Open Access

YY1 affects the levels and function of fibulin‑5 in ox‑LDL‑treated vascular smooth muscle cells

  • Authors:
    • Leijing Wang
    • Cuiyan Li
    • Chunli Feng
    • Yuanbo Zang
  • View Affiliations

  • Published online on: April 21, 2022     https://doi.org/10.3892/etm.2022.11334
  • Article Number: 407
  • Copyright: © Wang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Fibulin‑5 is reportedly involved in the pathological process of atherosclerosis (AS) where low expression has been frequently observed in ruptured atherosclerotic plaques. The aim of the present study was to determine the effects of fibulin‑5 on the responses of vascular smooth muscle cells (VSMC) to oxidized low‑density lipoprotein (ox‑LDL). The expression of fibulin‑5 was studied in human aortic‑VSMCs (HA‑VSMCs) treated with ox‑LDL. Fibulin‑5 was first overexpressed by the transfection of Ov‑Fibulin‑5 plasmids in HA‑VSMCs challenged with ox‑LDL to investigate its influence on cell proliferation, migration and invasion using Cell Counting Kit‑8, wound healing and Transwell assays. Yin Yang‑1 (YY1) was bioinformatically predicted to bind to the promoter sites of fibulin‑5, which was subsequently confirmed by dual‑luciferase reporter gene assay. Fibulin‑5 overexpression was able to suppress cell proliferation, invasion and migration, which was effectively reversed by YY1 silencing by the transfection of siRNA‑Fibulin‑5 plasmids which could induced fibulin‑5 silencing. YY1 binding sites in the promoter region of fibulin‑5 were identified and confirmed in vitro by chromatin immunoprecipitation assay and dual‑luciferase reporter gene assay. The present results suggested that as a modulator of fibulin‑5, YY1 alleviated ox‑LDL‑induced proliferation, invasion, migration and phenotypic transition from differentiated contractile phenotype to dedifferentiated phenotype in VSMCs. However, the mechanism underlying the YY1‑mediated regulation of fibulin‑5 expression needs to be confirmed further in vivo. Nevertheless, targeting fibulin‑5 and YY1 could be further developed for AS therapy.

Introduction

Atherosclerosis (AS) is the common pathological cause of cardiovascular and cerebrovascular diseases (1). Activation and injury of vascular endothelial cells, sub-endothelial deposition of oxidized low-density lipoprotein (ox-LDL), formation of foam cells under the vascular intima and proliferation and migration of vascular smooth muscle cells (VSMCs) are the main events in the pathogenesis of AS (2). Aberrant proliferation and migration of VSMCs are common pathological features of AS, restenosis after stenting and hypertension (3,4).

Fibulin-5 is a glycoprotein that consists of 448 amino acids and is involved in regulating extracellular matrix (5). Recent data showed that fibulin-5 expression was decreased in atherosclerotic plaque tissues in human aneurysmatic aortas compared with healthy vessels (6). Another previous study suggested that fibulin-5 overexpression reduces MMP levels by inhibiting the Wnt/β-catenin signaling pathway (7). Yin Yang-1 (YY1) is a widely expressed transcription factor belonging to the Gli-Kruppel class of zinc finger proteins that can bind transcriptional initiation repeats to alter the activities of target promoters (8,9). Its overexpression can cause the abnormal transcription of downstream genes, participating in disease occurrence, such as diabetes and B lymphoma (8,10,11). YY1 was reported to serve an important role in AS by mediating the proliferation and migration of VSMCs (12). In addition, a previous study indicated that YY1 was involved in the formation of foam cells in response to oxLDL (13). The present study was therefore designed to investigate the potential interaction between YY1 and the promoter region of fibulin-5, and the role of fibulin-5 in ox-LDL-treated VSMCs.

Materials and methods

Collection of clinical samples

In the present study, five patients with coronary heart disease (CHD; male, n=2; female, n=3), with an average age of 65.1±2.4 years, were diagnosed by coronary angiography in The Laixi Municipal Hospital (Laixi, China) between June 2019 and August 2020. The control group was also composed of five individuals (male, n=2; female, n=3) with an average age of 62.3±3.1 years, who were present in the Laixi Municipal Hospital for routine physical examination. The inclusion criterion for coronary heart disease cases was the diameter stenosis of at least one major coronary artery of patients with stable angina (>80%). The exclusion criteria were as follows: i) Unstable angina pectoris or myocardial infarction; ii) comorbidity with other organic heart diseases, such as rheumatic heart disease and myocardiopathy; iii) comorbidity with severe liver and kidney disease; iv) comorbidity with familial hypercholesterolemia; v) comorbidity with malignancies; and vi) comorbidity with inflammatory diseases. A total of 3 ml venous blood was collected from each participant. Oral informed consent was obtained from all participants prior to enrollment in the present study. The present study was approved by the Ethics Committee of Laixi Municipal Hospital (approval no. ky-2020-041-03).

Human aortic (HA)-VSMC culture

HA-VSMCs were purchased from the American Type Culture Collection (cat. no. CRL-1999) and cultured in DMEM (Gibco; Thermo Fisher Scientific, Inc.) containing 10% FBS (Gibco; Thermo Fisher Scientific, Inc.) at 37˚C with 5% CO2. Different concentrations of ox-LDL (Yeasen Biotechnology (Shanghai) Co., Ltd., Shanghai, China) dissolved into DMEM medium (0, 50, 100 and 150 µg/ml) were added to induce cell calcification at 37˚C for 48 or 72 h.

Reverse transcription-quantitative PCR (RT-qPCR) assay

Total RNA from VSMCs was extracted with TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.) according to the instructions of the supplier. A total of 1 µg mRNA per sample was reverse transcribed into cDNA using PrimeScript™ RT Reagent kit (Takara Bio, Inc.) at 37˚C for 15 min and 85˚C for 5 sec. Subsequently, SYBR Green kit (cat. no. 208054; Qiagen GmbH) was used to prepare a 20 µl qPCR reaction mix and qPCR was performed on the StepOne Real Time PCR instrument (Thermo Fisher Scientific, Inc.). The primer sequences were: Fibulin-5 Forward: 5'-CTCACTGTTACCATTCTGGCTC-3', Reverse: 5'-GACTGGCGATCCAGGTCAAAG-3'. YY1 Forward: 5'-ACGGCTTCGAGGATCAGATTC-3', Reverse: 5'-TGACCAGCGTTTGTTCAATGT-3'. Smooth muscle α-actin (α-SMA) Forward: 5'-AAAAGACAGCTACGTGGGTGA-3', Reverse: 5'-GCCATGTTCTATCGGGTACTTC-3'. GAPDH: 5'-GGAGCGAGATCCCTCCAAAAT-3', Reverse: 5'-GGCTGTTGTCATACTTCTCATGG-3'. PCR was performed at 95˚C for 30 sec, followed by 45 cycles of 95˚C for 5 sec and 60˚C for 30 sec. GAPDH was used as an internal reference. The relative levels of mRNA were calculated with the 2-ΔΔCq method (14).

Cell Counting Kit-8 (CCK-8) assay

VSMCs (1x104 cells/well) were seeded into 96-well plates and then incubated for 24 h at 37˚C. Following ox-LDL treatment or transfection of Ov-Fibulin-5 plasmids and ox-LDL treatment, the medium was replaced with serum-free DMEM medium. A total of 10 µl CCK-8 solution (Beyotime Institute of Biotechnology) was added to each well for incubation at 37˚C for 1 h. The absorbance at 450 nm was subsequently detected using a microplate reader (Thermo Fisher Scientific, Inc.).

Western blotting

Following the x-LDL treatment or transfection of Ov-Fibulin-5 plasmids plus ox-LDL treatment, or the co-treatment of transfection of Ov-YY1 and siRNA-Fibulin-5 plus ox-LDL treatment, VSMCs were collected and lysed by adding RIPA buffer (Beyotime Institute of Biotechnology) on ice for 30 min. The supernatant was collected after centrifugation at 4˚C at 14,000 x g for 20 min. The protein concentration of each group was detected by a BCA kit (Abcam). A total of 20 µg each protein sample was separated by 10% SDS-PAGE and then transferred into PVDF membrane. The membranes were then blocked with 5% BSA (MilliporeSigma) at room temperature for 2 h and incubated with primary antibodies (Fibulin-5; cat. no. ab109428, 1:1,000; cyclin D1; cat. no. ab16663, 1:200; CDK2; cat. no. ab32147, 1:1,000; MMP2; cat. no. ab92536, 1:1,000; MMP9; cat. no. ab76003, 1:1,000; GAPDH; cat. no. ab8245, 1:5,000. Abcam) at 4˚C overnight. After incubation with a secondary antibody at 37˚C for 2 h (Goat Anti-Rabbit IgG; cat. no. ab7090, 1:10,000. Abcam, England), a ECL color development reagent (cat. no. ab133409; Abcam) was added to the membranes. The density of the protein bands was analyzed using the ImageJ software (version 1.53b; National Institutes of Health).

Wound healing assay

HA-VSMCs in a logarithmic proliferation phase were seeded into six-well plates at 2x104 cells per well. After routine overnight culture, the cells received transfection of Ov-Fibulin-5 plasmids and ox-LDL treatment, or the transfection of Ov-YY1 and siRNA- Fibulin-5 and ox-LDL treatment, with three replicates per group. When 80% cell confluence was reached, a scratch was performed using a horizontal line perpendicular to the back with a 20-µl pipette tip. The cells were washed by phosphoric acid buffer three times and cultured in serum-free DMEM medium. PBS was used to rinse the plates and serum-free DMEM medium was added at 37˚C. The cell migration was observed under a light microscope at 0 and 24 h (Olympus Corporation; magnification, x100). The relative migration was calculated using following formula: (Initial width at 0 h-final width at 24 h)/width at 0 h.

Transwell assay

The HA-VSMCs received transfection with Ov-Fibulin-5 plasmids and ox-LDL treatment, or transfection of Ov-YY1 and siRNA-Fibulin-5 and ox-LDL treatment and their concentration was adjusted to 2x108 cells/ml. Diluted Matrigel (25 µl; BD Biosciences) with DMEM medium without serum was added to upper chamber at 37˚C. A total of 200 µl cell suspension prepared with serum-free medium was added to the upper chambers of a Transwell insert (cat. no. 3422; 8-µm; Corning). DMEM with 10% FBS was added to the lower chamber. After 24 h of incubation at 37˚C, the cells that have gone through the membrane were fixed using 4% methanol and stained with crystal violet at room temperature for 10 min and images were captured under an inverted microscope (Olympus Corporation, magnification, x100).

Plasmid transfection

Lipofectamine® 3000 transfection reagent (Thermo Fisher Scientific, Inc.) was used according to the manufacturer's instructions to transfect YY1-overexpressing plasmids (0.5 µg; pcDNA-YY1; Ov-YY1; Shanghai GenePharma Co., Ltd.) or its control (0.5 µg; pcDNA), or siRNA targeting fibulin-5 (15 pmol; 5'-GAUUGUUGUGAGGAGUCUAGC-3') or its negative control (15 pmol; siRNA-NC, scrambled RNA, 5'-GUGAUUGCAUGAUUGUGCGGA-3') into HA-VSMCs, which were constructed and purchased from Shanghai GenePharma Co., Ltd. After 24 h at 37˚C, the cells were treated with 100 µg/ml ox-LDL (100 µg/ml ox-LDL significantly promoted cell proliferation) for 24 h at 37˚C for further experiments.

Bioinformatics

Using PROMO (http://alggen.lsi.upc.es/cgi-bin/promo_v3/promo/promoinit.cgi?dirDB=TF_8.3, access date: 10.09.2020) and Transcription Factor DataBase (TFDB; http://bioinfo.life.hust.edu.cn/HumanTFDB#!/, access date: 10.09.2020) tools, YY1 was predicted to bind to the promoter region of fibulin-5.

Chromatin immunoprecipitation (ChIP)

After chromatin was subjected to ultrasonication, magnetic beads (60 µl) coated with anti-YY1 or anti-IgG (EMD Millipore) were added to form immune complex. The immune complex was washed with wash buffer (low salt wash buffer, high salt wash buffer, LiCl wash buffer and TE buffer, in sequence) and then was centrifuged at 4˚C at 16,000 x g for 2 min. Next, collected immunoprecipitation was eluted with eluent including 10% SDS, NaHCO3 and ddH2O at 65˚C for 15 min. 20 µl NaCl was added for incubation at 65˚C overnight. After de-crosslink, 1 µl RNaseA was added for incubation for 1 h at 37˚C. RT-qPCR was performed to analyze precipitated DNA fragments.

Dual-luciferase reporter gene assay

Luciferase activity was detected after co-transfection of fibulin-5 promoter site 1 (TCCCAGCCCCCGCACC) with mutations or wild-type sequence luciferase reporter genes (0.1 µg; pGL3-basic; Promega Corporation) and YY1 overexpressing plasmids (0.2 µg)or its control plasmids (empty plasmids) into 293T cells (ATCC, USA) with Lipofectamine® 3000 following the manufacturer's protocol. Renilla luciferase was used as control reporter gene. After 48 h, luciferase activity was detected using Dual luciferase reporter gene assay kit according to the manufacturer's instructions (cat. no. RG028; Beyotime Institute of Biotechnology).

Statistical analysis

All data were statistically analyzed by GraphPad Prism 8 software (GraphPad Software, Inc.). The measurement data were represented by the mean ± SD and one-way ANOVA was used for comparison between multiple groups, followed by Tukey's test. Chi-square test was used to compare the two rates of age, smoking, drinking, or hypertension between CHD and control. Each experiment was repeated at least three times. P<0.05 was considered to indicate a statistically significant difference.

Results

Ox-LDL induces low fibulin-5 expression in HA-VSMCs

The level of high-density lipoprotein in the peripheral blood samples of patients with CHD was significantly lower than that in the control group (Table I). However, there was no statistical significance in the comparison of all other indicators between the two groups (Table I).

Table I

Clinical characteristics of patients with CHD.

Table I

Clinical characteristics of patients with CHD.

CharacteristicsCHD (n=5)Control (n=5)P-value
Age, years65.1±2.462.3±3.1>0.05
Smoking, %2020>0.05
Drinking, %2020>0.05
Hypertension, %6060>0.05
Cholesterol, mmol/l3.98±0.664.01±0.42>0.05
High-density lipoprotein, mmol/l0.94±0.121.35±0.29<0.01
Low density lipoprotein, mmol/l2.12±0.632.15±0.64>0.05

[i] CHD, coronary heart disease.

To explore the role of fibulin-5 in AS, fibulin-5 mRNA expression was quantified in the peripheral blood of patients with AS. Fibulin-5 mRNA expression was significantly increased in patients with AS compared with that in healthy individuals (P<0.001; Fig. 1). VSMCs have been previously reported to serve an important role in atherogenesis (15). As indicated in Fig. 2A, 100 µg/ml ox-LDL was the concentration that increased VSMC viability the strongest (P<0.001). This concentration was therefore selected for further experiments. RT-qPCR and western blotting analyses of fibulin-5 expression in VSMCs indicated that the mRNA and protein levels of fibulin-5 were significantly reduced after ox-LDL treatment compared with those in the control group (P<0.001; Fig. 2B and C). To further investigate the role of fibulin-5, HA-VSMCs treated with ox-LDL were transfected with a fibulin-5-overexpressing plasmid.

Fibulin-5 overexpression suppresses HA-VSMC proliferation in response to ox-LDL

Since ox-LDL decreased fibulin-5 expression, it was hypothesized that fibulin-5 serve a role in HA-VSMCs in response to ox-LDL. As a transfection control, a vector overexpressing (Ov)-fibulin-5 significantly increased the mRNA and protein expression levels of fibulin-5 compared with that in the Ov-negative control (NC) group (P<0.001; Fig. 3A and B). Analyzes of fibulin-5 expression in ox-LDL-treated HA-VSMCs demonstrated that the reduced fibulin-5 expression was significantly reversed by fibulin-5 overexpression (P<0.001; Fig. 3C and D). In VSMCs cycle progression, cyclin D1 and CDK2 are involved in promoting Rb phosphorylation, the inhibition of which can lead to cycle arrest (16). VSMCs were subsequently subjected to CCK-8 analysis to measure cell viability (P<0.001; Fig. 3E) whereas western blotting was used to quantify protein levels of cell cycle proteins cyclin D1 and CDK2 (P<0.001; Fig. 3F). The present results demonstrated that the overexpression of fibulin-5 in HA-VSMCs reversed the increase in cell viability induced by ox-LDL treatment, in addition to reversing the increased protein expression of cyclin D1 and CDK2 induced by ox-LDL treatment (P<0.001; Fig. 3E and F).

Cell migration and invasion were studied by performing wound healing and Transwell assays, respectively. The present results demonstrated that HA-VSMCs exhibited significantly increased migration and invasion after ox-LDL treatment, which were significantly reversed following fibulin-5 overexpression (all P<0.001; Fig. 4A-D). MMP-2 and MMP-9 expressed in VSMCs, modulated the collagen degradation of the extracellular matrix and served a vital role in AS progression (17).

Additionally, the protein expression of MMP2 and MMP9, which participate in phenotype transition (18,19), was quantified by western blotting, whilst the mRNA and protein expression of α-SMA was measured by RT-qPCR and western blotting, respectively. MMP2 and MMP9 protein expression was significantly increased after ox-LDL treatment, an effect that was significantly reversed by fibulin-5 overexpression (all P<0.001; Fig. 4E). Similarly, α-SMA mRNA and protein expression were significantly decreased after ox-LDL treatment (P<0.001), which was significantly reversed by fibulin-5 overexpression (P<0.01 and P<0.001; Fig. 4E-G).

YY1 binds to the promoter of fibulin-5 to induce its upregulation in HA-VSMCs

To further explore the role of fibulin-5 in AS, PROMO and TFDB databases were used to predict whether YY1 can bind to the promoter region of fibulin-5 to regulate fibulin-5 expression in ox-LDL-treated HA-VSMCs. The mRNA and protein expression levels of YY1 were significantly decreased in ox-LDL-treated HA-VSMCs compared with those in the control group (P<0.01 and P<0.001; Fig. 5A and B). Next, the two promoter sites and the overall promoter region level of fibulin-5, were detected by qPCR in an immunoprecipitation complex formed by using YY1 antibody. Promoter site 1 was shown to be markedly enriched (Fig. 5C). The luciferase activity of the pGL4 luciferase reporter vectors containing the promoter region of fibulin-5 with binding site (site 1) for YY1 was significantly increased when YY1-overexpressing plasmids were co-transfected into HA-VSMCs, compared with that in cells co-transfected with the Ov-NC plasmid (P<0.001, Fig. 5D). When the pGL4 luciferase reporter vectors with muted sites and Ov-NC or Ov-YY1 were co-transfected into 293T cells, there was no significant difference in luciferase activities between Ov-NC and Ov-YY1 (Fig. 5D). These results suggest that YY1 can bind to site 1 of the fibulin-5 promoter. The transfection efficiency of the YY1-overexpressing plasmid was tested in Fig. 5E and F. As shown in Fig. 5E and F, cells transfected with Ov-YY1 showed significantly higher YY1 expression levels compared with those in cells transfected with Ov-NC (P<0.01 and P<0.001). As indicated in Fig. 5G and H, overexpression of YY1 in the HA-VSMCs significantly reversed the decreased mRNA and protein expression of YY1 induced by ox-LDL treatment (P<0.001). Similarly, YY1 overexpression also significantly restored the expression of fibulin-5, which was decreased after ox-LDL treatment (P<0.01 and P<0.001; Fig. 5I and J).

YY1 mediates the effects of fibulin-5 on proliferation, migration and invasion in HA-VSMCs stimulated with ox-LDL

The present study next aimed to determine whether fibulin-5 mediated the effects of YY1 in cultured HA-VSMCs stimulated with ox-LDL. First, the transfection efficiency of small interfering (si)RNA-fibulin-5 was tested in the HA-VSMCs. Analyzes of fibulin-5 expression in HA-VSMCs demonstrated that it was effectively knocked down by siRNA-Fibulin-5 transfection compared with cell transfected with siRNA-NC (P<0.001; Fig. 6A and B). Since it was the most efficient in knocking down fibuin-5 expression, si-fibulin-1 was selected for further experiments. Based on the present results, it was hypothesized that YY1 interacted with the promoter sites of fibulin-5 to regulate cell proliferation, migration and invasion in VSMCs treated with ox-LDL. First, cell viability was compared between VSMCs transfected with siRNA-NC and with siRNA-fibulin-5. Fibulin-5 silencing induced a significant reversal of the potentiating effects due to YY1 overexpression on cell viability, cyclin D1 and CDK2 expression (P<0.01 and P<0.001; Fig. 6C-E). Significant differences were also observed in the cell migration and invasion of HA-VSMCs, and the expression of MMP2, MMP9 and α-SMA between Ov-YY1 vs. Ov-NC and between Ov-YY1+siRNA-NC and with Ov-YY1+siRNA-fibulin-5 (P<0.05, P<0.01 and P<0.001; Fig. 7A-G). This suggests that YY1 participated in mediating the effects of fibulin-5 on cell viability, migration and invasion induced by ox-LDL.

Discussion

The present study demonstrated that fibulin-5 participated in ox-LDL-induced cell viability, invasion and migration in VSMCs. A previous study on the fibulin-5 mechanism revealed that fibulin-5 was involved in arterial stiffening in patients with AS (20). The present study indicated that YY1 regulated the promoter activity and expression levels of fibulin-5 in VSMCs under ox-LDL treatment, which provided a new insight in the understanding of the role of fibulin-5 in AS.

The role of fibulin-5 in AS was found to involve the binding of extracellular superoxide dismutase to the vascular tissue to modulate vascular superoxide production, which was identified to serve a vital role in AS (21). As for the regulatory mechanism of fibulin-5 promoter activity, a previous report indicated that SOX9 could regulate the fibulin-5 promoter activity in VSMCs under the stimulus of TNF-α (22). In the present study, a YY1 binding sequence on the fibulin-5 promoter was identified and predicted, where it was verified further that YY1 could directly bind to the fibulin-5 promoter and regulate its expression. This translated downstream to the regulation of the biological processes of VSMCs in response to ox-LDL. However, the molecular mechanism by which YY1 binding activates fibulin-5 transcription was not uncovered in the present study, which was predicted to be associated with YY1 involvement in transcriptional activation by forming complexes with some proteins, such as INO80 or Sp1(23) in the promoter of fibulin-5. YY1 has been reported to be a transcriptional activator of some genes, such as Xist in ES cells and CDC6 in HEK293 cells, (24,25). For instance, YY1 initiated the activity of the X-inactive specific transcript (Xist) promoter by directly binding to the Xist 5' region in ES cells (24). Downstream, YY1 was also revealed to be involved in the pathophysiology of AS by regulating cell proliferation and apoptosis (12,26,27). For example, a previous study reported that YY1 was involved in the promoting effects of miR-544 on the maturation and antioxidative effects of stem cell-derived endothelial-like cells (28). A previous mechanistic analysis of YY1 in ox-LDL-treated macrophages of AS demonstrated that YY1 can translocate into the nucleus and increase the expression of miR-29a by binding to its transcriptional promoter region (27).

In addition to its regulatory role in cell viability, invasion and migration, fibulin-5 overexpression markedly reduced the expression levels of cyclin D1 and CDK2, which regulate G1-phase cell cycle progression and subsequently proliferation in VSMCs (29,30). A previous study found that the expression levels of YY1, which is a negative regulator of cyclin D1, could be regulated by miR-29a (31). The present results suggest that cyclin D1 and CDK2 can participate in the regulation of YY1/fibulin-5 during cell proliferation. Although the present study revealed that fibulin-5/YY1 affected the viability, invasion and migration of HA-VSMCs stimulated with ox-LDL, whether this axis is involved in the pathogenesis of AS requires further study, which is a limitation of the present study.

In conclusion, the present study presented evidence that fibulin-5, regulated by YY1, can suppress the viability, invasion and migration of HA-VSMCs stimulated with ox-LD. The present results provided a new insight for the further investigation of the mechanism of AS. Targeting fibulin-5/YY1 can be a potential therapeutic option for AS. However, further studies should be performed, mainly on the molecular mechanism by which the binding of YY1 can activate fibulin-5 transcription. In addition, the mechanism of YY1/fibulin-5 would need to be validated in vivo.

Acknowledgements

Not applicable.

Funding

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

LW and YZ made substantial contributions to the conception and design of the study. LW collected clinical samples and interpreted the patient data regarding fibulin-5 expression. LW, CL, CF and YZ performed other experiments and collected data. LW and YZ interpreted the data, drafted and revised the manuscript for important intellectual content. LW and YZ confirm the authenticity of the raw data. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The present study was approved by the Ethics Committee of Laixi Municipal Hospital (approval no. ky-2020-041-03). Oral informed consent was obtained from all participants prior to enrollment in the present study.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Cao Q, Guo Z, Du S, Ling H and Song C: Circular RNAs in the pathogenesis of atherosclerosis. Life Sci. 255(117837)2020.PubMed/NCBI View Article : Google Scholar

2 

Kattoor AJ, Goel A and Mehta JL: LOX-1: Regulation, signaling and its role in atherosclerosis. Antioxidants (Basel). 8(218)2019.PubMed/NCBI View Article : Google Scholar

3 

Cheng YC, Sheen JM, Hu WL and Hung YC: Polyphenols and oxidative stress in atherosclerosis-related ischemic heart disease and stroke. Oxid Med Cell Longev. 2017(8526438)2017.PubMed/NCBI View Article : Google Scholar

4 

Lu QB, Wan MY, Wang PY, Zhang CX, Xu DY, Liao X and Sun HJ: Chicoric acid prevents PDGF-BB-induced VSMC dedifferentiation, proliferation and migration by suppressing ROS/NF-κB/mTOR/P70S6K signaling cascade. Redox Biol. 14:656–668. 2018.PubMed/NCBI View Article : Google Scholar

5 

Ito S, Yokoyama U, Nakakoji T, Cooley MA, Sasaki T, Hatano S, Kato Y, Saito J, Nicho N, Iwasaki S, et al: Fibulin-1 integrates subendothelial extracellular matrices and contributes to anatomical closure of the ductus arteriosus. Arterioscler Thromb Vasc Biol. 40:2212–2226. 2020.PubMed/NCBI View Article : Google Scholar

6 

Xu BF, Liu R, Huang CX, He BS, Li GY, Sun HS, Feng ZP and Bao MH: Identification of key genes in ruptured atherosclerotic plaques by weighted gene correlation network analysis. Sci Rep. 10(10847)2020.PubMed/NCBI View Article : Google Scholar

7 

Gao JB, Lin L, Men XQ, Zhao JB, Zhang MH, Jin LP, Gao SJ, Zhao SQ and Dong JT: Fibulin-5 protects the extracellular matrix of chondrocytes by inhibiting the Wnt/β-catenin signaling pathway and relieves osteoarthritis. Eur Rev Med Pharmacol Sci. 24:5249–5258. 2020.PubMed/NCBI View Article : Google Scholar

8 

Meliala ITS, Hosea R, Kasim V and Wu S: The biological implications of Yin Yang 1 in the hallmarks of cancer. Theranostics. 10:4183–4200. 2020.PubMed/NCBI View Article : Google Scholar

9 

Liu H, Schmidt-Supprian M and Shi Y, Hobeika E, Barteneva N, Jumaa H, Pelanda R, Reth M, Skok J, Rajewsky K and Shi Y: Yin Yang 1 is a critical regulator of B-cell development. Genes Dev. 21:1179–1189. 2007.PubMed/NCBI View Article : Google Scholar

10 

Blättler SM, Cunningham JT, Verdeguer F, Chim H, Haas W, Liu H, Romanino K, Rüegg MA, Gygi SP, Shi Y and Puigserver P: Yin Yang 1 deficiency in skeletal muscle protects against rapamycin-induced diabetic-like symptoms through activation of insulin/IGF signaling. Cell Metab. 15:505–517. 2012.PubMed/NCBI View Article : Google Scholar

11 

Sui G, Affar el B and Shi Y, Brignone C, Wall NR, Yin P, Donohoe M, Luke MP, Calvo D, Grossman SR and Shi Y: Yin Yang 1 is a negative regulator of p53. Cell. 117:859–872. 2004.PubMed/NCBI View Article : Google Scholar

12 

Yue Y, Lv W, Zhang L and Kang W: miR-147b influences vascular smooth muscle cell proliferation and migration via targeting YY1 and modulating Wnt/β-catenin activities. Acta Biochim Biophys Sin (Shanghai). 50:905–913. 2018.PubMed/NCBI View Article : Google Scholar

13 

Tian FJ, An LN, Wang GK, Zhu JQ, Li Q, Zhang YY, Zeng A, Zou J, Zhu RF, Han XS, et al: Elevated microRNA-155 promotes foam cell formation by targeting HBP1 in atherogenesis. Cardiovasc Res. 103:100–110. 2014.PubMed/NCBI View Article : Google Scholar

14 

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.PubMed/NCBI View Article : Google Scholar

15 

Grootaert MOJ, Moulis M, Roth L, Martinet W, Vindis C, Bennett MR and De Meyer GRY: Vascular smooth muscle cell death, autophagy and senescence in atherosclerosis. Cardiovasc Res. 114:622–634. 2018.PubMed/NCBI View Article : Google Scholar

16 

Kavurma MM and Khachigian LM: Sp1 inhibits proliferation and induces apoptosis in vascular smooth muscle cells by repressing p21WAF1/Cip1 transcription and cyclin D1-Cdk4-p21WAF1/Cip1 complex formation. J Biol Chem. 278:32537–32543. 2003.PubMed/NCBI View Article : Google Scholar

17 

Nasiri-Ansari Ν, Dimitriadis GK, Agrogiannis G, Perrea D, Kostakis ID, Kaltsas G, Papavassiliou AG, Randeva HS and Kassi E: Canagliflozin attenuates the progression of atherosclerosis and inflammation process in APOE knockout mice. Cardiovasc Diabetol. 17(106)2018.PubMed/NCBI View Article : Google Scholar

18 

Terzuoli E, Nannelli G, Giachetti A, Morbidelli L, Ziche M and Donnini S: Targeting endothelial-to-mesenchymal transition: The protective role of hydroxytyrosol sulfate metabolite. Eur J Nutr. 59:517–527. 2020.PubMed/NCBI View Article : Google Scholar

19 

Viedma-Rodríguez R, Martínez-Hernández MG, Martínez-Torres DI and Baiza-Gutman LA: Epithelial mesenchymal transition and progression of breast cancer promoted by diabetes mellitus in mice are associated with increased expression of glycolytic and proteolytic enzymes. Horm Cancer. 11:170–181. 2020.PubMed/NCBI View Article : Google Scholar

20 

Paapstel K, Zilmer M, Eha J, Tootsi K, Piir A and Kals J: Association between Fibulin-1 and aortic augmentation index in male patients with peripheral arterial disease. Eur J Vasc Endovasc Surg. 51:76–82. 2016.PubMed/NCBI View Article : Google Scholar

21 

Nguyen AD, Itoh S, Jeney V, Yanagisawa H, Fujimoto M, Ushio-Fukai M and Fukai T: Fibulin-5 is a novel binding protein for extracellular superoxide dismutase. Circ Res. 95:1067–1074. 2004.PubMed/NCBI View Article : Google Scholar

22 

Orriols M, Varona S, Martí-Pàmies I, Galán M, Guadall A, Escudero JR, Martín-Ventura JL, Camacho M, Vila L, Martínez-González J and Rodríguez C: Down-regulation of Fibulin-5 is associated with aortic dilation: role of inflammation and epigenetics. Cardiovasc Res. 110:431–442. 2016.PubMed/NCBI View Article : Google Scholar

23 

Sarvagalla S, Kolapalli SP and Vallabhapurapu S: The two sides of YY1 in cancer: A friend and a foe. Front Oncol. 9(1230)2019.PubMed/NCBI View Article : Google Scholar

24 

Makhlouf M, Ouimette JF, Oldfield A, Navarro P, Neuillet D and Rougeulle C: A prominent and conserved role for YY1 in Xist transcriptional activation. Nat Commun. 5(4878)2014.PubMed/NCBI View Article : Google Scholar

25 

Cai Y, Jin J, Yao T, Gottschalk AJ, Swanson SK, Wu S, Shi Y, Washburn MP, Florens L, Conaway RC and Conaway JW: YY1 functions with INO80 to activate transcription. Nat Struct Mol Biol. 14:872–874. 2007.PubMed/NCBI View Article : Google Scholar

26 

Wei SY, Shih YT, Wu HY, Wang WL, Lee PL, Lee CI, Lin CY, Chen YJ, Chien S and Chiu JJ: Endothelial Yin Yang 1 phosphorylation at S118 induces atherosclerosis under flow. Circ Res. 129:1158–1174. 2021.PubMed/NCBI View Article : Google Scholar

27 

Jian D, Dai B, Hu X, Yao Q, Zheng C and Zhu J: ox-LDL increases microRNA-29a transcription through upregulating YY1 and STAT1 in macrophages. Cell Biol Int. 41:1001–1011. 2017.PubMed/NCBI View Article : Google Scholar

28 

Guo J, Xiang Q, Xin Y, Huang Y, Zou G and Liu T: miR-544 promotes maturity and antioxidation of stem cell-derived endothelial like cells by regulating the YY1/TET2 signalling axis. Cell Commun Signal. 18(35)2020.PubMed/NCBI View Article : Google Scholar

29 

Qin L, Yang YB, Yang YX, Gong YZ, Li XL, Li GY, Luo HD, Xie XJ, Zheng XL and Liao DF: Inhibition of smooth muscle cell proliferation by ezetimibe via the cyclin D1-MAPK pathway. J Pharmacol Sci. 125:283–291. 2014.PubMed/NCBI View Article : Google Scholar

30 

Vadiveloo PK, Filonzi EL, Stanton HR and Hamilton JA: G1 phase arrest of human smooth muscle cells by heparin, IL-4 and cAMP is linked to repression of cyclin D1 and cdk2. Atherosclerosis. 133:61–69. 1997.PubMed/NCBI View Article : Google Scholar

31 

Zhang Y, Li YH, Liu C, Nie CJ, Zhang XH, Zheng CY, Jiang W, Yin WN, Ren MH, Jin YX, Liu SF, et al: miR-29a regulates vascular neointimal hyperplasia by targeting YY1. Cell Prolif. 50(e12322)2017.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

June-2022
Volume 23 Issue 6

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang L, Li C, Feng C and Zang Y: YY1 affects the levels and function of fibulin‑5 in ox‑LDL‑treated vascular smooth muscle cells. Exp Ther Med 23: 407, 2022
APA
Wang, L., Li, C., Feng, C., & Zang, Y. (2022). YY1 affects the levels and function of fibulin‑5 in ox‑LDL‑treated vascular smooth muscle cells. Experimental and Therapeutic Medicine, 23, 407. https://doi.org/10.3892/etm.2022.11334
MLA
Wang, L., Li, C., Feng, C., Zang, Y."YY1 affects the levels and function of fibulin‑5 in ox‑LDL‑treated vascular smooth muscle cells". Experimental and Therapeutic Medicine 23.6 (2022): 407.
Chicago
Wang, L., Li, C., Feng, C., Zang, Y."YY1 affects the levels and function of fibulin‑5 in ox‑LDL‑treated vascular smooth muscle cells". Experimental and Therapeutic Medicine 23, no. 6 (2022): 407. https://doi.org/10.3892/etm.2022.11334