Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
2.
Cell Death Dis ; 13(2): 121, 2022 02 08.
Article in English | MEDLINE | ID: mdl-35136037

ABSTRACT

Upon vascular injury, vascular smooth muscle cells (VSMCs) change from a contractile phenotype to a synthetic phenotype, thereby leading to atherogenesis and arterial restenosis. Myocardin (MYOCD) is essential for maintaining the contractile phenotype of VSMCs. Deletion of MYOCD in VSMCs triggers autophagy. However, the molecular mechanism underlying the effect of MYOCD on autophagy is not clear. In this study, knockdown of MYOCD in human aortic VSMCs (HA-VSMCs) triggered autophagy and diminished the expression of SMC contractile proteins. Inhibition of autophagy in MYOCD-knockdown cells restored the expression of contractile proteins. MYOCD activated the transcription of miR-30a by binding to the CArG box present in its promoter, as confirmed by luciferase reporter and chromatin immune coprecipitation assays, while miR-30a decreased the expression of autophagy protein-6 (ATG6, also known as beclin1) by targeting its 3'UTR. Restoring the expression of miR-30a in MYOCD-knockdown cells upregulated the levels of contractile proteins. Treatment of VSMCs with platelet-derived growth factor type BB (PDGF-BB) resulted in the transformation of VSMCs to a proliferative phenotype. A low level of miR-30a was observed in PDGF-BB-treated HA-VSMCs, and re-expression of miR-30a led to a decrease in proliferative marker expression. Furthermore, using a wire injury mouse model, we found that miR-30a expression was significantly downregulated in the arterial tissues of mice and that restoration of miR-30a expression at the injured site abolished neointimal formation. Herein, MYOCD could inhibit autophagy by activating the transcription of miR-30a and that miR-30a-mediated autophagy defects could inhibit intimal hyperplasia in a carotid arterial injury model.


Subject(s)
Beclin-1 , MicroRNAs , Muscle, Smooth, Vascular , Nuclear Proteins , Trans-Activators , Autophagy/genetics , Becaplermin/pharmacology , Beclin-1/metabolism , Cell Proliferation , Cells, Cultured , Contractile Proteins/genetics , Humans , MicroRNAs/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Nuclear Proteins/metabolism , Phenotype , Trans-Activators/metabolism
3.
Acta Biochim Biophys Sin (Shanghai) ; 52(11): 1236-1246, 2020 Dec 11.
Article in English | MEDLINE | ID: mdl-33079978

ABSTRACT

Arterial marker genes EphrinB2 and HEY2 are essential for cardiovascular development and postnatal neovascularization. Our previous study confirmed that E2F1 could activate the transcription of EphrinB2 and HEY2 in human mesenchymal stem cells; however, the detailed mechanism has not been resolved yet. In this study, we focused on the interaction between E2F1 and DNMT3A, a de novo DNA methyltransferase, on regulating the expression of EphrinB2 and HEY2, and explored the potential mechanisms. Gain- and loss-of-function experiments implicated the positive effect of E2F1 on the expression of EphrinB2 and HEY2 and tube formation in human umbilical artery endothelial cells. Accumulation of DNMT3A decreased the levels of EphrinB2 and HEY2, and impaired tube formation induced by E2F1, while inhibiting DNMT3A by RNA interference augmented their expression and angiogenesis in E2F1-trasfected cells. We then asked whether the low expressions of EphrinB2 and HEY2 induced by DNMT3A are related to the methylation status of their promoters. Surprisingly, the methylation status of the CpG islands in the promoter region was not significantly affected by overexpression of exogenous DNMT3A. Furthermore, the interaction between E2F1 and DNMT3A was confirmed by co-immunoprecipitation. DNMT3A could inhibit the transcription of EphrinB2 and HEY2 promoters by affecting the binding of E2F1 to its recognition sequences as revealed by luciferase reporter assay and chromatin immunoprecipitation. These results identified a novel mechanism underlying the cooperation of DNMT3A with E2F1 on regulating target gene expression, and revealed their roles in the angiogenic process.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/metabolism , E2F1 Transcription Factor/antagonists & inhibitors , Neovascularization, Physiologic , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Chlorocebus aethiops , Chromatin Immunoprecipitation , CpG Islands , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA Methyltransferase 3A , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/physiology , Endothelial Cells/metabolism , Ephrin-B2/metabolism , Gene Expression Regulation/genetics , Gene Knockdown Techniques , Humans , Primary Cell Culture , Promoter Regions, Genetic , Repressor Proteins/metabolism , Umbilical Arteries/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...