Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Prostaglandins Other Lipid Mediat ; 167: 106740, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37119935

RESUMO

Epoxyeicosatrienoic acids (EETs), which are synthesized from arachidonic acid by cytochrome P450 epoxygenases, function primarily as autocrine and paracrine effectors in the cardiovascular system. So far, most research has focused on the vasodilatory, anti-inflammatory, anti-apoptotic and mitogenic properties of EETs in the systemic circulation. However, whether EETs could suppress tissue factor (TF) expression and prevent thrombus formation remains unknown. Here we utilized in vivo and in vitro models to investigate the effects and underlying mechanisms of exogenously EETs on LPS induced TF expression and inferior vein cava ligation induced thrombosis. We observed that the thrombus formation rate and the size of the thrombus were greatly reduced in 11,12-EET treated mice,accompanied by decreased TF and inflammatory cytokines expression. Further in vitro studies showed that by enhancing p38 MAPK activation and subsequent tristetraprolin (TTP) phosphorylation, LPS strengthened the stability of TF mRNA and induced increased TF expression. However, by strengthening PI3K-dependent Akt phosphorylation, which acted as a negative regulator of p38-TTP signaling pathway,11,12-EET reduced LPS-induced TF expression in monocytes. In addition, 11,12-EET inhibited LPS-induced NF-κB nuclear translocation by activating the PI3K/Akt pathway. Further study indicated that the inhibitory effect of 11,12-EET on TF expression was mediated by antagonizing LPS-induced activation of thromboxane prostanoid receptor. In conclusion, our study demonstrated that 11,12-EET prevented thrombosis by reducing TF expression and targeting the CYP2J2 epoxygenase pathway may represent a novel approach to mitigate thrombosis related diseases.


Assuntos
Proteínas Proto-Oncogênicas c-akt , Trombose , Animais , Camundongos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Lipopolissacarídeos/farmacologia , Tromboplastina/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Sistema Enzimático do Citocromo P-450/metabolismo , Transdução de Sinais , Citocromo P-450 CYP2J2 , Ácido 8,11,14-Eicosatrienoico/metabolismo , Trombose/tratamento farmacológico , Estabilidade de RNA
2.
Am J Physiol Heart Circ Physiol ; 321(2): H353-H368, 2021 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-34142887

RESUMO

Arterial stiffness, a consequence of smoking, is an underlying risk factor of cardiovascular diseases. Epoxyeicosatrienoic acids (EETs), hydrolyzed by soluble epoxide hydrolase (sEH), have beneficial effects against vascular dysfunction. However, the role of sEH knockout in nicotine-induced arterial stiffness was not characterized. We hypothesized that sEH knockout could prevent nicotine-induced arterial stiffness. In the present study, Ephx2 (the gene encodes sEH enzyme) null (Ephx2-/-) mice and wild-type (WT) littermate mice were infused with or without nicotine and administered with or without nicotinamide [NAM, sirtuin-1 (SIRT1) inhibitor] simultaneously for 4 wk. Nicotine treatment increased sEH expression and activity in the aortas of WT mice. Nicotine infusion significantly induced vascular remodeling, arterial stiffness, and SIRT1 deactivation in WT mice, which was attenuated in Ephx2 knockout mice (Ephx2-/- mice) without NAM treatment. However, the arterial protective effects were gone in Ephx2-/- mice with NAM treatment. In vitro, 11,12-EET treatment attenuated nicotine-induced matrix metalloproteinase 2 (MMP2) upregulation via SIRT1-mediated yes-associated protein (YAP) deacetylation. In conclusion, sEH knockout attenuated nicotine-induced arterial stiffness and vascular remodeling via SIRT1-induced YAP deacetylation.NEW & NOTEWORTHY We presently show that sEH knockout repressed nicotine-induced arterial stiffness and extracellular matrix remodeling via SIRT1-induced YAP deacetylation, which highlights that sEH is a potential therapeutic target in smoking-induced arterial stiffness and vascular remodeling.


Assuntos
Aorta/efeitos dos fármacos , Epóxido Hidrolases/genética , Niacinamida/farmacologia , Nicotina/farmacologia , Agonistas Nicotínicos/farmacologia , Sirtuína 1/metabolismo , Rigidez Vascular/efeitos dos fármacos , Complexo Vitamínico B/farmacologia , Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Ácido 8,11,14-Eicosatrienoico/farmacologia , Proteínas Adaptadoras de Transdução de Sinal/efeitos dos fármacos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Aorta/metabolismo , Aorta/fisiopatologia , Metaloproteinase 2 da Matriz/efeitos dos fármacos , Metaloproteinase 2 da Matriz/genética , Camundongos , Camundongos Knockout , Sirtuína 1/antagonistas & inibidores , Sirtuína 1/efeitos dos fármacos , Rigidez Vascular/genética , Vasodilatadores/farmacologia , Proteínas de Sinalização YAP
3.
Clin Cardiol ; 43(12): 1478-1493, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33026120

RESUMO

BACKGROUD: The association between underlying comorbidities and cardiac injury and the prognosis in coronavirus disease 2019 (COVID-19) patients was assessed in this study. HYPOTHESIS: The underlying comorbidities and cardiac injury may be associated with the prognosis in COVID-19 patients. METHODS: A systematic search was conducted in PubMed, EMBASE, Web of science, and The Cochrane library from December 2019 to July 2020. The odds ratio (OR) and 95% confidence intervals (95% CI) were used to estimate the probability of comorbidities and cardiac injury in COVID-19 patients with or without severe type, or in survivors vs nonsurvivors of COVID-19 patients. RESULTS: A total of 124 studies were included in this analysis. A higher risk for severity was observed in COVID-19 patients with comorbidities. The pooled result in patients with hypertension (OR 2.57, 95% CI: 2.12-3.11), diabetes (OR 2.54, 95% CI: 1.89-3.41), cardiovascular diseases (OR 3.86, 95% CI: 2.70-5.52), chronic obstractive pulmonary disease (OR 2.71, 95% CI: 1.98-3.70), chronic kidney disease (OR 2.20, 95% CI: 1.27-3.80), and cancer (OR 2.42, 95% CI: 1.81-3.22) respectively. All the comorbidities presented a higher risk of mortality. Moreover, the prevalence of acute cardiac injury is higher in severe group than in nonsevere group, and acute cardiac injury is associated with an increased risk for in-hospital mortality. CONCLUSION: Comorbidities and acute cardiac injury are closely associated with poor prognosis in COVID-19 patients. It is necessary to continuously monitor related clinical indicators of organs injury and concern comorbidities in COVID-19 patients.


Assuntos
COVID-19/mortalidade , Índice de Gravidade de Doença , COVID-19/fisiopatologia , Doenças Cardiovasculares/mortalidade , Causas de Morte , Comorbidade , Diabetes Mellitus/mortalidade , Feminino , Humanos , Hipertensão/mortalidade , Masculino , Neoplasias/mortalidade , Pandemias/estatística & dados numéricos , Prognóstico , Insuficiência Renal Crônica/mortalidade
5.
Clin Cardiol ; 43(3): 235-241, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31777973

RESUMO

The optimal antiplatelet therapy after percutaneous coronary intervention (PCI) remains to be elucidated. Monotherapy with a P2Y12 inhibitor may be inferior to dual antiplatelet therapy in patients after PCI. PubMed, EMBASE (by Ovidsp), Web of Science, and The Cochrane Library were searched from database inception to 2 October 2019. The composite of cardiovascular outcomes, all-cause mortality, myocardial infarction (MI), stroke, stent thrombosis, and major bleeding were evaluated. Pooled outcomes were presented as relative risk (RR) and 95% confidence intervals (CIs). A total of four trials randomizing 29 089 participants were included. Compared with the dual antiplatelet therapy group (n = 14 559), the P2Y12 inhibitor monotherapy group (n = 14 530) significantly decreased the incidence of bleeding events (2.0% vs 3.1%; RR: 0.60; 95% CI: 0.43-0.84; P = .005). There were no significant differences in all-cause mortality (1.3% vs 1.5%; RR: 0.87; 95% CI, 0.71-1.06; P = .16), myocardial infarction (2.1% vs 1.9%; RR, 1.06; 95% CI, 0.90-1.25; P = .46), stroke (0.6% vs 0.5%; RR, 1.18; 95% CI, 0.67-2.07; P = .57), or stent thrombosis (0.5% vs 0.4%; RR, 1.14; 95% CI, 0.81-1.61; P = .44) between the two groups. P2Y12 inhibitor monotherapy did not show any significant difference in the adverse cardiac and cerebrovascular events, but markedly decreased the risk of bleeding among patients after PCI vs dual antiplatelet therapy. However, it still needs to be further confirmed due to limited data.


Assuntos
Plaquetas/efeitos dos fármacos , Doença da Artéria Coronariana/terapia , Intervenção Coronária Percutânea , Inibidores da Agregação Plaquetária/uso terapêutico , Antagonistas do Receptor Purinérgico P2Y/uso terapêutico , Receptores Purinérgicos P2Y12/efeitos dos fármacos , Idoso , Plaquetas/metabolismo , Doença da Artéria Coronariana/diagnóstico , Doença da Artéria Coronariana/mortalidade , Trombose Coronária/mortalidade , Trombose Coronária/prevenção & controle , Feminino , Hemorragia/induzido quimicamente , Humanos , Masculino , Pessoa de Meia-Idade , Infarto do Miocárdio/mortalidade , Infarto do Miocárdio/prevenção & controle , Intervenção Coronária Percutânea/efeitos adversos , Intervenção Coronária Percutânea/instrumentação , Intervenção Coronária Percutânea/mortalidade , Inibidores da Agregação Plaquetária/efeitos adversos , Antagonistas do Receptor Purinérgico P2Y/efeitos adversos , Ensaios Clínicos Controlados Aleatórios como Assunto , Receptores Purinérgicos P2Y12/sangue , Medição de Risco , Fatores de Risco , Stents , Acidente Vascular Cerebral/mortalidade , Acidente Vascular Cerebral/prevenção & controle , Fatores de Tempo , Resultado do Tratamento
6.
Hum Mutat ; 39(9): 1238-1245, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29920840

RESUMO

Idiopathic pulmonary fibrosis (IPF) is a genetic heterogeneous disease with high mortality and poor prognosis. However, a large fraction of genetic cause remains unexplained, especially in sporadic IPF (∼80% IPF). By systemically reviewing related literature and potential pathogenic pathways, 92 potentially IPF-related genes were selected and sequenced in genomic DNAs from 253 sporadic IPF patients and 125 matched health controls using targeted massively parallel next-generation sequencing. The identified risk variants were confirmed by Sanger sequencing. We identified two pathogenic and 10 loss-of-function (LOF) candidate variants, accounting for 4.74% (12 out of 253) of all the IPF cases. In burden tests, rare missense variants in three genes (CSF3R, DSP, and LAMA3) were identified that have a statistically significant relationship with IPF. Four common SNPs (rs3737002, rs2296160, rs1800470, and rs35705950) were observed to be statistically associated with increased risk of IPF. In the cumulative risk model, high risk subjects had 3.47-fold (95%CI: 2.07-5.81, P = 2.34 × 10-6 ) risk of developing IPF compared with low risk subjects. We drafted a comprehensive map of genetic risks (including both rare and common candidate variants) in patients with IPF, which could provide insights to help in understanding mechanisms, providing genetic diagnosis, and predicting risk for IPF.


Assuntos
Desmoplaquinas/genética , Fibrose Pulmonar Idiopática/genética , Laminina/genética , Receptores de Fator Estimulador de Colônias/genética , Feminino , Predisposição Genética para Doença , Genoma Humano/genética , Estudo de Associação Genômica Ampla , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Mutação de Sentido Incorreto/genética , Polimorfismo de Nucleotídeo Único/genética , Fatores de Risco , Transdução de Sinais/genética
7.
Biochem Biophys Res Commun ; 433(4): 519-25, 2013 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-23524260

RESUMO

Pulmonary hypertension (PH) is an important pathophysiological process in the development of many diseases. However, the mechanism responsible for the development of PH remains unknown. The objective of the study was to explore the possible impact of sulfur dioxide (SO2) on the endogenous hydrogen sulfide (H2S) pathway in rats with PH induced by high pulmonary blood flow. Compared with sham group, the systolic pulmonary artery pressure (SPAP) in the shunt group was significantly increased, along with the increased percentage of muscularized arteries and partially muscularized arteries of small pulmonary arteries. Compared with the shunt group, SPAP in the shunt+SO2 group was significantly decreased, and the percentage of muscularized pulmonary arteries was also decreased. Additionally, rats that developed PH had significantly lower levels of SO2 concentration, aspartate aminotransferase (AAT) activity, protein and mRNA expressions of AAT2 in pulmonary tissues. Administration of an SO2 donor could alleviate the elevated pulmonary arterial pressure and decrease the muscularization of pulmonary arteries. At the same time, it increased the H2S production, protein expression of cystathionine-γ-lyase (CSE), mRNA expression of CSE, mercaptopyruvate transsulphurase (MPST) and cystathionine-ß-synthase (CBS) in the pulmonary tissue of the rats. The results suggested that endogenous SO2/AAT2 pathway and the endogenous H2S production were downregulated in rats with PH induced by high pulmonary blood flow. However, SO2 could reduce pulmonary arterial pressure and improve the pulmonary vascular pathological changes in association with upregulating endogenous H2S pathway.


Assuntos
Sulfeto de Hidrogênio/metabolismo , Hipertensão Pulmonar/fisiopatologia , Artéria Pulmonar/efeitos dos fármacos , Dióxido de Enxofre/farmacologia , Regulação para Cima , Animais , Aspartato Aminotransferases/metabolismo , Pressão Sanguínea , Western Blotting , Cistationina beta-Sintase/genética , Cistationina beta-Sintase/metabolismo , Cistationina gama-Liase/genética , Cistationina gama-Liase/metabolismo , Ativação Enzimática , Hipertensão Pulmonar/enzimologia , Hipertensão Pulmonar/metabolismo , Imuno-Histoquímica , Pulmão/irrigação sanguínea , Pulmão/enzimologia , Pulmão/fisiopatologia , Masculino , Artéria Pulmonar/metabolismo , Artéria Pulmonar/fisiopatologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Sulfurtransferases/genética , Sulfurtransferases/metabolismo
8.
Biochem Biophys Res Commun ; 415(1): 61-7, 2011 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-22020076

RESUMO

While sulfur dioxide (SO(2)) has been previously known for its toxicological effects, it is now known to be produced endogenously in mammals from sulfur-containing amino acid L-cysteine. L-cysteine is catalyzed by cysteine dioxygenase (CDO) to L-cysteinesulfinate, which converts to ß-sulfinylpyruvate through transamination by aspartate aminotransferase (AAT), and finally spontaneously decomposes to pyruvate and SO(2). The present study explored endogenous SO(2) production, and AAT and CDO distribution in different rat tissue. SO(2) content was highest in stomach, followed by tissues in the right ventricle, left ventricle, cerebral gray matter, pancreas, lung, cerebral white matter, renal medulla, spleen, renal cortex and liver. AAT activity and AAT1 mRNA expression were highest in the left ventricle, while AAT1 protein expression was highest in the right ventricle. AAT2 and CDO mRNA expressions were both highest in liver tissue. AAT2 protein expression was highest in the renal medulla, but CDO protein expression was highest in liver tissue. In all tissues, AAT1 and AAT2 were mainly distributed in the cytoplasm rather than the nucleus. These observed differences among tissues endogenously generating SO(2) and associated enzymes are important in implicating the discovery of SO(2) as a novel endogenous signaling molecule.


Assuntos
Dióxido de Enxofre/metabolismo , Animais , Aspartato Aminotransferases/genética , Aspartato Aminotransferases/metabolismo , Cisteína Dioxigenase/genética , Cisteína Dioxigenase/metabolismo , Imuno-Histoquímica , Masculino , Reação em Cadeia da Polimerase , Ratos , Ratos Wistar , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA