Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 2 de 2
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Int Immunopharmacol ; 100: 108066, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34492536

RESUMEN

Severe acute lung injury (ALI) cause significant morbidity and mortality worldwide. MicroRNAs (miRNAs) are possible biomarkers and therapeutic targets for ALI. We aimed to explore the role of miR-762, a known oncogenic factor, in the pathogenesis of ALI. Levels of miR-762 in lung tissues of LPS-treated ALI mice and blood cells of patients with lung injury were measured. Injury of human lung epithelial cell line A549 was induced by LPS stimulation. A downstream target of miR-762, NFIX, was predicted using online tools. Their interactions were validated by luciferase reporter assay. Effects of targeted regulation of the miR-762/NFIX axis on cell proliferation, apoptosis, and inflammatory responses were tested in vitro in A549 cells in vivo with an ALI mouse model. We found that upregulation of miR-762 expression and downregulation of NFIX expression were associated with lung injury. Either miR-762 inhibition or NFIX overexpression in A549 lung cells significantly attenuated LPS-mediated impairment of cell proliferation and viability. Notably, increasing expressions of miR-762 inhibitor or NFIX in vivo via airway lentivirus infection alleviated the LPS-induced ALI in mice. Further, targeted downregulation of miR-762 expression or upregulation of NFIX expression in A549 cells markedly down-regulates NF-κB/IRF3 activation, and substantially reduces the production of inflammatory factors, including TNF-α, IL-6, and IL-8. This study reveals a novel role for the miR-762/NFIX pathway in ALI pathogenesis and sheds new light on targeting this pathway for diagnosis, prevention, and therapy.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , MicroARNs/metabolismo , Factores de Transcripción NFI/genética , Complicaciones Posoperatorias/inmunología , Transducción de Señal/genética , Células A549 , Lesión Pulmonar Aguda/sangre , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/patología , Animales , Puente de Arteria Coronaria/efectos adversos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo/inmunología , Técnicas de Silenciamiento del Gen , Células HEK293 , Voluntarios Sanos , Humanos , Factor 3 Regulador del Interferón/metabolismo , Lipopolisacáridos/inmunología , Pulmón/inmunología , Pulmón/patología , Masculino , Ratones , MicroARNs/genética , FN-kappa B/metabolismo , Complicaciones Posoperatorias/sangre , Complicaciones Posoperatorias/genética , Complicaciones Posoperatorias/patología , Transducción de Señal/inmunología
2.
Cell Biol Int ; 45(3): 642-653, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33289184

RESUMEN

Diabetic cardiomyopathy (DCM) is characterized by myocardial hypertrophy and fibrosis. This study aimed to investigate the effects of microRNA (miR)-34a on myocardial fibrosis in DCM and its potential mechanism of targeting Pin-1 signaling. Vimentin and Pin-1 proteins in mouse cardiac tissues were detected by immunohistochemical staining. Locked nucleic acid in situ hybridization was used to measure miR-34a expression in cardiac tissues. Primary mouse cardiac fibroblasts (CFs) were transfected with a mimics control/miR-34a mimics or Pin-1 plasmid and cultured in high-glucose (HG) Dulbecco's modified Eagle's medium. The miR-34a levels were measured by quantitative polymerase chain reaction. The apoptosis and viability of transfected cells were detected by the terminal deoxynucleotidyl transferase dUTP nick end labeling and Cell Counting Kit-8 assays respectively. A cell migration experiment and dual-luciferase reporter assay were also performed. The body weight and fasting blood glucose of DCM mice were significantly higher than those in the control (CTL) group. In addition, DCM mice had decreased serum insulin levels and impaired cardiac function. The number of CFs in the DCM group was higher than in the CTL group and Pin-1 expression was upregulated. The expression level of miR-34a in the cardiac tissue of mice in the DCM group was obviously downregulated compared with the CTL group. The HG stimulation of CFs for 48 h significantly downregulated the expression level of miR-34a and was associated with increased Type I collagen expression, cell viability, and migration and decreased apoptosis. However, these effects could be reversed by overexpressing miR-34a in HG-induced CFs. Furthermore, we found that Pin-1 was a direct target of miR-34a. Our results suggest that miR-34a can attenuate myocardial fibrosis in DCM by reducing Type I collagen production, cell viability, and migration and increasing the apoptosis of CFs by targeting Pin-1 signaling.


Asunto(s)
Cardiomiopatías Diabéticas/genética , Cardiomiopatías Diabéticas/patología , MicroARNs/metabolismo , Miocardio/metabolismo , Miocardio/patología , Peptidilprolil Isomerasa de Interacción con NIMA/metabolismo , Animales , Apoptosis/genética , Glucemia/metabolismo , Movimiento Celular/genética , Colágeno Tipo I/metabolismo , Cardiomiopatías Diabéticas/diagnóstico por imagen , Cardiomiopatías Diabéticas/fisiopatología , Electrocardiografía , Fibroblastos/metabolismo , Fibrosis , Regulación de la Expresión Génica , Ventrículos Cardíacos/patología , Masculino , Ratones Endogámicos C57BL , MicroARNs/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA