Effect of MicroRNA-26a Targeting mCTGF on Calcification of Vascular Smooth Muscle Cells Through Regulating Osteoprotegerin/Receptor Activator of NF-kB/Receptor Activator of Nuclear Factor Kappa-B Ligand Signaling Pathway

2020 ◽  
Vol 10 (12) ◽  
pp. 1813-1819
Author(s):  
Zhe Zhang ◽  
Jing Cheng ◽  
Zhen Yang

To explore the effect of microRNA-26a targeting mCTGF on inhibiting the calcification of vascular smooth muscle cells (VSMCs) through OPG/RANK/RANKL signaling pathway. VSMCs were isolated and cultured by adherent method. The morphology of VSMCs was observed under microscope and the surface antigen was identified by flow cytometry. The calcification of VSMCs was detected using alizarin red S staining and oil red O staining. microRNA-26a level in supernatant of cultured VSMCs was detected by ultracentrifugation method. The binding site between microRNA-26a and 3′-UTR of mCTGF was predicted by bioinformatics software; VSMCs were induced to be calcified and verified by the luciferase reporter gene assay. The mRNA and protein expressions of OPG/RANK/RANKL signaling pathway were measured by qRT-PCR and Western blotting. Normal VSMCs were assigned into normal group (normal VSMCs were treated with the exosomes secreted by normal VSMCs), CKD group (normal VSMCs were treated with the exosomes secreted by VSMCs in mice with chronic kidney disease (CKD)) and CKD/overexpressed mCTGF group (normal VSMCs were treated with the exosomes secreted by VSMCs in CKD mice and meanwhile treated with overexpressed mCTGF) followed by analysis of OPG/RANK/RANKL signaling level by qRT-PCR and ALP activity. VSMCs grew against the wall and showed a long fusiform shape. Calcification and adipogenesis of VSMCs was found under different induction conditions. microRNA-26a level in the supernatant of VSMCs in CKD group was significantly higher than normal group; after microRNA-26a bound to of mCTGF, mCTGF level in the CKD/overexpressed mCTGF group was significantly lower. The expression of OPG/RANK/RANKL signaling pathway was significantly decreased in the CKD/overexpressed mCTGF group. Meanwhile, OPG/RANK/RANKL signaling protein was increased significantly after treatment of exosomes secreted by VSMCs in CKD mice. mCTGF is the target gene of microRNA-26a and microRNA-26a can target mCTGF to regulate exosomes secretion by VSMCs and OPG/RANK/RANKL signaling to inhibit the calcification of VSMCs.

Pathobiology ◽  
2021 ◽  
pp. 1-12
Author(s):  
Hang Dong ◽  
Guangyu Jiang ◽  
Jiayue Zhang ◽  
Yuming Kang

<b><i>Background:</i></b> The dysregulation of proliferation and migration of vascular smooth muscle cells (VSMCs) is one of the major causes of atherosclerosis (AS). Accumulating studies confirm that Kruppel-like factor 4 (KLF4) can regulate the proliferation and differentiation of VSMCs through multiple signaling pathways. However, the mechanism of KLF4 dysregulation remains unknown. <b><i>Methods:</i></b> Apolipoprotein E-knockout (ApoE<sup>−/−</sup>) mice and human VSMCs were used to establish AS animal model and cell model, respectively. qRT-PCR was employed to determine the expressions of miR-506-3p and KLF4. Cell Counting Kit -8, Transwell, TUNEL assays, and flow cytometry were performed to measure the proliferation, migration, and apoptosis of VSMCs. The upstream miRNAs of KLF4 were predicted by microT, miRanda, miRmap, and TargetScan databases. The interaction between KLF4 and miR-506-3p was confirmed using qRT-PCR, Western blot, and luciferase reporter gene assay. <b><i>Results:</i></b> KLF4 expression was significantly decreased in the VSMCs of ApoE<sup>−/−</sup> mice fed with high-fat diet and in human VSMCs treated with oxidized low-density lipoprotein in time-dependent and dose-dependent manners. The transfection of miR-506-3p mimics or KLF4 shRNA promoted the proliferation and migration of VSMCs but inhibited the apoptosis while miR-506-3p inhibitors and pcDNA3.1-KLF4 exerted opposite effects. Additionally, KLF4 was confirmed as a target gene of miR-506-3p and could be negatively regulated by miR-506-3p. <b><i>Conclusion:</i></b> MiR-506-3p can promote the proliferation and migration of VSMCs via targeting KLF4, which can probably contribute to the pathogenesis of AS.


Author(s):  
Qianqian Lu ◽  
Ying Li ◽  
Jiaping Lou ◽  
Pingzhen Li ◽  
Yi Gu ◽  
...  

Circular RNAs (circRNAs) are associated with the pathogenesis of human diseases, including atherosclerosis. Here, we undertook to investigate the biological role and mechanism of circRNA E3 ubiquitin-protein ligase (circ-CHFR) in atherosclerosis. The expression levels of circ-CHFR, miR-214-3p, and pregnancy-associated plasma protein A (PAPPA) were measured by real-time quantitative polymerase chain reaction (RT-qPCR) and western blot in human aorta vascular smooth muscle cells (HA-VSMCs) exposed to oxidized low-density lipoprotein (ox-LDL). Cell proliferation, migration, and invasion capabilities were assessed by 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyl-2H-tetrazol-3-ium bromide (MTT), and transwell assays, respectively. The relationship between miR-214-3p and circ-CHFR or PAPPA was confirmed by dual-luciferase reporter and RNA immunoprecipitation (RIP) assays. Our data showed that circ-CHFR was upregulated in HA-VSMCs after stimulation with ox-LDL. Downregulation of circ-CHFR inhibited the proliferation, migration, and invasion of HA-VSMCs exposed to ox-LDL. Mechanistically, circ-CHFR acted as a miR-214-3p sponge, and miR-214-3p was a molecular mediator of circ-CHFR regulation in ox-LDL-stimulated HA-VSMCs. PAPPA was a miR-214-3p target, and circ-CHFR regulated the expression of PAPPA by sponging miR-214-3p. Moreover, overexpression of miR-214-3p repressed the proliferation, migration, and invasion of ox-LDL-induced HA-VSMCs by decreasing PAPPA expression. Our findings suggest that the circ-CHFR/miR-214-3p/PAPPA axis regulates ox-LDL-induced proliferation, migration, and invasion in HA-VSMCs.


2020 ◽  
Vol 2020 ◽  
pp. 1-10
Author(s):  
Baoliang Zhu ◽  
Jing Liu ◽  
Ying Zhao ◽  
Jing Yan

Coronary heart disease (CHD) is the most common cardiovascular disease with high prevalence, disability, and mortality. The balance between proliferation and apoptosis of vascular smooth muscle cells (VSMCs) plays a key role in the initiation of atherosclerosis. In this study, we found a significant decrease in the expression of lncRNA-SNHG14 in atherosclerotic plaque tissues of ApoE-/- mice. Overexpression of lncRNA-SNHG14 can inhibit VSMC proliferation while promoting apoptosis. There is a potential reciprocal regulatory relationship between lncRNASNHG14 and miR-19a-3p, which inhibit each other’s expression in vascular smooth muscle cells. In addition, the luciferase reporter gene analysis results showed that there was a direct interaction between miR-19a-3p and the 3′UTR of RORα. The results of qRT-PCR showed that the level of RORα mRNA was significantly increased in the aortas treated with miR-19a-3p and SNHG14 compared with that treated with miR-19a-3p alone. In conclusion, we demonstrated that lncRNA-SNHG14 regulates the apoptosis/proliferation balance of VSMCs in atherosclerosis.


2020 ◽  
Vol 98 (2) ◽  
pp. 249-257
Author(s):  
Qiang Xue ◽  
Xiaoli Wang ◽  
Xiaohui Deng ◽  
Yue Huang ◽  
Wei Tian

In this study we investigated the regulatory role of cell-migration-inducing and hyaluronan-binding protein (CEMIP) in the proliferation and migration of vascular smooth muscle cells (VSMCs). The mRNA and protein levels of CEMIP were upregulated in the plasma samples from patients with atherosclerosis, and in VSMCs stimulated with platelet-derived growth factor-BB (PDGF-BB), compared with plasma from healthy subjects and untreated VSMCs. Silencing CEMIP suppressed PDGF-BB-induced cell migration and proliferation in VSMCs, as determined using a Cell Counting Kit-8 assays, 5-ethynyl-2′-deocyuridine (EDU) assays, flow cytometry, wound healing assays, and Transwell assays. Overexpression of CEMIP promoted the proliferation and migration of VSMCs via activation of the Wnt–β-catenin signaling pathway and the upregulation of its target genes, including matrix metalloproteinase-2, matrix metalloproteinase-7, cyclin D1, and c-myc, whereas CEMIP deficiency showed the opposite effects. The knockdown of CEMIP in ApoE−/− mice by intravenous injection of lentiviral vector expressing si-CEMIP protected against high-fat-diet-induced atherosclerosis, as shown by the reduced aortic lesion areas, aortic sinus lesion areas, and the concentration of blood lipids compared with mice normally expressing CEMIP. These results demonstrated that CEMIP regulates the proliferation and migration of VSMCs in atherosclerosis by activating the WNT–β-catenin signaling pathway, which suggests the therapeutic potential of CEMIP for the management of atherosclerosis.


Sign in / Sign up

Export Citation Format

Share Document