Atorvastatin regulates vascular smooth muscle cell phenotypic transformation by epigenetically modulating contractile proteins and mediating Akt/FOXO4 axis

  • Authors:
    • Xinping Du
    • Zhiyuan Sun
    • Zhongnan Cao
    • Xiuhong Zhou
    • Dong Wang
    • Kuan Wang
    • Xuebin Li
    • Guoxing Zuo
  • View Affiliations

  • Published online on: March 16, 2022     https://doi.org/10.3892/mmr.2022.12683
  • Article Number: 167
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Atherosclerosis (AS) is a prevalent cardiovascular disease with severe morbidity and high mortality. Phenotypic regulation of vascular smooth muscle cells (VSMCs) from the contractile and quiescent phenotype to the synthetic type is a critical step for the vascular remodeling of AS. Atorvastatin, as a 3‑hydroxy‑3‑methyl‑glutaryl coenzyme A reductase inhibitor, presents an anti‑inflammatory effect to improve vascular endothelial functions. The aim of the present study was to examine the effect of atorvastatin on VSMCs phenotypic transformation and the underlying mechanism. The rat primary VSMCs were isolated and identified. The protein expression of contractile proteins, such as α‑SMA, SM‑MHC, and SM22α, was reduced by angiotensin II (AngII) and enhanced by atorvastatin, in which atorvastatin could reverse the effect of AngII in the VSMCs. The treatment of HDAC inhibitor trichostatin A was able to enhance AngII‑inhibited expression of α‑SMA and SM‑MHC. Atorvastatin regulated AngII‑associated VSMCs phenotypic transformation by epigenetically regulating contractile proteins. Moreover, atorvastatin modulated platelet‑derived growth factor‑BB (PDGF‑BB)‑induced VSMC phenotypic transformation by modulating the Akt/forkhead Box O4 (FOXO4) axis. Immunofluorescence analysis revealed that PDGF‑BB enhanced the accumulation of FOXO4 in the VSMCs, while the treatment of atorvastatin was able to attenuate this effect and the co‑treatment of Akt inhibitor LY294002 could further inhibit the phenotype. The treatment of PDGF‑BB enhanced the interaction of SRF with FOXO4 and myocardin in the VSMCs, in which the co‑treatment of atorvastatin and LY294002 could reverse the effect of PDGF‑BB in the system. Thus, atorvastatin regulates VSMCs phenotypic transformation by epigenetically modulating contractile proteins and mediating the Akt/FOXO4 axis. Findings of the present study provide new insights into the mechanism by which atorvastatin modulates VSMCs, providing valuable evidence for the application of atorvastatin in the treatment of AS.

Introduction

Cardiovascular disease may lead to death and affects approximately 17.9 million individuals annually (13). Atherosclerosis (AS) is a prevalent cardiovascular disease and a leading cause of vascular-related death (4,5). The critical function of vascular smooth muscle cells (VSMCs) phenotypic transformation during AS progression has been previously identified (6,7). VSMC phenotypic transformation from a contractile to a synthetic status in vessel walls, stimulated by adverse microenvironmental provocations, accompanied by VSMC proliferation and migration, is crucial to AS development (8,9). Phenotypic regulation of VSMCs from the contractile and quiescent phenotype to the synthetic type is a critical step for the vascular remodeling of AS (10). In addition, it has been identified that VSMCs also experience notable phenotypic transformation stimulated by some signals, such as platelet-derived growth factor-BB (PDGF-BB) and angiotensin II (AngII) (11). Accordingly, the exploration of the potential candidate that is able to regulate VSMCs phenotypic transformation may benefit the treatment of AS.

Atorvastatin is a 3-hydroxy-3-methyl-glutaryl coenzyme A (HMG-CoA) reductase inhibitor and is commonly applied for the treatment of AS patients (12). Atorvastatin can enhance plaque instability and relieve the coronary artery inflammation response, decreasing the morbidity and mortality incidences of cardiovascular disorder (13). Besides, atorvastatin has several other functions, such as vascular endothelial function improvement, anti-oxidation, and anti-inflammation (14). Moreover, it has been reported that histone deacetylases (HDACs) are involved in the modulation of VSMCs (15). In addition, forkhead box O4 (FOXO4) and Akt signaling play a critical role in VSMCs phenotypic transformation during AS (16,17). In addition, FOXO4 activity is regulated by the PI3K/Akt signaling-mediated phosphorylation (18). Moreover, it has been reported that serum response factor (SRF)/myocardin signaling participates in the regulation of VSMCs shiftiness (19). However, the effect of atorvastatin on these factors in the development of VSMCs phenotypic transformation remains obscure.

In this study, the role and underlying mechanism of atorvastatin in the modulation of VSMCs phenotypic transformation was investigated. A novel function of atorvastatin in regulating VSMCs phenotypic transformation by epigenetically modulating contractile proteins and mediating Akt/FOXO4 axis.

Materials and methods

Cell culture and treatment

Primary VSMCs were isolated from normal rat aortas (N=5, male, 8 weeks) by applying the explant method as previously reported (20). Briefly, the aortas were isolated and the endothelial cells (ECs) were removed, the smooth muscle layer was stripped and chopped into small fragments (~1 mm3) in 0.5 ml of fetal bovine serum. The fragments, together with the fetal bovine serum, were transferred to a 25 cm2 flask and maintained upside down in an incubator at 37°C for 4 h. The flask was turned over gently and incubated for 4–7 days after the addition of 2 ml of DMEM. VSMCs migrated out of the explants 4–7 days later, and passage was performed 10–14 days after isolation. The VSMCs were cultured in the medium of DMEM (Gibco; Thermo Fisher Scientific, Inc.) containing 10% fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc.), 0.1 mg/ml streptomycin (Beijing Solarbio Science & Technology Co., Ltd.) and 100 U/ml penicillin (Beijing Solarbio Science & Technology Co., Ltd.) at a condition of 37°C with 5% CO2. The primary VSMCs were identified using α-SMA. For all the analyses, VSMCs (2–5 generations) were applied followed by quiescence for 12 h. The VSMCs were treated with AngII (10 nM; MilliporeSigma), trichostatin A (TSA, 0.01 ng/ml; MilliporeSigma), atorvastatin (30 µmol/l; MilliporeSigma), mevalonic acid (MVA, 500 µM; MilliporeSigma), PDGF-BB (20 ng/ml; MilliporeSigma), and LY294002 (10 µM, Sigma, USA) as indicated. The morphology of VSMCs was analyzed using a Nikon microscope (Tokyo).

The average weight of the rats at the start of the experiment was 187 g. The rats were fed in a condition of 25°C and 50% relative humidity with a 12-h light/dark cycle and free access to standard chow and tap water. The rats were euthanized with an overdose of pentobarbital sodium (150 mg/kg, iv). Animal care and method procedure were authorized by the Animal Ethics Committee of Tianjin Fifth Central Hospital (approval no. 2019-0619-37). The procedures were conformed to the Guide for the Care and Use of Laboratory Animal published by the US National Institutes of Health (NIH publication, 8th edition, 2011).

Reverse transcription-quantitative PCR (RT-qPCR)

The total RNAs from the mice and cells were extracted by TRIzol® (Invitrogen; Thermo Fisher Scientific, Inc.) from the VSMCs. The first-strand cDNA was manufactured as per the manufacturer's instructions (Thermo Fisher Scientific, Inc.). The RT-qPCR was carried out by applying SYBR Real-time PCR I kit (Takara Bio, Inc.). The standard control for mRNA was GAPDH. Quantitative determination of the RNA levels was conducted in triplicate independent experiments (n=3). The following thermocycling conditions were used for qPCR: Initial denaturation at 95°C for 30 sec; followed by 39 cycles of 95°C for 5 sec and 60°C for 30 sec; and a final extension at 72°C for 5 min. The 2−ΔΔCq method was used (21). The sequences were designed according to the references (Genscript, Nanjing, China). Primer sequences are listed in Table I.

Table I.

Primer sequences.

Table I.

Primer sequences.

Gene namePrimerGenebank accession no.Product sizes (bp)
α-SMA 5′-GAAGAGCTACGAACTGCCTGATG-3′NM_007392.3417
5′-TAGAAGCATTTGCGGTGGAC-3′
SM-MHC 5′-TTTGCCATTGAGGCCTTAGG-3′NG_009299427
5′-GTTCACACGGCTGAGAATCCA-3′
SM22α 5′-TTGTAATGCAGTGTGGCCCT-3′NM_003186369
5′-CAGGCTGTTCACCAACTTG-3′
GAPDH 5′-AAGAAGGTGGTGAAGCAGGC-3′XM_036165840.1203
5′-TCCACCACCCAGTTGCTGTA-3′
Calponin 5′-GTCTGGGCATGGAACACTGT-3′NM_031747.2477
5′-GAGGTACTTACTTGTGAGGGAAT-3′
Western blot analysis

Total proteins were extracted from the cells or mice tissues with RIPA buffer (Cell Signaling Technology, Inc.). Protein concentrations were measured by using the BCA Protein Quantification Kit. The same concentration of protein (25 µg) was divided by SDS-PAGE (12% polyacrylamide gels), and transferred to PVDF membranes (MilliporeSigma) in the subsequent step. The membranes were blocked with 5% milk and incubated overnight at 4°C with the primary antibodies for α-SMA (1:1,000; cat. no. ab7817; Abcam), SM-MHC (1:1,000; cat. no. ab133567; Abcam), SM22α (1:1,000; cat. no. ab14106; Abcam), FOXO4 (1:1,000; cat. no. ab128908; Abcam), p-FOXO4 (1:1,000; cat. no. ab126594; Abcam), Akt (1:1,000; cat. no. ab8805; Abcam), p-Akt (1:1,000; cat. no. ab38449; Abcam), SRF (1:1,000; cat. no. ab252868; Abcam), myocardin (1:1,000; cat. no. ab107301; Abcam), and β-actin (1:1,000; cat. no. ab7817; Abcam), in which β-actin served as the control. Then, the corresponding secondary antibodies (1:1,000; cat. no. ab96899/ab96879; Abcam) were used for incubating the membranes 1 h at room temperature, followed by the visualization by using an Odyssey CLx Infrared Imaging System. The experiments were conducted in triplicate independent experiments (n=3). The densitometry analysis was performed using ImageJ software (NIH, v1.8.0).

Immunofluorescence analysis

Cells were solidified at 4% paraformaldehyde for 30 min, treated with Triton X-100 (0.2%) for 10 min and treated with BSA (2%) for 30 min. The slides were hatched with the primary antibody overnight at 4°C, then hatched with secondary antibodies (Proteintech) for 1 h at 37°C. The slides were stained with DAPI (Beyotime Institute of Biotechnology) for 10 min at 25°C. A Nikon microscope (Tokyo) was utilized to analyze the immunofluorescence. The experiments were conducted in triplicate independent experiments (n=3).

ChIP analysis

Chromatin immunoprecipitation (ChIP) was performed using a SimpleChIP Enzymatic Chromatin IP Kit (Cell Signaling Technology, Inc.) according to the manufacturer's instructions. Chromatin prepared from the cells in a 15-cm dish was used to determine total DNA input and was incubated overnight with specific antibodies, H3K9ac (1:500; cat. no. ab32129; Abcam), H4Ac (1:500, 39243, Thermo Fisher, Cambridge, MA, USA), HDAC1 (1:500; cat. no. ab109411; Abcam), HDAC3 (1:500; cat. no. ab32369; Abcam), P300 (1:500; cat. no. ab275378; Abcam), or normal rabbit IgG. Then, the binding DNA was analyzed by qPCR assays and the primer sequences as indicated in Table I. The experiments were conducted as triplicate independent experiments (n=3).

HDAC activity analysis

The activities of HDAC were analyzed by applying an HDAC Activity Colorimetric Assay Kit (Biovision, Inc.) in the VSMCs according to the manufacturer's protocol. The experiments were conducted as triplicate independent experiments (n=3).

Immunoprecipitation assays

The interaction of the proteins was analyzed by immunoprecipitation in the VSMCs. IP was conducted by Pierce Co-Immunoprecipitation kit (Thermo Fisher Scientific, Inc., Germany) according to the manufacturer's instructions. The experiments were conducted in triplicate independent experiments (n=3).

Statistical analysis

Data are presented as mean ± SD, and the statistical analysis was performed by GraphPad prism 7 (GraphPad Software, Inc.). The unpaired Student's t-test was applied for comparing two groups, and the one-way analysis of variance (ANOVA) followed by a post hoc Tukey's test was applied for comparing among multiple groups. P<0.05 was considered statistically significant.

Results

The isolation and identification of rat primary VSMCs

During AS, VSMCs present a significant phenotypic transformation from the quiescent contractile status to the stimulated synthetic status and are featured by induced migration and proliferation ability. Firstly, we isolated the primary rat VSMCs and the morphology was shown (Fig. 1A). In addition, the purity of VSMCs was validated by the phenotypic transformation markers termed α-SMA in the VSMCs (Fig. 1B).

Atorvastatin regulates AngII-associated VSMCs phenotypic transformation by epigenetically regulating contractile proteins

Next, we explored the effect of atorvastatin on the AngII-associated VSMC phenotypic transformation. To this end, the VSMCs were treated with AngII, atorvastatin, or co-treated with AngII and atorvastatin, AngII and HDAC inhibitor termed trichostatin A (2). Significantly, the protein expression of contractile proteins, including α-SMA, SM-MHC, and SM22α, as the VSMCs-specific genes, was reduced by AngII and enhanced by atorvastatin, in which atorvastatin could reverse the effect of AngII in the VSMCs (Fig. 2A and B). In addition, the treatment of TSA was able to enhance the AngII-inhibited expression of α-SMA and SM-MHC (Fig. 2A and B). Furthermore, ChIP assays showed that the interaction of HDAC, P300, SRF, histone H3 lysine 9 acetylation (H3K9ac), histone H4 acetylation (H4ac) on the promoters of α-SMA and SM-MHC, but not SM22α, in which AngII could regulate the interaction while the co-treatment of atorvastatin or TSA could reverse the effect of AngII in the system (Fig. 2C). Consistently, the treatment of AngII reduced the HDAC activities and atorvastatin presented a reversed effect, in which atorvastatin could attenuate the effect of AngII in the VSMCs (Fig. 2D). Together these suggest that atorvastatin regulates VSMCs phenotypic transformation by modulating HDAC3.

Atorvastatin regulates PDGF-BB-induced VSMCs phenotypic transformation by modulating PI3K signaling

PDGF-BB is able to induce the phenotypic transformation of VSMCs and plays a critical role in the regulation of VSMCs phenotypes. Accordingly, we further evaluated the effect of atorvastatin on the PDGF-BB-induced VSMCs phenotypic transformation. For this purpose, the VSMCs were treated with PDGF-BB, atorvastatin, or co-treated with PDGF-BB and atorvastatin, PDGF-BB, atorvastatin, and PI3K inhibitor LY294002. The results showed that the mRNA expression of contractile proteins, such as α-SMA, SM-MHC, SM22α, and calponin, was inhibited by PDGF-BB but enhanced by atorvastatin, in which co-treatment of atorvastatin with PDGF-BB could rescue the PDGF-BB-reduced phenotypes in the VSMCs (Fig. 3). Meanwhile, LY294002 was able to enhance α-SMA, SM-MHC, SM22α, and calponin expression in the PDGF-BB and atorvastatin co-treated VSMCs (Fig. 3).

Atorvastatin modulates PDGF-BB-induced VSMC phenotypic transformation by modulating PI3K/FOXO4 axis

Moreover, the protein levels of α-SMA, SM-MHC, SM22α, and calponin, were reduced by PDGF-BB but upregulated by atorvastatin, in which the co-treatment of atorvastatin with PDGF-BB could rescue the PDGF-BB-inhibited phenotypes in the VSMCs (Fig. 4A and B). Furthermore, LY294002 induced α-SMA, SM-MHC, SM22α, and calponin expression in the PDGF-BB and atorvastatin co-treated VSMCs (Fig. 4A and B). In addition, the expression and phosphorylation of Akt were increased by PDGF-BB, in which atorvastatin could attenuate the effect of atorvastatin and LY294002 was able to further inhibit the phenotype (Fig. 4A and C). Significantly, the expression and phosphorylation of FOXO4, especially the serine 262 phosphorylation, was induced by PDGF-BB, while atorvastatin reduced the effect in the system and LY294002 could further reinforce the inhibitor phenotype in the VSMCs (Fig. 4A and D), suggesting that atorvastatin modulates PDGF-BB-induced VSMC phenotypic transformation by modulating the PI3K/FOXO4 axis.

Atorvastatin is able to regulate FOXO4/SRF/myocardin axis

The immunofluorescence analysis revealed that PDGF-BB enhanced the accumulation of FOXO4 in the VSMCs, while the treatment of atorvastatin was able to attenuate this effect and the co-treatment of LY294002 could further inhibit the phenotype (Fig. 5A). Subsequently, the treatment of PDGF-BB enhanced the interaction of SRF with FOXO4 and myocardin in the VSMCs, in which the co-treatment of atorvastatin and LY294002 could reverse the effect of PDGF-BB in the system (Fig. 5B), suggesting that atorvastatin is able to regulate FOXO4/SRF/myocardin axis.

Discussion

AS is a predominant type of cardiovascular disease with severe morbidity and high mortality (4). Atorvastatin has presented anti-inflammatory effects and improvements in vascular endothelial function. Nevertheless, the role and the potential mechanism of atorvastatin in the regulation of VSMCs phenotypic transformation is still unclear. In this study, we discovered a novel function of atorvastatin in modulating VSMCs phenotypic transformation by epigenetically modulating contractile proteins and mediating Akt/FOXO4 axis.

Previous findings identified the function of atorvastatin in AS and VSMCs regulation. It has been reported that atorvastatin upregulates ACE2 expression by epigenetic histone modifications in rabbit AS mode (22). Atorvastatin reduces pyroptosis via NEXN-AS1/NEXN signaling in vascular endothelial cells during AS (23). Atorvastatin represses the PDGF-ββ-stimulated migration and proliferation of VSMCs by G0/G1 cell cycle suppression and the arrest of PDGFRβ/PI3K/Akt signaling (24). Atorvastatin enhances apoptosis of VSMCs by downregulating Rho A prenylation and Bcl-2 expression (25). Results of the present study showed that, atorvastatin was able to regulate PDGF-BB/AngII-induced VSMCs phenotypic transformation. This is a novel function of atorvastatin in VSMCs regulation, providing valuable evidence for the fundamental role of atorvastatin in the development of AS.

Moreover, HDAC is known to participate in VSMCs and AS development (26,27). Protein kinase B/AKT regulates insulin-like growth factor 1-accociated phosphorylation and nuclear export of HDAC5 by activating NADPH oxidase 4 in VSMCs (26). TSA suppresses VSMCs proliferation by enhancing WAF1 (27). Additionally, FOXO4 plays a critical role in the progression of VSMCs and AS. MiR-23b downregulation enhances phenotypic switching of VSMCs by targeting FOXO4 (28). MiR-128-3p decreases VSMCs migration and proliferation by inhibiting FOXO4 signaling (29). In addition, Akt signaling is involved in the modulation of VSMCs and AS (18,30). In the present study, atorvastatin modulated VSMCs phenotypic transformation potentially by epigenetically regulating contractile proteins and regulating PI3K/FOXO4 signaling. These data identify the unreported correlation of atorvastatin with these critical factors in the modulation of VSMCs during AS development.

In conclusion, findings of the present study showed that atorvastatin regulated VSMCs phenotypic transformation by epigenetically modulating contractile proteins and mediating Akt/FOXO4 axis. This finding provides new insights into the mechanism by which atorvastatin modulates VSMCs, providing valuable evidence for the application of atorvastatin in the treatment of AS.

Acknowledgements

Not applicable.

Funding

This study was supported by the Sichuan Provincial Science and Technology Department Project (2018JY0403), Youth innovative Scientific Research Project of Sichuan Medical Association (Q16076), and Technology and Human resources' Bureau of Luzhou (17256).

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contributions

XD, XL and GZ designed the study; XD performed the experiments in figures 1,2,3,4. ZS, ZC, XZ, DW, and KW performed the experiments in figures 4 and 5. XD, XL and GZ wrote the manuscript. XD, XL and GZ confirm the authenticity of all the raw data. All authors reviewed and approved the final manuscript.

Ethics approval and consent to participate

Animal care and method procedure were authorized by the Animal Ethics Committee of Tianjin Fifth Central Hospital (approval no.: 2019-0619-37). The procedures were conformed to the Guide for the Care and Use of Laboratory Animal published by the US National Institutes of Health (NIH publication, 8th edition, 2011).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Peng J, Xiao X, Hu M and Zhang X: Interaction between gut microbiome and cardiovascular disease. Life Sci. 214:153–157. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Nitsa A, Toutouza M, Machairas N, Mariolis A, Philippou A and Koutsilieris M: Vitamin D in cardiovascular disease. In Vivo. 32:977–981. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Leong DP, Joseph PG, McKee M, Anand SS, Teo KK, Schwalm JD and Yusuf S: Reducing the global burden of cardiovascular disease, part 2: Prevention and treatment of cardiovascular disease. Circ Res. 121:695–710. 2017. View Article : Google Scholar : PubMed/NCBI

4 

Schaftenaar F, Frodermann V, Kuiper J and Lutgens E: Atherosclerosis: The interplay between lipids and immune cells. Curr Opin Lipidol. 27:209–215. 2016. View Article : Google Scholar : PubMed/NCBI

5 

Barrington WT and Lusis AJ: Atherosclerosis: Association between the gut microbiome and atherosclerosis. Nat Rev Cardiol. 14:699–700. 2017. View Article : Google Scholar : PubMed/NCBI

6 

He X, Lian Z, Yang Y, Wang Z, Fu X, Liu Y, Li M, Tian J, Yu T and Xin H: Long Non-coding RNA PEBP1P2 suppresses proliferative VSMCs phenotypic switching and proliferation in atherosclerosis. Mol Ther Nucleic Acids. 22:84–98. 2020. View Article : Google Scholar : PubMed/NCBI

7 

Xu J, Zhang Y, You S, Guo Y, Chen S, Chang Y, Zhang N and Sun Y: Paired box 9 regulates VSMC phenotypic transformation, proliferation, and migration via sonic hedgehog. Life Sci. 257:1180532020. View Article : Google Scholar : PubMed/NCBI

8 

Fu Y, Chang Y, Chen S, Li Y, Chen Y, Sun G, Yu S, Ye N, Li C and Sun Y: BAG3 promotes the phenotypic transformation of primary rat vascular smooth muscle cells via TRAIL. Int J Mol Med. 41:2917–2926. 2018.PubMed/NCBI

9 

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/NFĸB/mTOR/P70S6K signaling cascade. Redox Biol. 14:656–668. 2018. View Article : Google Scholar : PubMed/NCBI

10 

Zhang M, Li F, Wang X, Gong J, Xian Y, Wang G, Zheng Z, Shang C, Wang B, He Y, et al: MiR-145 alleviates Hcy-induced VSMC proliferation, migration, and phenotypic switch through repression of the PI3K/Akt/mTOR pathway. Histochem Cell Biol. 153:357–366. 2020. View Article : Google Scholar : PubMed/NCBI

11 

Song W, Gao K, Huang P, Tang Z, Nie F, Jia S and Guo R: Bazedoxifene inhibits PDGF-BB induced VSMC phenotypic switch via regulating the autophagy level. Life Sci. 259:1183972020. View Article : Google Scholar : PubMed/NCBI

12 

Taniguti EH, Ferreira YS, Stupp IJV, Fraga-Junior EB, Doneda DL, Lopes L, Rios-Santos F, Lima E, Buss ZS, Viola GG and Vandresen-Filho S: Atorvastatin prevents lipopolysaccharide-induced depressive-like behaviour in mice. Brain Res Bull. 146:279–286. 2019. View Article : Google Scholar : PubMed/NCBI

13 

Li S, Yu Y, Jin Z, Dai Y, Lin H, Jiao Z, Ma G, Cai W, Han B and Xiang X: Prediction of pharmacokinetic drug-drug interactions causing atorvastatin-induced rhabdomyolysis using physiologically based pharmacokinetic modelling. Biomed Pharmacother. 119:1094162019. View Article : Google Scholar : PubMed/NCBI

14 

Kogawa AC, Pires AEDT and Salgado HRN: Atorvastatin: A review of analytical methods for pharmaceutical quality control and monitoring. J AOAC Int. 102:801–809. 2019. View Article : Google Scholar : PubMed/NCBI

15 

Ginnan R, Sun LY, Schwarz JJ and Singer HA: MEF2 is regulated by CaMKIIδ2 and a HDAC4-HDAC5 heterodimer in vascular smooth muscle cells. Biochem J. 444:105–114. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Zhao G, Fu Y, Cai Z, Yu F, Gong Z, Dai R, Hu Y, Zeng L, Xu Q and Kong W: Unspliced XBP1 Confers VSMC homeostasis and prevents aortic aneurysm formation via FoxO4 Interaction. Circ Res. 121:1331–1345. 2017. View Article : Google Scholar : PubMed/NCBI

17 

Huang C, Huang W, Wang R and He Y: Ulinastatin inhibits the proliferation, invasion and phenotypic switching of PDGF-BB-Induced VSMCs via Akt/eNOS/NO/cGMP signaling pathway. Drug Des Devel Ther. 14:5505–5514. 2020. View Article : Google Scholar : PubMed/NCBI

18 

Lu W, Ni Z, Jiang S, Tong M, Zhang J, Zhao J, Feng C, Jia Q, Wang J, Yao T, et al: Resveratrol inhibits bile acid-induced gastric intestinal metaplasia via the PI3K/AKT/p-FoxO4 signalling pathway. Phytother Res. 35:1495–1507. 2021. View Article : Google Scholar : PubMed/NCBI

19 

Zhou N, Lee JJ, Stoll S, Ma B, Wiener R, Wang C, Costa KD and Qiu H: Inhibition of SRF/myocardin reduces aortic stiffness by targeting vascular smooth muscle cell stiffening in hypertension. Cardiovasc Res. 113:171–182. 2017. View Article : Google Scholar : PubMed/NCBI

20 

Qin X, Hou X, Zhang M, Liang T, Zhi J, Han L and Li Q: Relaxation of rat aorta by farrerol correlates with potency to reduce intracellular calcium of VSMCs. Int J Mol Sci. 15:6641–6656. 2014. View Article : Google Scholar : PubMed/NCBI

21 

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

22 

Tikoo K, Patel G, Kumar S, Karpe PA, Sanghavi M, Malek V and Srinivasan K: Tissue specific up regulation of ACE2 in rabbit model of atherosclerosis by atorvastatin: Role of epigenetic histone modifications. Biochem Pharmacol. 93:343–351. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Wu LM, Wu SG, Chen F, Wu Q, Wu CM, Kang CM, He X, Zhang RY, Lu ZF, Li XH, et al: Atorvastatin inhibits pyroptosis through the lncRNA NEXN-AS1/NEXN pathway in human vascular endothelial cells. Atherosclerosis. 293:26–34. 2020. View Article : Google Scholar : PubMed/NCBI

24 

Chen S, Dong S, Li Z, Guo X, Zhang N, Yu B and Sun Y: Atorvastatin calcium inhibits PDGF-ββ-induced proliferation and migration of VSMCs Through the G0/G1 cell cycle arrest and suppression of activated PDGFRbeta-PI3K-Akt signaling cascade. Cell Physiol Biochem. 44:215–228. 2017. View Article : Google Scholar : PubMed/NCBI

25 

Blanco-Colio LM, Villa A, Ortego M, Hernandez-Presa MA, Pascual A, Plaza JJ and Egido J: 3-Hydroxy-3-methyl-glutaryl coenzyme A reductase inhibitors, atorvastatin and simvastatin, induce apoptosis of vascular smooth muscle cells by downregulation of Bcl-2 expression and Rho A prenylation. Atherosclerosis. 161:17–26. 2002. View Article : Google Scholar : PubMed/NCBI

26 

Pietruczuk P, Jain A, Simo-Cheyou ER, Anand-Srivastava MB and Srivastava AK: Protein kinase B/AKT mediates insulin-like growth factor 1-induced phosphorylation and nuclear export of histone deacetylase 5 via NADPH oxidase 4 activation in vascular smooth muscle cells. J Cell Physiol. 234:17337–17350. 2019. View Article : Google Scholar : PubMed/NCBI

27 

Okamoto H, Fujioka Y, Takahashi A, Takahashi T, Taniguchi T, Ishikawa Y and Yokoyama M: Trichostatin A, an inhibitor of histone deacetylase, inhibits smooth muscle cell proliferation via induction of p21(WAF1). J Atheroscler Thromb. 13:183–191. 2006. View Article : Google Scholar : PubMed/NCBI

28 

Iaconetti C, De Rosa S, Polimeni A, Sorrentino S, Gareri C, Carino A, Sabatino J, Colangelo M, Curcio A and Indolfi C: Down-regulation of miR-23b induces phenotypic switching of vascular smooth muscle cells in vitro and in vivo. Cardiovasc Res. 107:522–533. 2015. View Article : Google Scholar : PubMed/NCBI

29 

Qu C, Liu X, Guo Y, Fo Y, Chen X, Zhou J and Yang B: MiR-128-3p inhibits vascular smooth muscle cell proliferation and migration by repressing FOXO4/MMP9 signaling pathway. Mol Med. 26:1162020. View Article : Google Scholar : PubMed/NCBI

30 

Gong C, Ai J, Fan Y, Gao J, Liu W, Feng Q, Liao W and Wu L: NCAPG promotes the proliferation of hepatocellular carcinoma through PI3K/AKT signaling. Onco Targets Ther. 12:8537–8552. 2019. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2022
Volume 25 Issue 5

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Du X, Sun Z, Cao Z, Zhou X, Wang D, Wang K, Li X and Zuo G: Atorvastatin regulates vascular smooth muscle cell phenotypic transformation by epigenetically modulating contractile proteins and mediating Akt/FOXO4 axis. Mol Med Rep 25: 167, 2022
APA
Du, X., Sun, Z., Cao, Z., Zhou, X., Wang, D., Wang, K. ... Zuo, G. (2022). Atorvastatin regulates vascular smooth muscle cell phenotypic transformation by epigenetically modulating contractile proteins and mediating Akt/FOXO4 axis. Molecular Medicine Reports, 25, 167. https://doi.org/10.3892/mmr.2022.12683
MLA
Du, X., Sun, Z., Cao, Z., Zhou, X., Wang, D., Wang, K., Li, X., Zuo, G."Atorvastatin regulates vascular smooth muscle cell phenotypic transformation by epigenetically modulating contractile proteins and mediating Akt/FOXO4 axis". Molecular Medicine Reports 25.5 (2022): 167.
Chicago
Du, X., Sun, Z., Cao, Z., Zhou, X., Wang, D., Wang, K., Li, X., Zuo, G."Atorvastatin regulates vascular smooth muscle cell phenotypic transformation by epigenetically modulating contractile proteins and mediating Akt/FOXO4 axis". Molecular Medicine Reports 25, no. 5 (2022): 167. https://doi.org/10.3892/mmr.2022.12683