Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Br J Pharmacol ; 179(22): 5132-5147, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35764296

RESUMEN

BACKGROUND AND PURPOSE: Pulmonary arterial hypertension (PAH) is a pulmonary vasculature obstructive disease that leads to right heart failure and death. Maresin 1 is an endogenous lipid mediator known to promote inflammation resolution. However, the effect of Maresin 1 on PAH remains unclear. EXPERIMENTAL APPROACH: The serum Maresin 1 concentration was assessed using UPLC. A mouse model of PAH was established by combining the Sugen 5416 injection and hypoxia exposure. After treatment with Maresin 1, the right ventricular systolic pressure (RVSP) and right ventricular function were measured by haemodynamic measurement and echocardiography, respectively. Vascular remodelling was evaluated by histological staining. Confocal microscopy and western blot were used to test related protein expression. In vitro cell migration, proliferation and apoptosis assays were performed in primary rat pulmonary artery smooth muscle cells (PASMCs). Western blotting and siRNA transfection were used to clarify the mechanism of Maresin 1. KEY RESULTS: Endogenous serum Maresin 1 was decreased in PAH patients and mice. Maresin 1 treatment decreased RVSP and attenuated right ventricular dysfunction (RVD) in the murine PAH model. Maresin 1 reversed abnormal changes in pulmonary vascular remodelling, attenuating endothelial to mesenchymal transformation and enhancing apoptosis of α-SMA positive cells. Furthermore, Maresin 1 inhibited PASMC proliferation and promoted apoptosis by inhibiting STAT, AKT, ERK, and FoxO1 phosphorylation via LGR6. CONCLUSION AND IMPLICATIONS: Maresin 1 improved abnormal pulmonary vascular remodelling and right ventricular dysfunction in PAH mice, targeting aberrant PASMC proliferation. This suggests Maresin 1 may have a potent therapeutic effect in vascular disease.


Asunto(s)
Hipertensión Pulmonar , Hipertensión Arterial Pulmonar , Disfunción Ventricular Derecha , Animales , Proliferación Celular , Ácidos Docosahexaenoicos/farmacología , Ratones , Miocitos del Músculo Liso , Proteínas Proto-Oncogénicas c-akt/metabolismo , Hipertensión Arterial Pulmonar/tratamiento farmacológico , Arteria Pulmonar , ARN Interferente Pequeño/farmacología , Ratas , Remodelación Vascular , Disfunción Ventricular Derecha/metabolismo , Disfunción Ventricular Derecha/patología
2.
J Cell Mol Med ; 26(4): 1034-1049, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34989130

RESUMEN

The proliferation, migration and apoptotic resistance of pulmonary artery smooth muscle cells (PASMCs) are central to the progression of pulmonary arterial hypertension (PAH). Our previous study identified that fibroblast growth factor 21 (FGF21) regulates signalling pathway molecules, such as peroxisome proliferator-activated receptor gamma (PPARγ), to play an important role in PAH treatment. However, the biological roles of miRNAs in these effects are not yet clear. In this study, using miRNA sequencing and real-time PCR, we found that FGF21 treatment inhibited miR-130 elevation in hypoxia-induced PAH in vitro and in vivo. Dual luciferase reporter gene assays showed that miR-130 directly negatively regulates PPARγ expression. Inhibition of miR-130 expression suppressed abnormal proliferation, migration and apoptotic resistance in hypoxic PASMCs, and this effect was corrected upon PPARγ knockdown. Both the ameliorative effect of FGF21 on pulmonary vascular remodelling and the inhibitory effect on proliferation, migration and apoptotic resistance in PASMCs were observed following exogenous administration of miR-130 agomir. In conclusion, this study revealed the protective effect and mechanism of FGF21 on PAH through regulation of the miR-130/PPARγ axis, providing new ideas for the development of potential drugs for PAH based on FGF21.


Asunto(s)
MicroARNs , Hipertensión Arterial Pulmonar , Proliferación Celular/genética , Células Cultivadas , Regulación hacia Abajo/genética , Factores de Crecimiento de Fibroblastos , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Miocitos del Músculo Liso/metabolismo , PPAR gamma/genética , PPAR gamma/metabolismo , Arteria Pulmonar/metabolismo
3.
Int J Mol Med ; 47(6)2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33907846

RESUMEN

Pulmonary arterial hypertension (PAH), is a chronic and progressive disorder characterized by pulmonary vascular remodeling, including endothelial cell dysfunction and inflammation. MicroRNAs (miRNAs or miRs) play an important role in the development of PAH. In addition, fibroblast growth factor 21 (FGF21) has been found to have marked anti-dysfunction and anti­inflammatory properties. Therefore, the present study aimed to investigate the latent effects of FGF21 against PAH through the miR­27b/peroxisome proliferator­activated receptor γ (PPARγ) axis. Human pulmonary arterial endothelial cells (HPAECs) subjected to hypoxia were used as PAH models. The results revealed that PPARγ expression was downregulated and miR­27b expression was upregulated in the HPAECs exposed to hypoxia. Luciferase assay suggested that PPARγ was a target gene of miR­27b. Furthermore, miR­27b inhibited the expression of the PPARγ gene, thereby aggravating hypoxia­induced HPAEC dysfunction. Moreover, miR­27b activated the nuclear factor­κB signaling pathway and the expression of inflammatory factors [interleukin (IL)­1ß, IL­6 and tumor necrosis factor­α] by targeting PPARγ. In addition, the expression of miR­27b decreased following treatment of the hypoxia­exposed HPAECs with FGF21. Furthermore, FGF21 alleviated hypoxia­induced HPAEC dysfunction and inflammation by inhibiting miR­27b expression and thereby promoting PPARγ expression. On the whole, the findings of the present study suggest that FGF21 may serve as a therapeutic target for managing PAH through the miR­27b­mediated PPARγ pathway.


Asunto(s)
Células Endoteliales/metabolismo , Células Endoteliales/patología , Factores de Crecimiento de Fibroblastos/farmacología , Inflamación/patología , MicroARNs/metabolismo , PPAR gamma/metabolismo , Arteria Pulmonar/patología , Transducción de Señal , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/genética , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Células Endoteliales/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Inflamación/genética , MicroARNs/genética , PPAR gamma/genética , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
4.
Mol Carcinog ; 58(11): 2026-2039, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31397502

RESUMEN

Carboxypeptidase A4 (CPA4) is a member of the metallocarboxypeptidase family. A previous study indicated that CPA4 may participate in the modulation of peptide hormone activity and hormone-regulated tissue growth and differentiation. However, the role of CPA4 in lung tumorigenesis remains unclear. Our study revealed that CPA4 expression was higher in both lung cancer cells and tumor tissues. We performed 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assays, colony-formation assays, and Cellomics ArrayScan Infinity analysis to demonstrate that CPA4 knockdown inhibited non small-cell lung cancer (NSCLC) cell proliferation. Conversely, ectopic expression of CPA4 enhanced lung cancer cell proliferation. Consistent with these observations, we generated xenograft tumor models to confirm that CPA4 downregulation suppressed NSCLC cell growth. Mechanistically, we revealed that CPA4 downregulation may induce apoptosis and G1-S arrest by suppressing the protein kinase B/c-MYC pathway. These results suggest that CPA4 has an oncogenic effect on lung cancer growth. Taken together, we identified a novel gene in lung cancer that might provide a basis for new therapeutic targets.


Asunto(s)
Carboxipeptidasas A/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Proteína Oncogénica v-akt/genética , Proteínas Proto-Oncogénicas c-myc/genética , Animales , Apoptosis/genética , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Xenoinjertos , Humanos , Ratones , Transducción de Señal/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA