Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oxid Med Cell Longev ; 2022: 5759626, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35509835

RESUMO

Background: Arachidonic acid (ARA) metabolites are involved in the pathogenesis of epithelial-mesenchymal transformation (EMT). However, the role of ARA metabolism in the progression of EMT during pulmonary fibrosis (PF) has not been fully elucidated. The purpose of this study was to investigate the role of cytochrome P450 oxidase (CYP)/soluble epoxide hydrolase (sEH) and cyclooxygenase-2 (COX-2) metabolic disorders of ARA in EMT during PF. Methods: A signal intratracheal injection of bleomycin (BLM) was given to induce PF in C57BL/6 J mice. A COX-2/sEH dual inhibitor PTUPB was used to establish the function of CYPs/COX-2 dysregulation to EMT in PF mice. In vitro experiments, murine alveolar epithelial cells (MLE12) and human alveolar epithelial cells (A549) were used to explore the roles and mechanisms of PTUPB on transforming growth factor (TGF)-ß1-induced EMT. Results: PTUPB treatment reversed the increase of mesenchymal marker molecule α-smooth muscle actin (α-SMA) and the loss of epithelial marker molecule E-cadherin in lung tissue of PF mice. In vitro, COX-2 and sEH protein levels were increased in TGF-ß1-treated alveolar epithelial cells (AECs). PTUPB decreased the expression of α-SMA and restored the expression of E-cadherin in TGF-ß1-treated AECs, accompanied by reduced migration and collagen synthesis. Moreover, PTUPB attenuated TGF-ß1-Smad2/3 pathway activation in AECs via Nrf2 antioxidant cascade. Conclusion: PTUPB inhibits EMT in AECs via Nrf2-mediated inhibition of the TGF-ß1-Smad2/3 pathway, which holds great promise for the clinical treatment of PF.


Assuntos
Fibrose Pulmonar , Fator de Crescimento Transformador beta1 , Animais , Camundongos , Células Epiteliais Alveolares/metabolismo , Caderinas/metabolismo , Ciclo-Oxigenase 2/metabolismo , Células Epiteliais/metabolismo , Transição Epitelial-Mesenquimal , Camundongos Endogâmicos C57BL , Fator 2 Relacionado a NF-E2/metabolismo , Fibrose Pulmonar/patologia , Pirazóis , Sulfonamidas , Fator de Crescimento Transformador beta1/metabolismo
2.
Mol Med Rep ; 25(1)2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34812475

RESUMO

Schisandrin A (Sch A) has a protective effect on cardiomyocytes. Circulating miR­155 levels are related to chronic heart failure (CHF). The present study aimed to clarify the role and the molecular mechanism of Sch A in CHF. C57BL/6JGpt mice were used for an isoproterenol (ISO)­induced CHF model to collect heart samples. Echocardiography was employed to detect heartbeat indicators. The degree of myocardial hypertrophy was evaluated based on the measurement of heart weight (HW), body weight (BW) and tibia length (TL) and the observation using hematoxylin­eosin staining. Sprague­Dawley rats were purchased for the separation of neonatal rat ventricular myocytes (NRVMs), which were treated with ISO for 24 h. Transfection regulated the level of miR­155. The viability of NRVMs was detected via MTT assay. The mRNA and protein levels were measured via reverse transcription­quantitative PCR and western blotting and immunofluorescence was used to detect the content of α­smooth muscle actin (α­SMA). Treatment with ISO resulted in rising left ventricular posterior wall thickness, intra­ventricular septum diastole, left ventricular end diastolic diameter, left ventricular end systolic diameter, HW/BW, HW/TL and falling ejection fraction and fractional shortening, the trend of which could be reversed by Sch A. Sch A ameliorated myocardial hypertrophy in CHF mice. In addition, Sch A inhibited ISO­induced upregulated expressions of atrial natriuretic peptide, B­type natriuretic peptide, B­myosin heavy chain and miR­155 in myocardial tissue. Based on the results in vitro, Sch A had no significant effect on the viability of NRVMs when its concentration was <24 µmol/l. Sch A inhibited the levels of miR­155, α­SMA and the phosphorylation levels of AKT and cyclic AMP response­element binding protein (CREB) in ISO­induced NRVMs, which was reversed by the upregulation of miR­155. Schisandrin A mediated the AKT/CREB signaling pathway to prevent CHF by regulating the expression of miR­155, which may shed light on a possible therapeutic target for CHF.


Assuntos
Ciclo-Octanos/farmacologia , Insuficiência Cardíaca/tratamento farmacológico , Isoproterenol/efeitos adversos , Lignanas/farmacologia , MicroRNAs/metabolismo , Compostos Policíclicos/farmacologia , Animais , Fator Natriurético Atrial/metabolismo , Cardiomegalia/metabolismo , Ecocardiografia , Insuficiência Cardíaca/induzido quimicamente , Ventrículos do Coração/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Miocárdio , Miócitos Cardíacos/metabolismo , Peptídeo Natriurético Encefálico , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Função Ventricular Esquerda
3.
J Healthc Eng ; 2021: 7404671, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34900201

RESUMO

Objective: To assess whether miR-204 and HA affect A549 cell injury induced by lipopolysaccharide. Material and Methods. A549 cells were treated with hirsutanol A, and cell damage was induced by LPS followed by analysis of cell proliferation by CCK-8, cell apoptosis by flow cytometry, apoptosis-related protein expression by western blot, downstream target of miR-20 by dual-luciferase reporter gene, and inflammatory factors by ELISA and PCR. Results: LPS can significantly inhibit the viability of A549 cells, induce cell apoptosis, and promote the release of IL-6, IL-1ß, and TNF-α, while HA pretreatment can target FOXK2 by upregulating miR-204 levels, thereby alleviating apoptosis and promoting cell viability and at the same time inhibiting the release of inflammatory factors by inhibiting the activation of NF-κB. Conclusions: miR-204 participates in the protection of HA acute lung injury by targeting FOXK2.


Assuntos
Lipopolissacarídeos , MicroRNAs , Células A549 , Apoptose/genética , Fatores de Transcrição Forkhead , Humanos , Lipopolissacarídeos/farmacologia , MicroRNAs/genética , MicroRNAs/metabolismo , NF-kappa B/metabolismo
4.
Biochem Biophys Res Commun ; 556: 87-92, 2021 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-33839419

RESUMO

Virus-induced cytokine storm has been a devastating actuality in clinic. The abnormal production of type I interferon (IFN-1) and upregulation of multiple cytokines induced strong inflammation and thus lead to shock and organ failure. As an E3 ubiquitin ligase, tripartite motif-containing 37 (TRIM37) regulates the ubiquitination of multiple proteins including TRAFs. RNA sequencing was performed to investigated the alteration of transcriptional profile of H1N1-infected patients. qRT-PCR assay was performed to investigate the RNA levels of certain genes. The group of immune cells was examined by the Flow cytometry analysis. H&E staining was applied to evaluate lung inflammation of WT and TRIM37-KO mice. ELISA assay was performed to demonstrate the alteration of multiple cytokines. The protein levels in NF-kB signaling was estimated by western blotting and immunoprecipitation assays were applied to demonstrate the direct interaction between TRIM37 and TRAF-6. The RNA level of TRIM37 decreased in CD11b+ cells of Flu-infected patients. Knockout of TRIM37 inhibited the immune responses of H1N1-infected mice. TRIM37 deficiency reduced the levels of virous proinflammatory cytokines in bone marrow derived macrophages (BMDMs). Mechanically, TRIM37 promoted the K63-linked ubiquitination of TRAF6. TRIM37 negatively regulated inflammatory responses induced by virus infection via promoting TRAF6 ubiquitination at K63.


Assuntos
Inflamação/metabolismo , Infecções por Orthomyxoviridae/metabolismo , Infecções por Orthomyxoviridae/virologia , Fator 6 Associado a Receptor de TNF/metabolismo , Proteínas com Motivo Tripartido/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Animais , Feminino , Humanos , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/imunologia , Vírus da Influenza A Subtipo H1N1/imunologia , Influenza Humana/genética , Influenza Humana/imunologia , Influenza Humana/metabolismo , Influenza Humana/virologia , Camundongos , Camundongos Endogâmicos C57BL , Infecções por Orthomyxoviridae/genética , Infecções por Orthomyxoviridae/imunologia , Fator 6 Associado a Receptor de TNF/química , Proteínas com Motivo Tripartido/deficiência , Proteínas com Motivo Tripartido/genética , Ubiquitina-Proteína Ligases/deficiência , Ubiquitina-Proteína Ligases/genética
5.
Biochem Biophys Res Commun ; 517(4): 648-654, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31395340

RESUMO

The objective of this study was to investigate the exact therapeutic effects of Verapamil on lipopolysaccharide (LPS)-induced acute lung injury (ALI) and the molecular mechanism involved, through using LPS-induced animal models as well as LPS-stimulated mouse primary peritoneal macrophages models. Our results demonstrated that Verapamil reduced LPS-induced pathological damage of the lung tissue, infiltration of inflammatory cells and the production of IL-1ß, TNF-α, and MCP-1 in the serum. The MPO activity, MDA content, lung wet/dry ratio and LDH activity were also attenuated by Verapamil. In addition, Verapamil attenuated LPS-induced inflammatory cytokine production and oxidative stress in primary murine peritoneal macrophages in vitro. Moreover, we confirmed that NF-κB/NLRP3 pathway was involved in the therapeutic effect of Verapamil against LPS-induced injury in vivo and in vitro. In conclusion, these findings indicate that Verapamil has a therapeutic effect on LPS-induced ALI in mice. The mechanism may be related to the inhibition of NF-κB and NLRP3 signaling pathways. Verapamil may be a potential therapeutic agent for the treatment of ALI.


Assuntos
Lesão Pulmonar Aguda/induzido quimicamente , Lesão Pulmonar Aguda/tratamento farmacológico , Verapamil/uso terapêutico , Animais , Células Cultivadas , Citocinas/metabolismo , Modelos Animais de Doenças , Inflamação/patologia , Mediadores da Inflamação/metabolismo , Lipopolissacarídeos , Macrófagos Peritoneais/efeitos dos fármacos , Macrófagos Peritoneais/metabolismo , Macrófagos Peritoneais/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Verapamil/farmacologia
7.
PLoS One ; 12(5): e0175807, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28459804

RESUMO

BACKGROUND: Skeletal myoblast transplantation seems a promising approach for the repair of myocardial infarction (MI). However, the low engraftment efficacy and impaired angiogenic ability limit the clinical efficiency of the myoblasts. Gene engineering with angiogenic growth factors promotes angiogenesis and enhances engraftment of transplanted skeletal myoblasts, leading to improved infarction recovery in myocardial ischemia. The present study evaluated the therapeutic effects of hepatocyte growth factor (HGF) gene-engineered skeletal myoblasts on tissue regeneration and restoration of heart function in a rat MI model. METHODS AND RESULTS: The skeletal myoblasts were isolated, expanded, and transduced with adenovirus carrying the HGF gene (Ad-HGF). Male SD rats underwent ligation of the left anterior descending coronary artery. After 2 weeks, the surviving rats were randomized into four groups and treated with skeletal myoblasts by direct injection into the myocardium. The survival and engraftment of skeletal myoblasts were determined by real-time PCR and in situ hybridization. The cardiac function with hemodynamic index and left ventricular architecture were monitored; The adenovirus-mediated-HGF gene transfection increases the HGF expression and promotes the proliferation of skeletal myoblasts in vitro. Transplantation of HGF-engineered skeletal myoblasts results in reduced infarct size and collagen deposition, increased vessel density, and improved cardiac function in a rat MI model. HGF gene modification also increases the myocardial levels of HGF, VEGF, and Bcl-2 and enhances the survival and engraftment of skeletal myoblasts. CONCLUSIONS: HGF engineering improves the regenerative effect of skeletal myoblasts on MI by enhancing their survival and engraftment ability.


Assuntos
Transplante de Células , Fator de Crescimento de Hepatócito/genética , Mioblastos Esqueléticos/metabolismo , Mioblastos Esqueléticos/transplante , Isquemia Miocárdica/terapia , Adenoviridae/genética , Animais , Sobrevivência Celular/fisiologia , Colágeno/metabolismo , Modelos Animais de Doenças , Feminino , Fibrose/patologia , Fibrose/fisiopatologia , Fibrose/terapia , Engenharia Genética , Vetores Genéticos , Ventrículos do Coração/patologia , Ventrículos do Coração/fisiopatologia , Hemodinâmica/fisiologia , Fator de Crescimento de Hepatócito/metabolismo , Masculino , Isquemia Miocárdica/patologia , Isquemia Miocárdica/fisiopatologia , Neovascularização Fisiológica/fisiologia , Distribuição Aleatória , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/fisiologia , Regeneração/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...