethylation induced by OVX could also be ameliorated by E2 replacement. CONCLUSION: Estrogens maintain CRHR2 expression in myocardium, which may through an epigenetic mechanism, and enhance UCN-induced cardioprotective effects against I/R injury.


Assuntos
Estradiol/administração & dosagem , Regulação da Expressão Gênica , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Miocárdio/patologia , RNA Mensageiro/genética , Receptores de Hormônio Liberador da Corticotropina/genética , Urocortinas/administração & dosagem , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Modelos Animais de Doenças , Terapia de Reposição de Estrogênios/métodos , Estrogênios/administração & dosagem , Feminino , Frequência do Gene , Marcação In Situ das Extremidades Cortadas , Injeções Subcutâneas , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/metabolismo , Miocárdio/metabolismo , Ovariectomia/efeitos adversos , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Hormônio Liberador da Corticotropina/biossíntese
15.
Menopause ; 20(10): 1084-91, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23571523

RESUMO

OBJECTIVE: Hydrogen sulfide (H2S), generated in the myocardium predominantly via cystathionine-γ-lyase (CSE), is cardioprotective. The objectives of the present study were to investigate the effects of estrogens on CSE expression and H2S generation in the myocardium and to examine whether serum 17ß-estradiol (E2) level is associated with CSE activity and H2S generation and whether H2S or E2 level is associated with proinflammatory cytokines and oxidative stress status. METHODS: Ovariectomized Sprague-Dawley rats received subcutaneous E2 (30 µg/kg/d) or vehicle for 12 weeks. At the end of the 12-week treatment, CSE expression, H2S generation, reduced glutathione/oxidized glutathione (GSH/GSSG) ratio, total antioxidant capacity (T-AOC), superoxide dismutase (SOD) activity, catalase (CAT) activity, interleukin (IL)-6 concentration, and tumor necrosis factor-α (TNF-α) concentration in the left ventricle were determined. RESULTS: E2 increased CSE expression and H2S generation in the myocardium of ovariectomized rats. H2S production rate and serum E2 were positively correlated. E2 increased GSH/GSSG ratio, T-AOC, CAT, and SOD activity but decreased IL-6 and TNF-α levels. Serum E2 level was positively correlated with GSH/GSSG ratio, T-AOC, CAT, and SOD activity, and inversely correlated with IL-6 and TNF-α levels. H2S generation rate was positively correlated with T-AOC and GSH/GSSG ratio, and inversely correlated with IL-6 and TNF-α levels. CONCLUSIONS: E2 increases CSE expression and endogenous H2S generation in the myocardium. The effects of E2 are associated with decreased oxidative stress and inflammatory status. Our data suggest that estrogens might exert cardioprotective effects through up-regulation of CSE expression and H2S generation.


Assuntos
Cistationina gama-Liase/análise , Estradiol/farmacologia , Inflamação/tratamento farmacológico , Miocárdio/enzimologia , Ovariectomia , Estresse Oxidativo/efeitos dos fármacos , Animais , Antioxidantes/análise , Cistationina gama-Liase/genética , Cistationina gama-Liase/metabolismo , Citocinas/análise , Estradiol/sangue , Feminino , Sulfeto de Hidrogênio/metabolismo , Miocárdio/química , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/metabolismo , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley
16.
Endocrinology ; 153(4): 1915-24, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22315451

RESUMO

Urocortin (Ucn), a member of CRH family, has been implicated to be one of the endogenous regulators in the cardiovascular system and exerts its effects locally via an autocrine/paracrine fashion. Previous studies have shown the gender difference in CRH-induced vasodilation in human skin, which is related to the concentration of estrogens during the menstrual cycle. The aim of this study was to investigate whether estrogens modulate Ucn/CRH receptor type 2 (CRHR2) expression in vascular smooth muscle, thereby leading to vasodilation. We performed sham operation or bilateral ovariectomy (OVX) on female Sprague Dawley rats. OVX rats were sc administered 17ß-estradiol (E2) at a dose of 30 µg/kg·d or with placebo for 12 wk. Primary smooth muscle cells of aorta were used for the in vitro study. It was found that the Ucn-induced vasodilation and CRHR2 expression were decreased in OVX rats and restored by E2 replacement treatment for 12 wk. E2 increased the expression of CRHR2 in cultured smooth muscle cells, which was blocked by estrogen receptor-ß antagonist. Ucn significantly suppressed the phenylephrine-induced phospholipase Cß3 activation, inositol 1,4,5-trisphosphate (IP3) production, and intracellular Ca²âº elevation. Ucn stimulated the expression of active GTP-bound Gαs protein and cAMP production. The suppressive effects of Ucn on phenylephrine-induced IP3 production and intracellular Ca²âº elevation were blocked by the inhibitors of adenylate cyclase and protein kinase A. Our results demonstrate that estrogen maintains the expression of CRHR2 in aorta smooth muscle, thereby enhancing vasodilator actions of Ucn. Ucn exerts its vasorelaxant effects via Gαs-cAMP-protein kinase A signaling, leading to down-regulation of the phospholipase Cß-IP3-Ca²âº signaling pathway.


Assuntos
Aorta Torácica/efeitos dos fármacos , Aorta Torácica/metabolismo , Estrogênios/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Animais , Aorta Torácica/citologia , Sinalização do Cálcio/fisiologia , Células Cultivadas , Estradiol/farmacologia , Receptor beta de Estrogênio/antagonistas & inibidores , Feminino , Técnicas In Vitro , Modelos Animais , Músculo Liso Vascular/citologia , Ovariectomia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/fisiologia , Urocortinas/metabolismo , Vasodilatação/efeitos dos fármacos , Vasodilatação/fisiologia
17.
Purinergic Signal ; 7(1): 73-83, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21484099

RESUMO

There is evidence that gonadal hormones may affect the perception of painful stimulation, although the underlying mechanisms remain unclear. This investigation was undertaken to determine whether the adenosine 5'-triphosphate (ATP) receptor subunit, P2X(3), is involved in the modulatory action of estrogen in peripheral pain signal transduction in dorsal root ganglion (DRG). The mechanical pain behavior test, real-time quantitative reverse transcription-polymerase chain reaction analysis, and Western blot methods were used to determine the mean relative concentrations and functions of P2X(3) receptors in DRG in sham, ovariectomized (OVX), and estradiol replacement (OVX+E(2)) female rats and in sham and orchiectomized male rats. The mechanical hyperalgesia appeared after ovariectomy, which was subsequently reversed after estradiol replacement, whereas it was not observed after orchiectomy in male rats. Plantar injection of 2'(3')-O-(2,4,6-trinitrophenyl) ATP (TNP-ATP), a P2X(3) and P2X(2/3) receptor antagonist, resulted in an increase of the pain threshold force in OVX rats while had no effect on sham rats. Furthermore, A-317491, a selective P2X(3)/P2X(2/3) receptor antagonist, significantly reversed the hyperalgesia of OVX rats. Injection of ATP into the plantars also caused a significant increase of the paw withdrawal duration in OVX rats compared with that seen in the sham group, which became substantially attenuated by TNP-ATP. P2X(3) receptors expressed in DRG were significantly increased in both mRNA and protein levels after ovariectomy and then reversed after estrogen replacement, while a similar increase was not observed after orchiectomy in male rats. Furthermore, P2X(3) mRNA was significantly decreased 24 h after the application of 17ß-estradiol in a concentration-dependent manner in cultured DRG neurons. ICI 182,780, an estrogen receptor antagonist, blocked the reduction in the protein level. These results suggest that the female gonadal hormone, 17ß-estradiol, might participate in the control of peripheral pain signal transduction by modulating P2X(3) receptor-mediated events in primary sensory neurons, probably through genomic mechanisms.

18.
Reprod Biol Endocrinol ; 7: 43, 2009 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-19432998

RESUMO

BACKGROUND: Corticotropin-releasing hormone (CRH) and CRH-related peptide are shown to modulate uterine contractility through two CRH receptor subtype, CRH-R1 and CRH-R2 during pregnancy. Through different signaling pathways, CRH-R1 maintains myometrial quiescence whereas CRH-R2 promotes smooth muscle contractility. We hypothesized that the expression of CRH receptors in myometrium might be changed during pregnancy and labour. METHOD: Immunohistochemistry, Western blot and RT-PCR were used to quantify the cellular localization, the protein levels and the mRNA variants of both CRH-R1 and CRH-R2 in upper segment (US) and lower segment (LS) myometrium from nonpregnant and pregnant women at term before or after labour. RESULTS: CRH-R1 and CRH-R2 were predominately localized to myometrial smooth muscle cells in US and LS. The protein level of CRH-R1 in US was significantly down-regulated in pregnancy, with a further decrease at the onset of labour. However, the expression of CRH-R1 in LS remained unchanged during pregnancy and labour. No significant changes in CRH-R2 expression were observed in US or LS. Six variants of CRH-R1, CRH-R1alpha,-R1beta,-R1c, -R1e,-R1f and -R1g, were identified in nonpregnant and pregnant myometrium. CRH-R2alpha was identified in pregnant myometrium, whereas CRH-R2beta was identified in nonpregnant myometrium CONCLUSION: CRH-R1 and CRH-R2 are expressed in nonpregnant and pregnant US and LS myometrium. Changed expression of CRH receptors during labour may underlie the initiation of uterine contractility during parturition.


Assuntos
Trabalho de Parto/fisiologia , Miométrio/fisiologia , Receptores de Hormônio Liberador da Corticotropina/genética , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Processamento Alternativo/fisiologia , Western Blotting , Feminino , Expressão Gênica/fisiologia , Humanos , Imuno-Histoquímica , Gravidez , RNA Mensageiro/metabolismo , Transdução de Sinais/fisiologia , Contração Uterina/fisiologia
19.
Am J Physiol Endocrinol Metab ; 295(4): E938-46, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18713960

RESUMO

Corticotropin-releasing hormone (CRH) has been shown to exhibit various functions in hippocampus. In the present study, we examined the effect of CRH on the expression of serum/glucocorticoid-inducible protein kinase-1 (SGK-1), a novel protein kinase, in primary cultured hippocampal neurons. A dose-dependent increase in mRNA and protein levels of SGK-1 as well as frequency of SGK-1-positive neurons occurred upon exposure to CRH (1 pmol/l to 10 nmol/l). These effects can be reversed by the specific CRH-R1 antagonist antalarmin but not by the CRH-R2 antagonist astressin 2B. Blocking adenylate cyclase (AC) activity with SQ22536 and PKA with H89 completely prevented CRH-induced mRNA and protein expression of SGK-1. Blockage of PLC or PKC did not block CRH-induced SGK-1 expression. Our results suggest that CRH act on CRH-R1 to stimulate SGK-1 mRNA and protein expression in cultured hippocampal neurons via a mechanism that is involved in AC/PKA signaling pathways.


Assuntos
Hormônio Liberador da Corticotropina/farmacologia , Hipocampo/enzimologia , Proteínas Imediatamente Precoces/biossíntese , Neurônios/enzimologia , Proteínas Serina-Treonina Quinases/biossíntese , Receptores de Hormônio Liberador da Corticotropina/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Western Blotting , Células Cultivadas , AMP Cíclico/biossíntese , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Inibidores Enzimáticos/farmacologia , Imunofluorescência , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Proteínas Imediatamente Precoces/genética , Neurônios/efeitos dos fármacos , Fosfolipase C beta/antagonistas & inibidores , Fosfolipase C beta/biossíntese , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Proteínas Serina-Treonina Quinases/genética , RNA/biossíntese , RNA/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estimulação Química
20.
Auton Neurosci ; 141(1-2): 112-116, 2008 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-18541457

RESUMO

TRPV(1) and P2X(3) receptors are cation channels known to modulate responses to noxious stimuli. In the nervous system, these receptors are preferentially expressed in the pathways that transmit pain signal from the periphery to the brain. The aim of this study is to determine whether prenatal exposure to glucocorticoids alters the expression of P2X(3) and TRPV(1) in the dorsal root ganglia (DRG) and spinal cord (SC) during early postnatal development. Time-pregnant rats received daily subcutaneous injection of dexamethasone (100 microg/kg/day) or a vehicle from prenatal days 9 to 20. The DRG and lumbar/sacral SC of the newborn rats were harvested on postnatal days 1, 7, and 42 for a quantitative real-time PCR analysis of messenger RNAs. In the control rats, mRNA level of P2X(3) and TRPV(1) receptors from DRG remained relatively constant from postnatal days 1 to 42 while those from SC were significantly higher on postnatal day 42 than days 1 and 7. Prenatal treatment with dexamethasone significantly decreased P2X(3) receptor mRNA level in the DRG and SC on postnatal day 1. Such an effect was no longer statistically significant on postnatal day 7, and disappeared completely on postnatal day 42. Expression of TRPV 1 was not altered by dexamethasone regardless of anatomical localization or developmental stages. Therefore, prenatal exposure to glucocorticoids leads to a transient inhibition of P2X(3) expression in the DRG and SC, suggesting a potential involvement of P2X(3) receptors in the unique profile of pain perception in neonates.


Assuntos
Glucocorticoides/toxicidade , Efeitos Tardios da Exposição Pré-Natal/induzido quimicamente , Receptores Purinérgicos P2/genética , Canais de Cátion TRPV/genética , Fatores Etários , Animais , Animais Recém-Nascidos , Dexametasona/administração & dosagem , Dexametasona/toxicidade , Feminino , Gânglios Espinais/efeitos dos fármacos , Gânglios Espinais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Glucocorticoides/administração & dosagem , Injeções Subcutâneas , Região Lombossacral/inervação , Masculino , Gravidez , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores Purinérgicos P2X3 , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Medula Espinal/efeitos dos fármacos , Medula Espinal/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...