miR‑132 inhibits high glucose‑induced vascular smooth muscle cell proliferation and migration by targeting E2F5

  • Authors:
    • Qun Xu
    • Ying Liang
    • Xiangjuan Liu
    • Chunmei Zhang
    • Xiaoqian Liu
    • Hong Li
    • Jiangjiu Liang
    • Guang Yang
    • Zhiming Ge
  • View Affiliations

  • Published online on: June 12, 2019     https://doi.org/10.3892/mmr.2019.10380
  • Pages: 2012-2020
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The dysregulated behavior of vascular smooth muscle cells (VSMCs) serves an important role in the pathogenesis of cardiovascular diseases in diabetes. The present study aimed to investigate the effects of microRNA (miR)‑132 on the proliferation and migration of VSMCs under high glucose conditions to mimic diabetes. We observed that the expression of miR‑132 was significantly decreased and that of E2F transcription factor 5 (E2F5) was upregulated in high glucose (HG)‑treated VSMCs or those obtained from diabetic rats. A dual luciferase reporter gene assay revealed that miR‑132 could specifically bind to the 3'‑untranslated region of E2F5 and significantly suppress the luciferase activity. The proliferation and migration of diabetic rat or HG‑treated VSMCs were increased compared with non‑diabetic rat VSMCs and those under normal glucose conditions. Upregulation of miR‑132 significantly inhibited the proliferation and migration of diabetic rat VSMCs; similar effects were observed following E2F5 downregulation. The inhibitory effects of miR‑132 on the proliferation and migration of HG‑treated VSMCs could be reversed by E2F5 overexpression. In conclusion, miR‑132 was proposed to inhibit the proliferation and migration of diabetic rat or high‑glucose‑treated VSMCs by targeting E2F5. The findings of the present study suggested that increasing the expression of miR‑132 may serve as a novel therapeutic approach to inhibit the progression of cardiovascular disease in diabetes.

Introduction

Diabetes mellitus (DM), as a form of chronic metabolic disease, is characterized by the lack of insulin and/or insulin resistance that results in hyperglycemia and abnormal metabolism (1,2). Long-term hyperglycemia can lead to various complications, including cardiovascular disease (CVD) that accounts for approximately a third of all mortalities worldwide (2,3). In addition, DM is considered as an independent risk factor for CVD, excluding other factors such as obesity, hypertension and age (4,5). Vascular smooth muscle cells (VSMCs) serve a key role in vascular remodeling (6). Accumulating evidence has indicated that the proliferation and migration of VSMCs were important features of numerous types of CVD, including atherosclerosis, hypertension and restenosis (79). It has been reported that high glucose (HG) can induce the excessive proliferation and migration of VSMCs, which promote the progression of diabetic vascular diseases (10,11). Thus, understanding the mechanism of HG-mediated VSMC proliferation and migration is important for improving current intervention strategies for the treatment of vascular diseases in diabetes.

MicroRNAs (miRNAs/miRs) are a type of small noncoding single-stranded RNAs (~21–23 nucleotides) that negatively regulate the expression of their target genes by complete or partial complementary binding with the 3′-untranslated region (3′UTR) of these target genes, to modulate various physiological and pathological processes, including cancer, DM, CVD and other diseases (1214). Several studies have demonstrated that miRNAs are involved in regulating the dysfunction of VSMC behavior (1517), including miR-145 (18), miR-155 (19), miR-146 (20), miR-504 (21), miR-24 (22) and miR-126 (23).

miR-132 originates from the miR-212/132 cluster that is located on chromosome 17 in humans (24). The majority of studies of miR-132 revealed its roles in cancer and the regulation of neurons (2426). A recent study indicated that miR-132 regulated brain vascular integrity (27). Additionally, the expression levels of miR-132 are downregulated in VSMCs in diabetic rat models (21); however, the role and mechanism of miR-132 in diabetes-induced VSMC dysfunction are not clear. The present study aimed to investigate the effects of miR-132 on the proliferation and migration of VSMCs under HG conditions to mimic diabetes, which may provide insight into the biological mechanism for the development of novel approaches to treat diabetic vascular complications.

Materials and methods

Animals

The diabetic animal model was generated as previously described (28). In total, 24 male Sprague-Dawley rats (age, 4–6 weeks; weight, 180–200 g) were obtained from Beijing Vital River Laboratory Animal Technology Co., Ltd. (Beijing, China). A single dose of streptozotocin (STZ; 65 mg/kg, citrate buffer) was injected into 12 rats. An equal volume of citrate buffer was used as the control treatment (n=12). When blood glucose levels were >250 mg/dl, the animals were defined as diabetic rats. After 2 days following treatment, the animals became hyperglycemic. All rats were sacrificed by cervical dislocation under ketamine administration. Thoracic aorta samples from non-diabetic and diabetic rats were collected and isolated; mRNA and protein were extracted for further analysis. All the procedures and experimental protocols were approved by the Institutional Animal Care and Use Committee of Qianfoshan Hospital of Shandong (Jinan, China).

Cell culture and treatment

The isolation of primary VSMCs from the thoracic aorta of non-diabetic and diabetic rats was performed according to a previous study (29). Morphological and immunohistochemical analyses of VSMCs were performed as previously described (29). The 293 cells were purchased from the Shanghai Institute of Chinese Academy of Sciences (Shanghai, China). VSMCs and 293 cells were maintained in DMEM (HyClone; Thermo Fisher Scientific, Inc.) with 10% FBS (HyClone; Thermo Fisher Scientific, Inc.), as well as penicillin (100 U/ml; Bio-sciences Ltd., Dublin, Ireland) and streptomycin (100 U/ml; Bio-sciences, Ltd.) at 37°C within 5% CO2. Experiments were conducted on transduced and non-transduced VSMCs of passages 3–8. For HG treatment, VSMCs were cultured in normal media (5 mM glucose) for 24 h, and then incubated with HG (25 mM glucose) or 20 mM mannitol plus 5 mM glucose (normal glucose, NG) for 24 h at 37°C with 5% CO2.

Plasmid construction and infection

miR-132 precursor and the corresponding miR-control were designed and synthesized by Shanghai GeneChem Co., Ltd. (Shanghai, China), and were cloned into pCDH-CMV-MCS-EF1-coGFP (System Biosciences, LLC, Palo Alto, CA, USA). The gene encoding E2F transcription factor 5 (E2F5) lacking the 3′UTR was amplified from rat genomic DNA by PCR using the Phusion High-Fidelity PCR Kit (Thermo Fisher Scientific, Inc., Waltham, MA, USA) according to the manufacturer's protocol. PCR thermocycling conditions were as follows: Initial denaturation at 95°C for 30 sec, followed by 40 cycles of 95°C for 15 sec, 60°C for 30 sec, 72°C for 1 min and 4°C for 1 min. The following primers were used: E2F5 forward, AGAATTCATGGCGGCGGCGGAGCCCA and reverse, ATGGATCCCTAATAATTTAGTATCTGAACATCAA. Then, the PCR product was cloned into pCDH-CMV-MCS-EF1-Puro (Addgene). The production and purification of lentivirus and cell infection (multiplicity of infection, 100) were performed as previously described (30,31). Empty lentiviral vector pCDH-CMV-MCS-EF1-Puro (vector) was used as the control for lenti-E2F5 transduction. VSMCs from non-diabetic rats were infected with lenti-E2F5 or vector. Small interfering (si)RNAs targeting two different sites (cat. nos. RSS350386 and RSS350386) of the rat E2F5 gene and the negative control (cat. no. 12935100; scramble) were purchased from Thermo Fisher Scientific Inc. Cells were transfected with E2F5 siRNAs (100 nM) or scramble (100 nM) using Lipofectamine® 2000 (Thermo Fisher Scientific Inc.) according to the manufacturer's protocols. miR-132 inhibitor (cat. no. 4464084) and corresponding negative control (inhibitor-NC; cat. no. 4464076) as well as miR-132 mimic (cat. no. 4464066) and corresponding negative control (miR-NC; cat. no. 4464058) were obtained from Thermo Fisher Scientific Inc. miR-132 mimic (100 nM) and miR-NC (100 nM) were used to transfect diabetic VSMCs. Normal VSMCs were transfected with miR-132 inhibitor (100 nM) and inhibitor-NC (100 nM) using Lipofectamine 2000 (Thermo Fisher Scientific Inc.) according to the manufacturer's protocol. Cells were collected and used for analysis at 72 h after infection or transfection. All constructions were confirmed by plasmid DNA sequencing.

Dual luciferase assays

miR-132 mimic, miR-132 inhibitor and corresponding negative control (miR-NC and inhibitor-NC, respectively) were obtained from Shanghai GenePharma Co., Ltd. (Shanghai, China). The wild-type E2F5 3′UTR and mutant-type E2F5 3′UTR were cloned into the reporter plasmid pGL3 (Promega Corporation, Madison, WI, USA) downstream of the luciferase reporter gene according to as previously described (26). For the dual luciferase assay, 293 cells were seeded into a 24-well plate at a density of 2×104 cells per well. Cells were co-transfected with 1 µg of wild-type or mutant luciferase vector and miR-132mimics (100 nM), miR-132 inhibitor (100 nM) or their NCs. After co-transfection for 48 h, luciferase activity was detected using the dual luciferase Reporter Assay System (Promega Corporation) according to the manufacturer's instructions. Firefly luciferase activity was normalized to Renilla luciferase activity.

VSMC proliferation assay

A bromodeoxyuridine (BrdU) incorporation assay was employed to detect the proliferation of VSMCs (22). VSMCs were transfected with E2F5 siRNAs (100 nM) or scramble (100 nM) using Lipofectamine 2000 (Thermo Fisher Scientific Inc.) according to the manufacturer's protocols. miR-132 mimic was used to transfect diabetic VSMCs. Normal VSMCs were transfected with miR-132 inhibitor using Lipofectamine 2000 (Thermo Fisher Scientific Inc.) according to the manufacturer's protocols. Following transfection, stable infection or co-infection of miR-132, miR-NC, lenti-E2F5 and lenti-vector, cells were seeded into 96-well plates at 3×103 cells/well. Cells were cultured with NG or HG media for 24 h, and were then starved in FBS-free DMEM containing the aforementioned concentrations of glucose (NG or HG) for 24 h at 37°C with 5% CO2. Subsequently, BrdU (10 µmol/l) was added into each well and incubated at 37°C for 30 min. The cells were fixed in 4% paraformaldehyde for 15 min at room temperature, and then detected using the cell proliferation ELISA kit (Roche Diagnostics, Mannheim, Germany) according to the manufacturer's instructions. Absorbance values were detected at a wavelength of 450 nm using a microplate reader (Thermo Fisher Scientific, Inc.).

VSMC migration assay

VSMC migration was detected by Transwell migration assay (22). VSMCs from non-diabetic rats were infected with lenti-E2F5 or vector. Following transfection, stable infection or co-infection of miR-132, miR-control, lenti-E2F5 and lenti-vector, cells were treated with NG or HG media for 48 h. Cells (5×104) were plated into the upper chamber with 8-mm pore size (BD Bioscience, Franklin Lakes, NJ, USA), while 800 µl DMEM containing 1% FBS was placed in the lower chamber. After incubation for 6 h at 37°C, the cells in the upper membrane were removed with cotton swabs, and cells on the lower membrane were fixed in 4% paraformaldehyde for 15 min at room temperature and stained with crystal violet (1%) for 1 h at room temperature (Beyotime Institute of Biotechnology, Haimen, China). Cells were analyzed in 10 random fields per well (magnification, ×100) using a phase contrast light microscope.

Protein extraction and western blotting

Thoracic aorta tissue or VSMC was lysed with radioimmunoprecipitation assay lysis buffer (Beyotime Institute of Biotechnology) containing protease inhibitors (Roche Diagnostics) according to the manufacturer's instructions. The protein concentrations were measured with a BCA Protein Assay kit (Applygen Technologies, Inc., Beijing, China) according to the manufacturer's protocols. Protein samples (20 µg for each sample) were fractionated by SDS-PAGE on 10% gels, and then transferred to a polyvinylidene fluoride membrane (EMD Millipore, Bedford, MA, USA). The PVDF membranes were incubated with anti-E2F5 antibody (1:1,500; cat. no. ab176017; Abcam, Cambridge, UK) or anti-β-actin antibody (1:6,000; cat. no. ab8226; Abcam) at 4°C overnight. Then, the membranes were washed and incubated with horseradish peroxidase-conjugated secondary antibodies (1:4,000; cat. no. ab205718; Abcam) for 1 h at room temperature. Finally, the protein bands were detected using an enhanced chemiluminescence detection kit (Pierce; Thermo Fisher Scientific, Inc.) and a ChemoDoc XRS detection system (Bio-Rad Laboratories, Inc.). The results were analyzed with ImageJ software (version 1.34e; National Institutes of Health, Bethesda, MD, USA).

RNA extraction and reverse-transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA from thoracic aorta tissue or VSMCs was isolated using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocols. miRNAs were reversely transcribed into cDNA using the miScript Reverse Transcription Kit (Qiagen GmbH; cat. no. 218060) according to the manufacturer's protocol. For mRNA detection, cDNA was generated using the PrimeScript RT reagent kit (Takara Bio, Inc.) according to the manufacturer's protocol. miR-132 and E2F5 expression levels were quantified using the SYBR Green PCR Master Mix kit (Applied Biosystems; cat. no. 4309155). The sequences of the primers used are as follows: miR-132 forward, 5′-GGGTAACAGTCTACAGCCAT-3′ and miR-132 reverse, 5′-GGCAATTGCACTGGATAC-3′; U6 forward, 5′-GCTTCGGCAGCACATATACTAAAAT-3′ and U6 reverse, 5′-CGCTTCACGAATTTGCGTGTCAT-3′; rat E2F5 forward, 5′-GTACTTCCTTTGGCCTTAGTTT-3′ and reverse, 5′-CTGGCACCATCACACCGACATA-3′; rat β-actin forward, 5′-TGTCACCAACTGGGACGATATG-3′ and reverse, 5′-GGCTGGGGTGTTGAAGGTCTC-3′. The expression levels of miR-132 and E2F5 were quantified using the 2−ΔΔCq method (32). RT-qPCR experiments were performed in triplicate.

Databases and bioinformatics analysis

The predicted target genes of miR-132 were identified using TargetScan (version 7.2; http://www.targetscan.org).

Statistical analyses

Data were expressed as the mean ± SD. One-way ANOVA followed by a Tukey's post-hoc test was conducted to assess significant differences. All statistical calculations were performed using SPSS 19.0 software (IBM Corp. Armonk, NY, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

Expression of miR-132 and E2F5 in thoracic aorta samples and VSMCs from non-diabetic and diabetic rats

To investigate the potential roles of miR-132 and E2F5 in VSMCs under diabetic conditions, the expression of miR-132 and E2F5 in thoracic aorta samples and VSMCs non-diabetic and diabetic rats was detected by RT-qPCR and western blotting. Compared with the non-diabetic control rats, the expression levels of miR-132 in thoracic aorta samples and VSMCs of diabetic rats were significantly decreased compared with the control (P<0.001; Fig. 1A and C). Conversely, the mRNA and protein expression levels of E2F5 in thoracic aorta samples and VSMCs of diabetic rats were upregulated than in non-diabetic control rats (P<0.001; Fig. 1A-D). Taken together, our results revealed the opposing expression profiles of miR-132 and E2F5, which suggested their conflicting roles in the abnormal behavior of VSMCs mediated by diabetic conditions.

Expression of miR-132 and E2F5 in HG-treated VSMCs

We investigated whether HG stimulation altered expression of miR-132 and E2F5. The results revealed that miR-132 was significantly downregulated (P<0.001), while the mRNA and protein expression levels of E2F5 were increased (P<0.001) in VSMCs incubated with HG media for 24 h compared with the control (Fig. 2). These findings further suggested the opposing expression profiles of miR-132 and E2F5 in HG-treated VSMCs.

E2F5 is a target of miR-132

Generally, miRNAs regulate gene expression by targeting the 3′UTR of these genes. Using TargetScan (http://www.targetscan.org), it was predicted that miR-132 binds to the 3′UTR of E2F5. As presented in Fig. 3A, the paired sequences between miR-132 and E2F5 3′UTR, and the mutant were determined. This prediction was confirmed by a dual luciferase reporter assay. The relative luciferase activity of the reporter containing the wild-type 3′UTR was significantly decreased by miR-132 overexpression compared with the control, but increased following transfection with miR-132 inhibitor (P<0.001; Fig. 3B). However, the luciferase activity of the mutant reporter was markedly affected by miR-132 overexpression and knockdown (Fig. 3B). These results suggested that miR-132 could directly bind to the 3′UTRs of E2F5 which were mutated in this study. We also overexpressed miR-132 in VSCMs from diabetic rats. The expression levels of miR-132 and E2F5 were detected by RT-qPCR and western blotting, respectively. The results showed the successful transfection of cells with miR-132 mimics (Fig. 3C); the expression of E2F5 was significantly decreased by miR-132 overexpression in the VSCMs from diabetic rats compared with the control (Fig. 3D). Additionally, downregulation of miR-132 (Fig. 3E) significantly increased the expression of E2F5 in VSCMs compared with the control (Fig. 3F). Our findings suggested that E2F5 was a target of miR-132.

Effects of miR-132 and E2F5 on VSMC proliferation and migration

Excessive cell proliferation and migration of VSMCs are fundamental mechanisms underlying CVDs in diabetes (10). This prompted us to investigate the functions of miR-132 and E2F5 in the proliferation and migration of VSMCs from diabetic rats by the nuclear incorporation of BrdU (DNA synthesis) and Transwell migration assays. We reported that the proliferation and migration of VSMCs from diabetic rats were significantly increased than that of VSMCs of non-diabetic rats (P<0.001; Fig. 4A and B). Furthermore, miR-132 was overexpressed in VSMCs from diabetic rats (Fig. 3C and D), then the proliferation and migration of cells were analyzed. The results showed that diabetic rat-derived VSMCs transfected with miR-132 exhibited significantly reduced proliferation and migration compared with the control group (P<0.001; Fig. 4C and D). In addition, VSMCs from non-diabetic rats were infected with lenti-E2F5 or vector. The results demonstrated that the expression of E2F5 significantly increased 5.2-fold in the lenti-E2F5 infection group compared the control group (P<0.001; Fig. 4E). Overexpression of E2F5 significantly increased the proliferative and migration abilities of normal rat VSMCs compared with the control (P<0.001; Fig. 4F and G). Thus, these data suggested that miR-132 and E2F5 serve opposing roles in the proliferation and migration of VSMCs.

Downregulation of E2F5 inhibits diabetic rat VSMC proliferation and migration

To further confirm the function of E2F5 in VSMCs from diabetic rats, E2F5 expression was knocked down by delivery of E2F5 siRNAs, and the proliferation and migration of these VSMCs were analyzed. As determined by western blot analysis, the expression of E2F5 was significantly knocked down compared with the control (Fig. 5A). Of note, downregulation of E2F5 significantly inhibited the proliferation of diabetic rat VSMCs compared with those transfected with scramble siRNA (Fig. 5B). Additionally, compared with the control, downregulation of E2F5 significantly suppressed migration of diabetic VSMCs (Fig. 5C). Taken together, these results suggested that downregulation of E2F5 inhibited VSMC proliferation and migration similar to miR-132 overexpression.

E2F5 overexpression rescues the inhibitory effects of miR-132 on the proliferation and migration of VSMCs treated with HG

We further examined whether E2F5 was involved in the inhibitory effects of miR-132 on the HG-induced proliferation and migration of VSMCs, overexpression of E2F5 was conducted by infecting miR-132 overexpressed VSMCs; cell proliferation and migration were detected by the nuclear incorporation of BrdU and a Transwell migration assay, respectively. The results showed that miR-132 was successfully overexpressed under HG conditions compared with the corresponding control (Fig. 6A). In addition, miR-132 inhibited HG-induced upregulation of E2F5 in VSMCs (Fig. 6B). Furthermore, transduction with lenti-E2F5 exhibited non-significant effect on the expression of miR-132, but could rescue downregulated E2F5 expression induced by miR-132 (Fig. 6A and B). Furthermore, our results showed that E2F5 overexpression could significantly rescue the inhibitory effects of miR-132 on the proliferation and migration of VSMCs, compared with VSMCs that were co-infected with miR-132 and vector (P<0.001; Fig. 6C-E). Thus, our findings suggested that miR-132 suppressed the proliferation and migration of HG-treated VSMCs by targeting E2F5.

Discussion

CVD-associated complications, such as restenosis, hypertension and atherosclerosis are the leading causes of mortality in patients with diabetes (2). The pathological development of diabetes-related CVDs is characterized by dysfunction of VSMCs (10). Previous studies have reported that miRNAs exhibit a pathophysiological role in VSMCs in diabetic CVD (33). It was demonstrated that miR-132 was downregulated in VSMCs from diabetic mice (21). Accordingly, our results showed that the expression levels of miR-132 were decreased in thoracic aorta tissue and VSMCs of diabetic rats. Furthermore, the expression of miR-132 was suppressed in HG-treated VSMCs. These findings suggested that miR-132 may serve a regulatory role in the dysfunction of VSMCs in diabetes.

It has been reported that miR-132 is involved in diabetic complications and CVDs, including diabetic cardiac microangiopathy, neointimal hyperplasia, cardiac hypertrophy endothelial cell function and angiogenesis (3436). Rawal et al (36) have reported downregulation of proangiogenic miRNA-132 as an early modulator of diabetic cardiac microangiopathy. In addition, miR-132 improved the repair of infarcted heart tissue via angiogenic activation (37). A previous study (37) reported that miR-132 promotes the proliferation of endothelial cells and facilitates pathological angiogenesis. Additionally, miR-132 induced myofibroblast proliferation (38); however, accumulating evidence has indicated that miR-132 inhibited the proliferation of VSMC (34,39). These opposing effects of miR-132 on cell proliferation may be associated with the various function of miRNAs in different cell types and microenvironments. Hyperglycemia is harmful to VSMCs (40); the present study reported that HG-treatment induced the proliferation and migration of VSMCs, which is consistent with recent studies (22,28,41). VSMC proliferation and migration are fundamental processes of vascular dysfunction in diabetes. Therefore, CVD-associated VSMC dysfunction could be disrupted by regulating their proliferation and migration (11,42). In the present study, it was reported that miR-132 significantly inhibited the proliferation and migration of diabetic and HG-treated VSMCs.

miR-132 regulates biological functions by binding to the 3′UTR of target genes, inhibiting their expression. Previous studies have identified many downstream mRNA targets of miR-132, including p120 RasGAP in endothelial cells, RING finger protein 51 in cervical cancer cells and LRR binding FLII interacting protein 1 in VSMCs (34,43,44). TargetScan analysis revealed E2F5 as a potential target of miR-132. Furthermore, our results indicated that miR-132 interacted with the 3′UTR of E2F5, inhibiting its expression. In line with our observations, it has been reported that miR-132 suppresses ovarian cancer cell proliferation and migration by targeting E2F5 (26). E2F5 is a key member of the E2F family that controls the transcription of proliferation-related regulatory genes (45,46). In the present study, diabetic rat-derived and HG-treated VSMCs exhibited high proliferative and migration potentials. In addition, E2F5 was observed to be upregulated in VSMCs from diabetic rats of and HG-treated VSMCs. VSMCs from non-diabetic rats exhibited increased proliferation and migration associated with E2F5 overexpression. In addition, the anti-proliferative and anti-migratory effects of miR-132 in HG-induced or diabetic rat VSMCs could be reversed by E2F5 overexpression. Numerous studies have proposed that E2F5 suppresses the migration of several cancer cells, including breast cancer, ovarian cancer and prostate cancer cells (26,47,48). A previous study (48) has reported that E2F5 binds to the promoter of heme oxygenase 1 (HMOX1) and inhibits its expression. Of note, it has been demonstrated that HMOX1 inhibits VSMC proliferation and migration by inhibiting the MAPK and AKT signaling pathways (49). Combined with the findings of these previous studies, we proposed that the anti-proliferative and anti-migratory effects of miR-132 may be associated with its inhibitory effects on E2F5 expression, which may activate the MAPK and AKT signaling pathways via the suppression of HMOX1; however, further investigation is required.

In summary, our results revealed that miR-132 was downregulated in diabetic rat-derived and HG-treated VSMCs. Furthermore, miR-132 inhibited the proliferation and migration of diabetic rat-derived and HG-treated VSMCs by targeting E2F5. The findings of the present study suggest miR-132 and E2F5 as potentially effective therapeutic targets for treating VSMC dysfunction and CVDs in diabetes.

Acknowledgements

Not applicable.

Funding

This study was supported by the National Natural Science Foundation of China (grant no. 81470404), Natural Science Foundation of Shandong Province, China (grant no. ZR2014HM107) and Projects of Medical and Health Technology Development Program in Shandong province (grant no. 2017WSB04092).

Availability of data and materials

The analyzed datasets generated during the study are available from the corresponding author on reasonable request.

Authors' contributions

QX, GY and ZG contributed to the design of the experiment. CZ, XJL, HL, JL, QX, YL and XQL performed the experiments, analyzed the data and wrote the manuscript. All of the authors read and approved the final manuscript.

Ethics approval and consent to participate

All the procedures and experimental protocols were approved by the Institutional Animal Care and Use Committee of Qianfoshan Hospital of Shandong Province. All the mice were treated in accordance with the Guidelines for the Care and Use of Laboratory Animals published by the U.S. National Institutes of Health (50).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Kahn SE: Clinical review 135: The importance of beta-cell failure in the development and progression of type 2 diabetes. J Clin Endocrinol Metab. 86:4047–4058. 2001. View Article : Google Scholar : PubMed/NCBI

2 

Ding Y, Sun X and Shan PF: MicroRNAs and cardiovascular disease in diabetes mellitus. Biomed Res Int. 2017:40803642017. View Article : Google Scholar : PubMed/NCBI

3 

Quan A, Pan Y, Singh KK, Polemidiotis J, Teoh H, Leong-Poi H and Verma S: Cardiovascular inflammation is reduced with methotrexate in diabetes. Mol Cell Biochem. 432:159–167. 2017. View Article : Google Scholar : PubMed/NCBI

4 

Vamos EP, Millett C, Parsons C, Aylin P, Majeed A and Bottle A: Nationwide study on trends in hospital admissions for major cardiovascular events and procedures among people with and without diabetes in England, 2004–2009. Diabetes Care. 35:265–272. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Nichols GA, Hillier TA, Erbey JR and Brown JB: Congestive heart failure in type 2 diabetes: Prevalence, incidence, and risk factors. Diabetes Care. 24:1614–1619. 2001. View Article : Google Scholar : PubMed/NCBI

6 

Li SL, Reddy MA, Cai Q, Meng L, Yuan H, Lanting L and Natarajan R: Enhanced proatherogenic responses in macrophages and vascular smooth muscle cells derived from diabetic db/db mice. Diabetes. 55:2611–2619. 2006. View Article : Google Scholar : PubMed/NCBI

7 

Doran AC, Meller N and McNamara CA: Role of smooth muscle cells in the initiation and early progression of atherosclerosis. Arterioscler Thromb Vasc Biol. 28:812–819. 2008. View Article : Google Scholar : PubMed/NCBI

8 

Gomez D and Owens GK: Smooth muscle cell phenotypic switching in atherosclerosis. Cardiovasc Res. 95:156–164. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Wall VZ and Bornfeldt KE: Arterial smooth muscle. Arterioscler Thromb Vasc Biol. 34:2175–2179. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Wu WY, Yan H, Wang XB, Gui YZ, Gao F, Tang XL, Qin YL, Su M, Chen T and Wang YP: Sodium tanshinone IIA silate inhibits high glucose-induced vascular smooth muscle cell proliferation and migration through activation of AMP-activated protein kinase. PLoS One. 9:e949572014. View Article : Google Scholar : PubMed/NCBI

11 

Shi L, Ji Y, Jiang X, Zhou L, Xu Y, Li Y, Jiang W, Meng P and Liu X: Liraglutide attenuates high glucose-induced abnormal cell migration, proliferation, and apoptosis of vascular smooth muscle cells by activating the GLP-1 receptor, and inhibiting ERK1/2 and PI3K/Akt signaling pathways. Cardiovasc Diabetol. 14:182015. View Article : Google Scholar : PubMed/NCBI

12 

Ambros V: The functions of animal microRNAs. Nature. 431:350–355. 2004. View Article : Google Scholar : PubMed/NCBI

13 

Ha M and Kim VN: Regulation of microRNA biogenesis. Nat Rev Mol Cell Biol. 15:509–524. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Natarajan R and Nadler JL: Lipid inflammatory mediators in diabetic vascular disease. Arterioscler Thromb Vasc Biol. 24:1542–1548. 2004. View Article : Google Scholar : PubMed/NCBI

15 

Wu H, Kong L, Zhou S, Cui W, Xu F, Luo M, Li X, Tan Y and Miao L: The role of microRNAs in diabetic nephropathy. J Diabetes Res. 2014:9201342014. View Article : Google Scholar : PubMed/NCBI

16 

Alexander MR and Owens GK: Epigenetic control of smooth muscle cell differentiation and phenotypic switching in vascular development and disease. Annu Rev Physiol. 74:13–40. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Maegdefessel L, Rayner KJ and Leeper NJ: MicroRNA regulation of vascular smooth muscle function and phenotype: Early career committee contribution. Arterioscler Thromb Vasc Biol. 35:2–6. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Davis-Dusenbery BN, Chan MC, Reno KE, Weisman AS, Layne MD, Lagna G and Hata A: Down-regulation of Kruppel-like factor-4 (KLF4) by microRNA-143/145 is critical for modulation of vascular smooth muscle cell phenotype by transforming growth factor-beta and bone morphogenetic protein 4. J Biol Chem. 286:28097–28110. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Zhang RN, Zheng B, Li LM, Zhang J, Zhang XH and Wen JK: Tongxinluo inhibits vascular inflammation and neointimal hyperplasia through blockade of the positive feedback loop between miR-155 and TNF-alpha. Am J Physiol Heart Circ Physiol. 307:H552–H562. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Sun SG, Zheng B, Han M, Fang XM, Li HX, Miao SB, Su M, Han Y, Shi HJ and Wen JK: miR-146a and Krüppel-like factor 4 form a feedback loop to participate in vascular smooth muscle cell proliferation. EMBO Rep. 12:56–62. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Reddy MA, Das S, Zhuo C, Jin W, Wang M, Lanting L and Natarajan R: Regulation of vascular smooth muscle cell dysfunction under diabetic conditions by miR-504. Arterioscler Thromb Vasc Biol. 36:864–873. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Yang J, Chen L, Ding J, Fan Z, Li S, Wu H, Zhang J, Yang C, Wang H, Zeng P and Yang J: MicroRNA-24 inhibits high glucose-induced vascular smooth muscle cell proliferation and migration by targeting HMGB1. Gene. 586:268–273. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Zhou J, Li YS, Nguyen P, Wang KC, Weiss A, Kuo YC, Chiu JJ, Shyy JY and Chien S: Regulation of vascular smooth muscle cell turnover by endothelial cell-secreted microRNA-126: Role of shear stress. Circ Res. 113:40–51. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Nudelman AS, DiRocco DP, Lambert TJ, Garelick MG, Le J, Nathanson NM and Storm DR: Neuronal activity rapidly induces transcription of the CREB-regulated microRNA-132, in vivo. Hippocampus. 20:492–498. 2010.PubMed/NCBI

25 

Zhang S, Liang Z, Sun W and Pei L: Repeated propofol anesthesia induced downregulation of hippocampal miR-132 and learning and memory impairment of rats. Brain Res. 1670:156–164. 2017. View Article : Google Scholar : PubMed/NCBI

26 

Tian H, Hou L, Xiong YM, Huang JX, Zhang WH, Pan YY and Song XR: miR-132 targeting E2F5 suppresses cell proliferation, invasion, migration in ovarian cancer cells. Am J Transl Res. 8:1492–1501. 2016.PubMed/NCBI

27 

Xu B, Zhang Y, Du XF, Li J, Zi HX, Bu JW, Yan Y, Han H and Du JL: Neurons secrete miR-132-containing exosomes to regulate brain vascular integrity. Cell Res. 27:882–897. 2017. View Article : Google Scholar : PubMed/NCBI

28 

Qin B, Liu J, Liu S, Li B and Ren J: MiR-20b targets AKT3 and modulates vascular endothelial growth factor-mediated changes in diabetic retinopathy. Acta Biochim Biophys Sin (Shanghai). 48:732–740. 2016. View Article : Google Scholar : PubMed/NCBI

29 

Yang J, Chen L, Ding J, Rong H, Dong W and Li X: High mobility group box-1 induces migration of vascular smooth muscle cells via TLR4-dependent PI3K/Akt pathway activation. Mol Biol Rep. 39:3361–3367. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Yuan Y, Shen Y, Xue L and Fan H: miR-140 suppresses tumor growth and metastasis of non-small cell lung cancer by targeting insulin-like growth factor 1 receptor. PLoS One. 8:e736042013. View Article : Google Scholar : PubMed/NCBI

31 

Zheng F, Liao YJ, Cai MY, Liu YH, Liu TH, Chen SP, Bian XW, Guan XY, Lin MC, Zeng YX, et al: The putative tumour suppressor microRNA-124 modulates hepatocellular carcinoma cell aggressiveness by repressing ROCK2 and EZH2. Gut. 61:278–289. 2012. View Article : Google Scholar : PubMed/NCBI

32 

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

33 

Torella D, Iaconetti C, Tarallo R, Marino F, Giurato G, Veneziano C, Aquila I, Scalise M, Mancuso T, Cianflone E, et al: MicroRNA regulation of the hyper-proliferative phenotype of vascular smooth muscle cells in diabetes mellitus. Diabetes. 67:2018. View Article : Google Scholar : PubMed/NCBI

34 

Choe N, Kwon JS, Kim JR, Eom GH, Kim Y, Nam KI, Ahn Y, Kee HJ and Kook H: The microRNA miR-132 targets Lrrfip1 to block vascular smooth muscle cell proliferation and neointimal hyperplasia. Atherosclerosis. 229:348–355. 2013. View Article : Google Scholar : PubMed/NCBI

35 

Ucar A, Gupta SK, Fiedler J, Erikci E, Kardasinski M, Batkai S, Dangwal S, Kumarswamy R, Bang C, Holzmann A, et al: The miRNA-212/132 family regulates both cardiac hypertrophy and cardiomyocyte autophagy. Nat Commun. 3:10782012. View Article : Google Scholar : PubMed/NCBI

36 

Rawal S, Munasinghe PE, Shindikar A, Paulin J, Cameron V, Manning P, Williams MJ, Jones GT, Bunton R, Galvin I and Katare R: Down-regulation of proangiogenic microRNA-126 and microRNA-132 are early modulators of diabetic cardiac microangiopathy. Cardiovasc Res. 113:90–101. 2017. View Article : Google Scholar : PubMed/NCBI

37 

Katare R, Riu F, Mitchell K, Gubernator M, Campagnolo P, Cui Y, Fortunato O, Avolio E, Cesselli D, Beltrami AP, et al: Transplantation of human pericyte progenitor cells improves the repair of infarcted heart through activation of an angiogenic program involving micro-RNA-132. Circ Res. 109:894–906. 2011. View Article : Google Scholar : PubMed/NCBI

38 

Bijkerk R, de Bruin RG, van Solingen C, van Gils JM, Duijs JM, van der Veer EP, Rabelink TJ, Humphreys BD and van Zonneveld AJ: Silencing of microRNA-132 reduces renal fibrosis by selectively inhibiting myofibroblast proliferation. Kidney Int. 89:1268–1280. 2016. View Article : Google Scholar : PubMed/NCBI

39 

Jin W, Reddy MA, Chen Z, Putta S, Lanting L, Kato M, Park JT, Chandra M, Wang C, Tangirala RK and Natarajan R: Small RNA sequencing reveals microRNAs that modulate angiotensin II effects in vascular smooth muscle cells. J Biol Chem. 287:15672–15683. 2012. View Article : Google Scholar : PubMed/NCBI

40 

Yerneni KK, Bai W, Khan BV, Medford RM and Natarajan R: Hyperglycemia-induced activation of nuclear transcription factor kappaB in vascular smooth muscle cells. Diabetes. 48:855–864. 1999. View Article : Google Scholar : PubMed/NCBI

41 

Chen M, Zhang Y, Li W and Yang J: MicroRNA-145 alleviates high glucose-induced proliferation and migration of vascular smooth muscle cells through targeting ROCK1. Biomed Pharmacother. 99:81–86. 2018. View Article : Google Scholar : PubMed/NCBI

42 

He M, Xue ZM, Li J and Zhou BQ: Breviscapine inhibits high glucose-induced proliferation and migration of cultured vascular smooth muscle cells of rats via suppressing the ERK1/2 MAPK signaling pathway. Acta Pharmacol Sin. 33:606–614. 2012. View Article : Google Scholar : PubMed/NCBI

43 

Anand S, Majeti BK, Acevedo LM, Murphy EA, Mukthavaram R, Scheppke L, Huang M, Shields DJ, Lindquist JN, Lapinski PE, et al: MicroRNA-132-mediated loss of p120RasGAP activates the endothelium to facilitate pathological angiogenesis. Nat Med. 16:909–914. 2010. View Article : Google Scholar : PubMed/NCBI

44 

Liu GF, Zhang SH, Li XF, Cao LY, Fu ZZ and Yu SN: Overexpression of microRNA-132 enhances the radiosensitivity of cervical cancer cells by down-regulating Bmi-1. Oncotarget. 8:80757–80769. 2017.PubMed/NCBI

45 

Chen HZ, Tsai SY and Leone G: Emerging roles of E2Fs in cancer: An exit from cell cycle control. Nat Rev Cancer. 9:785–797. 2009. View Article : Google Scholar : PubMed/NCBI

46 

Dimova DK and Dyson NJ: The E2F transcriptional network: Old acquaintances with new faces. Oncogene. 24:2810–2826. 2005. View Article : Google Scholar : PubMed/NCBI

47 

Zheng Y, Zhu C, Ma L, Shao P, Qin C, Li P, Cao Q, Ju X, Cheng G, Zhu Q, et al: miRNA-154-5p inhibits proliferation, migration and invasion by targeting E2F5 in prostate cancer cell lines. Urol Int. 98:102–110. 2017. View Article : Google Scholar : PubMed/NCBI

48 

Cai C, Huo Q, Wang X, Chen B and Yang Q: SNHG16 contributes to breast cancer cell migration by competitively binding miR-98 with E2F5. Biochem Biophys Res Commun. 485:272–278. 2017. View Article : Google Scholar : PubMed/NCBI

49 

Chen S, Ding Y, Tao W, Zhang W, Liang T and Liu C: Naringenin inhibits TNF-alpha induced VSMC proliferation and migration via induction of HO-1. Food Chem Toxicol. 50:3025–3031. 2012. View Article : Google Scholar : PubMed/NCBI

50 

Portaluppi F, Smolensky MH and Touitou Y: Ethics and methods for biological rhythm research on animals and human beings. Chronobiol Int. 27:1911–1929. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2019
Volume 20 Issue 2

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
Xu Q, Liang Y, Liu X, Zhang C, Liu X, Li H, Liang J, Yang G and Ge Z: miR‑132 inhibits high glucose‑induced vascular smooth muscle cell proliferation and migration by targeting E2F5. Mol Med Rep 20: 2012-2020, 2019
APA
Xu, Q., Liang, Y., Liu, X., Zhang, C., Liu, X., Li, H. ... Ge, Z. (2019). miR‑132 inhibits high glucose‑induced vascular smooth muscle cell proliferation and migration by targeting E2F5. Molecular Medicine Reports, 20, 2012-2020. https://doi.org/10.3892/mmr.2019.10380
MLA
Xu, Q., Liang, Y., Liu, X., Zhang, C., Liu, X., Li, H., Liang, J., Yang, G., Ge, Z."miR‑132 inhibits high glucose‑induced vascular smooth muscle cell proliferation and migration by targeting E2F5". Molecular Medicine Reports 20.2 (2019): 2012-2020.
Chicago
Xu, Q., Liang, Y., Liu, X., Zhang, C., Liu, X., Li, H., Liang, J., Yang, G., Ge, Z."miR‑132 inhibits high glucose‑induced vascular smooth muscle cell proliferation and migration by targeting E2F5". Molecular Medicine Reports 20, no. 2 (2019): 2012-2020. https://doi.org/10.3892/mmr.2019.10380