Inhibitory effects of roscovitine on proliferation and migration of vascular smooth muscle cells in vitro

Shuang-shuang Zhang , Wei Wang , Chong-qiang Zhao , Min-jie Xie , Wen-yu Li , Xiang-li Yang , Jia-gao Lv

Current Medical Science ›› 2014, Vol. 34 ›› Issue (6) : 791 -795.

PDF
Current Medical Science ›› 2014, Vol. 34 ›› Issue (6) : 791 -795. DOI: 10.1007/s11596-014-1354-5
Article

Inhibitory effects of roscovitine on proliferation and migration of vascular smooth muscle cells in vitro

Author information +
History +
PDF

Abstract

Abnormal proliferation and migration of vascular smooth muscle cells (VSMCs) are the major cause of in-stent restenosis (ISR). Intervention proliferation and migration of VSMCs is an important strategy for antirestenotic therapy. Roscovitine, a second-generation cyclin-dependent kinase inhibitor, can inhibit cell cycle of multiple cell types. We studied the effects of roscovitine on cell cycle distribution, proliferation and migration of VSMCs in vitro by flow cytometry, BrdU incorporation and wound healing assay, respectively. Our results showed that roscovitine increased the proportion of G0/G1 phase cells after 12 h (69.57±3.65 vs. 92.50±1.68, P=0.000), 24 h (80.87±2.24 vs. 90.25±0.79, P=0.000) and 48 h (88.08±3.86 vs. 88.87±2.43, P=0.427) as compared with control group. Roscovitine inhibited proliferation and migration of VSMCs in a concentration-dependent way. With the increase of concentration, roscovitine showed increased capacity for growth and migration inhibition. Roscovitine (30 μmol/L) led to an almost complete VSMCs growth and migration arrest. Combined with its low toxicity and selective inhibition to ISR-VSMCs, roscovitine may be a potential drug in the treatment of vascular stenosis diseases and particularly useful in the prevention and treatment of ISR.

Keywords

roscovitine / vascular smooth muscle cells / cell cycle / cell proliferation / cell migration / restenosis

Cite this article

Download citation ▾
Shuang-shuang Zhang, Wei Wang, Chong-qiang Zhao, Min-jie Xie, Wen-yu Li, Xiang-li Yang, Jia-gao Lv. Inhibitory effects of roscovitine on proliferation and migration of vascular smooth muscle cells in vitro. Current Medical Science, 2014, 34(6): 791-795 DOI:10.1007/s11596-014-1354-5

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

DorrosG, CowleyMJ, SimpsonJ, et al. . Percutaneous transluminal coronary angioplasty: report of complications from the National Heart, Lung, and Blood Institute PTCA Registry. Circulation, 1983, 67(4): 723-730 PMID: 6218938

[2]

GrechED. ABC of interventional cardiology: percutaneous coronary intervention. I: history and development. BMJ, 2003, 326(7398): 1080-1082 PMCID: 1125993 PMID: 12750213

[3]

FischmanDL, LeonMB, BaimDS, et al. . A randomized comparison of coronary-stent placement and balloon angioplasty in the treatment of coronary artery disease. Stent Restenosis Study Investigators. N Engl J Med, 1994, 331(8): 496-501 PMID: 8041414

[4]

GershlickAH. Role of stenting in coronary revascularisation. Heart, 2001, 86(1): 104-112 PMCID: 1729822 PMID: 11410576

[5]

KomatsuR, UedaM, NarukoT, et al. . Neointimal tissue response at sites of coronary stenting in humans: macroscopic, histological, and immunohistochemical analyses. Circulation, 1998, 98(3): 224-233 PMID: 9697822

[6]

BennettMR, O’sullivanM. Mechanisms of angioplasty and stent restenosis: implications for design of rational therapy. Pharmacol Ther, 2001, 91(2): 149-166 PMID: 11728607

[7]

ColomboA, DrzewieckiJ, BanningA, et al. . Randomized study to assess the effectiveness of slow- and moderate-release polymer-based paclitaxel-eluting stents for coronary artery lesions. Circulation, 2003, 108(7): 788-794 PMID: 12900339

[8]

DawkinsKD, GrubeE, GuagliumiG, et al. . Clinical efficacy of polymer-based paclitaxel-eluting stents in the treatment of complex, long coronary artery lesions from a multicenter, randomized trial: support for the use of drug-eluting stents in contemporary clinical practice. Circulation, 2005, 112(21): 3306-3313 PMID: 16286586

[9]

GrubeE, SilberS, HauptmannKE, et al. . TAXUS I: six- and twelve-month results from a randomized, double-blind trial on a slow-release paclitaxel-eluting stent for de novo coronary lesions. Circulation, 2003, 107(1): 38-42 PMID: 12515740

[10]

MoriceMC, ColomboA, MeierB, et al. . Sirolimus- vs paclitaxel-eluting stents in de novo coronary artery lesions: the REALITY trial: a randomized controlled trial. JAMA, 2006, 295(8): 895-904 PMID: 16493102

[11]

MoriceMC, SerruysPW, SousaJE, et al. . A randomized comparison of a sirolimus-eluting stent with a standard stent for coronary revascularization. N Engl J Med, 2002, 346(23): 1773-1780 PMID: 12050336

[12]

MosesJW, LeonMB, PopmaJJ, et al. . Sirolimus-eluting stents versus standard stents in patients with stenosis in a native coronary artery. N Engl J Med, 2003, 349(14): 1315-1323 PMID: 14523139

[13]

SchampaertE, CohenEA, SchluterM, et al. . The Canadian study of the sirolimus-eluting stent in the treatment of patients with long de novo lesions in small native coronary arteries (C-SIRIUS). J Am Coll Cardiol, 2004, 43(6): 1110-1115 PMID: 15028375

[14]

StoneGW, EllisSG, CannonL, et al. . Comparison of a polymer-based paclitaxel-eluting stent with a bare metal stent in patients with complex coronary artery disease: a randomized controlled trial. JAMA, 2005, 294(10): 1215-1223 PMID: 16160130

[15]

StoneGW, EllisSG, CoxDA, et al. . One-year clinical results with the slow-release, polymer-based, paclitaxel-eluting TAXUS stent: the TAXUS-IV trial. Circulation, 2004, 109(16): 1942-1947 PMID: 15078803

[16]

StoneGW, EllisSG, CoxDA, et al. . A polymer-based, paclitaxel-eluting stent in patients with coronary artery disease. N Engl J Med, 2004, 350(3): 221-231 PMID: 14724301

[17]

WeiszG, LeonMB, HolmesDRJr., et al. . Two-year outcomes after sirolimus-eluting stent implantation: results from the Sirolimus-Eluting Stent in de Novo Native Coronary Lesions (SIRIUS) trial. J Am Coll Cardiol, 2006, 47(7): 1350-1355 PMID: 16580520

[18]

WindeckerS, JuniP. Safety of drug-eluting stents. Nat Clin Pract Cardiovasc Med, 2008, 5(6): 316-328 PMID: 18414453

[19]

IakovouI, SchmidtT, BonizzoniE, et al. . Incidence, predictors, and outcome of thrombosis after successful implantation of drug-eluting stents. JAMA, 2005, 293(17): 2126-2130 PMID: 15870416

[20]

ImanishiT, KobayashiK, KukiS, et al. . Sirolimus accelerates senescence of endothelial progenitor cells through telomerase inactivation. Atherosclerosis, 2006, 189(2): 288-296 PMID: 16455087

[21]

JonerM, FinnAV, FarbA, et al. . Pathology of drug-eluting stents in humans: delayed healing and late thrombotic risk. J Am Coll Cardiol, 2006, 48(1): 193-202 PMID: 16814667

[22]

OngAT, McfaddenEP, RegarE, et al. . Late angiographic stent thrombosis (LAST) events with drug-eluting stents. J Am Coll Cardiol, 2005, 45(12): 2088-2092 PMID: 15963413

[23]

GoodyearS, SharmaMC. Roscovitine regulates invasive breast cancer cell (MDA-MB231) proliferation and survival through cell cycle regulatory protein cdk5. Exp Mol Pathol, 2007, 82(1): 25-32 PMID: 17081516

[24]

McclueSJ, BlakeD, ClarkeR, et al. . In vitro. Int J Cancer, 2002, 102(5): 463-468 PMID: 12432547

[25]

MeijerL, BorgneA, MulnerO, et al. . Biochemical and cellular effects of roscovitine, a potent and selective inhibitor of the cyclin-dependent kinases cdc2, cdk2 and cdk5. Eur J Biochem, 1997, 243(1–2): 527-536 PMID: 9030781

[26]

RaynaudFI, WhittakerSR, FischerPM, et al. . In vitro and in vivo pharmacokinetic-pharmacodynamic relationships for the trisubstituted aminopurine cyclin-dependent kinase inhibitors olomoucine, bohemine and CYC202. Clin Cancer Res, 2005, 11(13): 4875-4887 PMID: 16000586

[27]

Wesierska-GadekJ, GueorguievaM, WojciechowskiJ, et al. . Cell cycle arrest induced in human breast cancer cells by cyclin-dependent kinase inhibitors: a comparison of the effects exerted by roscovitine and olomoucine. Pol J Pharmacol, 2004, 56(5): 635-641 PMID: 15591654

[28]

BensonC, WhiteJ, De BonoJ, et al. . A phase I trial of the selective oral cyclin-dependent kinase inhibitor seliciclib (CYC202; R-Roscovitine), administered twice daily for 7 days every 21 days. Br J Cancer, 2007, 96(1): 29-37 PMCID: 2360206 PMID: 17179992

[29]

Le TourneauC, FaivreS, LaurenceV, et al. . Phase I evaluation of seliciclib (R-roscovitine), a novel oral cyclin-dependent kinase inhibitor, in patients with advanced malignancies. Eur J Cancer, 2010, 46(18): 3243-3250 PMID: 20822897

[30]

O’sullivanM, ScottSD, MccarthyN, et al. . Differential cyclin E expression in human in-stent stenosis smooth muscle cells identifies targets for selective anti-restenosis therapy. Cardiovasc Res, 2003, 60(3): 673-683 PMID: 14659813

[31]

QiaoM, ShapiroP, FosbrinkM, et al. . Cell cycle-dependent phosphorylation of the RUNX2 transcription factor by cdc2 regulates endothelial cell proliferation. J Biol Chem, 2006, 281(11): 7118-7128 PMID: 16407259

[32]

ThompsonCC, AshcroftFJ, PatelS, et al. . Pancreatic cancer cells overexpress gelsolin family-capping proteins, which contribute to their cell motility. Gut, 2007, 56(1): 95-106 PMCID: 1856675 PMID: 16847067

[33]

Milovanceva-PopovskaM, KunterU, OstendorfT, et al. . R-roscovitine (CYC202) alleviates renal cell proliferation in nephritis without aggravating podocyte injury. Kidney Int, 2005, 67(4): 1362-1370 PMID: 15780088

[34]

WuPC, TaiMH, HuDN, et al. . Cyclin-dependent kinase inhibitor roscovitine induces cell cycle arrest and apoptosis in rabbit retinal pigment epithelial cells. J Ocul Pharmacol Ther, 2008, 24(1): 25-33 PMID: 18370874

[35]

LjungmanM, PaulsenMT. The cyclin-dependent kinase inhibitor roscovitine inhibits RNA synthesis and triggers nuclear accumulation of p53 that is unmodified at Ser15 and Lys382. Mol Pharmacol, 2001, 60(4): 785-789 PMID: 11562441

[36]

SrokaIM, HeissEH, HavlicekL, et al. . A novel roscovitine derivative potently induces G1-phase arrest in platelet-deried growth factor-BB-activated vascular smooth muscle cells. Mol Pharmacol, 2010, 77(2): 255-261 PMID: 19903826

AI Summary AI Mindmap
PDF

87

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/