Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Am J Transl Res ; 14(9): 6123-6136, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36247250

RESUMEN

OBJECTIVES: In vascular remodeling diseases, proliferation and inflammation of vascular smooth muscle cells (VSMCs) constitute the basic pathologic processes. Dehydroepiandrosterone (DHEA) exerts a protective effect on the cardiovascular system, but the molecular mechanism is unclear. METHODS: The plasma DHEA was measured using enzyme-linked immunosorbent assay (ELISA) kits. The neointima hyperplasia was assessed by hematoxylin/eosin staining. MiRNA microarray analysis was used to compare the influence of Ang II and DHEA on miRNA expression profiles in VSMCs. Cell counting and MTS assay were used to evaluate the effect of Ang II, DHEA and miR-486a-3p on VSMCs proliferation. qRT-PCR was performed to detect the expression of miR-486a-3p, PCNA, IL-1ß and NLRP3. Western blot analysis was performed to detect the expressions of PCNA, IL-1ß and NLRP3 after miR-486a-3p was knocked down or overexpressed in VSMCs. RESULTS: DHEA suppressed neointimal and VSMCs proliferation and inflammation. Using miRNA microarray analysis, we found that DHEA upregulated the expression of miR-486a-3p in VSMCs. Further experiments indicated that DHEA promoted miR-486a-3p expression in VSMCs and in the vascular intima. Gain- and loss-of-function experiments revealed that transfection of miR-486a-3p mimic inhibited proliferation and inflammation of VSMCs, which improved intimal hyperplasia. On the contrary, deletion of miR-486a-3p promoted VSMCs proliferation and inflammation. Furthermore, DHEA suppressed NOD-like receptor family pyrin domain containing 3 (NLRP3) expression and reduced VSMCs proliferation and inflammation. Importantly, DHEA inhibited NLRP3 expression via miR-486a-3p in VSMCs. CONCLUSIONS: DHEA inhibited VSMCs and vascular intimal proliferation and inflammation by regulating the miR-486a-3p/NLRP3 axis. Therefore, DHEA might be a candidate cardiovascular protective agent in the future.

2.
Adv Clin Exp Med ; 30(9): 933-939, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34498815

RESUMEN

BACKGROUND: Radiotherapy has been proven to be an effective treatment strategy for inhibiting head-and-neck cancer. However, side effects are common when using high-dosage irradiation, and the mechanism of action of this therapy has not been fully clarified. OBJECTIVES: To discover targeting molecules involved in an electron radiation-induced xerostomia murine model. MATERIAL AND METHODS: The xerostomia model mice were divided into Gy-3 (n = 5), Gy-7 (n = 5), and Gy-21 (n = 5) groups, and were compared to a negative control (NC) group. Drinking water amount, saliva volume, submandibular gland weight, and body weight were recorded. Real-time polymerase chain reaction (RT-PCR) was performed to amplify gene transcription. Hematoxylin and eosin (H&E) staining was used to identify submandibular gland damage. The dual-luciferase assay was used to observe the interaction between the Cdkn1a gene and miR-486a-3p. RESULTS: Electron radiation significantly increased the drinking water amount, and decreased saliva volume and body weight compared to mice without radiation treatment (p < 0.05). The H&E staining showed that electron radiation damaged the submandibular gland. Electron radiation also triggered significantly higher transcription of the Cdkn1a gene in the submandibular gland of xerostomia mice compared to those without radiation treatment (p < 0.05). The dual-luciferase assay demonstrated that miR-486a-3p interacted with the Cdkn1a gene (miRNA-mRNA). CONCLUSIONS: Radiation was found to induce damage of the submandibular gland and affect Cdkn1a expression by regulating the expression of miR-486a-3p in a xerostomia murine model. Therefore, modulation of miR-486a-3p and the Cdkn1a gene in a xerostomia murine model might improve damage of the submandibular gland.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , MicroARNs/genética , Xerostomía , Animales , Modelos Animales de Enfermedad , Ratones , Glándula Submandibular , Xerostomía/genética
3.
Am J Physiol Cell Physiol ; 320(3): C355-C364, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33296289

RESUMEN

Podocytes are epithelial cells adhering glomerular capillaries, which regulate the integrity of glomerular filtration barrier. Irreversible podocyte injury induces glomerular inflammation and causes chronic renal diseases. Kcnq1ot1, a long noncoding RNA, participates in the pathogenesis of diabetic retinopathy and cardiomyopathy. However, its function in podocyte injury is elusive. Pyroptosis of murine podocyte MPC5 was triggered by sublytic complement C5b-9 (sC5b-9) for subsequent in vitro functional and mechanistic investigation. Gain/loss-of-function analysis was conducted to examine the functional role of Kcnq1ot1 in podocyte pyroptosis. Meanwhile, the molecular mechanism of Kcnq1ot1's effect on podocyte injury was explored by identifying downstream molecules and their intermediate interactions. Kcnq1ot1 was upregulated in sC5b-9-induced podocytes, and silencing Kcnq1ot1 could inhibit sC5b-9's effect on podocyte pyroptosis. We also identified the interaction between Kcnq1ot1 and miR-486a-3p, through which Kcnq1ot1 mediated miR-486a-3p inhibition by sC5b-9. Furthermore, miR-486a-3p reduced the transcriptional activity of NLRP3, while the overexpression of NLRP3 enhanced sC5b-9's effect on podocyte pyroptosis through activating NLRP3 inflammasome. sC5b-9 induces pyroptosis in podocytes through modulating the Kcnq1ot1/miR-486a-3p/NLRP3 regulatory axis, and these uncovered key molecules might facilitate podocyte-targeted treatment for renal inflammatory diseases.


Asunto(s)
Complejo de Ataque a Membrana del Sistema Complemento/farmacología , MicroARNs/genética , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Podocitos/efectos de los fármacos , Canales de Potasio con Entrada de Voltaje/genética , Piroptosis/genética , ARN Largo no Codificante/genética , Regulación hacia Arriba/genética , Animales , Células Cultivadas , Ratones , Piroptosis/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Transcripción Genética/genética , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA