Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Front Pharmacol ; 13: 1019755, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36618911

RESUMEN

Introduction: Damage to alveolar epithelial cells caused by uncontrolled inflammation is considered to be the main pathophysiological change in acute lung injury. FGF10 plays an important role as a fibroblast growth factor in lung development and lung diseases, but its protective effect against acute lung injury is unclear. Therefore, this study aimed to investigate protective effect and mechanism of FGF10 on acute lung injury in mice. Methods: ALI was induced by intratracheal injection of LPS into 57BL/6J mice. Six hours later, lung bronchoalveolar lavage fluid (BALF) was acquired to analyse cells, protein and the determination of pro-inflammatory factor levels, and lung issues were collected for histologic examination and wet/dry (W/D) weight ratio analysis and blot analysis of protein expression. Results: We found that FGF10 can prevent the release of IL-6, TNF-α, and IL-1ß, increase the expression of BMP4 and autophagy pathway, promote the regeneration of alveolar epithelial type Ⅱ cells, and improve acute lung injury. BMP4 gene knockdown decreased the protective effect of FGF10 on the lung tissue of mice. However, the activation of autophagy was reduced after BMP4 inhibition by Noggin. Additionally, the inhibition of autophagy by 3-MA also lowered the protective effect of FGF10 on alveolar epithelial cells induced by LPS. Conclusions: These data suggest that the protective effect of FGF10 is related to the activation of autophagy and regeneration of alveolar epithelial cells in an LPS-induced ALI model, and that the activation of autophagy may depend on the increase in BMP4 expression.

2.
Int Immunopharmacol ; 101(Pt B): 108230, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34655850

RESUMEN

Maintaining the pulmonary endothelial barrier that prevents the exudation of inflammatory factors and proteins is the key to the treatment of acute lung injury (ALI). Apelin-13 plays an important role in vascular diseases; however, the protective effects of Apelin-13 on ALI with pulmonary endothelial barrier are unknown. Therefore, mice and human umbilical vein endothelial cells (HUVECs) were injured by LPS following Apelin-13 administration. ALI mice showed reduced pulmonary vascular permeability, adhesion junction, mitochondrial function, mitochondrial biogenesis, and autophagy compared to the control group. Apelin-13 administration in ALI mice ameliorated LPS-induced lung injury, pulmonary vascular permeability, mitochondrial function, and promoted autophagic flux in mice and HUVECs. However, the effect of Apelin-13 was reduced after AMPK inhibition using Compound C. These data suggest that Apelin-13 ameliorates pulmonary vascular permeability in mice with ALI induced by LPS, which may be related to enhanced phosphorylation of AMPK to regulate mitochondrial function and autophagy.


Asunto(s)
Endotelio Vascular/efectos de los fármacos , Péptidos y Proteínas de Señalización Intercelular/farmacología , Mitocondrias/efectos de los fármacos , Animales , Autofagia/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Lipopolisacáridos/toxicidad , Lesión Pulmonar , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Distribución Aleatoria
3.
Zhongguo Ying Yong Sheng Li Xue Za Zhi ; 36(5): 390-393, 2020 Sep.
Artículo en Chino | MEDLINE | ID: mdl-33629549

RESUMEN

Objective: To investigate the effects of Apelin-13 on barrier function injury of human umbilical vein endothelial cells (HUVECs) induced by LPS. Methods: The HUVECs cultured in vitro were divided into 4 groups: Control group, LPS group, Apelin-13+LPS group, Apelin-13 group. HUVECs were treated by 5 µg/ml LPS for 24 h to replicate the model with endothelial barrier impaired. Apelin-13 at the concentration of 1 µmol/L was given 30 min before LPS treatment. The cell viabillity of HUVECs was measured by CCK-8 assay. Protein expressions of VE-cadherin and F-actin were measured by Western blot and immunofluorescence. Nuclear factor κB p65(NF-κB p65) was detected by immunofluorescence. Results: Compared with the control group, the cell viabillity of HUVECs and protein expression of VE-cadherin were decreased by LPS, but the protein expression of F-actin and activation of NF-κB p65 were increased by LPS. These effects were attenuated by Apelin-13 administration. Conclusion: Apelin-13 ameliorates LPS-induced barrier function injury of HUVECs, which may be related to the inhibition of inflammation.


Asunto(s)
Lipopolisacáridos , FN-kappa B , Células Cultivadas , Células Endoteliales de la Vena Umbilical Humana , Humanos , Péptidos y Proteínas de Señalización Intercelular
4.
Int J Biol Sci ; 15(7): 1440-1451, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31337974

RESUMEN

Chronic obstructive pulmonary disease (COPD) is a common disease characterized by persistent airflow limitation. Pulmonary vascular endothelial barrier injury and inflammation are increasingly considered to be important pathophysiological processes in cigarette smoke extract (CSE)-induced COPD, but the mechanism remains unclear. To identify the cellular mechanism of endothelial barrier injury and inflammation in CSE-treated human umbilical vein endothelial cells (HUVECs), we investigated the effect of the mitochondrion-targeting antioxidant mitoquinone (MitoQ) on endothelial barrier injury and inflammation. We demonstrated that MitoQ restored endothelial barrier integrity by preventing VE-cadherin disassembly and actin cytoskeleton remodeling, as well as decreased inflammation by the NF-κB and NLRP3 inflammasome pathways in endothelial cells. In addition, MitoQ also maintained mitochondrial function by reducing the production of ROS and excess autophagy. Inhibition of autophagy by 3-MA protected against cytotoxicity that was induced by CSE in HUVECs. Overall, our study indicated that mitochondrial damage is a key promoter in the induction of endothelial barrier dysfunction and inflammation by CSE. The protective effect of MitoQ is related to the inhibition of ROS and excess autophagy in CSE-induced HUVEC injury.


Asunto(s)
Antioxidantes/farmacología , Endotelio Vascular/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Compuestos Organofosforados/farmacología , Especies Reactivas de Oxígeno/metabolismo , Humo/efectos adversos , Ubiquinona/análogos & derivados , Actinas/metabolismo , Antígenos CD/metabolismo , Autofagia , Cadherinas/metabolismo , Supervivencia Celular , Endotelio Vascular/lesiones , Endotelio Vascular/metabolismo , Humanos , Inflamasomas/metabolismo , Inflamación/prevención & control , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/inducido químicamente , Enfermedad Pulmonar Obstructiva Crónica/prevención & control , Productos de Tabaco/efectos adversos , Ubiquinona/farmacología
5.
Cell Physiol Biochem ; 49(5): 1918-1932, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30235451

RESUMEN

BACKGROUND/AIMS: Acute lung injury (ALI) is induced by a variety of external and internal factors and leads to acute progressive respiratory failure. Previous studies have shown that apelin-13 can decrease the acute lung injury induced by LPS, but the specific mechanism is unclear. Therefore, a mouse lung injury model and a cell model were designed to explore the mechanism of how apelin-13 alleviates the acute lung injury caused by LPS. METHODS: The effect of apelin-13 on LPS-induced structural damage was determined by H&E staining and by the wet/dry weight ratio. The related inflammatory factors in BALF were examined by ELISA. The apoptotic pathway and the NF-κB and NLRP3 inflammasome pathways were evaluated by using Western blotting and immunofluorescence staining. RESULTS: LPS induced the structural damage and the production of inflammatory cytokines in the lung tissues of mice. These deleterious effects were attenuated by apelin-13 administration. The protective effects of apelin-13 were associated with decreased reactive oxygen species (ROS) formation and the inhibition of the activation of the NF-κB and NLRP3 inflammasome pathways in mice and in Raw264.7 cells. CONCLUSION: Taken together, these data suggest that apelin-13 administration ameliorates LPS-induced acute lung injury by suppressing ROS formation, as well as by inhibiting the NF-κB pathway and the activation of the NLRP3 inflammasome in the lungs.


Asunto(s)
Lesión Pulmonar Aguda/etiología , Apelina/farmacología , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Sustancias Protectoras/farmacología , Transducción de Señal/efectos de los fármacos , Lesión Pulmonar Aguda/prevención & control , Animales , Apelina/uso terapéutico , Apoptosis/efectos de los fármacos , Líquido del Lavado Bronquioalveolar/química , Citocinas/análisis , Modelos Animales de Enfermedad , Lipopolisacáridos/toxicidad , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Sustancias Protectoras/uso terapéutico , Células RAW 264.7 , Especies Reactivas de Oxígeno/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA