用户名: 密码: 验证码:
重组Psilencer1.0-U6-siRNA-STAT3对动脉化静脉壁内膜异常增生抑制作用的实验研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
临床上,冠状动脉旁路移植手术(CABG)是挽救冠状动脉粥样硬化性心脏病病人的重要措施。和自体的乳内动脉(IMA)相比,自体静脉中的大隐静脉(SV)是目前冠状动脉搭桥手术中的第二选择,但其缺点是远期阻塞率较高,术后阻塞发生率逐年增加,至术后10年全部阻塞率为35-40%。研究者发现其根本原因是由于血管重塑的结果以及新生内膜的形成,血管重塑包括细胞增殖,细胞凋亡,细胞迁移和降解以及细胞外基质的产生。
     转录信号传导子及激活子通路(STATs)代表一条从膜到核的信号传导系统, STAT作用于细胞周期蛋白D1(CyclinD1)、B淋巴细胞瘤(Bcl-xL)等下游靶基因,调节细胞增殖、分化及凋亡。蛋白酪氨酸激酶(JAK)/STAT是一类具有复合效应和功能的信号通路,与其他细胞内信号通路有着复杂的交互关系,研究其与血管平滑肌细胞(VSMCs)病理生理功能之间的关系,可能为揭示以VSMCs增殖、迁移为发病核心环节的血管新生内膜形成、血管重塑等血管增殖性疾病的病理机制提供重要的实验基础和理论依据并指导临床治疗。
     本研究利用经RNA干扰技术构建的针对STAT3通路的特异性质粒-Psilencer1.0-U6- siRNA-STAT3,来验证STAT3通路在VSMCs增殖中的作用;一、体外将该质粒通过脂质体Lipofectamine2000转染培养的大鼠主动脉平滑肌细胞(RSMCs),分为三组;对照组、空质粒组和实验组。共培养72小时可见,对照组和空质粒组的RSMC细胞数量明显高于实验组,细胞贴壁生长,状况良好,增殖旺盛,细胞间相互融合,呈“铺路石”样;细胞形态呈现梭形;而实验组RSMCs 48小时后则出现生长缓慢,细胞皱缩;部分细胞失去原有形态,细胞数目明显减少,折光性差。MTT检测显示实验组抑制率明显高于对照组和空质粒组。实验组的STAT3及其下游靶基因CyclnD1和Bcl-2的mRNA及蛋白的表达明显低于对照组。二、建立大鼠颈静脉移植模型,将其分为2组,对照组和实验组;并应用生物蛋白胶将质粒-脂质体混合物均匀的涂布于移植血管外膜。对照组血管内膜在术后明显增生,而实验组则增生不明显。同时实验组内膜增生的阳性细胞比例明显少于对照组。实验组移植血管STAT3及其下游靶基因CyclnD1和Bcl-2的mRNA及蛋白的表达明显低于对照组。该质粒确实通过对STAT3基因片段的转录翻译过程的抑制作用,阻断了其对下游靶基因CyclinD1和Bcl-2的作用,抑制了VSMCs的增殖,减轻了动脉化静脉壁血管的内膜增生。
Cardiovascular diseases due to atherosclerosis are the main reasons for deaths in the western world. For many patients with severe coronary disease,bypass-surgery is required. Veins are frequently used and have far better results compared with synthetic conduits. But Veins are not ideal bypass conduits which are reflected by the incidence of vein graft failure. Recent studies demonstrated that the primary patency of vein grafts was not better than 60-80 % within the first year after surgery treatment,and by the time when in the tenth year,the failure rate is 50%. A large amount of clinical and experimental studies have demonstrated that intimal thickening may lead to the restenosis of the vein graft. Proliferation and migration of vescular smooth muscle cells(VSMCs) may be a key contributor to neointimal formation. It was demonstrated in a canine vein graft model that the SMCs were transformed from contrictile to a secretory phenotype in response to vessel wall injury. By the switch of phenotypes the SMCs enter a state enabling proliferation,migration,and gain synthetic capacity,which is a pre-requisite for neointimal formation. It has been a well- supported opinion,that the source for the intimal cell population is SMCs from the media. Accumulating evidence sμggests that neointima formation is mediated by a complex interaction of a variety of growth regulatory molecules, such as growth factors,vasoactive peptides,inflammatory cytokines and chemokines.
     However,it remains largely unknown which signaling pathways are responsible for neointima formation. STAT3,as one of the candidate signaling pathways,plays an important role in the SMCs proliferation and migration. STAT3 contains a conserved amino-terminal,aDNA-binding domain that binds specific interferon-activated DNA sequences,a SH-2 domain for receptor recruitment and STAT dimerization,and a transactivation domain. STAT3 protein is activated by a variety of stimuli. It is also involved in contradictory responses such as proliferation and apoptosis. Following activation,STAT3 protein changes conformation,forms homo- or hetero-dimers,and translocated from the cytoplasm to the nucleus. In the nucleus,STAT3 binds cis elements,which induce growth responsive and inflammatory genes; including junB,IRF-1,CyclinD1 and anti-apoptotic factors Bcl-xL and Bcl-2. In cultured rat SMCs , the STAT3 activation is substantially involved in the angiotensin II(AII)-or platelet-derived growth factor (PDGF)–induced proliferation.
     To prevent restenosis of vein graft,extensive research has been conducted including standard technique of vein harvest and drug therapy et al,but these methods were not an ideal therapy. Two decades ago,gene therapy emerged since the first description of the site-directed transfer of exogenous genetic material into the vascular system .In initial experiments,vascular gene transfer were used to evaluate hypothesis by targeted overex- pression of selected proteins in the vasculature.Therefore,gene therapy as a potential strategy for the treatment of restenosis after angioplasty and vascular bypass graft has been extensively studied. Although effective delivery tools are remain to be solved,these reports sparked the anticipation that such approaches would be used for the therapeutic modulation of vascular responses,including stenosis after injury and restenosis.
     In this study,the potential anti-proliferation effects of the recombinant plasmid- Psilencer1.0-U6-siRNA-STAT-3 on VSMCs in vitro and in vivo were investigated to find potential novel strategies for the treatment of restenosis after vein grafting. This plasmid is constructed by RNAi technique to aim at inhibiting mRNA expression and phosphorylation of STAT3. Following this process,Psilencer1.0-U6-siRNA-STAT-3 block the tranicription and translation of the downstream target genes-CyclnD1 and Bcl-2. By this mechanism,proliferation of VSMCs and the neointima formation are inhibited,and restenosis of transpl- anted vein graft is lessen or even blocked. Our study afford a theory base for anti-restenosis after coronary artery bypass graft on clinic.
     Psilencer1.0-U6-siRNA-STAT-3 inhibited the proliferation of rat vascular smooth muscle cells in vitro
     Methods:RSMCs (2×105 cells per well) seeded in 6-well culture plates were divided into 3 groups: the control group that no transfection were done; the scramble plasmid group in which the scramble plasmid transfected RSMCs and recombinant plasmid group in which RSMCs were transfected with Psilencer1.0-U6-siRNA-STAT-3. The lipofectamine 2000,as the vector,was mixed with plasmid (recombinant plasmid group or scramble)or itself at a proportionality of 0.5μl:0.2μg and added to the RSMCs .After coincubation for 72h,we use contrast phase microscope to observe the shape change of RSMCs; we added MTT solution (5 mg/mL in phosphate-buffered saline (PBS) to observe the proliferation capability of RSMCs; RT-PCR were examined to observe the mRNA expression of STAT3 and its downstream target gene CyclnD1 and Bcl-2; Western-blot were performed to observe expression of their fuctional products-protein.
     Results:Observed with contrast phase microscope,Most RSMCs of the control group and scramble plasmid group are clostridial、anomalous triangle or fan shape with clear and intermediate nucleis and proliferate vigorous;however,RSMCs of Psilencer1.0-U6-siRNA- STAT3 group lost their intrinsical form and proliferated slowly with a pycnosis nucleis and some of them even floated in the culture bottle. By MTT assays,the proliferation of RSMCs in Psilencer1.0-U6-siRNA-STAT-3 group was significantly inhibited compared with the control group. RT-PCR detection show significantly decreased mRNA expression of STAT3、CyclnD1 and Bcl-2 in the Psilencer1.0-U6-siRNA-STAT-3 group,whereas the mRNA exepression of them were indistinguishable between the control and scramble plasmid group. Western-blot detection show that STAT3、Bcl-2 and CyclinD1 phosphorylation in the Psilencer1.0-U6-siRNA-STAT-3 group significantly declined compared with those of the control group.
     Psilencer1.0-U6-siRNA-STAT-3 inhibited the proliferation of vascular smooth muscle cells in vivo
     Methods:we constructed vein graft model and the Psilencer1.0-U6- siRNA- STAT-3/ lipofectamine2000 mixture were mixed with bioprotain gel (volume 80 ul) and sprayed to adventitia of vein graft. 168 free male wistar rats (250-350g) were divided into two groups: the control group (n=84) and recombinant plasmid(experiment)group (n = 84). On day 3、7、14 and 28 after operative procedure,the rats were killed. 5 of these interposed vein graft containing an adjacent section of the left carotid artery were harvested for morphological examination(HE) and immuno- histochemistry(PCNA); 8 of these interposed vein graft were harvested for RT-PCR detection and 8 for Western-blot assay.
     Results:The intimal thickening on 3d post operative procedure had not much difference in the two groups,and it peaked on day 7 in the scramble plasmid group,while that in the recombinant plasmid group on day 7 had only scabrosity,on day 14 and 28,it had even no thickening in the experiment group. The thickness of intima had significant difference on day 7(12.3±0.21 and 6.7±0.25 respectively) and 14(7.2±0.31 and 4.6±0.14 respectively); PCNA index in the control group on day 7 and 14 were 31.3±4.7 and 27.2±5.7 respectively and that of experiment group were 23.3±2.8 and 15.6±0.4 respectively; there are significant deviation between the control and experiment group on day 7 and 14 of PCNA positive cells. RT-PCR detection shows that from day 3 after operative procedure,mRNA exepression of STAT3 and its downstream target gene CyclnD1 and Bcl-2 were showed in both the control and exeperiment group,and all peaked at 14d and declined to insignificant level at 28d. Compared with those of the control group,the mRNA expression of the experiment group on 7d decreased significantly. Immunoblotting studies showed that a transient STAT3 induction was accompanied with constitutive expression in the control group,The constitutive STAT3 phosphorylation was detected in the arterialized vein graft after day 3,peaked at day 7,and declined to insignificant levels at day 14. while in the experiment group,although STAT3 phosphorylation detected on day 3 was nearly the same as that in the control group,it was far lower than that of the control group on the 7d; and also that on day 7,CyclnD1 and Bcl-2 proteins level in the experiment group declined significantly compared with that of the control group.
引文
[1]Pieter Klinkert, Abbey Schepers, Desire′e H. C. Burger, et al. Vein versus polytetra- fluoroethylene in above-knee femoropopliteal bypass grafting:five-year results of a randomized controlled trial[J]. J Vasc Surg, 2003, 37(1):149-155.
    [2]Loop, F.D.,et al. Influence of the internal-mammary-artery graft on 10-year survival and other cardiac events[J]. N Engl J Med, 1986, 314(1):1-6.
    [3]Lytle, B.W., et al. Two internal thoracic artery grafts are better than one[J]. J Thorac Cardiovasc Surg, 1999, 117(5):855-872.
    [4]Goldman, S., et al. Long-term patency of saphenous vein and left internal mammary artery grafts after coronary artery bypass surgery: results from a Department of Veterans Affairs Cooperative Study[J]. J Am Coll Cardiol, 2004, 44(11):2149-2156.
    [5]Conte, M.S., et al. Results of PREVENT III: a multicenter, randomized trial of edifoligide for the prevention of vein graft failure in lower extremity bypass surgery[J]. J Vasc Surg, 2006, 43(4):742-751.
    [6]Lundell, A., et al. Femoropopliteal-crural graft patency is improved by an intensive surveillance program: a prospective randomized study[J]. J Vasc Surg, 1995, 21(1):26-33.
    [7]Alexander, J.H., et al. Efficacy and safety of edifoligide, an E2F transcription factor decoy, for prevention of vein graft failure following coronary artery bypass graft surgery: PREVENT IV: a randomized controlled trial[J]. JAMA, 2005, 294(19):2446-2454.
    [8]Mills, J.L.,et al. The characteristics and anatomic distribution of lesions that cause reversed vein graft failure: a five-year prospective study[J]. J Vasc Surg, 1993, 17(1):195- 204.
    [9]Fitzgibbon, G.M., et al. Coronary bypass graft fate and patient outcome: angiographic follow-up of 5065 grafts related to survival and reoperation in 1388 patients during 25 years[J]. J Am Coll Cardiol, 1996, 28(3):616-626.
    [10]Cho, K.R., et al. Serial angiographic follow-up of grafts one year and five years after coronary artery bypass surgery[J]. Eur J Cardiothorac Surg, 2006, 29 (4):511-516.
    [11]Widimsky, P., et al. One-year coronary bypass graft patency: a randomized comparison between off-pump and on-pump surgery angiographic results of the PRAGUE-4 trial[J]. Circulation, 2004, 110(22):3418-3423.
    [12]Kenagy R D, Fukai N, Min SK, et al. Proliferative capacity of vein graft smooth muscle cells and fibroblasts in vitro correlates with graft stenosis[J]. J Vasc Surg, 2009, 49(5): 1282-1288
    [13]Conte MS. Boston mass. Molecular engineerig of vein bypass grafts[J]. J Vasc Surg, 2007, 45(10):74A-81A.
    [14]Regisa G, Pensaa S, Boselli D,et al. The STAT1:STAT3 seesaw of Interferon and gp130 receptor signaling[J]. Semi in Cell Develop Bio, 2008, 19(6):351-359.
    [15]Kishore R, W. Losordo D. Gene therapy for restenosis: Biological solution to a biological problem[J]. J Mole Cell Cardio, 2007, 42(12):461-468.
    [16]Shrager JB. The Vineberg procedure:the innediate forerunner of coronary bypass grafting[J]. Ann Thorac Surg, 1994, 57:1354-1364.
    [17]Johnson WD,Flemma RJ,Lepley D. Direct coronary surgery utilizing multiple -vein bypass grafts[J]. Ann Thorc Surg, 1970, 9:436-444.
    [18]Perault L,Pelletier LC,Carrier M. Are there other alternative conduits to consider for coronary artery graft? Austin:R G Landes Company, 1993, 74-84
    [19]Krabatsch T,Grauhan O,Hetzer R. Unilateral arterial graft with patency of 30 years[J]. Circulation, 2000, 102:1724-1725
    [20]Gertler, J.P. and W.M. Abbott. Prothrombotic and fibrinolytic function of normal and perturbed endothelium[J]. J Surg Res, 1992,52(1):89-95.
    [21]Davies, P.F. and S.C. Tripathi. Mechanical stress mechanisms and the cell. An endothelial paradigm[J]. Circ Res, 1993, 72(2):239-245.
    [22]Cines, D.B., et al. Endothelial cells in physiology and in the pathophysiology of vascular disorders[J]. Blood, 1998, 91(10):3527-3561.
    [23]K. ?sterberg E. Mattsson. Intimal hyperplasia in mouse vein grafts is regulated by flow[J]. J Vasc Res, 2005, 42(1):13-20.
    [24]Corson, J.D., et al. Relationship between vasa vasorum and blood flow to vein bypass endothelial morphology[J]. Arch Surg, 1985, 120(3):386-388.
    [25]Gnasso, A., et al. Association between wall shear stress and flow-mediated vasodilation in healthy men[J]. Atherosclerosis, 2001, 156(1):171-176.
    [26]Pyke, K.E. and M.E. Tschakovsky. The relationship between shear stress and flow-mediated dilatation: implications for the assessment of endothelial function[J]. J Physiol, 2005, 568(2):357-369.
    [27]Rubanyi, G.M., et al. Flow-induced release of endothelium-derived relaxing factor[J]. Am J Physiol, 1986, 250(6):H1145-1149.
    [28]Nagel, T., et al. Vascular endothelial cells respond to spatial gradients in fluid shear stress by enhanced activation of transcription factors[J]. Arterioscler Thromb Vasc Biol, 1999, 19(8):1825-1834.
    [29]Ihnat, D.M., et al. The correlation of early flow disturbances with the development of infrainguinal graft stenosis: a 10-year study of 341 autogenous vein grafts[J]. J Vasc Surg, 1999, 30(1):8-15.
    [30]Frangos, S.G., et al. Localization of atherosclerosis: role of hemodynamics[J]. Arch Surg, 1999, 134(10):1142-9.
    [31]Schwartz, S.M., et al. The intima. Soil for atherosclerosis and restenosis[J]. Circ Res, 1995, 77(3):445-65.
    [32]Newby, A.C. and A.B. Zaltsman. Molecular mechanisms in intimal hyperplasia[J]. J Pathol, 2000, 190(3):300-309.
    [33]Berman, S.S., et al. Impact of nonpenetrating clips on intimal hyperplasia of vascular anastomoses[J]. Cardiovasc Surg, 2001, 9(6):540-547.
    [34]Zwolak, R.M., et al. Kinetics of vein graft hyperplasia: association with tangential stress[J]. J Vasc Surg, 1987, 5(1):126-136.
    [35]Fingerle, J., et al. Intimal lesion formation in rat carotid arteries after endothelial denudation in absence of medial injury[J]. Arteriosclerosis, 1990, 10(6):1082-1087.
    [36]Clowes, A.W., et al. Kinetics of cellular proliferation after arterial injury. V. Role of acute distension in the induction of smooth muscle proliferation[J]. Lab Invest, 1989, 60(3):360- 364.
    [37]Shi, Y., et al. Adventitial myofibroblasts contribute to neointimal formation in injured porcine coronary arteries[J]. Circulation, 1996, 94(7):1655-1664.
    [38]Morinaga, K., et al. Effect of wall shear stress on intimal thickening of arterially transplanted autogenous veins in dogs[J]. J Vasc Surg, 1985, 2(3):430-433.
    [39]Hμghes, P.E. and T.V. How,. Effects of geometry and flow division on flow structures in models of the distal end-to-side anastomosis[J]. J Biomech, 1996, 29(7):855-872.
    [40]Leask, R.L., et al. Human saphenous vein coronary artery bypass graft morphology, geometry and hemodynamics[J]. Ann Biomed Eng, 2005, 33(3):301-309.
    [41]Quist, W.C. and F.W. LoGerfo,. Prevention of smooth muscle cell phenotypic modulation in vein grafts: a histomorphometric study[J]. J Vasc Surg, 1992, 16(2):225-231.
    [42]Owens, G.K., et al. Molecular regulation of vascular smooth muscle cell differentiation in development and disease[J]. Physiol Rev, 2004, 84(3):767-801.
    [43]Yamamura, S., et al. Blood flow and kinetics of smooth muscle cell proliferation in canine autogenous vein grafts: in vivo BrdU incorporation[J]. J Surg Res, 1994, 56(2):155-161.
    [44]Angelini, G.D., et al. Time-course of medial and intimal thickening in pig venous arterial grafts: relationship to endothelial injury and cholesterol accumulation[J]. J ThoracCardiovasc Surg, 1992, 103(6):1093-1103.
    [45]Davies, M.G. and P.O. Hagen,. Pathophysiology of vein graft failure: a review[J]. Eur J Vasc Endovasc Surg, 1995, 9(1):7-18.
    [46]Lemson, M.S., et al. Intimal hyperplasia in vascular grafts[J]. Eur J Vasc Endovasc Surg, 2000, 19(4):336-350.
    [47]Faries, P.L., et al. Immunolocalization and temporal distribution of cytokine expression during the development of vein graft intimal hyperplasia in an experimental model[J]. J Vasc Surg, 1996, 24(3):463-471.
    [48]Ferns, G.A., et al. Inhibition of neointimal smooth muscle accumulation after angioplasty by an antibody to PDGF[J]. Science, 1991, 253(5024):1129-1132.
    [49]Lindner, V. and M.A. Reidy,. Proliferation of smooth muscle cells after vascular injury is inhibited by an antibody against basic fibroblast growth factor[J]. Proc Natl Acad Sci U S A, 1991, 88(9):3739-3743.
    [50]Yamashita, A., et al. Antisense basic fibroblast growth factor alters the time course of mitogen-activated protein kinase in arterialized vein graft remodeling[J]. J Vasc Surg, 2003, 37(4):866-873.
    [51]Morishita, R., et al. A gene therapy strategy using a transcription factor decoy of the E2F binding site inhibits smooth muscle proliferation in vivo[J]. Proc Natl Acad Sci U S A, 1995, 92(13):5855-5859.
    [52]Hillebrands, J., et al. Recipient origin of neointimal vascular smooth muscle cells in cardiac allografts with transplant arteriosclerosis[J]. J Heart Lung Transplant, 2000, 19(12): 1183-1192.
    [53]Hu, Y., et al. Smooth muscle cells in transplant atherosclerotic lesions are originated from recipients, but not bone marrow progenitor cells[J]. Circulation, 2002, 106(14):1834-1839.
    [54]Li, J., et al. Vascular smooth muscle cells of recipient origin mediate intimal expansion after aortic allotransplantation in mice[J]. Am J Pathol, 2001, 158(6):1943-1947.
    [55]Shimizu, K., et al. Host bone-marrow cells are a source of donor intimal smooth-muscle- like cells in murine aortic transplant arteriopathy[J]. Nat Med, 2001, 7(6):738-741.
    [56]Han, C.I., et al. Circulating bone marrow cells can contribute to neointimal formation[J]. J Vasc Res, 2001, 38(2):113-9.
    [57]Sata, M., et al. Hematopoietic stem cells differentiate into vascular cells that participate in the pathogenesis of atherosclerosis[J]. Nat Med, 2002, 8(4):403-409.
    [58]Darnell, J. E., Jr,. STATs and gene regulation[J]. Science, 1997, 277:1630-1635.
    [59]Pan, J., Fukuda, K., et al. Mechanical stretch activates the JAK STAT pathway in ratcardiomyocytes[J]. Circ Res, 1999, 84:1127-1136.
    [60]Xuan,Y. T., Guo, Y., et al. An essential role of the JAK-STAT pathway in ischemic precon- ditioning[J]. Proc Natl Acad Sci USA, 2001, 98:9050-9055.
    [61]Buttmann, M., Berberich-Siebelt.,et al. Interferon-beta is a potent inducer of interferon regulatory factor-1/2-dependent IP-10/CXCL10 expression in primary human endothelial cells[J]. J Vasc Res, 2007, 44:51-60.
    [62]Dumler, I., Kopmann, A.,et al. Urokinase induces activation and formation of Stat4 and Stat1–Stat2 complexes in human vascular smooth muscle cells[J].J Biol Chem, 1999, 274:24059-24065.
    [63]Zurney, J., Howard, et al. Basal expression levels of IFNAR and Jak-STAT components are determinants of cell-type-specific differences in cardiac antiviral responses[J]. J Virol, 2007, 81:13668-13680.
    [64]Lim, C. P., Cao, X. M, et al. Structure, function, and regulation of STAT proteins[J]. Mol Biosyst, 2006, 2:536-550.
    [65]Maritano, D., Sμgrue, et al The STAT3 isoforms and have unique and specific functions[J]. Nat Immunol, 2004, 5:401-409.
    [66]Fischer, P., Hilfiker-Kleiner, et al. Survival pathways in hypertrophy and heart failure: The gp130-STAT3 axis[J]. Basic Res Cardiol, 2007, 102:279-297.
    [67]Hilfiker-Kleiner, D., Hilfiker, et al. Signal transducer and activator of transcription 3 isrequired formyocardial capillary growth, control of interstitialmatrix deposition, andheart protection from ischemic injury[J]. Circ Res, 2004, 95:187-195.
    [68]Hilfiker-Kleiner, D., Hilfiker, et al. Many good reasons to have STAT3 in the heart[J]. Pharmacol Therapeu, 2005, 107:131-137.
    [69]Snyder,M., Huang,et al. Identification of novel direct STAT3 target genes for control of growth and differentiation[J]. J Biol Chem, 2008, 283:3791-3798.
    [70]Wang, R., Cherukuri, et al. Activation of STAT3 sequence-specific DNA binding and transcription by p300/CREB-binding protein-mediated acetylation[J]. J Biol Chem, 2005, 280:11528-11534.
    [71]Kwon, M. C., Koo, et al. Crif1 is a novel transcriptional coactivator of STAT3[J]. EMBO J, 2008, 27:642-653.
    [72]Haspel,R. L., SaldittGeorgieff, et al. The rapid inactivation of nuclear tyrosine phosp- horylated Stat1 depends upon a protein tyrosine phosphatase[J]. EMBO J, 1996, 15:6262-6268.
    [73]Cooney, R. N., et al. Suppressors of cytokine signaling (SOCS): inhibitors of the JAK/STAT pathway[J]. Shock, 2002, 17:83-90.
    [74]Heinrich, P. C., Behrmann, et al. Principles of interleukin (IL)-6-type cytokine signalling and its regulation[J]. Biochem J, 2003, 374:1-20.
    [75]Terrell, A. M., Crisostomo,et al. JAK/STAT/SOCS signaling circuits and associated cytokine-mediated inflammation and hypertrophy in the heart[J]. Shock, 2006, 26: 226-234.
    [76]Barry, S. P., Townsend,et al. Role of the JAK-STAT pathway in myocardial injury[J]. Trends Mol Med, 2007, 13:211-215.
    [77]Dronadula N, Liu Z, Wang C, et al. STAT-3-dependent cytosolic phospholipase A2 expression is required for thrombin-induced vascular smooth muscle cell motility[J ]. J Biol Chem, 2005, 280 (4):3112-3120.
    [78]Hilfiker-Kleiner, D., Kaminski, et al. A cathepsin D-cleaved 16 kDa form of prolactin mediates postpartum cardiomyopathy[J]. Cell, 2007, 128:589-600.
    [79]Chandrasekar, B., Mitchell, et al. Regulation of CCAAT/enhancer binding protein, interleukin-6, interleukin-6 receptor, and gp130 expression during myocardial ischemia/reperfusion[J]. Circulation, 1999, 99:427-433.
    [80]McCormick, J., Barry, et al. Free radical scavenging inhibits STAT phosphorylation following in vivo ischemia/ reperfusion injury[J]. FASEB J, 2006, 20:2115-2117.
    [81]Negoro, S., Kunisada, et al. Activation of JAK/STAT pathway transduces cytoprotective signal in rat acute myocardial infarction[J]. Cardiovasc Res, 2000, 47:797-805.
    [82]Foshay, K., Rodriguez, et al. JAK2/STAT3 directs cardiomyogenesis within murine embryonic stem cells in vitro[J]. Stem Cells, 2005, 23:530-543.
    [83]Oshima, Y., Fujio, et al. STAT3 mediates cardioprotection against ischemia/ reperfusion injury throμgh metallothionein induction in the heart[J]. Cardiovasc Res, 2005, 65:428- 435.
    [84]Hilfiker-Kleiner, D., Hilfiker, et al. Signal transducer and activator of transcription 3 is required formyocardial capillary growth, control of interstitial matrix deposition, and heart protection from ischemic injury[J]. Circ Res, 2004, 95:187-195.
    [85]Boengler, K., Buechert, et al. Cardioprotection by ischemic postconditioning is lost in aged andSTAT3-deficient mice[J]. Circ Res, 2008, 102:131-135.
    [86]Smith, R. M., Suleman, et al. Genetic depletion of cardiac myocyte STAT-3 abolishes classical preconditioning[J]. Cardiovasc Res, 2004, 63:611-616.
    [87]Wang, M. J., Zhang, et al. Endothelial STAT3 plays a critical role in generalized myocardial oinflammatory and proapoptotic signaling[J]. Am J Physiol Heart CircPhysiol, 2007, 293:2101-2108.
    [88]Gross, E. R., Hsu, et al. The JAK/STAT pathway is essential for opioidinduced cardioprotection: JAK2 as a mediator of STAT3, Akt, and GSK-3?[J]. Am J Physiol Heart Circ Physiol, 2006, 291:H827-H834.
    [89]Ueda,K.,Takano, et al. Granulocyte colony stimulating factor directly inhibits myocardial ischemiareperfusion injury throμgh Akt-endothelial NO synthase pathway[J]. Arterioscler Thromb Vasc Biol, 2006, 26:E108-E113.
    [90]Boengler, K., Buechert, et al. Cardioprotection by ischemic postconditioning is lost in aged and STAT3-deficient mice[J]. Circ Res, 2008, 102:131-135.
    [91]Nicolosi, A. C.,Strande, et al. Gadolinium limits myocardial infarction in the rat: dose-response, temporal relations and mechanisms[J]. J Mol Cell Cardio, 2008, 44:345- 351.
    [92]Hwang, Y. Y. C., Shaw, et al. Aldose reductase pathway mediates JAK-STAT signaling: a novel axis in myocardial ischemic injury[J]. FASEB J, 2005, 19:795-797.
    [93]Mascareno, E., El-Shafei, et al. JAK/STAT signaling is associated with cardiac dysfunction during ischemia and reperfusion[J]. Circulation, 2001, 104:325-329.
    [94]Murry, C. E., Jennings,et al. Preconditioning with ischemia: a delay of lethal cell injury in ischemic myocardium[J]. Circulation, 1986, 74:1124-1136.
    [95]Cohen, M. V., Downey, J. M., et al. Adenosine: trigger and mediator of cardioprotection[J]. Basic Res Cardiol, 2008, 103:203-215.
    [96]Shi,Y., Rafiee, et al. Acute cardioprotective effects of erythropoietin in infant rabbits are mediated by activation of protein kinases and potassium channels[J]. Basic Res Cardiol, 2004, 99:173-182.。
    [97]Murphy, E., Steenbergen, C.,et al. Preconditioning: the mitochondrial connection[J]. Annu Rev Physiol, 2007, 69:51-67.
    [98]Bell, R. M., Cave, et al. Pivotal role of NOX-2-containing NADPH oxidase in early ischemic preconditoning[J]. FASEB J, 2005, 19:2037-2039.
    [99]Halestrap, A. P., Clarke, et al. The role of mitochondria in protection of the heart by preconditioning[J]. Biochim Biophys Acta, 2007, 1767:1007-1031.
    [100]Xuan, Y. T., Guo, et al. Endothelial nitric oxide synthase plays an obligatory role in the late phase of ischemic preconditioning by activating the protein kinase C epsilon p44/42 mitogen-activated protein kinase pSer-signal transducers and activators of transcription1/3 pathway[J]. Circulation, 2007, 116:535-544.
    [101]Hattori, R., Maulik, et al. Role of STAT3 in ischemic preconditioning[J]. J Mol CellCardiol, 2001, 33:1929-1936.
    [102]Lecour, S., Suleman, et al. Pharmacological preconditioning with tumor necrosis factor- alpha activates signal transducer and activator of transcription-3 at reperfusion without involving classic prosurvival kinases (Akt and extracellular signal-regulated kinase)[J]. Circulation, 2005, 112:3911-3918.
    [103]Huffman, L. C., Koch, et al. Coronary effluent from a preconditioned heart activates the JAK-STAT pathway and induces cardioprotection in a donor heart[J]. Am J Physiol Heart Circ Physiol, 2008, 294:H257-H262.
    [104]Rowland, R. T., Meng, et al. Cardioadaptation induced by cyclic ischemic precon- ditioning is mediated by translational regulation of de novo protein synthesis[J]. J Surg Res, 1997, 71:155-160.
    [105]Kuzuya, T., Hoshida, et al. Delayed effects of sublethal ischemia on the acquisition of tolerance to ischemia[J]. Circ Res, 1993, 72:1293-1299.
    [106]Singh, K., Balligand, et al. Regulation of cytokine-inducible nitric oxide synthase in cardiac myocytes and microvascular endothelial cells-role of extracellular signal-regulated kinases 1 and 2 (ERK1/ ERK2) and STAT1 alpha[J]. J Biol Chem, 1996, 271:1111- 1117.
    [107]Dawn, B., Xuan, et al. Il-6 plays an obligatory role in late preconditioning via JAK-STAT signaling and upregulation of iNOS and COX-2[J]. Cardiovasc Res, 2004, 64:61-71.
    [108]Oshima, Y., Fujio, et al. STAT3 mediates cardioprotection against ischemia/ reperfusion injury throμgh metallothionein induction in the heart[J]. Cardiovasc Res, 2005, 65:428- 435.
    [109]Neeli I, Liu Z, Dronadula N, et al. An essential role of the Jak-2/ STAT-3/cytosolic phospholipase A2 axis in platelet-derived growth factor BB-induced vascular smooth muscle cell motility[J]. J Biol Chem, 2004, 279(44):46122-46128.
    [110]Kakisis J D., Pradhan S., Cordova A., et al. The role of STAT-3 in the mediation of smooth muscle cell response to cyclic strain[J]. Int J Biochem Cell Biol, 2005, 37(7):1396-1406.
    [111]Madamanchi NR, Li S, Patterson C, et al. Thrombin regulates vascular smooth muscle cell growth and heat shock proteins via the JAK-STAT pathway[J]. J Biol Chem, 2001, 276(22):18915-18924.
    [112]Shu, J., Ren, et al. Increased levels of interleukin-6 and matrix metall oproteinase-9 are of cardiac origin in acute coronary syndrome[J]. Scand Cardiovasc J, 2007, 41:149-154.
    [113]Podewski, E.K., Hilfiker-Kleiner, et al. Alterations in janus kinase (JAK)-signal transducers and activators of transcription (STAT) signaling in patients with end-stagedilated cardiomyopathy[J]. Circulation, 2003, 107:798-802.
    [114]Turkson, J. STAT proteins as novel targets for cancer drμg discovery[J]. Exp Opin Therap Targ, 2004, 8:409-422.
    [115]Xi, S. C., Gooding, et al. In vivo antitumor efficacy of STAT3 blockade using a transcription factor decoy approach: implications for cancer therapy[J]. Oncogene, 2005, 24:970-979.
    [116]Zhang, X., Zhang, et al. STAT3-decoy oligodeoxynucleotide inhibits the growth of human lung cancer via down-regulating its target genes[J]. Oncol Rep, 2007, 17:1377- 1382.
    [117]Sun, X., Zhang, et al. Growth inhibition of human hepatocullular carcinoma cells by blocking STAT3 activation with decoy-ODN[J]. Cancer Lett, 2008, 262:201-213.
    [118]Yu, X. W, Kennedy, et al. JAK2/STAT3,not ERK1/2,mediates interleukin -6- induced activation of inducible nitric-oxide synthase and decrease in contractility of adult ventricular myocytes[J]. J Biol Chem, 2003, 278:16304-16309.
    [119]Espert L, Dusanter-Fourt I, Chelbi-Alix MK.,et al. Negative regulation of the JAK/ STAT:pathway implication in tumorigenesis[J]. Bull Cancer, 2005, 92(10):845- 857.
    [120]Giuseppe Vassalli, David A. Dichek. Gene therapy for arterial thrombosis[J]. Cardiovas Res, 1997, 35:459-469.
    [121]Nabel EG, Plautz G, Boyce FN, et al. Recombinant gene expression in vivo within endothelial cells of the arterial wall[J]. Science, 1989, 244:1342-1344.
    [122]Wilson JM, Birinyi LK, Salomon RN, et al. Implantation of vascular grafts lined with genetically modified endothelial cells[J]. Science, 1989, 244:1344-1346.
    [123]Dichek DA, Neville RF, Zwiebel JA, et al. Seeding of intravascular stents with genetically engineered endothelial cells[J]. Circulation, 1989, 80:1347-1353.
    [124]Nabel EG, Shum L, Pompili VJ, et al. Direct transfer of transforming growth factor b1 gene into arteries stimulates fibrocellular hyperplasia[J]. Proc Natl Acad Sci USA, 1993, 90:10759-10763.
    [125]Nabel EG, Yang Z, Liptay S, et al. Recombinant platelet-derived growth factor b gene expression in porcine arteries induces intimal hyperplasia in vivo[J]. J Clin Invest, 1993, 91:1822-1829.
    [126]Nabel EG, Yang Z-Y, Plautz G, et al. Recombinant fibroblast growth factor-1 promotes intimal hyperplasia and angiogenesis in arteries in vivo[J]. Nature, 1993, 362:844-846.
    [127]Chang MW, Barr E, Seltzer J, et al. Cytostatic gene therapy for vascular proliferative disorders with a constitutively active form of the retinoblastoma gene product[J]. Science,1995, 267:518-522.
    [128]Smith RC, Branellec D, Gorski DH, et al. p21CIP1-mediated inhibition of cell proliferation by overexpression of the gax homeodomain gene[J]. Genes Dev, 1997, 11: 1674-1689.
    [129]Indolfi C, Avvedimento EV, Rapacciuolo A, et al. Inhibition of cellular ras prevents smooth muscle cell proliferation after vascular injury in vivo[J]. Nat Med, 2003, 1:541- 545.
    [130]Ueno H, Yamamoto H, Ito S, et al. Takeshita A. Adenovirus-mediated transfer of a dominant-negative H-ras suppresses neointimal formation in balloon-injured arteries in vivo[J]. Arterioscler Thromb Vasc Biol, 1997, 17:898-904.
    [131]Shi Y, O’Brien JE, Fard A, et al. Adventitial myofibroblasts contribute to neointimal formation in injured porcine coronary arteries[J]. Circulation, 1996, 94:1655-1664.
    [132]Shi Y, O’Brien JE Jr, Fard A, et al. Transforming growth factor-beta 1 expression and myofibroblast formation during arterial repair[J]. Arterioscler Thromb Vasc Biol, 2003, 16:1298-1305.
    [133]Shi Y, Pieniek M, Fard A, et al. Adventitial remodeling after coronary arterial injury[J]. Circulation, 1996, 93:340-348.
    [134]Pepper MS, Montesano R, Mandriota SJ, et al. Angiogenesis: a paradigm for balanced extracellular proteolysis during cell migration and morphogenesis[J]. Enzyme Protein, 1996, 49:138-162.
    [135]Mandriota SJ, Seghezzi G, Vassalli JD, et al. Vascular endothelial growth factor increases urokinase receptor expression in vascular endothelial cells[J]. J Biol Chem, 2006, 270: 9709-9716.
    [136]Bendeck MP, Zempo N, Clowes AW, et al. Smooth muscle cell migration and matrix metalloproteinase expression after arterial injury in the rat[J]. Circ Res, 1994, 75:539- 545.
    [137]Zempo N, Kenagy RD, Au YP, et al. Matrix metalloproteinases of vascular wall cells are increased in balloon-injured rat carotid artery[J]. J Vasc Surg, 2004, 20:209-217.
    [138]Carmeliet P, Moons L, Ploplis V, et al. Impaired arterial neointima formation in mice with disruption of the plasminogen gene[J]. J Clin Invest, 1997, 99:200-208.
    [139]Dichek DA, Anderson J, Kelly AB, et al. Enhanced in vivo antithrombotic effects of endothelial cells expressing recombinant plasminogen activators transduced with retroviral vectors[J]. Circulation, 2005, 93:201-209.
    [140]Dunn PF, Newman KD, Jones M, et al. Seeding of vascular grafts with geneticallymodified endothelial cells: secretion of recombinant TPA results in decreased seeded cell retention in vitro and in vivo[J]. Circulation, 1996, 93:1439-1446.
    [141]Von der Leyen HE, Gibbons GH, Morishita R, et al. Gene therapy inhibiting neointimal vascular lesion:in vivo transfer of endothelial cell nitric oxide synthase gene[J]. Proc Natl Acad Sci USA, 1995, 92:1137-1141.
    [142]Varenne OH, Pislaru S, Gillijns H, et al. Adenoviral gene transfer of human constitutive endothelial nitric oxide synthase to injured coronary arteries[J]. J Am Coll Cardiol, 2002, 29:380A.
    [143]Barath P, Popov A, Michiels R., et al. Nipple balloon catheter[J]. Semin Intervent Cardiol, 1996, 1:43.
    [144]Pavlides GS, Barath P, Maginas A, et al. Intramural drμg delivery by direct injection within the arterial wall:first clinical experience with a novel intracoronary delivery- infiltrator system[J]. Cathet Cardiovasc Diagn, 1997, 41:287-292.
    [145]Massion PB, Feron O, Dessy C,et al. Nitric oxide and cardiac function :ten years after,and continuing[J]. Circ Res, 2003, 93:388-398.
    [146]Gaudino MD, Toesca B, Maggiano B, et al. Localization of nitric oxide synthase typeⅢin the internal thoracic and radial arteries and the great saphenous vein: A comparative immunohistochemical study[J]. J Thorac and Cardiovascular Surg, 2003, 125:1510-1514.
    [147]Yang Y, Vanin EF, Whitt MA, et al. Inducible, high-level production of infections murine leukemia retroviral vector particles pseudotyped with vesicular stomatitis virus G envelope protein[J]. Hum Gene Ther, 1999, 6:1203-1213.
    [148]Liu M-L, Winther BL, Kay MA., et al. Pseudotransduction of hepatocytes by using concentrated pseudotyped vesicular stomatitis virus G glycoprotein(VSV-G)-Moloney murine leukemia virus-derived retrovirus vectors: comparison of VSV-G and amphotropic vectors for hepatic gene transfer[J]. J Virol, 1996, 70:2497-2502.
    [149]Ory DS, Neμgeboren BA, Mulligan RC., et al. A stable human-derived packaging cell line for production of high titer retrovirus/vesicular stomatitis virus G pseudotypes[J]. Proc Natl Acad Sci USA, 1996, 93:11400-11406.
    [150]Flμgelman MY, Jaklitsch MT, Newman KD, et al. Low level in vivo gene transfer into the arterial wall throμgh a perforated balloon catheter[J]. Circulation, 2002, 85:1110-1117.
    [151]Guzman RJ, Lemarchand P, Crystal RG, et al. Efficient and selective adenovirus-mediated gene transfer into vascular neointima[J]. Circulation, 1993, 88:2838-2848.
    [152]Lee SW, Trapnell BC, Rade JJ, et al. In vivo adenoviral vector-mediated gene transfer into balloon-injured rat carotid arteries[J]. Circ Res, 1993, 73:797-807.
    [153]Lemarchand P, Jones M, Yamada I, et al. In vivo gene transfer and expression in normal uninjured blood vessels using replication-deficient recombinant adenovirus vectors[J]. Circ Res, 2005, 72:1132-1138.
    [154]Rome JJ, Shayani V, Newman KD, et al. Adenoviral vector-mediated gene transfer into sheep arteries using a double-balloon catheter[J]. Hum Gene Ther, 1994, 5:1249-1258.
    [155]Kessler PD, Podsakoff GM, Chen X, et al. Gene delivery to skeletal muscle results in sustained expression and systemic delivery of a therapeutic protein[J]. Proc Natl Acad Sci USA, 1996, 93:14082-14087.
    [156]Chapman GD, Lim CS, Gammon RS, et al. Gene transfer into coronary arteries of intact animals with a percutaneous balloon catheter[J]. Circ Res, 2001, 71:27-33.
    [157]Leclerc G, Gal D, Takeshita S, et al. Percutaneous arterial gene transfer in a rabbit model: efficiency in normal and balloon-dilated atherosclerotic arteries[J]. J Clin Invest, 1992, 90: 936-944.
    [158]Isner JM, Pieczek A, Schainfield R, et al. Clinical evidence of angiogenesis after arterial gene transfer of phVEGF165 in patient with ischemic limb[J]. Lancet, 1996, 348: 370-374.
    [159]Isner JM, Walsh K, Rosenfield K, et al. Arterial gene therapy for restenosis[J]. Hum Gene Ther, 2005, 7:989-1011.
    [160]Dzau VJ, Mann MJ, Morishita R, et al. Fusigenic viral liposome for gene therapy in cardiovascular diseases[J]. Proc Natl Acad Sci USA, 1996, 93:11421-11425.
    [161]Cable DG, O’Brien T, Schaff HV, et al. Recombinant endothelial nitric oxide synthase-transduced human saphenous veins: gene therapy to aμgment nitric oxide production in bypass conduits[J]. Circulation, 2003, 96(suppl 9):II-173-II-178.
    [162]Mann MJ, Whittemore AD, Donaldson MC, et al. Preliminary clinical experience with genetic engineering of human vein grafts: evidence for target gene inhibition [Abstract] [J]. Circulation, 2006, 96:1-4.
    [163]Zou Y, Dietrich H, Hu Y, et al. Mouse model of venous bypass artetiosclerosis [J]. Am J Pathol, 1998, 153(4):1301-310.
    [164]Kibbe MR , Nie S, Yoneyama T, et al. Optimization of ex vivo induceble nitric oxide synthase gene transfer to vein grafts[J]. Surgery, 1999, 126(2):323-329.
    [165]Nabel EG, Plautz G, Nabel GJ., et al. Site-specific gene expression in vivo by direct gene transfer into the arterial wall[J]. Science, 1990, 249:1285-1288.
    [166]Rome JJ, Shayani V, Flμgelman MY, et al. Anatomic barriers influence the distribution of in vivo gene transfer into the arterial wall. Modeling with microscopic tracer particles andverification with a recombinant adenoviral vector[J]. Arterioscler Thromb, 2002, 14:148- 161.
    [167]Ross R. The pathogenesis of atherosclerosis: a perspective for the 1990s[J]. Nature, 1993, 362:801-809.
    [168]Barinaga M. Gene therapy for clogged arteries passed test in pigs[J]. Science, 1994, 265: 738
    [169]Rios CD, Ooboshi H, Piegors D, et al. Adenovirus-mediated gene transfer to normal and atherosclerotic arteries. A novel approach[J]. Arterioscler Thromb Vasc Biol, 2006, 15: 2241-2245.
    [170]Scott NA, Cipolla GD, Ross CE, et al. Identification of a potential role for the adventitia in vascularlesion formation after balloon overstretch injury of porcine coronary arteries[J]. Circulation, 1996, 93:2178-2187.
    [171]Mintz GS, Pichard AD, Kent KM, et al. Intravascular ultrasound comparison of restenotic and de novo coronary artery narrowings[J]. Am J Cardiol,2005, 74:1278-1280.
    [172]Kullo IJ, Mozes G, Schwartz RS, et al. Adventitial gene transfer of recombinant endothelial nitric oxide synthase to rabbit carotid arteries alters vascular reactivity[J]. Circulation, 1997, 96:2254-2261.
    [173]Ooboshi H, Welsh MJ, Rios CD, et al. Adenovirus-mediated gene transfer in vivo to cerebral blood vessels and perivascular tissue[J]. Circ Res, 1995, 77:7-13.
    [174]Woody M, Mehdi K, Zipes DP, et al. High efficiency adenovirus-mediated pericardial gene transfer in vivo[J]. J Am Coll Cardiol, 1996, 27(suppl A):31A.
    [175]Mehdi K, Wilensky RL, Baek SH, et al. Efficient adenovirus-mediated perivascular gene transfer and protein delivery by a transvascular injection catheter[J]. J Am Coll Cardiol, 2004, 27(suppl A):164A.
    [176]Reed ML, Wu C, Kneller J, et al. Micromechanical devices for intravascular drμg delivery[J]. J Pharm Sci, 1998, 87(11):1387-394.
    [177]Nikol S, Huehns TY, Krausz E, et al. Needle injection catheter delivery of the gene for an antibacterial agent inhibits neointimal formation[J]. Gene Ther, 1999, 6:737-748.
    [178]Neschis DG, Sattord SD, Hanna AK, et al. Antisense basic fibroblast growth factor gene transfer reduces early intimal thickening in a rabbit femoral artery balloon injury model[J]. J Vasc Surg, 2004, 27:126-134.
    [179] Fulton GJ, Davies MG. Antisense olignucleotide to proto-oncogene c-cmb inhibits the formation of intimal hyperplasia in experimental vein grafts[J]. J Vasc Surg, 1997, 25: 453-463.
    [180]Mann MJ, Gibbons GH, Kernoff RS, et al. Genetic engineering of vein grafts resistant to atherosclerosis[J]. Proc Natl Acad Sci USA, 2005, 92:4502-4506.
    [181]Mann MJ, Gibbons GH, Tsao PS, et al. Cell cycle inhibition preserves endothelial function in genetically engineered rabbit vein grafts[J]. J Clin Invest, 1997, 99:1295- 1301.
    [182]Hector M. Dourron, Gary M. Jacobson, James L. Park, et al. Perivascular gene transfer of NADPH oxidase inhibitor suppresses angioplasty-induced neointimal proliferation of rat carotid artery[J]. Am J Physiol Heart Circ Physio, 2005, 288:H946-H953.
    [183]MARKS A R. Rapamycin:signaling in vascular smooth muscle[J]. Transplant Proc, 2003, 35(3 Suppl):231S-233S.
    [184]Boudn Y V, Kovarik J. JAK/STAT signaling pathways and cancer. Janus kinases/signal transducers and activators of transcription[J]. Neoplasma, 2002, 49:349-355.
    [185]Shibata R, Kai H, Seki Y, et al. Inhibition of STAT3 prevents neointima formation by inhibiting proliferation and promoting apoptosis of neointimal smooth muscle cells[J]. Hum Gene Ther, 2003, 14(7):601-610.
    [186]Melo LG, Ward CA, Dzau VJ., et al. Gene therapies and stem cell therapies[J]. J Cardiovascu Therap, 2007, 3(3):40-66.
    [187]Jun JI, FANG WH, Xia HE, et al. An antisense oligonucleatide targeting against PDGF-βmRNA inhibits proliferation of smooth muscle cells in injured rabbit arteries after denudation[J]. China Modern Med, 2008, 18(22):515-520.
    [188]Schachner T, Steger C, Heiss S, et al. Paclitaxel treatment reduces neointimal hyperplasia in cultured human saphenous veins[J]. Eur Cardio-thora Surg, 2007, 32(10):906-911.
    [189]Kawai-Kowase K, Owens GK. Multiple repressor pathways contribute to phenotypic switching of vascular smooth muscle cells[J]. Am J Physiol Cell Physiol, 2007, 292(9):C 59-69.
    [190]Ehret, G.B., Reichenbach, et al. DNA binding specificity of different STAT proteins. Comparison of in vitro specificity with natural target sites[J]. J Biol Chem, 2001, 276: 6675-6688.
    [191]Chen Z, Han ZC. STAT3: a critical transcription activator in angiogenesis[J]. Med Res, 2008, 28(4):185-200.

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700