Journal List > Korean J Physiol Pharmacol > v.26(3) > 1516078579

Ding, Pan, Qian, and Hu: Differentially expressed mRNAs and their upstream miR-491-5p in patients with coronary atherosclerosis as well as the function of miR-491-5p in vascular smooth muscle cells

Abstract

MicroRNAs (miRNAs) regulate gene expression and are biomarkers for coronary atherosclerosis (AS). A novel miRNA-mRNA regulation network of coronary AS still needs to be disclosed. The aim of this study was to analyze potential mRNAs in coronary AS patients and the role of their upstream miR-491-5p in vascular smooth muscle cells (VSMCs). We first confirmed top ten mRNAs according to the analysis from Gene Expression Omnibus database (GSE132651) and examined the expression levels of them in the plaques and serum from AS patients. Five mRNAs (UBE2G2, SLC16A3, POLR2C, PNO1, and AMDHD2) presented significantly abnormal expression in both plaques and serum from AS patients, compared with that in the control groups. Subsequently, they were predicted to be targeted by 11 miRNAs by bioinformatics analysis. Among all the potential upstream miRNAs, only miR-491-5p was abnormally expressed in the plaques and serum from AS patients. Notably, miR-491-5p overexpression inhibited viability and migration, and significantly increased the expression of contractile markers (α-SMA, calponin, SM22α, and smoothelin) in VSMCs. While silencing miR-491-5p promoted viability and migration, and significantly suppressed the expression of α-SMA, calponin, SM22α, and smoothelin. Overall, miR-491-5p targeted UBE2G2, SLC16A3, and PNO1 and regulated the dysfunctions in VSMCs.

INTRODUCTION

Coronary Atherosclerosis (AS) is a multifactorial chronic disease characterized by accumulation of lipids and fibrous components in the aorta [1,2]. In the aortic tunica media, vascular smooth muscle cells (VSMCs) are the main cell type [3,4], and the proliferation and migration of VSMCs in intima are associated with the formation of atherosclerotic plaques [5,6]. Due to the long clinical treatment cycle, poor prognosis, and frequent relapse of coronary AS, it is urgent to explore a novel therapeutic target for Coronary AS from the molecular level.
MicroRNAs (miRNAs) are small non-coding RNA molecules that can regulate gene expression at the post-transcriptional level [7-9]. MiRNAs can directly or indirectly modulate plaque formation, which involves the regulation of VCMCs, so that to affect the pathophysiology of coronary AS and cardiovascular disease [8,10]. Numerous miRNAs have been reported to participate in functionally regulating VSMCs during AS. For examples, miR-499a-3p and miR-135b-5p promote AS development by targeting MEF2C [11]. Overexpression of miR-448 promotes VSMC proliferation and migration via MEF2C [12]. MiR-365 participates in coronary AS through regulating IL-6 expression [13]. MiR-491-5p is abnormally expressed in cancers [14-16]. Notably, a previous study revealed that miR-491-5p is lowly expressed in the atherosclerotic plaque tissues and plasma samples of AS patients, and inhibited the growth and migration of VSMCs by targeting MMP-9 [17], suggesting its meaningful role in the AS development. Moreover, miR-491-5p also increases the risk of atherosclerotic cerebral infarction through modulation of MMP-9 gene, as reported in another study [18].
In this study, we examined the expression of ten potential mRNAs in plaque tissues and serum from coronary AS patients and identified miR-491-5p as an upstream regulator. We hypothesized that miR-491-5p regulated the dysfunctions of VSMCs, which may contribute to novel research strategies for the coronary AS.

METHODS

Clinical specimens

This studied included 22 coronary AS patients (16 males and 6 females; 9 cases aged ≤ 50; 13 cases aged > 50) with coronary endarterectomy at our hospital. Clinical characteristics of patients with coronary AS were provided in Table 1. The healthy control group included 13 patients (6 males and 7 females; 5 cases aged ≤ 50; 8 cases aged > 50) who underwent physical examinations in the same period at our hospital. The patients with complications or infections of liver or kidney, diabetes, or tumors were excluded. The Gensini scoring system was applied to evaluate the degree of AS. Written informed consent was obtained from all participants. The study complied with the Declaration of Helsinki and was approved by the Ethics Committee of The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University (IRB: 2016-045, Jiangsu, China).
Plaque tissues and adjacent intimal tissues were taken by coronary endarterectomy and then stored in liquid nitrogen. Peripheral blood was collected, and serum was separated by centrifugation at 400 g for 10 min.

Cell culture and transfection

VSMCs were dissected from perioperative aortic punch samples of participating patients as previous study described [19]. The dissected VSMCs were then cultured in Dulbecco's modified Eagle's medium (DMEM; Sigma-Aldrich, St. Louis, MO, USA) supplemented with 10% Gibco fetal bovine serum (FBS; Thermo Fisher Scientific, Inc., Waltham, MA, USA) and 100 µg/ml penicillin-streptomycin (Sigma-Aldrich) at 37°C and 5% CO2. The 10 nM miR-491-5p mimics were used for miR-491-5p overexpression and an equal quantity of miR-491-5p antagomir was used for miR-370 downregulation. Simultaneously, NC mimics and NC antagomir was used in the present study. To downregulate UBE2G2, SLC16A3, and PNO1 expression, short hairpin RNAs targeting UBE2G2, SLC16A3, and PNO1 (sh-UBE2G2, sh-SLC16A3, and sh-PNO1) were used. The miR-491-5p mimic, miR-491-5p antagomir, sh-UBE2G2, sh-SLC16A3, sh-PNO1, and their relevant controls were all synthesized by GenePharma (Shanghai, China). Cell transfections were conducted for 48 h using Lipofectamine 2000 (Invitrogen, Carlsbad, CA, USA).

Reverse transcription quantitative polymerase chain reaction

Total RNA was extracted using TRIzol reagent (Invitrogen). Synthetic spike-in miRNAs (MiRXES, Singapore) was prepared for miRNAs to obtain RNA samples, which was then reverse transcribed into cDNA. Reverse transcription quantitative polymerase chain reaction (RT-qPCR) analysis was performed using mSMRT-qPCR miRNA assay platform (MiRXES, Singapore). The relative expression levels of miRNAs were normalized to U6, and the relative expression levels of mRNAs were normalized to GAPDH, which were all quantified using the 2−ΔΔCq method. Sequences of primers are provided in Table 2.

Western blot analysis

Total protein extraction was performed, and the protein concentration was measured. Proteins (25 µg) were then loaded on 10% SDS-PAGE and transferred onto PVDF membranes. After blocking with non-fat milk (5%) at room temperature for 1 h, the membranes were then incubated with the primary antibodies below at 4°C overnight: UBE2G2 (ab174296; 1:1,000; Abcam, Shanghai, China), SLC16A3 (ab244385; 1:1,000; Abcam), POLR2C (ab182150; 1:100; Abcam), PNO1 (ab227978; 1:1,000; Abcam), AMDHD2 (ab133306; 1:1,000; Abcam), α-SMA (ab5831; 1:1,000; Abcam), calponin (ab46794; 1:5,000; Abcam), SM22α (ab14106; 1:1,000; Abcam), smoothelin (ab8969; 1:100; Abcam), GAPDH (as a loading control; ab8245; 1:500; Abcam). Subsequently, the secondary antibody of HRP-conjugated goat anti-rabbit IgG (1:3,000) was added and incubated at room temperature for 1 h. Finally, the protein bands were detected by the ECL Plus Kit (Pierce, Rockford, IL, USA), The relative protein expression was analyzed using ImageJ software 1.4.

RNA pull down assay

Bio miR-491-5p and Bio NC were co‐transfected to VSMCs for 2 days. VSMCs (1 × 107) were dissolved in the soft lysis buffer plus RNasin (80 U/ml; Promega, Madison, WI, USA). Subsequently, streptavidin agarose beads (Life Technologies, Rockville, MD, USA) were incubated with the reaction mixture for 1 h at room temperature. RT-qPCR was prepared to evaluate the co-precipitated RNAs.

Cell counting kit-8 assay

VSMCs were seeded in 96-well plates (5 × 103 cells/well) for 24 h at 37°C and then used to assess cell proliferation. After the transfection, the cell proliferation value was measured by a cell counting kit 8 (CCK-8 kit; Thermo Fisher Scientific, Inc.) at 24, 48, and 72 h. CCK-8 reagent (10 µl) was then added and incubated at 37°C for 1 h. Subsequently, the optical density was measured at 450 nm.

Wound healing assay

VSMCs were cultured in 6-well plates and cultured for 24 h. Subsequently, the parallel lines were scratched on the cell culture plate with a 200 µl pipette tip. Following washing with PBS three times, cells were cultured in serum-free DMEM for an additional 48 h. The wounded gaps were photographed at different time points (0 and 24 h).

Statistical analyses

Data are shown as the mean ± standard deviation. The Student’s t-test was used to assess the differences between two groups. Two-way ANOVA was conducted for difference comparison in groups with two independent variables. All data were statistically analyzed by the SPSS 21.0 (IBM Co., Armonk, NY, USA) statistical software. p-values of less than 0.05 was defined as significant.

RESULTS

Expression of 10 mRNAs in plaques and adjacent intimal tissues from AS patients

According to the gene profiling by array results of GSE132651 which determined differences in blood outgrowth endothelial cells from subjects with normal or abnormal coronary endothelial function, top ten genes were differentially expressed (Table 3). Subsequently, we measured the expression levels of UBE2G2, SNORD45C, SNORD45A, SNORD45B, RABGGTB, SLC16A3, POLR2C, PNO1, CEMP1, AMDHD2 using RT-qPCR. The levels of UBE2G2, SNORD45C, SNORD45A, SLC16A3, POLR2C, PNO1, AMDHD2 in plaques from coronary AS patients were significantly higher than those in adjacent intima. On the contrary, CEMP1 expression was downregulated in plaques from coronary AS patients, compared with that in adjacent intima. However, the expression levels of SNORD45B and RABGGTB had no significant difference in plaques and adjacent intima from coronary AS patients (Fig. 1A). Only mRNAs (UBE2G2, SNORD45C, SNORD45A, SLC16A3, POLR2C, PNO1, CEMP1, AMDHD2) with significant difference were listed in Fig. 1B.

Expression of 10 mRNAs in serum from AS patients and healthy controls

The levels of UBE2G2, SNORD45B, RABGGTB, SLC16A3, POLR2C, PNO1, and AMDHD2 in the serum of coronary AS patients were significantly higher than those in the serums of healthy patients. While the expression levels of SNORD45C, SNORD45A, and CEMP1 had no significant difference in the serum of coronary AS patients and healthy patients (Fig. 2A). In Fig. 2B, mRNAs (UBE2G2, SNORD45B, RABGGTB, SLC16A3, POLR2C, PNO1, and AMDHD2) with significant difference were listed.

Potential upstream miRNAs for the abnormally expressed mRNAs

We found that five mRNAs (UBE2G2, SLC16A3, POLR2C, PNO1, and AMDHD2) were significantly abnormally expressed in both plaques and serums from subjects with coronary AS. TargetScan was used to predict the potential upstream miRNAs of these five genes, and 11 common miRNAs (hsa-miR-491-5p, hsa-miR-4645-5p, hsa-miR-4673, hsa-miR-6791-5p, hsa-miR-6727-3p, hsa-miR-6747-3p, hsa-miR-4308, hsa-miR-4722-3p, hsa-miR-4292, hsa-miR-6852-5p, and hsa-miR-7160-5p) were identified (Fig. 3A). We them examined the expression levels of these miRNAs using RT-qPCR. Only miR-491-5p showed significantly downregulated expression in plaques from coronary AS patients, compared with that in adjacent intima (Fig. 3B). MiR-491-5p expression was also decreased in the serum of coronary AS patients, compared with that in the serums of healthy patients (Fig. 3C). Collectively, miR-491-5p was a potential upstream miRNA for abnormally expressed UBE2G2, SLC16A3, POLR2C, PNO1, and AMDHD2.

MiR-491-5p negatively regulates UBE2G2, SLC16A3, PNO1 expression in VSMCs

To confirm the predicted regulatory role of miR-491-5p on target genes, we conducted the subsequent experiments in VSMCs separated from the aortic wall sections of coronary AS patients. According to the results of RT-qPCR, miR-491-5p expression was higher in the VSMCs transfected with miR-491-5p mimics than that in the control group (Fig. 4A). mRNA expression and protein levels of UBE2G2, SLC16A3 and PNO1 were significant downregulated in the cells transfected with miR-491-5p mimics, compared with those in the control group, as evaluated by RT-qPCR and western blotting, respectively (Fig. 4B, C). However, the mRNA expression and protein levels of POLR2C and AMDHD2 had no significant difference in the cells transfected with miR-491-5p mimics and NC mimics. Similarly, RNA pull down assay revealed that the expression levels of UBE2G2, SLC16A3 and PNO1 were upregulated in VSMCs transfected with Bio miR-491-5p, compared with those in control groups (Fig. 4D). These results indicated that the expression levels of UBE2G2, SLC16A3 and PNO1 were negatively regulated by miR-491-5p.

MiR-491-5p inhibits viability, migration, and increased expression of contractile markers in VSMCs

The function of miR-491-5p in VSMCs was then explored. CCK-8 revealed that the viability was suppressed in VSMCs transfected with miR-491-5p mimics, compared with that in control group (Fig. 5A). According to the results of the wound healing assay, the migration capacity was decreased in VSMCs transfected with miR-491-5p mimics, compared with that in control group (Fig. 5B). Importantly, the protein levels of contractile markers in VSMCs were elevated by miR-491-5p mimics (Fig. 5C). These results suggested that miR-491-5p inhibited viability, migration, and increased expression of contractile markers in VSMCs.

MiR-491-5p antagomir promotes viability, migration, and increased expression of contractile markers in VSMCs

To further confirm the function of miR-491-5p and its target genes UBE2G2, SLC16A3, and PNO1 in VSMCs, miR-491-5p antagomir, sh-UBE2G2, sh-SLC16A3, and sh-PNO1 were transfected in VSMCs and NC antagomir and sh-NC were used as negative control. A CCK-8 assay revealed that silencing miR-491-5p in VSMCs significantly promoted cell proliferation when compared with the control group at 24, 48, and 72 h, while UBE2G2, SLC16A3 and PNO1 knockdown partially reversed the changes (Fig. 6A). Wound healing assay was performed to assess cell migration. The results indicated that the inhibition of miR-491-5p significantly upregulated VSMCs migration. However, the upregulation was then alleviated by UBE2G2, SLC16A3, and PNO1 knockdown (Fig. 6B). Moreover, the protein levels of α-SMA, calponin, SM22α, and smoothelin were suppressed by miR-491-5p antagomir, While UBE2G2, SLC16A3, and PNO1 knockdown partially restored the changes, as shown by western blotting (Fig. 6C). Taken together, these results suggested that miR-491-5p silencing promotes VSMCs proliferation and migration but inhibits the expression of contractile markers. However, UBE2G2, SLC16A3, and PNO1 knockdown reversed the changes induced by miR-491-5p silencing.

DISCUSSION

In our study, we first confirmed ten potential mRNAs that were differentially expressed in blood outgrowth endothelial cells from subjects with normal or abnormal coronary endothelial function, according to the gene profiling by array results of GSE132651. After evaluating their expression levels, we found that expression of five mRNAs (UBE2G2, SLC16A3, POLR2C, PNO1, and AMDHD2) had significant difference in both plaques and serum from coronary AS patients, compared with those in adjacent intimal tissues from coronary AS patients or serum from healthy controls. TargetScan then predicted 11 common upstream miRNAs of these mRNAs. Notably, only miR-491-5p showed significantly downregulated expression levels in plaques and serum from coronary AS patients, compared with those in adjacent intimal tissues from coronary AS patients or serum from healthy controls.
MiRNAs are short (approximately 24 nucleotides), noncoding RNAs that bind to the 3’ untranslated region of targeted mRNAs and induce degradation or translational inhibition [20]. To date, numerous studies have shown that miRNAs are effective regulators of VSMC differentiation [21-24]. These miRNAs regulate the VSMC differentiation and coronary AS mainly through the modulation of target gene expression [3,25,26]. Previous studies have reported several miRNA-mRNA regulatory networks in AS [27-29]. For instance, upregulated miR-93 contributes to coronary AS pathogenesis through targeting ABCA1 [30]. MiR-126 represses the progression of coronary AS in the mice by binding to S1PR2 [31]. MiR-491-5p has been once explored in VSMCs in a previous study, which demonstrates its inhibitory role on the growth and migration of VSMCs by binding with MMP-9 gene, indicating the involvement of miR-491-5p in AS development [17]. Particularly, we found three mRNAs UBE2G2, SLC16A3, and PNO1 that were negatively regulated by miR-491-5p, which were not mentioned in any previous research. Mechanically, UBE2G2, SLC16A3, and PNO1 were validated as the targets downstream miR-491-5p, indicating that the upregulation of UBE2G2, SLC16A3, and PNO1 in plaques and serum from AS patients was induced by the downregulation of miR-491-5p. Furthermore, the functions of miR-491-5p, UBE2G2, SLC16A3, and PNO1 in smooth muscle cells were assessed. Notably, we found that miR-491-5p overexpression inhibited viability, migration, and increased the expression of contractile markers α-SMA, calponin, SM22α, and smoothelin in VSMCs. The increase of these contraction markers suggests that miR-491-5p overexpression promoted the differentiation of phenotypic proteins, which is a key element in AS development because the synthetic phenotype of VSMCs can migrate, proliferate, and generate extracellular matrix proteins [5]. Inversely, silencing miR-491-5p significantly promoted VSMCs viability and migration, and inhibited the expression of α-SMA, calponin, SM22α, and smoothelin in VSMCs. However, UBE2G2, SLC16A3, and PNO1 knockdown reversed the changes induced by miR-491-5p antagomir.
In conclusion, this study revealed the differentially expressed mRNAs and their upstream miR-491-5p in patients with coronary atherosclerosis. We also found that miR-491-5p inhibited viability, migration, and increased expression of contractile markers via regulating target genes UBE2G2, SLC16A3, and PNO1 in VSMCs. These results might provide a novel insight into the molecular mechanisms underlying the miRNA-controlled VSMCs dysfunctions in diagnostic and therapeutic approaches against coronary AS.

ACKNOWLEDGEMENTS

None.

Notes

FUNDING

None to declare.

CONFLICTS OF INTEREST

The authors declare no conflicts of interest.

REFERENCES

1. Ou H, Liu C, Feng W, Xiao X, Tang S, Mo Z. 2018; Role of AMPK in atherosclerosis via autophagy regulation. Sci China Life Sci. 61:1212–1221. DOI: 10.1007/s11427-017-9240-2. PMID: 29656339.
crossref
2. Boudoulas KD, Triposciadis F, Geleris P, Boudoulas H. 2016; Coronary atherosclerosis: pathophysiologic basis for diagnosis and management. Prog Cardiovasc Dis. 58:676–692. DOI: 10.1016/j.pcad.2016.04.003. PMID: 27091673.
crossref
3. Bennett MR, Sinha S, Owens GK. 2016; Vascular smooth muscle cells in atherosclerosis. Circ Res. 118:692–702. DOI: 10.1161/CIRCRESAHA.115.306361. PMID: 26892967. PMCID: PMC4762053.
crossref
4. Wang J, Uryga AK, Reinhold J, Figg N, Baker L, Finigan A, Gray K, Kumar S, Clarke M, Bennett M. 2015; Vascular smooth muscle cell senescence promotes atherosclerosis and features of plaque vulnerability. Circulation. 132:1909–1919. DOI: 10.1161/CIRCULATIONAHA.115.016457. PMID: 26416809.
crossref
5. Johnson JL. 2014; Emerging regulators of vascular smooth muscle cell function in the development and progression of atherosclerosis. Cardiovasc Res. 103:452–460. DOI: 10.1093/cvr/cvu171. PMID: 25053639.
crossref
6. Hutcheson JD, Goettsch C, Bertazzo S, Maldonado N, Ruiz JL, Goh W, Yabusaki K, Faits T, Bouten C, Franck G, Quillard T, Libby P, Aikawa M, Weinbaum S, Aikawa E. 2016; Genesis and growth of extracellular-vesicle-derived microcalcification in atherosclerotic plaques. Nat Mater. 15:335–343. DOI: 10.1038/nmat4519. PMID: 26752654. PMCID: PMC4767675.
crossref
7. Jiang XI, Luo Y, Zhao S, Chen Q, Jiang C, Dai Y, Chen Y, Cao Z. 2015; Clinical significance and expression of microRNA in diabetic patients with erectile dysfunction. Exp Ther Med. 10:213–218. DOI: 10.3892/etm.2015.2443. PMID: 26170937. PMCID: PMC4486873.
crossref
8. Zhu N, Zhang D, Chen S, Liu X, Lin L, Huang X, Guo Z, Liu J, Wang Y, Yuan W, Qin Y. 2011; Endothelial enriched microRNAs regulate angiotensin II-induced endothelial inflammation and migration. Atherosclerosis. 215:286–293. DOI: 10.1016/j.atherosclerosis.2010.12.024. PMID: 21310411.
crossref
9. Ambros V. 2004; The functions of animal microRNAs. Nature. 431:350–355. DOI: 10.1038/nature02871. PMID: 15372042.
crossref
10. Ndrepepa G, Colleran R, Kastrati A. 2018; Gamma-glutamyl transferase and the risk of atherosclerosis and coronary heart disease. Clin Chim Acta. 476:130–138. DOI: 10.1016/j.cca.2017.11.026. PMID: 29175647.
crossref
11. Xu Z, Han Y, Liu J, Jiang F, Hu H, Wang Y, Liu Q, Gong Y, Li X. 2015; miR-135b-5p and miR-499a-3p promote cell proliferation and migration in atherosclerosis by directly targeting MEF2C. Sci Rep. 5:12276. DOI: 10.1038/srep12276. PMID: 26184978. PMCID: PMC4505325.
crossref
12. Zhang R, Sui L, Hong X, Yang M, Li W. 2017; miR-448 promotes vascular smooth muscle cell proliferation and migration in through directly targeting MEF2C. Environ Sci Pollut Res Int. 24:22294–22300. DOI: 10.1007/s11356-017-9771-1. PMID: 28799067.
crossref
13. Lin B, Feng DG, Wang F, Wang JX, Xu CG, Zhao H, Cheng ZY. 2016; miR-365 participates in coronary atherosclerosis through regulating IL-6. Eur Rev Med Pharmacol Sci. 20:5186–5192. PMID: 28051250.
14. Lu L, Cai M, Peng M, Wang F, Zhai X. 2019; miR-491-5p functions as a tumor suppressor by targeting IGF2 in colorectal cancer. Cancer Manag Res. 11:1805–1816. DOI: 10.2147/CMAR.S183085. PMID: 30863186. PMCID: PMC6391127.
15. Guo J, Luo C, Yang Y, Dong J, Guo Z, Yang J, Lian H, Ye C, Liu M. 2021; miR-491-5p, as a tumor suppressor, prevents migration and invasion of breast cancer by targeting ZNF-703 to regulate AKT/mTOR pathway. Cancer Manag Res. 13:403–413. DOI: 10.2147/CMAR.S279747. PMID: 33488122. PMCID: PMC7816048.
crossref
16. Sun R, Liu Z, Tong D, Yang Y, Guo B, Wang X, Zhao L, Huang C. 2017; miR-491-5p, mediated by Foxi1, functions as a tumor suppressor by targeting Wnt3a/β-catenin signaling in the development of gastric cancer. Cell Death Dis. 8:e2714. DOI: 10.1038/cddis.2017.134. PMID: 28358374. PMCID: PMC5386537.
crossref
17. He Z, Wang Y, He Q, Chen M. 2020; microRNA-491-5p protects against atherosclerosis by targeting matrix metallopeptidase-9. Open Med (Wars). 15:492–500. DOI: 10.1515/med-2020-0047. PMID: 33313408. PMCID: PMC7706122.
crossref
18. Yuan M, Zhan Q, Duan X, Song B, Zeng S, Chen X, Yang Q, Xia J. 2013; A functional polymorphism at miR-491-5p binding site in the 3'-UTR of MMP-9 gene confers increased risk for atherosclerotic cerebral infarction in a Chinese population. Atherosclerosis. 226:447–452. DOI: 10.1016/j.atherosclerosis.2012.11.026. PMID: 23257658.
crossref
19. Woo CC, Liu W, Lin XY, Dorajoo R, Lee KW, Richards AM, Lee CN, Wongsurawat T, Nookaew I, Sorokin V. 2019; The interaction between 30b-5p miRNA and MBNL1 mRNA is involved in vascular smooth muscle cell differentiation in patients with coronary atherosclerosis. Int J Mol Sci. 21:11. DOI: 10.3390/ijms21010011. PMID: 31861407. PMCID: PMC6982107.
crossref
20. Bartel DP. 2004; MicroRNAs: genomics, biogenesis, mechanism, and function. Cell. 116:281–297. DOI: 10.1016/S0092-8674(04)00045-5. PMID: 14744438.
21. Song Z, Li G. 2010; Role of specific microRNAs in regulation of vascular smooth muscle cell differentiation and the response to injury. J Cardiovasc Transl Res. 3:246–250. DOI: 10.1007/s12265-010-9163-0. PMID: 20543900. PMCID: PMC2883267.
crossref
22. Chen Q, Yang F, Guo M, Wen G, Zhang C, Luong le A, Zhu J, Xiao Q, Zhang L. 2015; miRNA-34a reduces neointima formation through inhibiting smooth muscle cell proliferation and migration. J Mol Cell Cardiol. 89(Pt A):75–86. DOI: 10.1016/j.yjmcc.2015.10.017. PMID: 26493107.
crossref
23. Davis-Dusenbery BN, Wu C, Hata A. 2011; Micromanaging vascular smooth muscle cell differentiation and phenotypic modulation. Arterioscler Thromb Vasc Biol. 31:2370–2377. DOI: 10.1161/ATVBAHA.111.226670. PMID: 22011749. PMCID: PMC4429757.
crossref
24. Albinsson S, Sessa WC. 2011; Can microRNAs control vascular smooth muscle phenotypic modulation and the response to injury? Physiol Genomics. 43:529–533. DOI: 10.1152/physiolgenomics.00146.2010. PMID: 20841497. PMCID: PMC3110893.
crossref
25. Tabas I. 2010; Macrophage death and defective inflammation resolution in atherosclerosis. Nat Rev Immunol. 10:36–46. DOI: 10.1038/nri2675. PMID: 19960040. PMCID: PMC2854623.
crossref
26. Libby P, Ridker PM, Hansson GK. 2011; Progress and challenges in translating the biology of atherosclerosis. Nature. 473:317–325. DOI: 10.1038/nature10146. PMID: 21593864.
crossref
27. Pordzik J, Pisarz K, De Rosa S, Jones AD, Eyileten C, Indolfi C, Malek L, Postula M. 2018; The potential role of platelet-related microRNAs in the development of cardiovascular events in high-risk populations, including diabetic patients: a review. Front Endocrinol (Lausanne). 9:74. DOI: 10.3389/fendo.2018.00074. PMID: 29615970. PMCID: PMC5869202.
crossref
28. Fasolo F, Di Gregoli K, Maegdefessel L, Johnson JL. 2019; Non-coding RNAs in cardiovascular cell biology and atherosclerosis. Cardiovasc Res. 115:1732–1756. DOI: 10.1093/cvr/cvz203. PMID: 31389987. PMCID: PMC7967706.
crossref
29. Hueso M, Mallén A, Casas Á, Guiteras J, Sbraga F, Blasco-Lucas A, Lloberas N, Torras J, Cruzado JM, Navarro E. 2020; Integrated miRNA/mRNA counter-expression analysis highlights oxidative stress-related genes CCR7 and FOXO1 as blood markers of coronary arterial disease. Int J Mol Sci. 21:1943. DOI: 10.3390/ijms21061943. PMID: 32178422. PMCID: PMC7139611.
crossref
30. He Y, Lin L, Cao J, Mao X, Qu Y, Xi B. 2015; Up-regulated miR-93 contributes to coronary atherosclerosis pathogenesis through targeting ABCA1. Int J Clin Exp Med. 8:674–681. PMID: 25785043. PMCID: PMC4358498.
31. Fan JL, Zhang L, Bo XH. 2020; miR-126 on mice with coronary artery disease by targeting S1PR2. Eur Rev Med Pharmacol Sci. 24:893–904. DOI: 10.26355/eurrev_202001_20074. PMID: 32016996.

Fig. 1

Expression of 10 mRNAs in plaques and adjacent intimal tissues from atherosclerosis (AS) patients.

(A) Expression levels of ten mRNAs (UBE2G2, SNORD45C, SNORD45A, SNORD45B, RABGGTB, SLC16A3, POLR2C, PNO1, CEMP1, AMDHD2) in plaques and adjacent intima from coronary AS patients were measured using RT-qPCR with pared t-test, as presented by the scatter diagram. (B) mRNAs (UBE2G2, SNORD45C, SNORD45A, SLC16A3, POLR2C, PNO1, CEMP1, AMDHD2) with significant difference were listed in the bar graph. *p < 0.05, **p < 0.01, NS indicates no significance.
kjpp-26-3-183-f1.tif
Fig. 2

Expression of 10 mRNAs in serum from atherosclerosis (AS) patients and healthy controls.

(A) Expression levels of ten mRNAs (UBE2G2, SNORD45C, SNORD45A, SNORD45B, RABGGTB, SLC16A3, POLR2C, PNO1, CEMP1, AMDHD2) in the serum of coronary AS patients and healthy patients were measured using RT-qPCR with unpaired t-test. (B) mRNAs (UBE2G2, SNORD45C, SNORD45A, SLC16A3, POLR2C, PNO1, CEMP1, AMDHD2) with significant difference were listed. *p < 0.05, **p < 0.01, ***p < 0.001, NS indicates no significance.
kjpp-26-3-183-f2.tif
Fig. 3

Potential upstream miRNAs for the abnormally expressed mRNAs.

(A) TargetScan predicted the potential upstream miRNAs of UBE2G2, SLC16A3, POLR2C, PNO1, and AMDHD2. (B) Expression levels of 11 miRNAs in plaques and adjacent intima from coronary AS patients were examined using RT-qPCR with pared t-test. (C) Expression levels of 11 miRNAs in the serum of coronary atherosclerosis (AS) patients and healthy patients were examined using RT-qPCR with unpaired t-test. *p < 0.05, **p < 0.01.
kjpp-26-3-183-f3.tif
Fig. 4

MiR-491-5p negatively regulates UBE2G2, SLC16A3, PNO1 expression in vascular smooth muscle cells (VSMCs).

(A) Overexpression efficiency of miR-491-5p in VSMCs was evaluated using RT-qPCR. (B) Expression levels of UBE2G2, SLC16A3, POLR2C, PNO1, and AMDHD2 in VSMCs transfected with miR-491-5p mimics and NC mimics were examined by RT-qPCR. (C) Protein levels of UBE2G2, SLC16A3, POLR2C, PNO1, and AMDHD2 in VSMCs transfected with miR-491-5p mimics and NC mimics were examined by Western blot. (D) RNA pull down assay was for detecting whether UBE2G2, SLC16A3, and PNO1 bind with miR-491-5p. *p < 0.05, **p < 0.01, ***p < 0.001.
kjpp-26-3-183-f4a.tifkjpp-26-3-183-f4b.tifkjpp-26-3-183-f4c.tif
Fig. 5

MiR-491-5p inhibited viability, migration, and increased expression of contractile markers in vascular smooth muscle cells (VSMCs).

(A) Cell counting kit 8 (CCK-8) revealed the viability in VSMCs with transfection of miR-491-5p mimics and NC mimics. (B) Wound healing was performed to detect the migration capacity of VSMCs with transfection of miR-491-5p mimics and NC mimics. (C) The protein levels of contractile markers (α-SMA, calponin, SM22α, and smoothelin) in VSMCs with transfection of miR-491-5p mimics and NC mimics were measured by Western blot. *p < 0.05, **p < 0.01.
kjpp-26-3-183-f5a.tifkjpp-26-3-183-f5b.tif
Fig. 6

MiR-491-5p antagomir promotes viability, migration, and increased expression of contractile markers in vascular smooth muscle cells (VSMCs).

(A) CCK-8 revealed the viability in VSMCs with transfection of NC antagomir, miR-491-5p antagomir, miR-491-5p antagomir+sh-NC, miR-491-5p antagomir+sh-UBE2G2, miR-491-5p antagomir+sh-SLC16A3, and miR-491-5p antagomir+sh PNO1. (B) Wound healing was performed to detect the migration capacity of VSMCs with transfection of NC antagomir, miR-491-5p antagomir, miR-491-5p antagomir+sh-NC, miR-491-5p antagomir+sh-UBE2G2, miR-491-5p antagomir+sh- SLC16A3, and miR-491-5p antagomir+sh PNO1. (C) The protein levels of contractile markers (α-SMA, calponin, SM22α, and smoothelin) in VSMCs with transfection of NC antagomir, miR-491-5p antagomir, miR-491-5p antagomir+sh-NC, miR-491-5p antagomir+sh-UBE2G2, miR-491-5p antagomir+sh- SLC16A3, and miR-491-5p antagomir+sh PNO1. *p < 0.05, **p < 0.01.
kjpp-26-3-183-f6a.tifkjpp-26-3-183-f6b.tif
Table 1
Clinical characteristics of patients with coronary atherosclerosis
Parameters Cases (n)
Age
≤ 50 9
> 50 13
Sex
Female 6
Male 16
Diabetes mellitus
Yes 10
No 12
Hypertension
Yes 11
No 11
Hyperlipidemia
Yes 16
No 6
Smoking
Yes 14
No 8
Ejection fraction
Good 12
Fair 8
Poor 2

p-values less than 0.05 indicate statistical significance. Chi-square test was performed.

Table 2
Primers used for quantitative RT-PCR
Name Sequence (5´-3´) Forward Sequence (5´-3´) Reverse
UBE2G2 GAAGGAATTGTAGCAGGCC CATCAGGGTAGATGTTGGGA
SNORD45C GATGAGTTGGCATGTATTCTG GGTCTCAGAGTAATTCTAGAGC
SNORD45A GGTCAATGATGTGTTGGCA GGTCTCAGAGTAATTCTAGAGCT
SLC16A3 CATCATCCAGGTCTACCTCAC GAAGTTGAGTGCCAAACCC
POLR2C CACAGGCTTGGATTAATTCCC ACTCCTCACATGTGCAGTC
PNO1 GAAAGTTCACATCCTTGGCTC GGAGGATTTCCCAAGATTAGGT
CEMP1 ACCTCTAACACATCGGCTG TAGTGTCATCCTGCCTGTG
AMDHD2 GAAGCTGTTCTTTGAGGAGC CCAAATCCACCGTTGATCTG
miR-491-5p GGAGTGGGGAACCCTTCC GTGCAGGGTCCGAGGT
miR-4645-5p GCGACCAGGCAAGAAATATTG AGACAGTAGTTCTTGCCTGGTCCT
miR-4673 ACACTCCAGCTGGGAGGTC TCGGCACTCAACTGGTCC
miR-6791-5p AGGCGGGCAGAGAA GGTCTGGCTCCGTCCG
miR-6727-3p GCCTGCCACCUCCUCCGCA CTCGGGGCAGGCGGCTGG
miR-6747-3p GTGGAAAGAGGCAGAACA GCCGCCTTCCTCTGCACCA
miR-4308 AGGTAAGAAGAAACTCCAGGGAA GGGTTCAGAGGGAACTCCATT
miR-4722-3p GGCAGGAGGGCTGTGCCAGGTTG CTGCAGGGAGGTGCTGGCAGGT
miR-4292 TTTAATACGTACTTTAGCTGAATTATTCTATGTCAATG AATAAATTTTGTCTTAACAAGGTAAGTTGTGAAAAG
miR-6852-5p CCCTGGGGTTCTGAGGACATG CATGTCCTCAGAACCCCAGGG
miR-7160-5p TGCTUGAGGTCCGGGCTGTGCC TGCTGAGGTCCGGGCTGTGCC
Table 3
Top 10 genes that are differentially expressed in blood outgrowth endothelial cells from subjects with normal or abnormal coronary endothelial function
Gene symbol Chromosome location Gene title p-value
UBE2G2 21q22.3 Ubiquitin conjugating enzyme E2 G2 0.0000145
SNORD45C 1p31.1 Small nucleolar RNA, C/D box 45C 0.0000151
SNORD45A 1p31.1 Small nucleolar RNA, C/D box 45A 0.0000151
SNORD45B 1p31.1 Small nucleolar RNA, C/D box 45B 0.0000151
RABGGTB 1p31 Rab geranylgeranyltransferase beta subunit 0.0000151
SLC16A3 17q25 Solute carrier family 16 member 3 0.0000213
POLR2C 16q13-q21 RNA polymerase II subunit C 0.0000223
PNO1 2p14 Partner of NOB1 homolog 0.0000265
CEMP1 16p13.3 Cementum protein 1 0.0000299
AMDHD2 16p13.3 Amidohydrolase domain containing 2 0.0000299

Data were downloaded from GSE132651.

TOOLS
Similar articles