Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Biochim Biophys Acta Mol Basis Dis ; 1865(9): 2441-2450, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31175931

RESUMO

Although macrophage migration inhibitory factor (MIF) is known to have antioxidant property, the role of MIF in cardiac fibrosis has not been well understood. We found that MIF was markedly increased in angiotension II (Ang-II)-infused mouse myocardium. Myocardial function was impaired and cardiac fibrosis was aggravated in Mif-knockout (Mif-KO) mice. Functionally, overexpression of MIF and MIF protein could inhibit the expression of fibrosis-associated collagen (Col) 1a1, COL3A1 and α-SMA, and Smad3 activation in mouse cardiac fibroblasts (CFs). Consistently, MIF deficiency could exacerbate the expression of COL1A1, COL3A1 and α-SMA, and Smad3 activation in Ang-II-treated CFs. Interestingly, microRNA-29b-3p (miR-29b-3p) and microRNA-29c-3p (miR-29c-3p) were down-regulated in the myocardium of Ang-II-infused Mif-KO mice but upregulated in CFs with MIF overexpression or by treatment with MIF protein. MiR-29b-3p and miR-29c-3p could suppress the expression of COL1A1, COL3A1 and α-SMA in CFs through targeting the pro-fibrosis genes of transforming growth factor beta-2 (Tgfb2) and matrix metallopeptidase 2 (Mmp2). We further demonstrated that Mif inhibited reactive oxygen species (ROS) generation and Smad3 activation, and rescued the decrease of miR-29b-3p and miR-29c-3p in Ang-II-treated CFs. Smad3 inhibitors, SIS3 and Naringenin, and Smad3 siRNA could reverse the decrease of miR-29b-3p and miR-29c-3p in Ang-II-treated CFs. Taken together, our data demonstrated that the Smad3-miR-29b/miR-29c axis mediates the inhibitory effect of macrophage migration inhibitory factor on cardiac fibrosis.


Assuntos
Fatores Inibidores da Migração de Macrófagos/metabolismo , MicroRNAs/metabolismo , Proteína Smad3/metabolismo , Regiões 3' não Traduzidas , Animais , Antígenos de Diferenciação de Linfócitos B/química , Antígenos de Diferenciação de Linfócitos B/genética , Antígenos de Diferenciação de Linfócitos B/metabolismo , Cardiomegalia/patologia , Cardiomegalia/veterinária , Colágeno Tipo I/metabolismo , Cadeia alfa 1 do Colágeno Tipo I , Fibroblastos/citologia , Fibroblastos/metabolismo , Fibrose , Antígenos de Histocompatibilidade Classe II/química , Antígenos de Histocompatibilidade Classe II/genética , Antígenos de Histocompatibilidade Classe II/metabolismo , Fatores Inibidores da Migração de Macrófagos/antagonistas & inibidores , Fatores Inibidores da Migração de Macrófagos/genética , Masculino , Metaloproteinase 2 da Matriz/química , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/química , MicroRNAs/genética , Miocárdio/citologia , Miocárdio/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Fator de Crescimento Transformador beta2/química , Fator de Crescimento Transformador beta2/genética , Fator de Crescimento Transformador beta2/metabolismo , Regulação para Cima
2.
Sci Rep ; 7(1): 11879, 2017 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-28928469

RESUMO

The molecular mechanisms underlying anthracyclines-induced cardiotoxicity have not been well elucidated. MiRNAs were revealed dysregulated in the myocardium and plasma of rats received Dox treatment. MicroRNA-34a-5p (miR-34a-5p) was verified increased in the myocardium and plasma of Dox-treated rats, but was reversed in rats received Dox plus DEX treatments. Human miR-34a-5p was also observed increased in the plasma of patients with diffuse large B-cell lymphoma after 9- and 16-week epirubicin therapy. Up-regulation of miR-34a-5p was observed in Dox-induced rat cardiomyocyte H9c2 cells. MiR-34a-5p could augment Bax expression, but inhibited Bcl-2 expression, along with the increases of the activated caspase-3 and mitochondrial potentials in H9C2 cells. MiR-34a-5p was verified to modulate Sirt1 expression post-transcriptionally. In parallel to Sirt1 siRNA, miR-34a-5p could enhance p66shc expression, accompanied by increases of Bax and the activated caspase-3 and a decrease of Bcl-2 in H9c2 cells. Moreover, enforced expression of Sirt1 alleviated Dox-induced apoptosis of H9c2 cells, with suppressing levels of p66shc, Bax, the activated caspase-3 and miR-34a-5p, and enhancing Bcl-2 expression. Therefore, miR-34a-5p enhances cardiomyocyte apoptosis by targeting Sirt1, activation of miR-34a-5p/Sirt1/p66shc pathway contributes to Dox-induced cardiotoxicity, and blockage of this pathway represents a potential cardioprotective effect against anthracyclines.


Assuntos
Cardiotoxicidade/metabolismo , Doxorrubicina/efeitos adversos , MicroRNAs/biossíntese , Miocárdio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sirtuína 1/biossíntese , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src/biossíntese , Animais , Cardiotoxicidade/patologia , Linhagem Celular , Doxorrubicina/administração & dosagem , Feminino , Humanos , Linfoma Difuso de Grandes Células B/tratamento farmacológico , Linfoma Difuso de Grandes Células B/metabolismo , Linfoma Difuso de Grandes Células B/patologia , Masculino , Miocárdio/patologia , Ratos , Ratos Sprague-Dawley
3.
Stem Cells Transl Med ; 4(12): 1415-24, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26564862

RESUMO

UNLABELLED: The reprogramming of fibroblasts to induced pluripotent stem cells raises the possibility that somatic cells could be directly reprogrammed to cardiac progenitor cells (CPCs). The present study aimed to assess highly efficient protein-based approaches to reduce or eliminate the genetic manipulations to generate CPCs for cardiac regeneration therapy. A combination of QQ-reagent-modified Gata4, Hand2, Mef2c, and Tbx5 and three cytokines rapidly and efficiently reprogrammed human dermal fibroblasts (HDFs) into CPCs. This reprogramming process enriched trimethylated histone H3 lysine 4, monoacetylated histone H3 lysine 9, and Baf60c at the Nkx2.5 cardiac enhancer region by the chromatin immunoprecipitation quantitative polymerase chain reaction assay. Protein-induced CPCs transplanted into rat hearts after myocardial infarction improved cardiac function, and this was related to differentiation into cardiomyocyte-like cells. These findings demonstrate that the highly efficient protein-transduction method can directly reprogram HDFs into CPCs. This protein reprogramming strategy lays the foundation for future refinements both in vitro and in vivo and might provide a source of CPCs for regenerative approaches. SIGNIFICANCE: The findings from the present study have demonstrated an efficient protein-transduction method of directly reprogramming fibroblasts into cardiac progenitor cells. These results have great potential in cell-based therapy for cardiovascular diseases.


Assuntos
Diferenciação Celular , Técnicas de Reprogramação Celular , Fibroblastos/metabolismo , Células-Tronco Pluripotentes Induzidas , Infarto do Miocárdio , Miócitos Cardíacos , Animais , Citocinas/farmacologia , Fibroblastos/citologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Células-Tronco Pluripotentes Induzidas/transplante , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/terapia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/transplante , Ratos , Fatores de Transcrição/metabolismo , Transdução Genética
4.
Metabolism ; 64(12): 1682-93, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26455966

RESUMO

OBJECTIVE: Evidence shows that both macrophage migration inhibitory factor (MIF) and GLUT4 glucose transporter are involved in diabetic cardiomyopathy (DCM), but it remains largely unknown whether and how MIF regulates GLUT4 expression in cardiomyocytes. The present study aims to investigate the mechanism underlying the modulation of GLUT4 by MIF in cardiomyocytes. MATERIAL AND METHODS: Activations of AKT and AMPK signaling, and expressions of MIF, GLUT4 and the candidate GLUT4 regulation associated transcription factors in the diabetic mouse myocardium were determined. The screened transcription factors mediating MIF-promoted GLUT4 expression were verified by RNA interference (RNAi) and electrophoretic mobility shift assay (EMSA), respectively. RESULTS: MIF was increased, but GLUT4 was decreased in the diabetic mouse myocardium. MIF could enhance glucose uptake and up-regulate GLUT4 expression in NMVCs. Expressions of transcription factor MEF2A, -2C, -2D and Zac1 were significantly up-regulated in MIF-treated neonatal mouse ventricular cardiomyocytes (NMVCs), and markedly reduced in the diabetic myocardium. Knockdown of MEF2A, -2C, -2D and Zac1 could significantly inhibit glucose uptake and GLUT4 expression in cardiomyocytes. Moreover, EMSA results revealed that transcriptional activities of MEF2 and Zac1 were significantly increased in MIF-treated NMVCs. AMPK signaling was activated in MIF-stimulated NMVCs, and AMPK activator AICAR could enhance MEF2A, -2C, -2D, Zac1 and GLUT4 expression. Additionally, MIF effects were inhibited by an AMPK inhibitor compound C and siRNA targeting MIF receptor CD74, suggesting the involvement of CD74-dependent AMPK activation. CONCLUSIONS: Transcription factor MEF2 and Zac1 mediate MIF-induced GLUT4 expression through CD74-dependent AMPK activation in cardiomyocytes.


Assuntos
Proteínas de Ciclo Celular/fisiologia , Genes Supressores de Tumor/fisiologia , Transportador de Glucose Tipo 4/genética , Oxirredutases Intramoleculares/fisiologia , Fatores de Transcrição MEF2/fisiologia , Fatores Inibidores da Migração de Macrófagos/fisiologia , Miócitos Cardíacos/metabolismo , Fatores de Transcrição/fisiologia , Proteínas Quinases Ativadas por AMP/fisiologia , Animais , Antígenos de Diferenciação de Linfócitos B/fisiologia , Células Cultivadas , Cardiomiopatias Diabéticas/fisiopatologia , Antígenos de Histocompatibilidade Classe II/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptor de Insulina/fisiologia , Função Ventricular Esquerda
5.
J Cell Mol Med ; 19(3): 608-19, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25583328

RESUMO

Cyclins/retinoblastoma protein (pRb) pathway participates in cardiomyocyte hypertrophy. MicroRNAs (miRNAs), the endogenous small non-coding RNAs, were recognized to play significant roles in cardiac hypertrophy. But, it remains unknown whether cyclin/Rb pathway is modulated by miRNAs during cardiac hypertrophy. This study investigates the potential role of microRNA-16 (miR-16) in modulating cyclin/Rb pathway during cardiomyocyte hypertrophy. An animal model of hypertrophy was established in a rat with abdominal aortic constriction (AAC), and in a mouse with transverse aortic constriction (TAC) and in a mouse with subcutaneous injection of phenylephrine (PE) respectively. In addition, a cell model of hypertrophy was also achieved based on PE-promoted neonatal rat ventricular cardiomyocyte and based on Ang-II-induced neonatal mouse ventricular cardiomyocyte respectively. We demonstrated that miR-16 expression was markedly decreased in hypertrophic myocardium and hypertrophic cardiomyocytes in rats and mice. Overexpression of miR-16 suppressed rat cardiac hypertrophy and hypertrophic phenotype of cultured cardiomyocytes, and inhibition of miR-16 induced a hypertrophic phenotype in cardiomyocytes. Expressions of cyclins D1, D2 and E1, and the phosphorylated pRb were increased in hypertrophic myocardium and hypertrophic cardiomyocytes, but could be reversed by enforced expression of miR-16. Cyclins D1, D2 and E1, not pRb, were further validated to be modulated post-transcriptionally by miR-16. In addition, the signal transducer and activator of transcription-3 and c-Myc were activated during myocardial hypertrophy, and inhibitions of them prevented miR-16 attenuation. Therefore, attenuation of miR-16 provoke cardiomyocyte hypertrophy via derepressing the cyclins D1, D2 and E1, and activating cyclin/Rb pathway, revealing that miR-16 might be a target to manage cardiac hypertrophy.


Assuntos
Cardiomegalia/genética , Ciclina D1/metabolismo , Ciclina D2/metabolismo , Ciclinas/metabolismo , MicroRNAs/genética , Animais , Aorta Abdominal/cirurgia , Linhagem Celular , Ciclina D1/biossíntese , Ciclina D2/biossíntese , Ciclinas/biossíntese , Modelos Animais de Doenças , Ativação Enzimática , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/biossíntese , Miócitos Cardíacos/patologia , Fenilefrina/farmacologia , Fosforilação , Proteínas Proto-Oncogênicas c-myc , Ratos , Ratos Sprague-Dawley , Proteína do Retinoblastoma/metabolismo , Fator de Transcrição STAT3/metabolismo
6.
Mol Cell Biochem ; 397(1-2): 7-16, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25060909

RESUMO

The beneficial effects of mesenchymal stem cells (MSCs) in cardiac cell therapy are greatly limited due to poor survival after transplantation into ischemic hearts. Here, we investigated whether caspase 8 small hairpin RNA (shRNA) modification enhance human MSCs (hMSCs) survival and improve infarcted heart function. Recombinant adenovirus encoding pre-miRNA-155-designed caspase 8 shRNA was prepared to inhibit caspase 8 expression in hMSCs. The effect of caspase 8 shRNA modification on protecting hMSCs from apoptosis under the conditions of serum deprivation and hypoxia was tested by Annexin V/PI staining and caspase 8 activity assay. The caspase 8 shRNA-modified and superparamagnetic iron oxide (SPIO)-labeled hMSCs were injected into the border zone of the infarcted region of rat heart. Echocardiography and Masson trichrome staining were performed to assess heart function and cardiac fibrosis. Our results showed that adenovirus-mediated caspase 8 shRNA could efficiently inhibit caspase 8 expression in hMSCs. Knock-down of caspase 8 expression lead to inhibition of hMSCs apoptosis, reduction of caspase 8 activity and up-regulations of HGF, IGF-1 and Bcl-2. Transplantation of caspase 8 shRNA-modified hMSCs could significantly improve infracted heart function, attenuate cardiac fibrosis. Consistently, the rate of cardiomyocyte apoptosis and caspase 8 activity were significantly decreased, and the survival rate of transplanted hMSCs was markedly elevated in the myocardium receiving caspase 8 shRNA-modified hMSCs transplantation. Together, our findings implicated the therapeutic potential of caspase 8 shRNA-modified hMSCs in improving the infarcted heart function.


Assuntos
Adenoviridae , Caspase 8 , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/enzimologia , MicroRNAs , Infarto do Miocárdio/terapia , Adulto , Animais , Apoptose/genética , Caspase 8/biossíntese , Caspase 8/genética , Feminino , Técnicas de Silenciamento de Genes/métodos , Humanos , Masculino , MicroRNAs/biossíntese , MicroRNAs/genética , Infarto do Miocárdio/enzimologia , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Ratos , Ratos Sprague-Dawley , Transdução Genética
7.
PLoS One ; 8(9): e75557, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24086569

RESUMO

Carvedilol, a nonselective ß-adrenoreceptor antagonist, protects against myocardial injury induced by acute myocardium infarction (AMI). The mechanisms underlying the anti-fibrotic effects of carvedilol are unknown. Recent studies have revealed the critical role of microRNAs (miRNAs) in a variety of cardiovascular diseases. This study investigated whether miR-29b is involved in the cardioprotective effect of carvedilol against AMI-induced myocardial fibrosis. Male SD rats were randomized into several groups: the sham surgery control, left anterior descending (LAD) surgery-AMI model, AMI plus low-dose carvedilol treatment (1 mg/kg per day, CAR-L), AMI plus medium-dose carvedilol treatment (5 mg/kg per day, CAR-M) and AMI plus high-dose carvedilol treatment (10 mg/kg per day, CAR-H). Cardiac remodeling and impaired heart function were observed 4 weeks after LAD surgery treatment; the observed cardiac remodeling, decreased ejection fraction, and fractional shortening were rescued in the CAR-M and CAR-H groups. The upregulated expression of Col1a1, Col3a1, and α-SMA mRNA was significantly reduced in the CAR-M and CAR-H groups. Moreover, the downregulated miR-29b was elevated in the CAR-M and CAR-H groups. The in vitro study showed that Col1a1, Col3a1, and α-SMA were downregulated and miR-29b was upregulated by carvedilol in a dose-dependent manner in rat cardiac fibroblasts. Inhibition of ROS-induced Smad3 activation by carvedilol resulted in downregulation of Col1a1, Col3a1, and α-SMA and upregulation of miR-29b derived from the miR-29b-2 precursor. Enforced expression of miR-29b significantly suppressed Col1a1, Col3a1, and α-SMA expression. Taken together, we found that smad3 inactivation and miR-29b upregulation contributed to the cardioprotective activity of carvedilol against AMI-induced myocardial fibrosis.


Assuntos
Carbazóis/farmacologia , Fibrose/tratamento farmacológico , MicroRNAs/genética , Miocárdio/metabolismo , Propanolaminas/farmacologia , Proteína Smad3/genética , Regulação para Cima/efeitos dos fármacos , Animais , Cardiotônicos/farmacologia , Carvedilol , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Fibrose/genética , Fibrose/metabolismo , Fibrose/patologia , Coração/efeitos dos fármacos , Coração/fisiologia , Masculino , MicroRNAs/metabolismo , Infarto do Miocárdio/tratamento farmacológico , Infarto do Miocárdio/genética , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Miocárdio/patologia , Ratos , Ratos Sprague-Dawley , Proteína Smad3/metabolismo , Regulação para Cima/genética , Função Ventricular Esquerda/efeitos dos fármacos , Função Ventricular Esquerda/genética , Função Ventricular Esquerda/fisiologia
8.
Mol Biol Rep ; 39(2): 1333-42, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21667244

RESUMO

We investigated whether transplantation of bone marrow mesenchymal stem cells (BMSC) with induced BMSC (iBMSC) or uninduced BMSC (uBMSC) into the myocardium could improve the performance of post-infarcted rat hearts. BMSCs were specified by flowcytometry. IBMSCs were cocultured with rat cardiomyocyte before transplantation. Cells were injected into borders of cardiac scar tissue 1 week after experimental infarction. Cardiac performance was evaluated by echocardiography at 1, 2, and 4 weeks after cellular or PBS injection. Langendorff working-heart and histological studies were performed 4 weeks after treatment. Myogenesis was detected by quantitative PCR and immunofluorescence. Echocardiography showed a nearly normal ejection fraction (EF) in iBMSC-treated rats and all sham control rats but a lower EF in all PBS-treated animals. The iBMSC-treated heart, assessed by echocardiography, improved fractional shortening compared with PBS-treated hearts. The coronary flow (CF) was decreased obviously in PBS and uBMSC-treated groups, but recovered in iBMSC-treated heart at 4 weeks (P < 0.01). Immunofluorescent microscopy revealed co-localization of Superparamagnetic iron oxide (SPIO)-labeled transplanted cells with cardiac markers for cardiomyocytes, indicating regeneration of damaged myocardium. These data provide strong evidence that iBMSC implantation is of more potential to improve infarcted cardiac performance than uBMSC treatment. It will open new promising therapeutic opportunities for patients with post-infarction heart failure.


Assuntos
Transplante de Medula Óssea , Coração/fisiologia , Transplante de Células-Tronco Mesenquimais , Infarto do Miocárdio/terapia , Animais , Diferenciação Celular/fisiologia , Primers do DNA/genética , Ecocardiografia , Citometria de Fluxo , Masculino , Microscopia de Fluorescência , Desenvolvimento Muscular/fisiologia , Miócitos Cardíacos/transplante , Reação em Cadeia da Polimerase , Ratos , Ratos Sprague-Dawley
9.
J Ethnopharmacol ; 137(1): 263-70, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21619920

RESUMO

AIM OF THIS STUDY: The panax notoginseng saponins (PNS) have been clinically used for the treatment of cardiovascular diseases and stroke in China. Evidences demonstrated that PNS could protect cardiomyocytes from injury induced by ischemia, but the underlying molecular mechanisms of this protective effect are still unclear. This study was aimed to investigate the protective effect and potential molecular mechanisms of PNS on apoptosis in H9c2 cells in vitro and rat myocardial ischemia injury model in vivo. MATERIALS AND METHODS: H9c2 cells subjected to serum, glucose and oxygen deprivation (SGOD) were used as in vitro models and SD rats subjected to left anterior descending (LAD) coronary artery ligation were used as in vivo models. The anti-apoptotic effect of PNS was evaluated by Annexin V/PI analysis or TUNEL assay. Mitochondrial membrane potential (Δψm) was detected by JC-1 analysis. The expression of Akt and phosphorylated Akt (p-Akt) were detected by western blot assay. RESULTS: PNS exhibited anti-apoptotic effect both in H9c2 cells and in ischemic myocardial tissues. However, the effect was blocked in vitro by LY294002, a specific PI3K inhibitor. The anti-apoptotic effect of PNS was mediated by stabilizing Δψm in H9c2 cells. Furthermore the indices of the left ventricular ejection fractions (EF), left ventricular fractional shortening (FS), left ventricular dimensions at end diastole (LVDd) and left ventricular dimensions at end systole (LVDs) suggested that PNS improved rats cardiac function. PNS significantly increased p-Akt both in H9c2 cells and in ischemic myocardial tissues and this effect was also blocked by LY294002 in H9c2 cells. CONCLUSION: Results of this study suggested that PNS could protect myocardial cells from apoptosis induced by ischemia in both the in vitro and in vivo models through activating PI3K/Akt signaling pathway.


Assuntos
Cardiotônicos/farmacologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Miócitos Cardíacos/efeitos dos fármacos , Panax notoginseng , Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Saponinas/farmacologia , Animais , Anexina A5/metabolismo , Apoptose/efeitos dos fármacos , Western Blotting , Cardiotônicos/isolamento & purificação , Linhagem Celular , Citoproteção , Modelos Animais de Doenças , Ativação Enzimática , Marcação In Situ das Extremidades Cortadas , Masculino , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Traumatismo por Reperfusão Miocárdica/enzimologia , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Miócitos Cardíacos/enzimologia , Miócitos Cardíacos/patologia , Panax notoginseng/química , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica , Saponinas/isolamento & purificação , Transdução de Sinais/efeitos dos fármacos , Volume Sistólico/efeitos dos fármacos , Fatores de Tempo , Função Ventricular Esquerda/efeitos dos fármacos
10.
Zhong Yao Cai ; 33(1): 89-92, 2010 Jan.
Artigo em Chinês | MEDLINE | ID: mdl-20518313

RESUMO

OBJECTIVE: To research the effects of Panax notoginseng saponins (PNS) on angiotensin-converting enzymes 2 ( ACE2) and tumor necrosis factor-alpha (TNF-alpha) in rats with post-myocardial infarction ventricular remodeling. METHODS: Models of acute myocardial infarction (AMI) were produced by ligation of left anterior descending coronary artery, 24 hours after operation the rats were randomly divided into control and experiment groups, then respectively administrated with NS, fosinopril and low, middle and high dosage of PNS for four consecutive weeks. To observe effects of PNS on malondialdehyde (MDA), nitric oxide (NO), glutathione peroxidase (GSH-Px), ACE2 and TNF-alpha in rats with post-myocardial infarction ventricular remodeling. RESULTS: Compared with NS group, MDA significantly decreased, the activity of GSH-Px significantly increased (P < 0.05 or P < 0.01), NO of the high-dose PNS group decreased (P < 0.05), Compared with the NS group, ACE2 increased and TNF-a significantly decreased in low-dose PNS group, middle and high-dose groups (P < 0.05). CONCLUSION: PNS can stimulate ACE2 to inhibit the expression of TNF-alpha and enhance the antioxidance. PNS can reduce pathological injury of cardiac myocytes in myocardial ischemia and cardiac muscle, which can improve ventricular remodeling.


Assuntos
Antioxidantes/farmacologia , Infarto do Miocárdio/tratamento farmacológico , Peptidil Dipeptidase A/sangue , Saponinas/farmacologia , Fator de Necrose Tumoral alfa/sangue , Remodelação Ventricular/efeitos dos fármacos , Enzima de Conversão de Angiotensina 2 , Animais , Modelos Animais de Doenças , Feminino , Fosinopril/farmacologia , Glutationa Peroxidase/sangue , Glutationa Peroxidase/metabolismo , Masculino , Malondialdeído/sangue , Malondialdeído/metabolismo , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miocárdio/metabolismo , Miocárdio/patologia , Óxido Nítrico/sangue , Óxido Nítrico/metabolismo , Panax notoginseng/química , Peptidil Dipeptidase A/metabolismo , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Fator de Necrose Tumoral alfa/metabolismo
11.
Zhong Yao Cai ; 33(10): 1592-5, 2010 Oct.
Artigo em Chinês | MEDLINE | ID: mdl-21355198

RESUMO

OBJECTIVE: To research the effects of Erigeron breviscapus injection (EBI) on TNF-alpha, PAI-1 and tPA in rats with acute myocardial infarction. METHODS: Models of acute myocardial infarction (AMI)were produced by ligation of left anterior descending coronary artery, then rats were randomly divided into control and experimental groups, then respectively gavaged NS, and the low, middle and high dosage of EBI for one week. The cardiac function index and the expression of TNF-alpha, tPA and PAI-1 were measured. RESULTS: Compared with the NS group, the cardiac function LVEDP, MAP, LVPmax, +/- dp/dt of AMI rat was improved by EBI in all dosage range, and the expression of TNF-alpha and PAI-1, LVEDP were decreased (P < 0.05), the expression of tPA, MAP, LVPmax and +/- dp/dt were increased obviously (P < 0.05) and had a dose-effect relationship. CONCLUSION: EBI can inhibit the expression of TNF-alpha and PAI-1, increase the expression of tPA,which can prevent the ongoing thrombopoiesis after AMI and improve the cardiac function. This maybe attributes to the inhibition of the overexpression of TNF-alpha.


Assuntos
Medicamentos de Ervas Chinesas/farmacologia , Erigeron , Infarto do Miocárdio/tratamento farmacológico , Miocárdio/metabolismo , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Doença Aguda , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Medicamentos de Ervas Chinesas/administração & dosagem , Medicamentos de Ervas Chinesas/uso terapêutico , Erigeron/química , Injeções , Masculino , Infarto do Miocárdio/etiologia , Infarto do Miocárdio/fisiopatologia , Miocárdio/patologia , Inibidor 1 de Ativador de Plasminogênio/sangue , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Ativador de Plasminogênio Tecidual/sangue , Ativador de Plasminogênio Tecidual/metabolismo , Fator de Necrose Tumoral alfa/sangue
12.
Zhonghua Yi Xue Za Zhi ; 90(48): 3431-4, 2010 Dec 28.
Artigo em Chinês | MEDLINE | ID: mdl-21223820

RESUMO

OBJECTIVE: To study the effects of B-type natriuretic peptide (BNP) preconditioning on the apoptosis and expressions of Bcl-2 and Bax in rat cardiomyocytes during myocardial ischemia-reperfusion. METHODS: Twenty-one male Sprague-Dawley rats weighing (250 ± 50) g were randomly divided into 3 groups of sham operation (SHAM), ischemia-reperfusion (I/R) and B-type natriuretic peptide (BNP). A rat model of in vivo myocardial ischemia-reperfusion injury was established by ligating the left anterior descending coronary artery for 35 minutes and then reperfusing for 240 minutes. The apoptosis of myocardial cell was determined by terminal deoxynucleotidyl transferase mediated dUTP-biotin nick end-labeling (TUNEL) method. Real-time polymerase chain reaction and Western blot were used to detect the expression changes of Bcl-2 and Bax in rat ischemia myocardium. RESULTS: The apoptotic indices of SHAM, BNP and I/R groups were 5.4% ± 4.2%, 22.5% ± 9.5% and 45.2% ± 13.0% respectively (P < 0.05). The Bcl-2 protein expression of SHAM, BNP and I/R groups were 0.87 ± 0.09, 0.70 ± 0.07 and 0.38 ± 0.09 respectively (P < 0.05). The Bax protein expression of SHAM, BNP and I/R groups were 0.08 ± 0.04, 0.39 ± 0.09 and 0.71 ± 0.18 respectively (P < 0.01). The Bcl-2/Bax mRNA ratio of SHAN, BNP and I/R groups were 0.763 ± 0.154, 0.099 ± 0.025 and 0.022 ± 0.024 respectively (P < 0.05). CONCLUSION: The BNP preconditioning can decrease the myocardial apoptosis induced by ischemia-reperfusion injury. The mechanisms may be associated with an elevated expression of Bcl-2, an increased ratio of Bcl-2/Bax and a lowered expression of Bax.


Assuntos
Apoptose/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Peptídeo Natriurético Encefálico/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteína X Associada a bcl-2/metabolismo , Animais , Precondicionamento Isquêmico Miocárdico/métodos , Masculino , Miócitos Cardíacos/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão
13.
Zhonghua Xin Xue Guan Bing Za Zhi ; 37(8): 680-4, 2009 Aug.
Artigo em Chinês | MEDLINE | ID: mdl-20021917

RESUMO

OBJECTIVE: To compare the efficacy of transplanting bone marrow mesenchymal stem cell (BMSC) or microenvironmental induced BMSC (iBMSC) into the ischemic myocardium of rats with myocardial infarction. METHODS: iBMSC was defined as BMSC co-cultured with myocardial cells for 2 weeks. The stem cells or equal volume PBS were injected into ischemic border zone 1 wk after experimental infarction. Cardiac performance was evaluated at 1, 2, and 4 wk after cell transplantation by echocardiography and analyzed histologically at 4 wk after cell transplantations. RESULTS: Compared with PBS group, both BMSC and iBMSC transplantations reduced infarct size. iBMSC enhanced the beneficial effects of BMSC on improving cardiac function (FS: 28.5% +/- 4.3% in PBS, 29.0% +/- 2.0% in BMSC and 45.1% +/- 3.1% in iBMSC group at 4 weeks post transplantation, iBMSC group vs. PBS group P < 0.05, iBMSC group vs. BMSC group P < 0.05). Immunofluorescence microscopy results revealed co-localization of SPIO-labeled transplanted cells with cardiac markers for cardiomyocytes, indicating regeneration of damaged myocardium. CONCLUSION: Our data suggest that iBMSC implantation is more effective on improving cardiac function than BMSC implantation in this model. iBMSC might serve as a new promising therapeutic cell source for regenerating ischemic myocardium in patients with post-infarction heart failure.


Assuntos
Transplante de Medula Óssea , Transplante de Células-Tronco Mesenquimais , Infarto do Miocárdio/cirurgia , Condicionamento Pré-Transplante , Animais , Diferenciação Celular , Células Cultivadas , Ratos , Ratos Sprague-Dawley
14.
Nan Fang Yi Ke Da Xue Xue Bao ; 29(10): 2048-50, 2009 Oct.
Artigo em Chinês | MEDLINE | ID: mdl-19861263

RESUMO

OBJECTIVE: To observe the effects of Panax notoginsenoside (PNS) on tumor necrosis factor-alpha (TNF-alpha) and matrix metalloproteinases-2 (MMP-2) expressions in rats with post-myocardial infarction ventricular remodeling and explore the mechanism. METHODS: Rat models of acute infarction ventricular (AMI) were established by ligation of the left anterior descending coronary artery. Twenty-four hours after the operation, the rats were randomized into control and experimental groups for intragastric administration of normal saline (control), fosinopril and PNS at the low, medium and high doses for 4 consecutive weeks. The effects of PNS on the cardiac function index including the left ventricular end-diastolic dimension (LVIDd), left ventricular end-systolic diameter (LVIDs), ejection fraction (EF), percentage of left ventricular systole (FS), mitral early diastolic flow velocity mouth (MV), and heart rate (HR) were observed, and the changes in TNF-alpha and MMP-2 expression were detected after post-myocardial infarction ventricular remodeling. RESULTS: Compared with the control group, PNS at the medium and high doses produced significant improvements in the EF, FS and MV of the rats (P<0.01 or 0.05). TNF-alpha and MMP-2 expressions were significantly decreased by PNS treatment at low, medium and high doses (P<0.01). CONCLUSION: PNS can inhibit or reduce the expression of TNF-alpha and MMP-2, thereby enhancing left ventricular systolic and diastolic functions, decreasing peripheral resistance, and improving the cardiac function of rats with post-myocardial infarction left ventricular remodeling.


Assuntos
Metaloproteinase 2 da Matriz/metabolismo , Infarto do Miocárdio/tratamento farmacológico , Panax notoginseng/química , Saponinas/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Remodelação Ventricular/efeitos dos fármacos , Animais , Feminino , Masculino , Infarto do Miocárdio/fisiopatologia , Ratos , Ratos Sprague-Dawley , Saponinas/uso terapêutico
15.
Zhonghua Xin Xue Guan Bing Za Zhi ; 36(7): 651-4, 2008 Jul.
Artigo em Chinês | MEDLINE | ID: mdl-19100098

RESUMO

OBJECTIVE: To investigate the ability of human bone marrow mesenchymal stem cells (hBMSCs), cocultured with semi-permeable membrane separated neonatal rat ventricular myocytes, to differentiate into cardiomyocytes. METHODS: hBMSCs were isolated and purified by density gradient centrifugation and adherence screening method. Cells were expanded as undifferentiated cells in culture for more than 3 passages and their phenotypes were identified with flow cytometer. hBMSCs were cocultured with neonatal rat ventricular myocytes in a rate of 1:10 separated by semi-permeable membrane. GATA4 mRNA was detected by RT-PCR; Immunocytochemistry, and Immunostaining were used to detect sarcomeric alpha-actinin, desmin, cTnT, and cTnI protein level. RESULTS: CD29 (98.64% +/- 0.80%) and CD44 (96.70% +/- 1.50%) were the major surface markers of hBMSCs. After coculturing with semi-permeable membrane separated neonatal rat ventricular myocytes, the first contraction of single cells was noted at day 7 and GATA4 expression was detected on these cells by RT-PCR after 1 to 3 weeks coculture. Desmin, sarcomeric alpha-actinin, cTnI and cTnT could be detected by immunocytochemistry and immunostaining on some of these cells. CONCLUSION: hBMSCs possess the potential to differentiate into myocardial cell phenotype in the cardiac microenvironment. Direct contact with cardiomyocytes was not necessary required for hBMSCs differentiation.


Assuntos
Células da Medula Óssea/citologia , Diferenciação Celular , Células-Tronco Mesenquimais/citologia , Miócitos Cardíacos/citologia , Animais , Técnicas de Cultura de Células , Células Cultivadas , Técnicas de Cocultura , Humanos , Ratos , Ratos Sprague-Dawley
16.
Nan Fang Yi Ke Da Xue Xue Bao ; 27(12): 1813-6, 2007 Dec.
Artigo em Chinês | MEDLINE | ID: mdl-18158990

RESUMO

OBJECTIVE: To investigate the expression of representative heart-specific primary microRNAs (pri-miRNAs) in the cardiomyocyte-like cells differentiated from human mesenchymal stem cells (hMSCs). METHODS: The phenotype of hMSCs isolated was identified by flow cytometry using monoclonal antibodies against FITC-conjugated CD29, CD34, and CD11b. The third-passage hMSCs were induced to differentiate into cardiomyocyte-like cells by 5-azacytidine and indirect coculture with neonatal rat myocytes, respectively. Immunocytochemical analysis was performed to detect the expression of the cardiac-specific proteins, namely cardiac troponin I (cTnI) and sarcomeric alpha-actinin, in the cardiomyocyte-like cells differentiated from hMSCs. RT-PCR and DNA sequencing were used to identify the expression of the 5 representative heart-specific pri-miRNAs. RESULTS: High hMSC marker CD29 expression rate (98.87%) and low hematopoietic cell markers CD34 (5%) and CD11b (0.4%) expression rates were identified in the hMSCs isolated. cTnI and sarcomeric alpha-actinin expression occurred in the hMSCs following induction with the 2 differentiation-inducing methods. miRNA-143 and -181 expressions were induced in the hMSCs by 5-azacytidine and miRNA-143, -181, -206, and -208 expressions were induced by indirect coculture with neonatal rat myocytes, but pri-miRNA-1-2 expression failed to be induced by these two induction methods. CONCLUSION: Expressions of the representative heart-specific pri-miRNAs in different patterns can be induced in cardiomyocyte-like cells differentiated from hMSCs by 5-azacytidine and indirect coculture with neonatal rat myocytes.


Assuntos
Diferenciação Celular , Células-Tronco Mesenquimais/citologia , MicroRNAs/metabolismo , Miócitos Cardíacos/metabolismo , Actinina/metabolismo , Animais , Técnicas de Cocultura , Humanos , Miócitos Cardíacos/citologia , Ratos , Troponina I/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA