Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Biochim Biophys Acta Mol Basis Dis ; 1870(3): 166944, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-37952827

RESUMO

Sema4D (CD100) is closely related to pathological and physiological processes, including tumor growth, angiogenesis and cardiac development. Nevertheless, the role and mechanism of Sema4D in cardiac hypertrophy are still unclear to date. To assess the impact of Sema4D on pathological cardiac hypertrophy, TAC surgery was performed on C57BL/6 mice which were transfected with AAV9-mSema4D-shRNA or AAV9-mSema4D adeno-associated virus by tail vein injection. Our results indicated that Sema4D knockdown mitigated cardiac hypertrophy, fibrosis and dysfunction when exposed to pressure overload, and Sema4D downregulation markedly inhibited cardiomyocyte hypertrophy induced by angiotensin II. Meanwhile, Sema4D overexpression had the opposite effect in vitro and in vivo. Furthermore, analysis of signaling pathways showed that Sema4D activated the MAPK pathway during cardiac hypertrophy induced by pressure overload, and the pharmacological mitogen-activated protein kinase kinase 1/2 inhibitor U0126 almost completely reversed Sema4D overexpression-induced deteriorated phenotype, resulting in improved cardiac function. Further research indicated that myocardial hypertrophy induced by Sema4D was closely related to the expression of the pyroptosis-related proteins PP65, NLRP3, caspase-1, ASC, GSDMD, IL-18 and IL-1ß. In conclusion, our study demonstrated that Sema4D regulated the process of pathological myocardial hypertrophy through modulating MAPK/NF-κB/NLRP3 pathway, and Sema4D may be the promising interventional target of cardiac hypertrophy and heart failure.


Assuntos
Antígenos CD , Miócitos Cardíacos , NF-kappa B , Semaforinas , Animais , Camundongos , Cardiomegalia/metabolismo , Camundongos Endogâmicos C57BL , Miócitos Cardíacos/metabolismo , NF-kappa B/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo
2.
Shock ; 60(3): 385-391, 2023 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-37548623

RESUMO

ABSTRACT: Background: Cardiogenic shock complicating acute myocardial infarction (AMICS) remains a high 30-day mortality. Mechanical circulatory support devices are increasingly used in AMICS, but their effects on mortality vary partly because of shock severity. Aims: This study aimed to evaluate the association between intra-aortic balloon pump (IABP) and 30-day mortality in patients with early-stage AMICS. Methods: We retrospectively analyzed patients with ST-segment elevation myocardial infarction (STEMI) based on a multicenter clinical trial (NCT04996901). Patients were stratified by IABP use, and shock severity was classified according to the Society for Cardiovascular Angiography and Interventions (SCAI) SHOCK stages. The primary outcome was 30-day all-cause mortality. The association between IABP and 30-day mortality was evaluated across shock stages using propensity score matching, weighting, and logistic regression. Results: Five thousand three hundred forty-three patients were included, and 299 received IABP. The SCAI SHOCK stage was associated with 30-day mortality (odds ratio [OR], 20.19; 95% confidence interval [CI], 13.60-29.97; P < 0.001). In the 580 matched patients, a significant interaction between IABP and 30-day mortality at different shock stages was observed ( P = 0.005). Intra-aortic balloon pump was associated with lower 30-day mortality among patients with shock stage A/B (5.8% vs. 1.2%; OR, 0.19; 95% CI, 0.03-0.73; P = 0.034) but not stage C/D/E (29.3% vs. 38.1%; OR, 1.49; 95% CI, 0.84-2.65; P = 0.172). These results were confirmed by sensitivity analyses of the weighted cohort. Conclusions: Intra-aortic balloon pump reduced 30-day mortality in patients with early-stage AMICS. The SCAI SHOCK stage provides risk stratification for patients with STEMI and helps identify those who may respond well to IABP.


Assuntos
Infarto do Miocárdio , Infarto do Miocárdio com Supradesnível do Segmento ST , Humanos , Choque Cardiogênico/etiologia , Infarto do Miocárdio com Supradesnível do Segmento ST/complicações , Infarto do Miocárdio com Supradesnível do Segmento ST/cirurgia , Estudos Retrospectivos , Infarto do Miocárdio/complicações , Infarto do Miocárdio/terapia , Balão Intra-Aórtico , Resultado do Tratamento
3.
ESC Heart Fail ; 10(5): 2914-2926, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37455355

RESUMO

AIMS: Suboptimal perfusion leading to heart failure (HF) often occurs after ST-segment elevation myocardial infarction (STEMI), despite restoration of epicardial coronary flow in primary percutaneous coronary intervention (PPCI) era. We determined the clinical implications of angio-based coronary functional assessment in evaluation of suboptimal perfusion and further outcomes among STEMI patients after successful PPCI. METHODS AND RESULTS: In this study, STEMI patients in the Chinese STEMI PPCI registry trial (NCT04996901) who achieved post-PPCI thrombolysis in myocardial infarction grade 3 flow were retrospectively screened. Post-procedural quantitative flow ratio (QFR), angio-based microvascular resistance (AMR), and coronary flow velocity (CFV) of the infarct-related artery were calculated. QFR and AMR measure epicardial stenosis severity and microvascular resistance, respectively. QFR+ was defined as QFR < 0.90 while QFR- was QFR ≥ 0.90. AMR+ was defined as AMR ≥ 250 mmHg*s/m while AMR- was AMR < 250 mmHg*s/m. The primary outcome was 30-day new-onset HF. The Kaplan-Meier curves were used to establish the associations between QFR, AMR, CFV, and HF incidences. The relationship between CFV and combined QFR and AMR indices was further assessed. Independent predictors were determined using Cox regression analysis. The receiver-operating characteristic curve was used to assess discriminant ability to predict HF. A total of 942 patients (mean age was 57.8 ± 11.7 years and 84.6% were men) were enrolled. Among them, 129 patients had new-onset HF episodes. Patients in the QFR-/AMR- group had a low risk of HF compared with those in the QFR+/AMR+ group (10.5% vs. 27.3%, P = 0.027). A higher CFV ≥ 17.4 cm/s was associated with low HF incidences as compared with CFV < 17.4 cm/s (10.3% vs. 16.8%, P = 0.005), whereas isolated QFR or AMR did not reveal any marked differences in HF incidences (P = 0.150 and 0.079, respectively). The highest and lowest medians of CFV were observed in the QFR-/AMR- and QFR+/AMR+ groups, respectively. CFV correlated well with the QFR/AMR ratio (adjusted R2  = 1, P < 0.001) and post-PPCI CFV was found to be an independent predictor of post-STEMI HF (adjusted hazard ratio: 0.61, 95% confidence interval: 0.41-0.90, P = 0.012). The area under curve estimate of the multivariable regression model was 0.749. CONCLUSIONS: CFV is an integrated coronary physiological assessment approach that incorporates epicardial and microcirculatory contributions. Patients with post-PPCI CFV < 17.4 cm/s were strongly associated with a high risk for post-STEMI HF, even achieving thrombolysis in myocardial infarction grade 3 flow. The immediate angio-based coronary functional assessment is a feasible tool for evaluating suboptimal perfusion and risk stratification.

4.
Food Funct ; 13(23): 12077-12092, 2022 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-36367287

RESUMO

Aims: The proliferation and migration of vascular smooth muscle cells (VSMCs) play vital roles in the pathological process of neointima formation after vascular injury. Galangin, an extract of the ginger plant galangal, is involved in numerous biological activities, including inhibiting the proliferation and migration of tumor cells, but its effect on VSMCs is unknown. This study focused on the role and mechanism of galangin in the neointima formation induced by vascular injury. Methods and results: In this study, we found that galangin restrained the PDGF-BB-induced proliferation, migration and phenotypic switching of VSMCs in a concentration-dependent manner. In vivo, we established a model of carotid artery balloon injury in rats, followed by intragastric administration of galangin (40 mg kg-1 day-1 or 80 mg kg-1 day-1) for 14 or 28 consecutive days. Then, the degree of neointima hyperplasia was evaluated by H&E staining, and the level of relevant protein expression was assessed by immunofluorescence and western blotting. In vitro, we isolated and grew primary rat aortic smooth muscle cells, which were treated with PDGF-BB and different doses of galangin, and then CCK-8 assay, wound healing assay, transwell assay, western blotting and immunofluorescence assays were performed. We found that galangin significantly inhibited PDGF-BB-induced proliferation, migration, and phenotypic switching of VSMCs and promoted autophagy in VSMCs in vitro, and galangin significantly inhibited neointimal hyperplasia after the common carotid artery balloon injury in rats. In terms of mechanisms, galangin inhibited the PI3K/AKT/mTOR pathway, thereby suppressing VSMC's switch from a contractile to a synthetic phenotype, inhibiting VSMC proliferation, migration and phenotypic switching and upregulating the Beclin1 protein expression levels and the ratio of LC3BII/I, promoting VSMC autophagy, and thereby inhibiting neointimal hyperplasia after vascular injury. Conclusion: Our study suggests that galangin inhibits neointimal hyperplasia after vascular injury by inhibiting smooth muscle cell proliferation, migration and phenotypic switching and by promoting autophagy, and that galangin may be a promising drug for the prevention and treatment of vascular restenosis after PCI.


Assuntos
Lesões das Artérias Carótidas , Intervenção Coronária Percutânea , Lesões do Sistema Vascular , Ratos , Animais , Neointima/tratamento farmacológico , Neointima/metabolismo , Neointima/patologia , Becaplermina/metabolismo , Becaplermina/farmacologia , Becaplermina/uso terapêutico , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Lesões do Sistema Vascular/tratamento farmacológico , Lesões do Sistema Vascular/genética , Lesões do Sistema Vascular/metabolismo , Músculo Liso Vascular , Hiperplasia/metabolismo , Hiperplasia/patologia , Movimento Celular , Proliferação de Células , Ratos Sprague-Dawley , Lesões das Artérias Carótidas/tratamento farmacológico , Lesões das Artérias Carótidas/metabolismo , Lesões das Artérias Carótidas/patologia , Miócitos de Músculo Liso , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Células Cultivadas
5.
Int J Mol Sci ; 23(20)2022 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-36293441

RESUMO

Increasing evidence has shown that vascular aging has a key role in the pathogenesis of vascular diseases. P300/CBP-associated factor (PCAF) is involved in many vascular pathological processes, but the role of PCAF in vascular aging is unknown. This study aims to explore the role and underlying mechanism of PCAF in vascular aging. The results demonstrated that the expression of PCAF was associated with age and aging, and remarkably increased expression of PCAF was present in human atherosclerotic coronary artery. Downregulation of PCAF could reduce angiotensin II (AngII)-induced senescence of rat aortic endothelial cells (ECs) in vitro. In addition, inhibition of PCAF with garcinol alleviated AngII-induced vascular senescence phenotype in mice. Downregulation of PCAF could alleviate AngII-induced oxidative stress injury in ECs and vascular tissue. Moreover, PCAF and nuclear factor erythroid-2-related factor 2 (Nrf2) could interact directly, and downregulation of PCAF alleviated vascular aging by promoting the activation of Nrf2 and enhancing the expression of its downstream anti-aging factors. The silencing of Nrf2 with small interfering RNA attenuated the protective effect of PCAF downregulation from vascular aging. These findings indicate that downregulation of PCAF alleviates oxidative stress by activating the Nrf2 signaling pathway and ultimately inhibits vascular aging. Thus, PCAF may be a promising target for aging-related cardiovascular disease.


Assuntos
Angiotensina II , Fator 2 Relacionado a NF-E2 , Animais , Humanos , Camundongos , Ratos , Envelhecimento , Angiotensina II/metabolismo , Regulação para Baixo , Células Endoteliais/metabolismo , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Fatores de Transcrição de p300-CBP/metabolismo , RNA Interferente Pequeno , Transdução de Sinais
6.
J Environ Manage ; 317: 115438, 2022 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-35653844

RESUMO

Health inequalities are globally widespread due to the regional socioeconomic inequalities. Myocardial infarction (MI) is a leading health problem causing deaths worldwide. Yet medical services for it are often inequitably distributed by region. Moreover, studies concerning MI's potential spatial risk factors generally suffer from difficulties in focusing on too few factors, inappropriate models, and coarse spatial grain of data. To address these issues, this paper integrates registered 1098 MI cases and urban multi-source spatio-temporal big data, and spatially analyses the risk factors for MI severity by applying an advanced interpretable model, the random forest algorithm (RFA)-based SHapley Additive exPlanations (SHAP) model. In addition, a community-scale model between spatio-temporal risk factors and MI cases is constructed to predict the MI severity of all communities in Wuhan, China. The results suggest that those risk factors (i.e., age of patients, medical quality, temperature changes, air pollution and urban habitat) affect the MI severity at the community scale. We found that Wuhan residents in the downtown area are at risk for high MI severity, and the surrounding suburb areas show a donut-shape pattern of risk for medium-to-high MI severity. These patterns draw our attention to the impact of spatial environmental risk factors on MI severity. Thus, this paper provides three recommendations for urban planning to reduce the risk and mortality from severe MI in the aspect of policy implication.


Assuntos
Poluição do Ar , Infarto do Miocárdio , Poluição do Ar/análise , China , Cidades , Humanos , Infarto do Miocárdio/epidemiologia , Fatores de Risco
7.
J Cell Mol Med ; 25(21): 10224-10235, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34601814

RESUMO

Cardiomyocyte apoptosis is the main reason of cardiac injury after myocardial ischaemia-reperfusion (I/R) injury (MIRI), but the role of p300/CBP-associated factor (PCAF) on myocardial apoptosis in MIRI is unknown. The aim of this study was to investigate the main mechanism of PCAF modulating cardiomyocyte apoptosis in MIRI. The MIRI model was constructed by ligation of the rat left anterior descending coronary vessel for 30 min and reperfusion for 24 h in vivo. H9c2 cells were harvested after induced by hypoxia for 6 h and then reoxygenation for 24 h (H/R) in vitro. The RNA interference PCAF expression adenovirus was transfected into rat myocardium and H9c2 cells. The area of myocardial infarction, cardiac function, myocardial injury marker levels, apoptosis, inflammation and oxidative stress were detected respectively. Both I/R and H/R remarkably upregulated the expression of PCAF, and downregulation of PCAF significantly attenuated myocardial apoptosis, inflammation and oxidative stress caused by I/R and H/R. In addition, downregulation of PCAF inhibited the activation of NF-κB signalling pathway in cardiomyocytes undergoing H/R. Pretreatment of lipopolysaccharide, a NF-κB pathway activator, could blunt these protective effects of PCAF downregulation on myocardial apoptosis in MIRI. These results highlight that downregulation of PCAF could reduce cardiomyocyte apoptosis by inhibiting the NF-κB pathway, thereby providing protection for MIRI. Therefore, PCAF might be a promising target for protecting against cardiac dysfunction induced by MIRI.


Assuntos
Apoptose/genética , Traumatismo por Reperfusão Miocárdica/etiologia , Traumatismo por Reperfusão Miocárdica/metabolismo , Miócitos Cardíacos/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais , Fatores de Transcrição de p300-CBP/genética , Animais , Biomarcadores , Linhagem Celular , Modelos Animais de Doenças , Suscetibilidade a Doenças , Regulação para Baixo , Regulação da Expressão Gênica , Testes de Função Cardíaca , Traumatismo por Reperfusão Miocárdica/diagnóstico , Estresse Oxidativo , Ratos , Fatores de Transcrição de p300-CBP/metabolismo
8.
BMC Cardiovasc Disord ; 21(1): 435, 2021 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-34521385

RESUMO

BACKGROUND: This study aimed to develop and validate a nomogram to predict probability of in-stent restenosis (ISR) in patients undergoing percutaneous coronary intervention (PCI). METHODS: Patients undergoing PCI with drug-eluting stents between July 2009 and August 2011 were retrieved from a cohort study in a high-volume PCI center, and further randomly assigned to training and validation sets. The least absolute shrinkage and selection operator (LASSO) regression model was used to screen out significant features for construction of nomogram. Multivariable logistic regression analysis was applied to build a nomogram-based predicting model incorporating the variables selected in the LASSO regression model. The area under the curve (AUC) of the receiver operating characteristics (ROC), calibration plot and decision curve analysis (DCA) were performed to estimate the discrimination, calibration and utility of the nomogram model respectively. RESULTS: A total of 463 patients with DES implantation were enrolled and randomized in the development and validation sets. The predication nomogram was constructed with five risk factors including prior PCI, hyperglycemia, stents in left anterior descending artery (LAD), stent type, and absence of clopidogrel, which proved reliable for quantifying risks of ISR for patients with stent implantation. The AUC of development and validation set were 0.706 and 0.662, respectively, indicating that the prediction model displayed moderate discrimination capacity to predict restenosis. The high quality of calibration plots in both datasets demonstrated strong concordance performance of the nomogram model. Moreover, DCA showed that the nomogram was clinically useful when intervention was decided at the possibility threshold of 9%, indicating good utility for clinical decision-making. CONCLUSIONS: The individualized prediction nomogram incorporating 5 commonly clinical and angiographic characteristics for patients undergoing PCI can be conveniently used to facilitate early identification and improved screening of patients at higher risk of ISR.


Assuntos
Doença da Artéria Coronariana/terapia , Reestenose Coronária/etiologia , Técnicas de Apoio para a Decisão , Nomogramas , Intervenção Coronária Percutânea/efeitos adversos , Intervenção Coronária Percutânea/instrumentação , Stents , Idoso , Tomada de Decisão Clínica , Angiografia Coronária , Doença da Artéria Coronariana/diagnóstico por imagem , Reestenose Coronária/diagnóstico por imagem , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Sistema de Registros , Reprodutibilidade dos Testes , Medição de Risco , Fatores de Risco , Fatores de Tempo , Resultado do Tratamento
9.
Curr Res Transl Med ; 69(1): 103270, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33268288

RESUMO

BACKGROUND: Hyperhomocysteinemia (HHcy) is one of the major risk factors of cardiovascular diseases. Metformin acts as a cardioprotective role in several cardiovascular diseases, including ischemia/reperfusion, atherosclerosis, and myocardial infarction. However, whether metformin protects against HHcy-induced cardiac hypertrophy is unclear. METHODS AND RESULTS: HHcy model was established in C57BL/6 mice with high L-methionine (L-MET) diet for 12 weeks. AC16 cells were exposed to homocysteine (Hcy) and then intervened with different concentrations of metformin in in vitro studies. The results showed that HHcy was able to induce cardiac hypertrophy, and metformin could abrogate this effect. HHcy increased the fibrosis area and induced apoptosis in the myocardium, whereas metformin could reverse the detrimental effects above. TUNEL assay showed that metformin was able to decrease Hcy-induced apoptosis in AC16 cells. Moreover, western blotting assay revealed that metformin could decrease Hcy-induced expression of Bax and cleaved caspase3, and increase the expression of Bcl-2. CONCLUSIONS: This study demonstrates that metformin is able to attenuate HHcy-induced cardiac hypertrophy by decreasing myocardial fibrosis and apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Cardiomegalia , Hiper-Homocisteinemia , Metformina/farmacologia , Miocárdio/patologia , Adulto , Animais , Cardiomegalia/tratamento farmacológico , Cardiomegalia/etiologia , Cardiomegalia/patologia , Células Cultivadas , Fibrose , Coração/efeitos dos fármacos , Humanos , Hiper-Homocisteinemia/complicações , Hiper-Homocisteinemia/tratamento farmacológico , Hiper-Homocisteinemia/patologia , Masculino , Metformina/uso terapêutico , Camundongos , Camundongos Endogâmicos C57BL
10.
J Cardiovasc Pharmacol ; 76(5): 540-548, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33170591

RESUMO

The outbreak of coronavirus disease 2019 (COVID-19) has rapidly spread worldwide. This study sought to share our experiences with in-hospital management and outcomes of acute myocardial infarction (AMI) during the COVID-19 pandemic. We retrospectively analyzed consecutive AMI patients, including those with ST-elevation myocardial infarction (STEMI) and non-STEMI (NSTEMI), from February 1, 2020, to April 15, 2020 (during the COVID-19 pandemic), and from January 1, 2019, to December 31, 2019 (before the COVID-19 pandemic), respectively. Fifty-three AMI patients (31 STEMI, 22 NSTEMI) during the COVID-19 pandemic were matched to 53 AMI patients before the pandemic. Baseline characteristics were comparable between the matched patients. STEMI patients during the COVID-19 pandemic had a longer delay time, less primary or remedial PCI and more emergency thrombolysis than those before the pandemic. Less coronary angiography and stenting were performed in AMI patients during the COVID-19 pandemic than before the pandemic. There were no statistically significant differences in the clinical outcomes between the matched patients. However, STEMI patients during the COVID-19 pandemic had a 4-fold (12.9% vs. 3.2%) increase in all-cause mortality rate compared with those before the pandemic. AMI combined with COVID-19 infection was associated with higher rates of mortality than AMI alone. This study demonstrates that the COVID-19 pandemic results in significant reperfusion delays in STEMI patients and has a marked impact on the treatment options selection in AMI patients. The mortality rate of STEMI patients exhibits an increasing trend during the pandemic of COVID-19.


Assuntos
Serviço Hospitalar de Cardiologia/tendências , Infecções por Coronavirus , Infarto do Miocárdio sem Supradesnível do Segmento ST/terapia , Avaliação de Processos e Resultados em Cuidados de Saúde/tendências , Pandemias , Intervenção Coronária Percutânea/tendências , Pneumonia Viral , Infarto do Miocárdio com Supradesnível do Segmento ST/terapia , Terapia Trombolítica/tendências , Tempo para o Tratamento/tendências , Idoso , COVID-19 , China , Angiografia Coronária/tendências , Infecções por Coronavirus/diagnóstico , Infecções por Coronavirus/mortalidade , Infecções por Coronavirus/transmissão , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Infarto do Miocárdio sem Supradesnível do Segmento ST/diagnóstico , Infarto do Miocárdio sem Supradesnível do Segmento ST/mortalidade , Admissão do Paciente , Intervenção Coronária Percutânea/efeitos adversos , Intervenção Coronária Percutânea/instrumentação , Pneumonia Viral/diagnóstico , Pneumonia Viral/mortalidade , Pneumonia Viral/transmissão , Estudos Retrospectivos , Fatores de Risco , Infarto do Miocárdio com Supradesnível do Segmento ST/diagnóstico , Infarto do Miocárdio com Supradesnível do Segmento ST/mortalidade , Terapia Trombolítica/efeitos adversos , Terapia Trombolítica/mortalidade , Fatores de Tempo , Resultado do Tratamento
11.
Int J Med Sci ; 17(9): 1196-1206, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32547315

RESUMO

Cardiomyocyte autophagy plays an important role in myocardial ischemia-reperfusion injury (MIRI). P300/CBP-associated factor (PCAF) was involved in the regulation of autophagy. However, the role of PCAF in MIRI is currently unknown. This study was to investigate whether downregulation of PCAF attenuate MIRI. The results showed that the expression of PCAF was significantly increased in MIRI in vivo and in vitro. Downregulation of PCAF not only inhibited autophagy and damage of H9c2 cells induced by hypoxia-reoxygenation, but also reduced autophagy and myocardial infarct size during myocardial ischemia-reperfusion in rats. In addition, downregulation of PCAF promoted activation of PI3K/Akt/mTOR signaling pathway in cardiomyocytes during hypoxia-reoxygenation. Wortmannin, a PI3K/Akt inhibitor, could abrogate the effects of downregulation of PCAF on cardiomyocytes autophagy. These results demonstrated that downregulation of PCAF alleviated MIRI by inhibiting cardiomyocyte autophagy through PI3K/Akt/mTOR signaling pathway. Thus, PCAF may be a potential target for prevention and treatment of MIRI.


Assuntos
Autofagia/fisiologia , Traumatismo por Reperfusão Miocárdica/metabolismo , Fatores de Transcrição de p300-CBP/metabolismo , Animais , Western Blotting , Linhagem Celular , Espectroscopia de Ressonância Magnética , Masculino , Microscopia Eletrônica de Transmissão , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Wortmanina/farmacologia , Fatores de Transcrição de p300-CBP/genética
12.
J Cell Mol Med ; 24(1): 973-983, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31736204

RESUMO

Patients with diabetes have an increased risk of vascular complications. Suv39h1, a histone methyltransferase, plays a protective role against myocardial injury in diabetes. Herein, we intend to explore whether Suv39h1 could affect neointimal formation after vascular injury in diabetic rats and reveal the underlying mechanism. In this study, we generated adenovirus expressing Suv39h1 as well as lentivirus expressing Suv39h1-targeting shRNA and evaluated the significance of Suv39h1 in vascular smooth muscle cells (VSMCs) under diabetic conditions. In vitro, we examined proliferative and migratory behaviours as well as the underlying signalling mechanisms in VSMCs in response to high glucose treatment. In vivo, we induced diabetes in SD rats with streptozocin and established the common carotid artery balloon injury model. Suv39h1 was found to be both necessary and sufficient to promote VSMC proliferation and migration under high glucose conditions. We observed corresponding changes in intracellular signalling molecules including complement C3 and phosphor-ERK1/2. However, either up-regulating or down-regulating Suv39h1, phosphor-p38 level was not significantly affected. Consistently, Suv39h1 overexpression led to accelerated neointima formation, while knocking down Suv39h1 reduced it following carotid artery injury in diabetic rats. Using microarray analyses, we showed that altering the Suv39h1 level in vivo dramatically altered the expression of myriad genes mediating different biological processes and molecular function. This study reveals the novel role of Suv39h1 in VSMCs of diabetes and suggests its potential role as a therapeutic target in diabetic vascular injury.


Assuntos
Lesões das Artérias Carótidas/complicações , Diabetes Mellitus Experimental/fisiopatologia , Regulação da Expressão Gênica , Metiltransferases/antagonistas & inibidores , Neointima/prevenção & controle , Proteínas Repressoras/antagonistas & inibidores , Animais , Movimento Celular , Proliferação de Células , Células Cultivadas , Perfilação da Expressão Gênica , Masculino , Metiltransferases/genética , Metiltransferases/metabolismo , Neointima/etiologia , Neointima/patologia , Ratos , Ratos Sprague-Dawley , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transdução de Sinais
13.
Atherosclerosis ; 288: 76-84, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31330382

RESUMO

BACKGROUND AND AIMS: Neointimal hyperplasia resulting from pathological vascular smooth muscle cells (VSMCs) activation is a common pathophysiological basis for numerous proliferative vascular diseases, such as restenosis. Suv39h1, an important transcription suppressor, may be involved in this process. Herein, we investigated the role of Suv39h1 in pathological intimal hyperplasia and its possible mechanisms in vitro and in vivo. METHODS: An adenovirus vector for Suv39h1 overexpression and a lentiviral vector for its downregulation were constructed and used to transfect cultured VSMCs in vitro. The functional changes in VSMCs stimulated by angiotensin II (Ang II) were observed and the possible mechanism was investigated. Additionally, rat carotid arteries with balloon injury were locally transfected with these viral vectors and changes in neointima formation, proliferating cell nuclear antigen (Pcna) expression and collagen deposition were examined. RESULTS: Upon Ang II stimulation, the expression of Suv39h1 and inhibitor of DNA binding 3 (Id3) was significantly increased. Suv39h1 downregulation inhibited Ang II-stimulated migration and proliferation of VSMCs, antagonized the production of Id3 and promoted p21 and p27Kip1 expression. In contrast, Suv39h1 overexpression had the opposite effects. Suv39h1 regulated the transcription of p21 and p27Kip1 by controlling H3K9me3 in the proximal promoter regions. Consistent with the VSMCs results, Suv39h1 and Id3 expression was significantly increased in blood vessels after balloon injury. Suv39h1 downregulation inhibited intimal hyperplasia, and attenuated Pcna expression and collagen synthesis in the intima, while Suv39h1 overexpression had the opposite effects. CONCLUSIONS: Suv39h1 downregulation effectively inhibited neointimal hyperplasia after vascular injury.


Assuntos
Metiltransferases/metabolismo , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Neointima , Proteínas Repressoras/metabolismo , Remodelação Vascular , Lesões do Sistema Vascular/metabolismo , Animais , Lesões das Artérias Carótidas , Artéria Carótida Primitiva/metabolismo , Artéria Carótida Primitiva/patologia , Artéria Carótida Primitiva/fisiopatologia , Movimento Celular , Proliferação de Células , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p21/genética , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/genética , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Modelos Animais de Doenças , Regulação para Baixo , Hiperplasia , Proteínas Inibidoras de Diferenciação/genética , Proteínas Inibidoras de Diferenciação/metabolismo , Masculino , Metiltransferases/genética , Músculo Liso Vascular/lesões , Músculo Liso Vascular/patologia , Músculo Liso Vascular/fisiopatologia , Miócitos de Músculo Liso/patologia , Ratos Sprague-Dawley , Proteínas Repressoras/genética , Transdução de Sinais , Lesões do Sistema Vascular/genética , Lesões do Sistema Vascular/patologia , Lesões do Sistema Vascular/fisiopatologia
14.
Biochem Biophys Res Commun ; 513(1): 41-48, 2019 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-30935684

RESUMO

P300/CBP-associated factor (PCAF) regulates vascular inflammation. This study was to explore the effect of PCAF on the proliferation and migrationof vascular smooth muscle cells (VSMCs) and neointimal hyperplasia in balloon-injured rat carotid artery. Downregulation of PCAF remarkably suppressed VSMCs proliferation and migration induced by lipopolysaccharide, and also significantly inhibit the nuclear translocation of nuclear factor-kappaB p65. Meanwhile, downregulation of PCAF inhibited the mRNA expression of tumor necrosis factor-α and interleukin-6, and also the levels in culture supernatants. Moreover, downregulation of PCAF profoundly reduced the intima area and the ratio of intima area to media area in balloon-injured rat carotid artery. In addition, the expression of PCNA and NF-κB p65 in intima were decreased by downregulation of PCAF. These results highlight that PCAF may be a potential target for prevention and treatment of neointimal hyperplasia and restenosis after angioplasty.


Assuntos
Inflamação/imunologia , Músculo Liso Vascular/imunologia , Fator de Transcrição RelA/imunologia , Fatores de Transcrição de p300-CBP/genética , Animais , Movimento Celular , Proliferação de Células , Células Cultivadas , Regulação para Baixo , Inflamação/genética , Inflamação/patologia , Masculino , Músculo Liso Vascular/citologia , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/imunologia , Miócitos de Músculo Liso/patologia , Ratos Sprague-Dawley , Fator de Transcrição RelA/genética , Túnica Íntima/imunologia , Túnica Íntima/patologia , Fatores de Transcrição de p300-CBP/imunologia
15.
Biol Pharm Bull ; 42(1): 34-42, 2019 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-30393274

RESUMO

Abnormal proliferation and migration of vascular smooth muscle cells (VSMCs) and the chronic inflammation regulated by various inflammatory factors are the major pathological processes in the development of neointimal hyperplasia and in-stent restenosis after angioplasty. Cantharidin is a potent and selective inhibitor of protein phosphatase 2A, which plays pivotal roles in cell cycle progression, cell fate, and inflammation. This study was to explore whether Cantharidin could inhibit VSMCs proliferation, migration and inflammation. Transwell migration assay, Cell Counting Kit 8 and flow cytometry were performed. Western blot, Quantitative real-time PCR, and enzyme-linked immunosorbent assay (ELISA) were used to detect the expression of the markers. Results showed that Cantharidin remarkably suppressed VSMCs proliferation and migration induced by platelet derived growth factor (PDGF)-BB. Meanwhile, Cantharidin could significantly inhibit the phosphorylation of Akt (P-AKT) and p38 mitogen-activated protein kinase (MAPK) (P-p38), the expression of p38 MAPK (p38), and also the phosphorylation level of nuclear factor-kappaB (NF-κB) p65 (p65). Cantharidin obviously inhibited the expression of interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α), and also the level of IL-6 and TNF-α in culture supernatants. Inhibitors for p38 MAPK, phosphatidylinositol 3-kinase (PI3K) /AKT and NF-κB signaling pathways did not affect the inhibition of Cantharidin on VSMCs proliferation, migration and inflammation. These findings indicated that Cantharidin could significantly inhibit the proliferation, migration and inflammatory response of VSMCs, which suggested that Cantharidin may be a potential inhibitor for neointimal hyperplasia and restenosis after angioplasty.


Assuntos
Anti-Inflamatórios/farmacologia , Cantaridina/farmacologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Mediadores da Inflamação/antagonistas & inibidores , Músculo Liso Vascular/efeitos dos fármacos , Animais , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Células Cultivadas , Relação Dose-Resposta a Droga , Mediadores da Inflamação/metabolismo , Masculino , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Ratos , Ratos Sprague-Dawley
16.
Mol Genet Genomics ; 293(4): 883-894, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29536180

RESUMO

MicroRNA-17-5p (miR-17-5p) was indicated to suppress the formation of blood vessels, which is associated with cardiac function after myocardial infarction. In this study, the relationship between miR-17-5p and cardiac function was researched. Human umbilical vein endothelial cells were infected with adenoviruses. Apoptosis was determined by Annexin V-7AAD/PI. Real-time RT-PCR was used to evaluate miR-17-5p and ERK levels. Western blotting was used to determine the levels of ERK, the anti-apoptosis protein bcl-2 and apoptosis proteins, including bax, caspase 3, and caspase 9. An in vivo acute myocardial infarction (AMI) model was established in SD male rats. Heart function was evaluated by echocardiography prior to inducing AMI and after 7 and 28 days later. The heart was removed to perform histological examination, real-time RT-PCR, and western blotting, as described above. The result indicated that the ERK pathway was activated by miR-17-5p downregulation and an increase in the level of the anti-apoptosis protein bcl-2; however, the levels of apoptosis proteins (bax/caspase 3/caspase 9) were decreased. The results were completely reversed when miR-17-5p was up-regulated. At 7 and 28 days after the induction of AMI, in the miR-17-5p inhibition group, the infarction areas and collagen fibers were decreased, apoptosis in cardiac tissues was inhibited, and the endothelial growth process was promoted. Therefore, MiR-17-5p silencing protects heart function after AMI through decreasing the rate of apoptosis and repairing vascular injury.


Assuntos
Apoptose , Regulação para Baixo , MicroRNAs/biossíntese , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Modelos Animais de Doenças , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miocárdio/patologia , Ratos , Ratos Sprague-Dawley
17.
Atherosclerosis ; 257: 152-163, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28135625

RESUMO

BACKGROUND AND AIMS: Deregulation of histone demethylase KDM3a, an important regulator for H3K9 methylation, is correlated with obesity and abnormal metabolism in rodent models. However, the function of KDM3a in vascular remodeling under diabetic condition is unknown. METHODS: Adenoviruses expressing KDM3a and lentiviruses expressing KDM3a-targeting siRNA were generated to study the role of KDM3a both in vivo and in vitro. The carotid artery balloon injury model was established in diabetic SD rats to evaluate the significance of KDM3a in vascular injury. RESULTS: Diabetic vessels were associated with sustained loss of histone H3 lysine 9 di-methylation (H3K9me2) and elevation of KDM3a. This phenomenon was induced by high glucose (HG) and was persistently present even after removal from diabetic condition and high glucose in vascular smooth muscle cells (VSMCs). After 28-day balloon injury, KDM3a overexpression accelerated while KDM3a knockdown reduced neointima formation, following vascular injury in diabetic rats without glucose control. Microarray analysis revealed KDM3a altered the expression of vascular remodeling genes; particularly, it mediated the Rho/ROCK and AngII/AGTR1 pathways. In the in vivo study, HG and Ang II-stimulated proliferation and migration of VSMCs were enhanced by KDM3a overexpression, whereas markedly prevented by KDM3a knockdown. KDM3a regulated the transcription of AGTR1 and ROCK2 via controlling H3K9me2 in the proximal promoter regions. CONCLUSIONS: Histone demethylase KDM3a promotes vascular neointimal hyperplasia in diabetic rats via AGTR1 and ROCK2 signaling pathways. Targeting KDM3a might represent a promising therapeutic approach for the prevention of coronary artery disease with diabetes.


Assuntos
Lesões das Artérias Carótidas/enzimologia , Proliferação de Células , Diabetes Mellitus Experimental/enzimologia , Histona Desmetilases/metabolismo , Histona Desmetilases com o Domínio Jumonji/metabolismo , Músculo Liso Vascular/enzimologia , Miócitos de Músculo Liso/enzimologia , Neointima , Animais , Artérias Carótidas/enzimologia , Artérias Carótidas/patologia , Lesões das Artérias Carótidas/genética , Lesões das Artérias Carótidas/patologia , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patologia , Regulação da Expressão Gênica , Células HEK293 , Histona Desmetilases/genética , Histonas/metabolismo , Humanos , Hiperplasia , Histona Desmetilases com o Domínio Jumonji/genética , Masculino , Metilação , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , Regiões Promotoras Genéticas , Interferência de RNA , Ratos Sprague-Dawley , Receptor Tipo 1 de Angiotensina/genética , Receptor Tipo 1 de Angiotensina/metabolismo , Transdução de Sinais , Fatores de Tempo , Transcrição Gênica , Transfecção , Quinases Associadas a rho/genética , Quinases Associadas a rho/metabolismo
18.
Int J Mol Med ; 37(1): 189-96, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26530537

RESUMO

The epigenetic modification of vascular smooth muscle cell (VSMC) phenotypic switching, proliferation, migration, apoptosis and extracellular matrix synthesis is known to occur in atherosclerosis. The aim of the present study was to investigate the effects of IOX1, a Jumonji domain-containing 2A (JMJD2A) inhibitor, on regulation of the cell cycle in angiotensin II (Ang II)-stimulated VSMCs and to elucidate the possible mechanisms involved. The proliferation and migration of the Ang II-stimulated VSMCs in the presence or absence of IOX1 were evaluated in vitro. Flow cytometric analysis was used to determine the effects of IOX1 on cell cycle progression. RT-qPCR and western blot analysis were carried out to measure the expression levels of cell cycle-related genes. The trimethylation of histone H3 lysine 9 (H3K9me3) at the promoters of these genes was detected by chromatin immunoprecipitation (ChIP) assay. We confirmed that the JMJD2A levels were increased, whereas the H3K9me3 levels were decreased in the Ang II-stimulated VSMCs. The inhibition of JMJD2A by IOX1 suppressed the Ang II-induced cell proliferation, migration and cell cycle progression by inhibiting cyclin D1 expression and increasing p21 expression. The underlying mechanisms were related to the restoration of the H3K9me3 levels at the promoters of these genes. In conclusion, the findings of our study indicate that IOX1 exerts its anti-proliferative and anti-migratory effects by regulating the expression of the cell cycle-related proteins, cyclin D1 and p21.


Assuntos
Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Histona Desmetilases/antagonistas & inibidores , Hidroxiquinolinas/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Miócitos de Músculo Liso/efeitos dos fármacos , Angiotensina II/metabolismo , Animais , Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/genética , Células Cultivadas , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , Ratos Sprague-Dawley
19.
Biol Pharm Bull ; 38(8): 1134-41, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26235577

RESUMO

Endothelial senescence has been proposed to be involved in endothelial dysfunction and atherogenesis. Curcumin, a natural phenol, possesses antioxidant and anti-inflammatory properties. However, the effect of curcumin on endothelial senescence is unclear. This study explores the effect of curcumin on hydrogen peroxide (H2O2)-induced endothelial premature senescence and the mechanisms involved. Human umbilical vein endothelial cells (HUVECs) were cultured, and premature senescence was induced with 100 µM H2O2. Results showed that pretreatment with curcumin significantly attenuated the H2O2-induced HUVECs' premature senescence, which was evidenced by a decreased percentage of senescence-associated ß-galactosidase positive cells, improved cell division and decreased expression of senescence-associated protein p21 (all p<0.05). Pretreatment with curcumin decreased oxidative stress and apoptosis in H2O2-treated HUVECs. Treatment of HUVECs with H2O2 also down-regulated the phosphorylation of endothelial nitric oxide synthase (eNOS), decreased the level of nitric oxide in the culture medium, and inhibited the protein expression and enzymatic activity of silent information regulator 1 (SIRT1), while pretreatment with curcumin partly reversed these effects (all p<0.05). Treatment with curcumin alone enhanced the enzymatic activity of SIRT1, but didn't affect cellular senescence, cell growth or apoptosis compared to the Control. The inhibition of SIRT1 using SIRT1 short interfering RNA (siRNA) could decrease the expression and phosphorylation of eNOS and abrogate the protective effect of curcumin on H2O2-induced premature senescence. These findings suggest that curcumin could attenuate oxidative stress-induced HUVECs' premature senescence via the activation of SIRT1.


Assuntos
Senescência Celular/efeitos dos fármacos , Curcuma/química , Curcumina/farmacologia , Endotélio Vascular/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Peróxido de Hidrogênio/metabolismo , Sirtuína 1/metabolismo , Antioxidantes/farmacologia , Apoptose , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Regulação para Baixo , Endotélio Vascular/citologia , Endotélio Vascular/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células Endoteliais da Veia Umbilical Humana/fisiologia , Humanos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Fosforilação , Extratos Vegetais/farmacologia , RNA Interferente Pequeno , beta-Galactosidase/metabolismo
20.
Exp Ther Med ; 8(1): 229-232, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24944626

RESUMO

High mobility group box 1 protein (HMGB1) has an important role in myocardial ischemia/reperfusion (I/R) injury. Sodium butyrate, an inhibitor of histone deacetylase, has been shown to inhibit HMGB1 expression. In the present study, the effect of sodium butyrate on myocardial I/R injury in rats was investigated. Anesthetized male rats were intraperitoneally administered sodium butyrate (100 or 300 mg/kg) 30 min prior to the induction of ischemia. The rats were then subjected to ischemia for 30 min followed by reperfusion for 4 h. Infarct size, lactate dehydrogenase (LDH), creatine kinase (CK) and superoxide dismutase (SOD) activity and malondialdehyde (MDA) levels were then measured. The expression of HMGB1 was assessed using western blot analysis. The results demonstrated that pretreatment with sodium butyrate (300 mg/kg) significantly reduced the infarct size, as well as the levels of LDH and CK (P<0.05). In addition, sodium butyrate (300 mg/kg) was shown to significantly inhibit the I/R-induced increase in the level of MDA and reduction in the level of SOD (P<0.05). Furthermore, treatment with sodium butyrate (300 mg/kg) was found to significantly inhibit the expression of TNF-α, IL-6 and HMGB1 induced by I/R injury (P<0.05). In conclusion, the results from the present study suggest that preconditioning with sodium butyrate may attenuate myocardial I/R injury by inhibition of the expression of inflammatory mediators during myocardial I/R.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA