miR‑541‑3p inhibits the viability and migration of vascular smooth muscle cells via targeting STIM1

  • Authors:
    • Zhiyu Lv
    • Dengliang Yi
    • Chi Zhang
    • Yujie Xie
    • He Huang
    • Zhongcai Fan
    • Xing Liu
  • View Affiliations

  • Published online on: March 2, 2021     https://doi.org/10.3892/mmr.2021.11951
  • Article Number: 312
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The transformation of vascular smooth muscle cells (VSMCs) into the proliferative migratory phenotype in the plaque area contributes to stable plaque formation and facilitates the pathogenesis of atherosclerosis. Stromal interaction molecule 1 (STIM1) has been identified to promote the proliferation of VSMCs, suggesting that STIM1 may be a potent target for the prevention and treatment of atherosclerosis. Bioinformatics analysis has previously predicted STIM1 as a target of microRNA (miR)‑541‑3p. The present study aimed to determine the effect of the miR‑541‑3p/STIM1 axis on the progression of atherosclerosis in vitro. Oxidized low‑density lipoprotein (ox‑LDL)‑treated VSMCs were used as an in vitro atherosclerosis model. Cell Counting Kit‑8 and Transwell migration assays were used to analyze cell viability and migration, respectively. Reverse transcription‑quantitative PCR and western blotting were applied to measure mRNA and protein expression levels, respectively. The association between miR‑541‑3p and STIM1 was detected using a dual luciferase gene reporter assay. The results of the present study revealed that ox‑LDL treatment significantly downregulated miR‑541‑3p expression levels and upregulated STIM1 expression levels in VSMCs. In addition, ox‑LDL stimulation enhanced cell viability and migration. The overexpression of miR‑541‑3p significantly reversed the ox‑LDL‑mediated increase in cell viability and migration, whereas the knockdown of miR‑541‑3p expression enhanced the ox‑LDL‑mediated effects. STIM1 was confirmed to be a target gene of miR‑541‑3p in VSMCs. The knockdown of STIM1 significantly impaired the stimulatory effects of miR‑541‑3p knockdown on cell viability and migration. In conclusion, the findings of the present study suggested that miR‑541‑3p may efficiently repress VSMC viability and migration by targeting STIM1 under the treatment of ox‑LDL. These results indicated that the miR‑541‑3p/STIM1 axis may represent a potent target to modulate VSMC viability and migration.

Introduction

Atherosclerosis is a chronic progressive inflammatory disease and is the leading cause of mortality worldwide due to the progression to myocardial infarction and stroke, which are major clinical consequences (1,2). It is widely accepted that the transformation of vascular smooth muscle cells (VSMCs) from a quiescent contractile phenotype to a proliferative migratory phenotype facilitates stable plaque formation, contributing to the pathogenesis of atherosclerosis (3). However, the molecular mechanisms underlying the proliferation and migration of VSMCs in the pathogenesis of atherosclerosis remain to be fully understood.

MicroRNAs (miRNAs/miRs) are evolutionarily conserved, non-coding small RNAs that can post-transcriptionally modulate gene expression by binding to the 3′-untranslated region (UTR) of target mRNA (4). miRNAs have been shown to play pivotal roles in tumorigenesis, biological metabolism and immune inflammation by modulating cell proliferation, apoptosis, migration, invasion and differentiation (58). Increasing evidence has revealed that miRNAs may be closely associated with the pathogenesis of atherosclerosis through the modulation of VSMC proliferation and migration (912). Therefore, targeting miRNAs may represent a potentially effective method for the treatment of atherosclerosis.

Sustained Ca2+ influx mediated by store-operated channels (SOCs) is triggered in response to Ca2+ depletion by the endoplasmic reticulum (ER), and has been discovered to serve an important role in numerous cellular functions, such as cell proliferation and apoptosis (13). Stromal interaction molecule 1 (STIM1) has been reported to modulate SOCs by functioning as an ER Ca2+ sensor and has also been reported to modulate the proliferation of VSMCs (14,15). Using bioinformatics technology, TargetScan (http://www.targetscan.org/vert_71) and miRDB (http://www.mirdb.org/index.html), STIM1 was predicted as a target gene of miR-541-3p in the present study. Thus, it was hypothesized that miR-541-3p may be involved in the pathogenesis of atherosclerosis by targeting STIM1.

The present study aimed to determine the role of the miR-541-3p/STIM1 axis in the biological behaviors of VSMCs. Oxidized low-density lipoprotein (ox-LDL)-treated VSMCs were used as an in vitro model of atherosclerosis, and VSMC viability and migration were detected to investigate the pathogenesis of atherosclerosis in vitro.

Materials and methods

Cell culture and treatment

Human VSMCs (cat. no. CRL-1999™) were purchased from the American Type Culture Collection and cultured in F-12K medium (Gibco; Thermo Fisher Scientific, Inc.), supplemented with 10% FBS (Gibco; Thermo Fisher Scientific, Inc.). The cells were maintained at 37°C in a humidified incubator with 5% CO2.

To construct the in vitro atherosclerosis model, VSMCs were treated with 100 mg/l ox-LDL (cat. no. H7950; Beijing Solarbio Science & Technology Co., Ltd.) for 24 or 48 h at 37°C. The untreated VSMCs served as a control.

Cell transfection

miR-541-3p mimic and inhibitor (used to overexpress and knockdown miR-541-3p expression in VSMCs, respectively), small interfering RNA (siRNA/si) targeting STIM1 (si-STIM1) and the negative controls (NCs), mimic-NC, inhibitor-NC and si-NC (non-targeting sequences) were all synthesized by Shanghai GenePharma Co., Ltd. The mimic (150 nM), inhibitor (100 nM), si-STIM1 (50 nM) and the same amount of control vectors were transfected into 1×106 cells using Lipofectamine® 2000 transfection reagent (Invitrogen; Thermo Fisher Scientific, Inc.) at room temperature according to the manufacturer's protocol. The transfection efficiency was detected by RT-qPCR and western blotting 48 h post-transfection. The sequences were as follows: Mimic, 5′-TGGTGGGCACAGAATCTGGACT-3′; mimic-NC, 5′-UUCUCCGAACGUGUCACGUTT-3′; inhibitor, 5′-AGTCCAGATTCTGTGCCCACCA-3′; inhibitor-NC, 5′-ACAGGAUUGAGGGGGGGCCCU-3′; si-STIM1, 5′-GAGGTGCAATATTACAACATCAAGA-3′; si-NC, 5′-GAGAACGTTATAACACTACATGAGA-3′.

Reverse transcription-quantitative PCR (RT-qPCR)

Total RNA was extracted from VSMCs using the RNApure Tissue & Cell kit (CoWin Biosciences), according to the manufacturer's protocol. Total RNA (1 µg) was reverse transcribed into cDNA for miRNA and mRNA detection using stem-loop primers and random primers using EasyScript® First-Strand cDNA Synthesis SuperMix (Beijing Transgen Biotech Co., Ltd.) according to the manufacturer's protocol, respectively. qPCR was subsequently performed using a TaqMan Universal Master mix II kit (Thermo Fisher Scientific, Inc.) on a Bio-Rad detection system (Bio-Rad Laboratories, Inc.). GAPDH and U6 served as the internal references for the normalization of mRNA and miRNA expression, respectively. The relative expression was calculated using the 2−ΔΔCq method (16). PCR thermocycling conditions were as follows: Initial denaturation at 95°C for 30 sec, followed by 39 cycles at 95°C for 15 sec and 60°C for 30 sec, and a final step at 72°C for 5 min. The following primer pairs were used: Stem-loop primer (used for RT of miR-541-3p), 5′-GTCGTATCCAGTGCAGGGTCCGAGGTATTCGCACTGGATACGACAGTCCA-3′; miR-541-3p forward, 5′-TGGTGGGCACAGAATC-3′ and reverse, 5′-GTGCAGGGTCCGAGGT-3′; U6 forward, 5′-CTCGCTTCGGCAGCACA-3′ and reverse, 5′-AACGCTTCACGAATTTGCGT-3′; STIM1 forward, 5′-GTTCTGAAGGCTACGGGACC-3′ and reverse, 5′-GTCAGAAGGGGGTGTGTCAG-3′; GAPDH forward, 5′-CCACTAGGCGCTCACTGTTCTC-3′ and reverse, 5′-ACTCCGACCTTCACCTTCCC-3′.

Western blotting

Total protein was extracted from VSMCs using RIPA lysis buffer (Sangon Biotech Co., Ltd.) supplemented with protease inhibitor (Beijing Solarbio Science & Technology Co., Ltd.). Total protein was quantified using a BCA protein kit (Bio-Rad Laboratories, Inc.) and 30 µg protein from each sample was loaded and separated via SDS-PAGE on 10% gels. The separated proteins were subsequently transferred onto polyvinylidene difluoride membranes (EMD Millipore) and blocked with 5% non-fat milk for 1 h at room temperature. The membranes were then incubated with the following primary antibodies overnight at 4°C: Anti-STIM1 (cat. no. 4916; Cell Signaling Technology, Inc.; 1:3,000 dilution), anti-VEGF (cat. no. 2463; Cell Signaling Technology, Inc. 1:3,000 dilution), anti-matrix metalloproteinase (MMP)9 (cat. no. 13667; Cell Signaling Technology, Inc.; 1:3,000 dilution), anti-phosphorylated (p)-PI3K p85 (cat. no. 17366; Cell Signaling Technology, Inc.; 1:2,000 dilution), anti-PI3K p85 (cat. no. 4292; Cell Signaling Technology, Inc.; 1:3,000 dilution), anti-p-AKT1 (cat. no. 9018; Cell Signaling Technology, Inc.; 1:2,000 dilution), anti-AKT1 (cat. no. 2938; Cell Signaling Technology, Inc.; 1:3,000 dilution) and anti-GAPDH (cat. no. 2118; Cell Signaling Technology, Inc.; 1:2,000 dilution). Following the primary antibody incubation, the membranes were incubated with the goat anti-rabbit IgG HRP-linked secondary antibodies (cat. no. sc-2004; Santa Cruz Biotechnology, Inc.; 1:5,000 dilution) for 1 h at room temperature. Protein expression levels were determined using a western blotting imaging and quantitative system (Gel Doc XR+; Bio-Rad Laboratories, Inc.) following incubation with the ECL western blotting substrate (EMD Millipore). Densitometric analysis was performed using ImageJ software (version 1.6.0; National Institutes of Health) after background subtraction, with GAPDH expression levels acting as the internal reference control.

Dual luciferase gene reporter assay

The putative binding sites between miR-541-3p and STIM1 were predicted using TargetScan (http://www.targetscan.org/vert_71) and miRDB (http://www.mirdb.org/index.html). The wild-type (WT) and mutant (MUT) type of the 3′-UTR of STIM1, which were cloned into the pGL3 vector (Promega Corporation), were constructed by Shanghai GenePharma Co., Ltd.; the resulting plasmids were denoted as STIM1-WT or STIM1-MUT reporter plasmids, respectively. VSMCs (1×105) were co-transfected with WT (0.2 µg) or MUT (0.2 µg) and miR-541-3p mimic (150 nM) or mimic-NC (150 nM), as well as the Renilla luciferase vector (0.2 µg; control) using Lipofectamine 2000 transfection reagent. A total of 48 h post-transfection, the relative luciferase activity normalized to Renilla luciferase activity was measured using a Dual Luciferase Reporter assay system (Promega Corporation) according to the manufacturer's protocol.

Cell Counting Kit-8 (CCK-8) assay

VSMCs were seeded into 96-well plates at a density of 3×103 cells/well and cultured at 37°C overnight, followed by transfection with different vectors as aforementioned. After incubation at 37°C for 24 or 48 h, cell culture medium was replaced with 10 µl CCK-8 reagent (Beyotime Institute of Biotechnology) and 90 µl fresh medium containing 10% FBS, and incubated at 37°C for a further 4 h. The optical density (OD) value was measured at a wavelength of 450 nm using a microplate reader (model 680; Bio-Rad Laboratories, Inc.). Cell viability (%) = OD (treatment group)/OD (control group) ×100.

Transwell migration assay

Transwell chambers (BD Biosciences) were used to determine the cell migration ability. Briefly, ~3×105 VSMCs were resuspended in serum-free medium and seeded into the upper chamber of the Transwell plates. The lower chambers were filled with 600 µl medium supplemented with 15% FBS. After incubation at 37°C for 48 h, the cells remaining on the top of the membranes were removed using cotton buds, while cells on the lower membrane were fixed with −20° C-precooled absolute methanol for 15 min and stained with 0.2% crystal violet (Beijing Solarbio Science & Technology Co., Ltd.) for 5 min all at room temperature. The stained cells were subsequently counted under a light microscope (magnification, ×200) to assess cell migration.

Statistical analysis

Data were presented as the mean ± SD. Statistical analysis was performed using SPSS 21.0 software (IBM Corp.). Each experiment was performed in triplicate. Statistical differences between two groups and multiple groups were performed using an unpaired Student's t-test or one-way ANOVA followed by a Bonferroni's post hoc test, respectively. P<0.05 was considered to indicate a statistically significant difference.

Results

Ox-LDL treatment downregulates the expression levels of miR-541-3p and upregulates STIM1 expression levels

To determine the role of the miR-541-3p/STIM1 axis in the pathogenesis of atherosclerosis, the expression levels of miR-541-3p and STIM1 in VSMCs following the treatment with ox-LDL were analyzed. The results demonstrated that ox-LDL treatment significantly downregulated miR-541-3p expression levels and significantly upregulated STIM1 expression levels compared with the untreated VSMCs in the control group in a time-dependent manner (Fig. 1A and B). VSMC viability was significantly increased following treatment with 100 mg/l ox-LDL for 24 and 48 h compared with the control group (Fig. 1C), as was the cell migratory ability (Fig. 1D). These results indicated that ox-LDL treatment may downregulate miR-541-3p expression and upregulate STIM1 expression, which may be involved in process of atherosclerosis.

Overexpression of miR-541-3p inhibits the viability and migration of VSMCs

The effects of miR-541-3p on cell viability and migration in ox-LDL (100 mg/l; 24 h)-treated VSMCs were analyzed using gain- and loss-of-function assays. Since ox-LDL treatment for 24 h caused significant differences in cell viability and migration, this timepoint was assessed in subsequent experiments. miR-541-3p expression levels were significantly downregulated following the transfection of VSMCs with the miR-541-3p inhibitor, and significantly upregulated following transfection with the miR-541-3p mimic compared with the respective NCs (Fig. 2A). The cell viability (Fig. 2B) and migratory abilities (Fig. 2C and D) were significantly weakened following the overexpression of miR-541-3p in VSMCs compared with the mimic-NC-transfected VSMCs, whereas knockdown of miR-541-3p expression exacerbated ox-LDL-induced stimulation of cell viability and migration compared with the inhibitor-NC-transfected VSMCs (Fig. 2B-D). These results suggested that miR-541-3p may suppress the ox-LDL-induced induction of cell viability and migration in VSMCs.

miR-541-3p targets STIM1 in VSMCs

As shown in Fig. 3A, STIM1 was a putative target of miR-541-3p, which was confirmed by the dual luciferase gene reporter assay using STIM1-WT or STIM1-MUT reporter plasmids (Fig. 3B). The results revealed that overexpression of miR-541-3p in VSMCs transfected with STIM1-WT induced a significant reduction in luciferase activity; however, the mutation in the binding site between miR-541-3p and the 3′UTR of STIM1 abrogated this effect (Fig. 3B). These results suggested that miR-541-3p may target STIM1 in VSMCs.

Knockdown of miR-541-3p promotes ox-LDL-induced VSMC viability and migration by targeting STIM1

CCK-8 and Transwell assays were subsequently performed to investigate the role of the miR-541-3p/STIM1 axis in ox-LDL-induced VSMC viability and migration. The transfection with si-STIM1 significantly downregulated STIM1 expression at both the mRNA and protein levels compared with the si-NC group (Fig. S1A and B). In addition, the expression levels of STIM1 were significantly upregulated in VSMCs in the miR-541-3p inhibitor + si-NC group as compared with in the inhibitor-NC + si-NC group, while the expression levels were significantly downregulated in cells transfected with si-STIM1 in the inhibitor-NC + si-STIM1 group (Fig. 4A and B). Knockdown of STIM1 in the miR-541-3p inhibitor + si-STIM1 group impaired the miR-541-3p inhibitor-induced stimulatory effect on cell viability and migration in ox-LDL (100 mg/l; 24 h)-treated VSMCs as compared with the inhibitor-NC + si-NC group (Fig. 4C and D), indicating that knockdown of miR-541-3p may promote ox-LDL-induced VSMC viability and migration by targeting STIM1.

In addition, the knockdown of miR-541-3p in the miR-541-3p inhibitor + si-NC group significantly upregulated the expression levels of VEGF and MMP9, and the p-AKT1:AKT1 and p-PI3K:PI3K ratios as compared with the inhibitor-NC + si-NC group. However, transfection with si-STIM1 in the inhibitor-NC + si-STIM1 group induced the opposite results and rescued the effect of the miR-541-3p inhibitor (miR-541-3p inhibitor + si-STIM1 group vs. miR-541-3p inhibitor + si-NC group) (Fig. 4E). These results suggested that the miR-541-3p/STIM1 axis may modulate VSMC viability and migration via PI3K/AKT signaling.

Discussion

The enhanced proliferation of VSMCs has been identified as the main mechanism of aberrant neointima formation in vascular diseases (17). During atherogenesis, VSMCs respond to several factors, including thrombin, platelet derived growth factor-BB, IFN-γ, endothelin-1 and IL-1, resulting in the migration of VSMCs to the intima (18,19). The migration of VSMCs leads to plaque formation and also facilitates the atherosclerosis hardening process (20). Thus, inhibition of VSMC proliferation and migration is a potential strategy for the prevention or treatment of atherosclerosis (21). The present study aimed to determine the role of the miR-541-3p/STIM1 axis in the viability and migration of VSMCs. The results illustrated that miR-541-3p suppressed ox-LDL-mediated VSMC viability and migration by targeting STIM1.

To date, numerous miRNAs have been reported to play an important role in the pathogenesis of atherosclerosis by modulating VSMC proliferation and migration (22). For example, miR-146a expression was revealed to be upregulated in proliferating VSMCs, and knockdown of miR-146a expression weakened the proliferative and migratory capacities of VSMCs in vitro (23). In addition, miR-92a expression was upregulated in the atherosclerotic plaques of mice and the overexpression of miR-92a significantly increased the proliferation of VSMCs (24). Xu et al (25) also reported that miR-647 expression was upregulated in the serum samples of patients with atherosclerosis and ox-LDL-treated VSMCs, and the upregulation of miR-647 expression levels promoted the proliferation and migration of ox-LDL-treated VSMCs via targeting PTEN. miR-541-3p expression levels were also shown to be downregulated in non-small cell lung cancer (NSCLC) tissues and plasma, and the overexpression subsequently suppressed NSCLC cell growth and metastasis (26). To the best of our knowledge, the present study was the first to demonstrate that miR-541-3p expression levels were downregulated in ox-LDL-treated VSMCs, and that the overexpression of miR-541-3p significantly suppressed ox-LDL-induced increases in VSMC viability and migration.

Using TargetScan and miRDB, the current study predicted that miR-541-5p was a regulator of STIM1, which has been previously identified to promote VSMC proliferation (14,15). Subsequently, dual luciferase gene reporter assays were used to verify that STIM1 was a target gene of miR-541-3p in VSMCs. Further in vitro experiments revealed that the genetic knockdown of STIM1 significantly weakened the miR-541-3p knockdown-induced stimulation of VSMC viability and migration, indicating that miR-541-3p may inhibit ox-LDL-induced VSMC viability and migration by targeting STIM1. miR-541-5p was found to negatively regulate STIM1 expression by binding to the 3′-UTR of STIM1 mRNA, leading to the inhibition of VSMC viability. The present results showed that STIM1 may serve a role in regulating cell viability, which was consistent with previous studies (2730), which reported that the dysregulation of STIM1 caused a deregulation in cell viability.

The PI3K/AKT signaling pathway, one of the most important intracellular pathways in the body, is involved in a variety of cellular processes, including cell proliferation, survival, differentiation and migration (31). Notably, PI3K/AKT signaling was also discovered to play a crucial role in the pathogenesis of atherosclerosis (32). It has previously been reported that the selective inhibition of PI3K/AKT signaling could potently suppress the progression of atherosclerosis (33,34), whereas the activation of PI3K/AKT signaling enhanced the proliferation, survival and migration of VSMCs and human umbilical vein endothelial cells (35). These findings indicated that PI3K/AKT signaling may be an important target for the treatment of atherosclerosis. Therefore, the present study investigated the effect of the miR-541-3p/STIM1 axis on the activation of PI3K/AKT signaling. The results revealed that the knockdown of miR-541-3p significantly increased the phosphorylation levels of PI3K and AKT; however, this trend was abolished following the silencing of STIM1 in ox-LDL-treated VSMCs. Consistent with these findings, a previous study reported that the knockdown of STIM1 inactivated PI3K/AKT signaling in human prostate cancer cells (36). These aforementioned results suggested that PI3K/AKT signaling may serve a role in the miR-541-5p/STIM1 axis-mediated inhibition of VSMC viability and migration.

There are two main limitations of the present study. Firstly, the specific role of the PI3K/AKT signaling pathway in the miR-541-5p/STIM1 axis-mediated inhibition of VSMC viability and migration was not clarified. Secondly, the role of the miR-541-3p/STIM1 axis was only investigated in vitro. Thus, in vivo experiments should be performed in future studies to determine the role of the miR-541-3p/STIM1 axis in the pathogenesis of atherosclerosis.

In conclusion, the findings of the present study suggested that miR-541-3p may efficiently suppress ox-LDL-induced increases in VSMC viability and migration by targeting STIM1, which may be associated with the suppression of PI3K/AKT signaling. These results indicated that the miR-541-3p/STIM1 axis may represent a potential target to modulate VSMC viability and migration.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

ZL, DY, CZ and YX designed the study, analyzed the data and interpreted the results. ZL, DY, YX and HH performed experiments. ZL, DY, CZ and YX wrote the manuscript and prepared the figures. ZF and XL analyzed data, and reviewed and edited the manuscript. XL coordinated and supervised the study. ZL and XL confirm the authenticity of all the raw data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Libby P, Ridker PM and Hansson GK: Progress and challenges in translating the biology of atherosclerosis. Nature. 473:317–325. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Tabas I, García-Cardeña G and Owens GK: Recent insights into the cellular biology of atherosclerosis. J Cell Biol. 209:13–22. 2015. View Article : Google Scholar : PubMed/NCBI

3 

Kolodgie FD, Burke AP, Farb A, Gold HK, Yuan J, Narula J, Finn AV and Virmani R: The thin-cap fibroatheroma: A type of vulnerable plaque: The major precursor lesion to acute coronary syndromes. Curr Opin Cardiol. 16:285–292. 2001. View Article : Google Scholar : PubMed/NCBI

4 

Bartel DP: MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell. 116:281–297. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Pal AS and Kasinski AL: Animal models to study MicroRNA function. Adv Cancer Res. 135:53–118. 2017. View Article : Google Scholar : PubMed/NCBI

6 

Bernardo BC, Ooi JY, Lin RC and McMullen JR: miRNA therapeutics: A new class of drugs with potential therapeutic applications in the heart. Future Med Chem. 7:1771–1792. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Karnati HK, Panigrahi MK, Gutti RK, Greig NH and Tamargo IA: miRNAs: Key players in neurodegenerative disorders and epilepsy. J Alzheimers Dis. 48:563–580. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Mehrgou A and Akouchekian M: Therapeutic impacts of microRNAs in breast cancer by their roles in regulating processes involved in this disease. J Res Med Sci. 22:1302017. View Article : Google Scholar : PubMed/NCBI

9 

Huang SC, Wang M, Wu WB, Wang R, Cui J, Li W, Li ZL, Li W and Wang SM: Mir-22-3p inhibits arterial smooth muscle cell proliferation and migration and neointimal hyperplasia by targeting HMGB1 in arteriosclerosis obliterans. Cell Physiol Biochem. 42:2492–2506. 2017. View Article : Google Scholar : PubMed/NCBI

10 

Chen Q, Yang F, Guo M, Wen G, Zhang C, Luong le A, Zhu J, Xiao Q and Zhang L: miRNA-34a reduces neointima formation through inhibiting smooth muscle cell proliferation and migration. J Mol Cell Cardiol. 89:75–86. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Zhu J, Liu B, Wang Z, Wang D, Ni H, Zhang L and Wang Y: Exosomes from nicotine-stimulated macrophages accelerate atherosclerosis through miR-21-3p/PTEN-mediated VSMC migration and proliferation. Theranostics. 9:6901–6919. 2019. View Article : Google Scholar : PubMed/NCBI

12 

Wang H, Jiang M, Xu Z, Huang H, Gong P, Zhu H and Ruan C: miR-146b-5p promotes VSMC proliferation and migration. Int J Clin Exp Pathol. 8:12901–12907. 2015.PubMed/NCBI

13 

Parekh AB and Putney JW Jr: Store-operated calcium channels. Physiol Rev. 85:757–810. 2005. View Article : Google Scholar : PubMed/NCBI

14 

Takahashi Y, Watanabe H, Murakami M, Ono K, Munehisa Y, Koyama T, Nobori K, Iijima T and Ito H: Functional role of stromal interaction molecule 1 (STIM1) in vascular smooth muscle cells. Biochem Biophys Res Commun. 361:934–940. 2007. View Article : Google Scholar : PubMed/NCBI

15 

Potier M, Gonzalez JC, Motiani RK, Abdullaev IF, Bisaillon JM, Singer HA and Trebak M: Evidence for STIM1- and Orai1-dependent store-operated calcium influx through ICRAC in vascular smooth muscle cells: Role in proliferation and migration. FASEB J. 23:2425–2437. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

17 

Giachini FR, Lima VV, Hannan JL, Carneiro FS, Webb RC and Tostes RC: STIM1/Orai1-mediated store-operated Ca2+ entry: The tip of the iceberg. Braz J Med Biol Res. 44:1080–1087. 2011. View Article : Google Scholar : PubMed/NCBI

18 

Lopes J, Adiguzel E, Gu S, Liu SL, Hou G, Heximer S, Assoian RK and Bendeck MP: Type VIII collagen mediates vessel wall remodeling after arterial injury and fibrous cap formation in atherosclerosis. Am J Pathol. 182:2241–2253. 2013. View Article : Google Scholar : PubMed/NCBI

19 

Rudijanto A: The role of vascular smooth muscle cells on the pathogenesis of atherosclerosis. Acta Med Indones. 39:86–93. 2007.PubMed/NCBI

20 

Pan J, Lu L, Wang X, Liu D, Tian J, Liu H, Zhang M, Xu F and An F: AIM2 regulates vascular smooth muscle cell migration in atherosclerosis. Biochem Biophys Res Commun. 497:401–409. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Zhang L, Cheng H, Yue Y, Li S, Zhang D and He R: H19 knockdown suppresses proliferation and induces apoptosis by regulating miR-148b/WNT/beta-catenin in ox-LDL-stimulated vascular smooth muscle cells. J Biomed Sci. 25:112018. View Article : Google Scholar : PubMed/NCBI

22 

Li L, Li Y and Tang C: The role of microRNAs in the involvement of vascular smooth muscle cells in the development of atherosclerosis. Cell Biol Int. 43:1102–1112. 2019. View Article : Google Scholar : PubMed/NCBI

23 

Dong S, Xiong W, Yuan J, Li J, Liu J and Xu X: miRNA-146a regulates the maturation and differentiation of vascular smooth muscle cells by targeting NF-κB expression. Mol Med Rep. 8:407–412. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Wang J, Zhang C, Li C, Zhao D, Li S, Ma L, Cui Y, Wei X, Zhao Y and Gao Y: MicroRNA-92a promotes vascular smooth muscle cell proliferation and migration through the ROCK/MLCK signalling pathway. J Cell Mol Med. 23:3696–3710. 2019. View Article : Google Scholar : PubMed/NCBI

25 

Xu CX, Xu L, Peng FZ, Cai YL and Wang YG: miR-647 promotes proliferation and migration of ox-LDL-treated vascular smooth muscle cells through regulating PTEN/PI3K/AKT pathway. Eur Rev Med Pharmacol Sci. 23:7110–7119. 2019.PubMed/NCBI

26 

Lu YJ, Liu RY, Hu K and Wang Y: miR-541-3p reverses cancer progression by directly targeting TGIF2 in non-small cell lung cancer. Tumour Biol. 37:12685–12695. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Mao YY, Wang JQ, Guo XX, Bi Y and Wang CX: Circ-SATB2 upregulates STIM1 expression and regulates vascular smooth muscle cell proliferation and differentiation through miR-939. Biochem Biophys Res Commun. 505:119–125. 2018. View Article : Google Scholar : PubMed/NCBI

28 

Ge C, Zeng B, Li R, Li Z, Fu Q, Wang W, Wang Z, Dong S, Lai Z, Wang Y, et al: Knockdown of STIM1 expression inhibits non-small-cell lung cancer cell proliferation in vitro and in nude mouse xenografts. Bioengineered. 10:425–436. 2019. View Article : Google Scholar : PubMed/NCBI

29 

Cheng H, Wang S and Feng R: STIM1 plays an important role in TGF-β-induced suppression of breast cancer cell proliferation. Oncotarget. 7:16866–16878. 2016. View Article : Google Scholar : PubMed/NCBI

30 

Kim JH, Lkhagvadorj S, Lee MR, Hwang KH, Chung HC, Jung JH, Cha SK and Eom M: Orai1 and STIM1 are critical for cell migration and proliferation of clear cell renal cell carcinoma. Biochem Biophys Res Commun. 448:76–82. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Aoki M and Fujishita T: Oncogenic roles of the PI3K/AKT/mTOR axis. Curr Top Microbiol Immunol. 407:153–189. 2017.PubMed/NCBI

32 

Linton MF, Moslehi JJ and Babaev VR: Akt signaling in macrophage polarization, survival, and atherosclerosis. Int J Mol Sci. 20:27032019. View Article : Google Scholar

33 

Zhai C, Cheng J, Mujahid H, Wang H, Kong J, Yin Y, Li J, Zhang Y, Ji X and Chen W: Selective inhibition of PI3K/Akt/mTOR signaling pathway regulates autophagy of macrophage and vulnerability of atherosclerotic plaque. PLoS One. 9:e905632014. View Article : Google Scholar : PubMed/NCBI

34 

Zhou M, Ren P, Zhang Y, Li S, Li M, Li P, Shang J, Liu W and Liu H: Shen-Yuan-Dan capsule attenuates atherosclerosis and foam cell formation by enhancing autophagy and inhibiting the PI3K/Akt/mTORC1 signaling pathway. Front Pharmacol. 10:6032019. View Article : Google Scholar : PubMed/NCBI

35 

Yao X, Yan C, Zhang L, Li Y and Wan Q: LncRNA ENST00113 promotes proliferation, survival, and migration by activating PI3K/Akt/mTOR signaling pathway in atherosclerosis. Medicine (Baltimore). 97:e04732018. View Article : Google Scholar : PubMed/NCBI

36 

Zhou Y, Gu P, Li J, Li F, Zhu J, Gao P, Zang Y, Wang Y, Shan Y and Yang D: Suppression of STIM1 inhibits the migration and invasion of human prostate cancer cells and is associated with PI3K/Akt signaling inactivation. Oncol Rep. 38:2629–2636. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2021
Volume 23 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lv Z, Yi D, Zhang C, Xie Y, Huang H, Fan Z and Liu X: miR‑541‑3p inhibits the viability and migration of vascular smooth muscle cells via targeting STIM1. Mol Med Rep 23: 312, 2021
APA
Lv, Z., Yi, D., Zhang, C., Xie, Y., Huang, H., Fan, Z., & Liu, X. (2021). miR‑541‑3p inhibits the viability and migration of vascular smooth muscle cells via targeting STIM1. Molecular Medicine Reports, 23, 312. https://doi.org/10.3892/mmr.2021.11951
MLA
Lv, Z., Yi, D., Zhang, C., Xie, Y., Huang, H., Fan, Z., Liu, X."miR‑541‑3p inhibits the viability and migration of vascular smooth muscle cells via targeting STIM1". Molecular Medicine Reports 23.5 (2021): 312.
Chicago
Lv, Z., Yi, D., Zhang, C., Xie, Y., Huang, H., Fan, Z., Liu, X."miR‑541‑3p inhibits the viability and migration of vascular smooth muscle cells via targeting STIM1". Molecular Medicine Reports 23, no. 5 (2021): 312. https://doi.org/10.3892/mmr.2021.11951