粉防己碱对动脉内膜损伤后血管再狭窄的防治作用及机制
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
第一部分建立家兔颈总动脉内膜球囊剥脱损伤后新生内膜增殖和血管重塑动态变化模型
     目的:目前普遍认为,血管内皮损伤诱发的血管平滑肌细胞(vascular smooth muscle cell,VSMC)增殖并且向内膜迁移,随之引起的内膜增殖和血管重塑被认为是经皮冠状动脉腔内成形术(percutaneous transluminal coronary angioplasty,PTCA)后再狭窄(restenosis,RS)的主要病理机制。为探索RS的发病机制并进行干预研究,我们模拟复制家兔颈总动脉内膜损伤后不同时间点的VSMC增殖和血管重塑动态变化模型。
     方法:选用家兔42只行左侧颈总动脉内膜(内皮细胞)球囊剥脱损伤,分别于损伤后1天(day,d)、3d、5d、7d、14d、28d、35d处死家兔,取损伤区域的血管段制备标本HE染色,进行形态学观察。同时应用医学图文分析系统测算损伤后不同时间点各组血管腔面积(lumen area,LA)、内膜和中膜厚度及面积、以及外弹力膜内横截面积(external elastic lamina area,EELA)与对侧未损伤血管比较。
     结果:
     (1)未损伤侧家兔颈总动脉内膜仅见单层内皮细胞;球囊损伤后1d可见动脉内皮剥脱,损伤后3d血管腔内表面可见增殖的VSMC,损伤后5~7d新生内膜(neointima,NI)形成并逐渐增厚,损伤14d以后NI厚度及面积继续增加,至35d达最大,同时细胞外基质(extracellular matrix,ECM)也逐渐增加。
     (2)损伤后3~14d中膜厚度及面积逐渐增加,其中14d中膜面积明显大于未损伤侧,28~35d趋于降低。LA于损伤后5~7d开始减少;14d以后明显小于未损伤侧。损伤后1~7d EELA逐渐增大,至14d达最大,28d后开始回缩。
     结论:
     (1)家兔颈总动脉内膜球囊损伤能较好的模拟RS形成过程,我们模拟复制的RS动态变化模型是成功的,具有合理性和科学性。
     (2)内膜增殖与血管重塑均是RS形成的主要病理机制,RS形成由内膜增殖与血管重塑的失衡所决定,两者协同导致管腔狭窄。
     第二部分
     实验一动脉内膜损伤后VSMC表型转化及P38 MAPK、MKP-1的表达变化
     目的:观察动脉内膜损伤后血管平滑肌细胞(VSMC)表型转化和P38丝裂原激活蛋白激酶(P38 MAPK)、丝裂原激活蛋白激酶磷酸酶-1(MKP-1)表达变化以及它们之间的关系。
     方法:本实验通过建立家兔颈总动脉内膜球囊损伤后再狭窄(restenosis,RS)模型,分别应用免疫组化、免疫印迹(Western blot)和逆转录-聚合酶链反应(RT-PCR)方法检测假损伤组(S组)和损伤后不同时间点血管壁中增殖细胞核抗原(PCNA)、平滑肌α肌动蛋白(SMα-actin)、P38 MAPK蛋白和MKP-1蛋白及其mRNA表达的变化。
     结果:
     (1) S组中膜VSMC及内皮细胞(内膜)PCNA免疫组化染色为阴性;中膜于损伤后1~14d、新生内膜(niointima,NI)于损伤后5~14d PCNA阳性细胞率逐渐增加,以14d达到高峰,28d后开始逐渐减少,但其阳性细胞率仍较高,且各时间点NI中的PCNA阳性细胞率略高于中膜。
     (2) S组中膜SMα-actin染色为阳性,内膜为阴性;中膜SMα-actin阳性表达面积于损伤后1d开始减少,3d最为明显,5d后开始逐渐增加,28d后接近于S组水平。损伤后5~35d NI中SMα-actin均有阳性表达,但阳性表达面积略少于中膜。
     (3) S组中膜P38 MAPK免疫组化染色呈阴性或弱阳性。中膜于损伤后1~14d、NI于损伤后5~14d P38 MAPK阳性表达面积显著增加,以14d达高峰,28d后开始逐渐减少,但至损伤后35d仍明显高于S组,且NI阳性表达面积高于中膜。Western blot检测损伤后1~35d血管壁中P38 MAPK呈持续性高表达,其中以14d升高最为明显,28d后开始逐渐减少。P38 MAPK与PCNA表达变化呈正相关。
     (4) S组中膜及内膜MKP-1免疫组化染色呈弱阳性或阳性。损伤后1~3d中膜阳性表达面积进行性减少,5~7d达到最低值,同时NI呈阴性或弱阳性着色。而损伤后14~28d中膜和NI中MKP-1阳性表达面积略有回升,至35d仍远未回到S组水平,且NI阳性表达面积稍低于中膜。血管壁中MKP-1 mRNA表达变化与其免疫组化结果基本一致,与PCNA表达变化呈负相关。
     结论:
     (1) VSMC增殖能力与其表型转化密切相关。
     (2) P38 MAPK和MKP-1可能参与了血管内膜损伤后VSMC表型转化的信号转导及其调节。
     第二部分
     实验二粉防己碱对血管内膜损伤后VSMC表型转化和P38 MAPK、MKP-1表达的影响
     目的:探讨粉防己碱(tetrandrine,Tet)对血管内膜损伤后血管平滑肌细胞(VSMC)表型转化和P38丝裂原激活蛋白激酶(P38 MAPK)及丝裂原激活蛋白激酶磷酸酶-1(MKP-1)表达的影响。
     方法:本实验通过建立家兔颈总动脉内膜球囊损伤模型,采用HE染色检测28天(day,d)假损伤组(sham-injured group,S组)、损伤组(injured group,Ⅰ组)和损伤+Tet治疗组(injured+Tet-treated group,Tet组)血管形态学改变;又分别使用免疫组化、Western blot和RT-PCR技术检测Ⅰ组和Tet组7d、14d和28d血管壁中增殖细胞核抗原(PCNA)、平滑肌α-肌动蛋白(SMα-actin)、P38 MAPK和MKP-1表达的变化。
     结果:
     (1) S组28d血管壁各层结构完整;Ⅰ组28d新生内膜面积(neointima area,NIA)显著增加,管腔面积(Lumen area,LA)显著缩小;而Tet组新生内膜增殖程度较同期Ⅰ组明显减轻,LA显著增加。
     (2)损伤后7d,Tet组与Ⅰ组比较血管壁中的SMα-actin、PCNA、P38 MAPK和MKP-1表达基本一致,新生内膜增殖程度亦基本相同。Tet组14d和28d,血管壁中的PCNA和P38 MAPK表达均分别显著低于同期Ⅰ组,而MKP-1表达均分别显著高于同期Ⅰ组,且Tet对上述各指标的影响均呈时间依赖性;损伤后14、28d SMα-actin表达均略高于同期Ⅰ组。
     结论:Tet能够不同程度地拮抗内膜损伤后P38 MAPK信号转导途径及其上调MKP-1的负性调节,抑制VSMC表型转化,继而减缓新生内膜增殖。
     第三部分血管内膜损伤后MMP9和TIMP2表达变化及Tet对其表达的影响
     目的:探讨血管内膜损伤后基质金属蛋白酶9(matrix metalloproteinases 9,MMP 9)和基质金属蛋白酶抑制剂2(tissue inhibitors of metalloproteinases 2,TIMP 2)在再狭窄细胞外基质(extracellular matrix,ECM)形成中的作用以及粉防己碱(Tetrandrine,Tet)对MMP9、TIMP2表达和新生内膜增殖的影响。
     方法:制作家兔颈总动脉内膜损伤后血管再狭窄模型,采用免疫组化、逆转录—聚合酶链反应(RT-PCR)和免疫印迹(Western blot)技术,分别检测假损伤组(S组)28 d时和损伤组(Ⅰ组)、损伤+Tet治疗组(Tet组)7 d、14 d、28 d时血管壁中的MMP9、TIMP2 mRNA及其蛋白表达的变化,以及新生内膜增殖的变化。
     结果:
     (1)损伤后7 d组血管中膜MMP9阳性表达面积较高,而14和28 d开始逐渐减少;新生内膜于损伤后7 d时MMP9阳性表达面积较低,14 d显著增加,而28d又明显减少。损伤7 d组中膜、新生内膜TIMP2阳性表达面积较低,而14 d和28 d均显著增加。
     (2)与同期Ⅰ组比较,Tet组7 d中膜及新生内膜中MMP9和TIMP2阳性表达面积无明显变化,而14 d和28 d两者阳性表达面积均分别显著减少,新生内膜增殖程度也有减轻和明显减轻。
     (3)S组动脉壁中MMP9 mRNA及其蛋白水平呈低表达,而TIMP2 mRNA及其蛋白则呈高表达。与S组比较,Ⅰ组7、14和28 d血管壁中MMP9 mRNA和蛋白呈强表达或高表达;Ⅰ组7 d血管壁中TIMP2 mRNA和蛋白呈低表达,而14和28 d又恢复甚至高于S组水平。
     (4)与同期Ⅰ组比较,Tet组7 d血管壁中MMP9、TIMP2 mRNA和蛋白表达均无显著变化;而治疗后14和28 d均分别显著减少,以TIMP2 mRNA及其蛋白减少更为明显。
     结论:
     (1)MMP9和TIMP2不平衡增加和/或比例失衡在血管内膜损伤后ECM形成中具有重要作用。
     (2)Tet可降低内膜损伤后血管壁中MMP9和TIMP2 mRNA及其蛋白的异常高表达,使二者比例趋向平衡,减少ECM大量分泌和积累,从而抑制新生内膜增厚和血管重塑。
Part I
    Dynamic Model of Neointimal Proliferation and Remodeling of Vascular Wall after Balloon Intimal Injury of Rabbit Carotid Common Arteries
    Objective: At present, many studies have reported that proliferation and migration of vascular smooth muscle cell ( VSMC ) in the media, and subsequent neointimal hyperplasia and remodeling of vascular wall is a major pathologic mechanism of vascular restenosis after injury of endothelial cells (intima ) resulted from percutaneous transluminal coronary angioplasty ( PTCA ). To study the mechanism of restenosis for the prevention and treatment following PTCA, we replicated the dynamic models of cellular proliferation and vascular remodeling after intimal denudation of rabbit carotid common arteries at different time points.
    Methods: The intima of left carotid common arteries in 42 rabbits was denudated by balloon injury. Rabbits were respectively killed after injury at different time points ( 1 d, 3d, 5 d, 7d, 14d, 28 d and 35 d ). The injured sections were taken out and made into specimens for HE staining. Morphological change of the arterial wall in injury sides and non-injury sides at different time points was observed by microscopy. In addition, Lumen area, thickness and area of the intima and the media, and cross-sectional area bounded by the external elastic lamina ( EELA) in injury sides and non-injury sides at different time points were measured by computer image analysis technology.
    Results: (1) Single layer endothelium was only showed in non-injury side arteries at different time points. Endothelial cells were denudated at day 1 after balloon injury. The proliferating of VSMC was spotted on the surface of lumen at 3 days after balloon injury. The neointima had been formed and continuously thicken at 5 ~7 days after injury. The neointimal thickness and area as well as extracellular matrix were gradually increased after 14 days, and they were maximal at 35 days. (2) The thickness and area of the media were gradually increased during 3-14 days, in which the medial area significantly increased at 14 days compared with non-injured vessel, and decreased after 28 days. Lumen area initially decreased at 5 ~ 7 days after injury. Lumen area after 14 days was significantly less than that of non-injured vessel. EELA gradually slightly increased at 1 ~ 7 days after injury, it was maximal at 14 days, but it declined gradually after 28 days.
    Conclusion: The models of vascular restenosis established by balloon intimal injury of rabbit carotid common arteries were successfully replicated, and proved to be rational and scientific. Intimal proliferation and vascular remodeling are important pathologic pathogenesis of vascular restenosis. The formation of vascular restenosis was determined by the unbalance of intimal proliferation and vascular remodeling. The both together resulted in lumen narrowing .
    
    Part II Experiment I
     Change of P38 MAKP and MKP-1 in Phenotypic Modulation of Vascular Smooth Muscle Cells after Intimal Injury
    Objective: To explore relationship among phenotypic modulation of vascular smooth muscle cell ( VSMC ) and change of P38 mitogen-activated protein kinase ( P38 MAPK ) as well as mitogen-activated protein kinase phosphatase-1 ( MKP-1 ) after vascular intimal injury.
    Methods: The model of vascular restenosis established by balloon injury of rabbit carotid common arteries was used. Immunohistochemistry, western blot and reverse transcriptase-polymerase chain reaction (RT - PCR) were used to detect the change of proliferation cell nuclear antigen ( PCNA ), smooth muscle α - actin ( SMα - actin ), P38 MAPK protein, and MKP-1 protein as well as its mRNA in vascular wall of sham-injured group and injury group at different time points.
    Results: (1) Immunohistochemical staining of PCNA was negative in the medium and endothelium of sham-injured group. Positive cell percentage of PCNA was gradually increased at 1 ~14 days in the medium and at 5 ~14 days in the neointima after injury, and it reached peak value at 14 days. Positive cell percentage of PCNA was still higher though it was gradually declined after 28 days. Positive cell percentage of PCNA in the neointima was slightly more than that in the medium. (2) Immunohistochemical staining of SMα - actin was positive in the medium, negative in the endothelium of sham- injured artery. Positive expression area of SMα - actin in the medium initially decreased at 1 day and was minimal at 3 days after injury, but it increased gradually after 5 days, and was close to level of sham-injured artery at 28 days after injury. SMα -actin staining was always positive in the neointima at 5 ~35 days after injury, but its positive cells area was slightly less than that in the medium at different time points. (3) Immunohistochemical staining of P38 MAKP was negative or feeble positive in the medium of sham-injured artery. Positive expression area of P38 MAKP was significantly increased in the medium at 1 ~14 days and in the neointima at 5 ~14 days, and it was maximal at 14 days after injury. However, positive expression area of P38 MAKP initially decreased after 28 days, but its positive area was still significantly higher than that in sham-injured artery at 35 days after injury. P38 MAKP positive area in the neointima was higher than that in the medium. P38 MAKP examined by Western blot was sustained increased in vascular wall at 1~35 days after injury, in which it reached peak value at 14 days, initially decreased after 28 days. There were positive relationships between the change of P38 MAKP and PCNA in vascular wall at different time points after injury. (4) Immunohistochemical staining of MKP-1 was feeble positive or positive in the medium and endothelium of sham-injured artery. Its positive expression area in the medium progressing decreased at 1 ~3 days, and it reached lowest value at 5 ~ 7 days. At the same time, staining of MKP-1 was negative or feeble positive in the neointima. However, MKP-1 positive expression area was slightly increased from 14 to 28 days in the medium and the neointima. Its positive expression area was still lower than that sham-injured group at days 35 after injury. MKP-1 positive area in the neointima was slightly lower than that in the medium. The change of MKP-1 mRNA in vascular wall was consistent with immunohistochemical results. There were negative relationships between the change of MKP-1 and PCNA in the vascular wall at different time point after injury.
    Conclusion: There was close relationship between phenotypic modulation and proliferating ability of VSMC. P38 MAPK and MKP-1 could participate in phenotypic modulation of VSMC and its regulation after intimal injury.
    
    Part II
    Experiment II
    Effects of Tetrandrine on Phenotypic Modulation of Vascular Smooth Muscle Cells and Expression of P38 MAPK as well as MKP-1 after Intimal Injury of Rabbit Carotid Arteries
    Objective: To study the effects of tetrandrine ( Tet ) on phenotypic modulation of vascular smooth muscle cells ( VSMC ) and P38 mitogen-activated protein kinase ( P38 MAPK) as well as mitogen - activated protein kinase phosphotase-1 ( MKP-1 ) after vascular intimal injury.
    Methods: The model of vascular restenosis established by balloon intimal injury of rabbit carotid common arteries was used. HE staining was used to analysis vascular morphology change of sham-injured, injured and Tet-treated group at 28 days. Immunohistochemistry, Western blot and RT - PCR were respectively used to detect the expression change of proliferation cell nuclear antigen ( PCNA ), smooth muscle α-actin ( SMα-actin), P38 MAPK and MKP-1 of vascular wall in injured group and Tet group at 7, 14 and 28 days after injury. Results: (1) The each layer structure in vascular wall was intact in sham-injured group arteries at day 28. The neointimal area was significantly increased and the lumen area notably decreased in injured group at day 28. Compared with injured-group, the neointima proliferation degree was evidently decreased, and lumen area was markedly increased in Tet treated-group at day 28. (2) Compared with injured-group, the expression of PCNA, SMα-actin, P38 MAPK and MKP-1 in vascular wall of Tet group had no difference and the neointimal proliferation degree was also basically same at day 7 after injury. The expression of PCNA and P38 MAKP in Tet group was all obviously lower than that in injured-group, whereas the expression of MKP-1 in Tet group was evidently higher than that in injured-group in vascular wall at days 14 and 28 after injury, and all had time dependent manner. The expression of SMα-actin in Tet group was slightly higher than that in injured-group at days 14 and 28 after injury. Conclusions: Tet could reduce neointimal proliferation by inhibiting phenotypic modulation and P38 MAPK signal transduction pathway as well as its negative regulation of VSMC after intimal injury.
    
    Part III
    Expressional Change of MMP9, TIMP2 and Effects of Tet on Their Expression in Vascular Wall after Intimal Injury of Rabbit Carotid
    Objective: To investigate (1) the effect of matrix metalloproteinases - 9 ( MMP9 ) and tissue inhibitors of metalloproteinases - 2 ( TIMP2 ) in extracellular matrix ( ECM ) formation of vascular restenosis; (2) the effects of tetrandrine (Tet) on MMP9, TIMP2 expression and vascular intimal proliferation. Methods: The model of restenosis established by balloon injury of rabbit carotid common arteries was performed. Immunohistochemistry, reverse transcriptase - polymerase chain reaction ( RT - PCR ) and Western blot were respectively used to detect the change of MMP9, TIMP2 mRNA and their protein expression in vascular wall as well as intimal proliferation in sham-injured group ( S group ) at 28 days, in injured group (I group ) and Tet-treated group ( Tet group ) at 7, 14 and 28 days after balloon injury. Results: (1) Positive expression area of MMP9 was higher in the medium at 7 days, and that it was gradually decreased at 14 and 28 days after injury; MMP9 positive expression area was lower relatively in the neointima at 7 days, and that it was significantly increased at 14 days, but it was obviously decreased at 28 days after injury. Positive expression area of TIMP2 in the medium and neointimal was lower at 7 days, and that it was significantly increased at 14 and 28 days after injury. (2) Compared with I group of same period, positive expression area of MMP9 and TIMP2 in the medium and neointimal of Tet group had no evident difference at 7 days, and that they were all respectively significantly decreased at 14 and 28 days after injury. At the same time, neointimal proliferation degree was also relieved and evidently relieved at 14 and 28 days after injury. (3) Expression of MMP9 mRNA and its protein was lower, and that the expression of TIMP2 mRNA and its protein was higher in vascular wall of S group. Compared with S group, expression of MMP9 mRNA and its protein in vascular wall of I group was stronger or higher at 7, 14, and 28 days. Expression of TIMP2 mRNA and its protein in vascular wall of I group was lower than that of S group at 7 days, and that it come back to or was higher than that of S group at 14 and 28 d. (4) Compared with I group of the same period, expression level of MMP9 mRNA and TIMP2 mRNA as well as their protein in vascular wall of Tet group had all no significant difference at 7 days, and that their expression of Tet group all had been significantly decreased at 14 days and 28 days after injury, in witch decrease of TIMP2 was more than that of MMP9. Conclusions: The lopsided increase and/or imbalance of MMP9 and TIMP2 expression played important role in ECM formation after vascular intimal injury. Tet could reduce abnormal higher expression of MMP9 and TIMP2 in vascular wall after intimal injury, ensure the equilibrium of MMP9 / TIMP2, and retard the accumulation and secretion of ECM, thereby inhibit neointimal hyperplasia and vascular remodeling.
引文
[1] Nishioka T. The incidence and mechanism of restenosis after PTCA[J]. Nippon Rinsho, 2003, 61(Suppl 4): 511-517.
    [2] Lafont A, Guzman LA, Whitlow PL, et al. Restenosis after experimental angioplasty. Intimal, medial and adventitial changes associated with constrictive remodeling[J]. Circ Res, 1995, 76(6): 996-1102.
    [3] Faxon DP, Williamo DO, Yeh W, et al. Improved in-hospital outcome with expanded use of coronary stents: results from the NHLBI dynamic registry [J]. J Am Coll Cardiol, 1999, 33 (suppl A):91A.
    [4] Lowe HC, Oesterle SN, Khachigian LM. Coronary in-stent restenosis: current status and future strategies [J]. J Am Coll Cardiol, 2002, 39 ( 2 ): 183 - 193.
    
    [5] Austin GE, Rathff NB, Hollinan J, et al. Intimal proliferation of smooth muscle cells as an explanation for recurrent coronary artery restenosis after percutaneous transluminal coronary angioplasty [J]. J Am Cell Cardiol, 1985, 6 (2): 369 - 375.
    
    [6] Kovach JA, Mintz GS, Kent KM, et al. Serial intravascular ultrasound studies indicate that chronic recoil is an important mechanism of restenosis following transcatheter therapy [J]. J Am Coll Cardiol, 1993,21 (suppl A) : 484A.
    [7] Mintz GS, Kovach JA, Saturnino PJ, et al. Geometric remodeling is the predominant mechanism of late lumen loss after coronary angioplasty [J]. Circulation, 1993, 88 : ( suppl 1 ) : I - 654.
    [8] Post MJ, Brost C, Kuntz RE. The relative importance of arterial remodelling compared with intima hyperplasia in lumen renarrowing after ballon angioplasty: a study in the normal rabbit and the hypercholesterolemic yucatan micropg [J]. Circulation, 1994, 89 : 2816 - 2821.
    [9] Feng AN, Chen YL, Chen YT, et al.Red wine inhibits monocyte chemotactic protein-1 expression and modestly reduces neointimal hyperplasia after balloon injury in cholesterol fed rabbits [J]. Circulation, 1999, 100 : 2254 - 2265.
    
    [10] Mark Fenton, Steve Barker, David J, et al.Cellular senescence after single and repeated balloon catheter denudations of rabbit carotid arteries [J]. Arterioscler Thromb Vasc Biol, 2001,21:220 -226.
    [11] Sims FH. A comparison of structural features of the walls of coronary arteries from 10 different species [J]. Pathology, 1989, 21 ( 2): 115 -124.
    [12] Sauvage LR, Berger KE, wood SJ, et al. Interspecies healing of porous arterial prostheses: observation from 1960 to 1974 [J]. Arch Surg, 1974, 109 (5): 698 - 705.
    
    [13] Myllarniemi M, Frosen J, Calderon Ramirez LG, et al. Selective tyrosine kinase inhibitor for the platelet-derived growth factor receptor in vitro inhibits smooth muscle cell proliferation after reinjury of arterial intima in vivo [J]. Cardiovasc Drugs Therp, 1999, 13 (2): 159 - 168.
    [14] Luan RH, Jia GL, Li W, et al. Inhibition of vascular smooth muscle cell proliferation by specific c-myc mRNA cleaving hammerhead ribozyme[J]. J Fourth Mil Med Univ, 2003, 24(6): 517-524.
    [15] Schwartz RS, Henry TD. Pathophysiology of coronary artery restenosis[J]. Rev Cardiovasc Med, 2002, 3(5): S4-S9.
    [16] Wiegman PJ, Barry WL, McPherson JA, et al. All-trans-retinoic acid limits restenosis after balloon angioplasty in the focally atherosclerotic rabbit: a favorable effect on vessel remodeling[J]. Arterioscler Thormb Vase Biol, 2000, 20(1): 89-95.
    [17] Wu XJ, Huang L, Song DL, et al. Effects of endothelial cell growth states on the proliferation and migration of vascular smooth muscle cells in vitro[J]. Sheng Li Xue Bao, 2003, 55: 554-559.
    [18] Van Exven L, Uelema E, Bos AN, et al. Thrombogenicity and intimal hyperplasia after conventional and thermal balloon dilation in normal rabbit iliac arteries[J]. Vase Res, 1992, 29: 426-434.
    [19] Mukherjee D, Yadav JS. Percutaneous treatment for carotid stenosis[J]. Cardiol Gin, 2002, 20: 589-597.
    [20] Keamey M, Pieczek A, Haley L, et al. Histopathology of in-stent restenosis in patients with peripheral artery disease[J]. Circulation, 1997, 95(8): 1998-2004.
    [21] Rutanen J, Puhakka H, Yla-Herttuala S. Post-intervention vessel remodeling[J]. Gene Ther, 2002, 9(22): 1487-1491.
    [22] Currier JW, Faxon DP. Restenosis after percutaneous transluminal coronary angioplasty: have we been aiming at the wrong target[J]? J Am Coll Cardiol, 1995, 25(2): 516-518.
    [23] Costa MA, Kozuma K, Gaster AI. Three dimensional intravascular ultrasonic assessment of the local mechanism of restenosis after balloon angioplasty[J]. Heart, 2001, 85(1): 73-79.
    [24] Webster WS, Bishop SP, Geer JC. Experimental aortic intimal thickening. 1. Morphology and source of intimal cells[J]. Am J Pathol, 1974, 76(2): 245-257.
    [25] Lowe HC, Oesterle SN, Khachigian LM. Coronary in-stent restenosis: current status and future strategies[J]. JAm Coll Cardiol, 2002, 39(2): 183-193.
    [26] Li Y, Li ZL, Jia GL, et al. Short-term protective effects of the heparincoated wiktor-1 stent on atherosclerotic coronary artery and its clinical significance[J]. J Fourth Mil Med Univ, 2001, 22(12): 1104-1107.
    [1] Austin GE, Rathff NB, Hollman J, et al. Intimal proliferation of smooth muscle cells as an explanation for recurrent coronary artery restenosis after percutaneous transluminal coronary angioplasty. J Am Cell Cardiol, 1985, 6(2): 369-375.
    [2] Gruberg L, Waksman R, Satler LF, et al. Novel approaches for the prevention of restenosis. Expert Opin Investig Drugs. 2000, 9(11): 2555-2578.
    [3] Kumar MS, Owens GK. Combinatorial control of smooth muscle specific gene expression. Arterioscler Thromb Vasc Biol, 2003, 23(5): 737-747.
    [4] Regan CP, Adam P J, Madsen CS, et al. Molecular mechanisms of decreased smooth muscle differentiation marker expression after vascular injury. J Clin Invest, 2000, 106(9): 1139-1147.
    [5] Crook MF, Akyurek LM. Gene transfer strategies to inhibit neointima formation. Trends Cardiovasc Med, 2003, 13(3): 102-106.
    [6] Lavigne MC, Ramwell PW, Clarke P. Growth and phenotypic characterization of porcine coronary artery smooth muscle cells. In Vitro Cell Dev Biol Anim, 1999, 35(3): 136-143.
    [7] 李琦,温进坤,韩梅.反义c-jun 表达质粒对血管平滑肌细胸表型转化标志的影响.中国动脉硬化杂志,2001,9:69-71.
    [8] Haapasalo HK, Sallinen PK, Helen PT, et al. Comparison of three quantitation methods for PCNA immunostaining: applicability and relation to survival in 83 astrocytic neoplasms. J Pathol, 1993, 171(3): 207-214.
    [9] Lange-Carter CA, Pleiman CM, Gardner AM, et al. A divergence in the MAP kinase regulatory network defined by MEK kinase and raf. Science, 1993, 260(5106): 315-319.
    [10] Price MA, Cruzalegui FH, Treisman R. The P38 and ERK MAP kinase pathways cooperate to activate Ternary Complex Factors and c-fos transcription in response to UV light. EMBO J. 1996, 15(23): 6552-6563.
    [11] Geng Y, Valbracht J, Lotz M. Selective activation of the mitogen-activated protein kinase subgroups c-jun NH_2 terminal kinase and P38 by IL-1 and TNF in human articular chondrocytes. J Clin Invest, 1996, 98: 2425-2430.
    [12] Morooka T, Nishida E. Requirement of P38 mitogen-activated protein kinase for neuronal differentiation in PC12 cell. J Biol Chem., 1998, 273(38): 24285-24288.
    [13] Zetseer A, Gredinger E, Bengal E. P38 mitogen-activated protein kinase pathway promotes skeletal muscle differentiation. J Biol Chem., 1999, 274(8): 5193-5200.
    [14] Metzler B, Li CH, Hu YH, et al. LDL stimulates mitogen-activated protein kinase phosphatase-1 expression, independent of LDL receptors, in vascular smooth muscle cells. Arterioscler Thromb Vasc Biol. 1999, 19: 1862-1871.
    [15] Brondello JM, Brunet A, Pouyssegur J, et al. The dual specificity mitogen-activated protein kinase phosphatase-1 and-2 are induced by the P42/P44 MAPK cascade. J Biol Chem, 1997, 272(2): 1368-1376.
    [16] Shehla P, Rajah S, William AF, et al. MKP-1-indueed dephosphorylation of extracellular signal-regulated kinase is essential for triggering nitric oxide-induced apoptosis in human breast cancer cell lines. Cancer Research, 2003, 15: 8853-8860.
    [17] Feldman LJ, Mazighi M, Scheuble A, et al. Differential expression of matrix metalloproteinases after stent implantation and balloon angioplasty in the hypercholesterolemic rabbit. Circulation, 2001, 103: 3117-3122.
    [18] Meena SK, Gary KO. Combinatorial control of smooth muscle specific gene expression. Arterioscier, Thrombosis, Vase Biol. 2003, 23: 737-747.
    [19] Hungerford JE. Developmental biology of the vascular smooth muscle cell: building a multilayered vessel wall. J Vase Res, 1999, 36(1): 2-27.
    [20] Seki Y, Kai H, Shibata R, et al. Role of the JAK/STAT pathway in rat carotid artery remodeling after vascular injury. Circ Res, 2000, 87: 12-18.
    [21] Lincoln TM, Dey N, Sellak H. cGMP-dependent protein kinase signaling mechanisms in smooth muscle: from the regulation of tone to gene expression. J Appl Physiol, 2001, 91: 1421-1430.
    [22] Hayashi K, Takahashi M, Nishida W, et al. Phenotypic modulation of vascular smooth muscle cells induced by unsaturated lysophosphatidic acids. Circ Res, 2001, 89(3): 251- 258.
    [23] Ohashi N, Matsumori A, Furukawa Y, et al. Role of P38 mitogen-activated protein kinase in neointimal hyperplasia after vascular injury. Arterioscler Thromb Vase Biol, 2000, 20(12): 2521-2526.
    [24] Haisong J, Sandhya N, Charles F, et al. Sustained activation of P38 MAPK mitogenactivated protein kinase contributes to the vascular response to injury. Pharmacology and Experimental Therapeutics. 2002, 301(1): 15-20.
    [25] Lai KY, Wang H, Lee WS, et al. Mitogen-activated protein kinase phosphatase-1 in rat arterial smooth muscle cell proliferation. Clin Invest, 1996, 98: 1560-1567.
    [1] Durand E, Mallat Z, Addad F, et al. Time courses of apoptosis and cell proliferation and their relationship to arterial remodeling and restenosis after angioplasty in an atherosclerotic rabbit model[J]. Am Coll Cardiol, 2002, 39(10): 1680-1685.
    [2] Schoenhagen P, Vince DG, Ziada KM, et al. Relation off matrix-metalloproteinase 3 found in coronary lesion samples retrieved by directional coronary atheroctomy to intravascular ultrasound observations on coronary remodeling[J]. Am J Cardiol, 2002, 89(12): 1354-1359.
    [3] Metzler B, Li CH, Hu YH, et al. LDL stimulates mitogen-activated protein kinase phosphatase-1 expression, independent of LDL receptors, in vascular smooth muscle eells[J]. Arterioscler Thromb Vase Biol, 1999, 19: 1862-1871.
    [4] Yamaguchi H, Igarashi M, Hirata A, et al. Characterization of platelet-derived growth factor induced P38 mitogen-activated protein kinase activation in vascular smooth muscle cells[J]. Eur J Clin Invest, 2001, 31(8): 672-680.
    [5] Samer MG, Philip H, Neal AS. Overview of therapies for prevention of restenosis after coronary interventions[J]. Pharmacol Ther, 2001, 92: 165-178.
    [6] Zhang YH, Fang LH, Ku BS. Fangchinoline inhibits rat aortic vascular smooth muscle cell proliferation and cell cycle progression through inhibition of ERK1/2 activation and c-fos expression[J]. Biochem Pharmacol, 2003, 66(9): 1853-1860.
    [7] Qianjia Qing. Cardiovascular pharmacological effects of bisbenzylisoquinoline alkaloid derivatives[J]. Acta Pharmacol Sin, 2002, 23(12): 1086-1092.
    [8] Xin Zhao, Xiangyu Cui, Baoqiong Chen, et al. Tetrandrine, a bisbenzylisoquinoline alkaloid from Chinese herb Radix, augmented the hypnotic effect of pentobarbital through serotonergic system[J]. European Journal of Pharmacology, 2004, 506(2): 101-105.
    [9] 熊一力,张露,姚伟星.Tet抑制血管平滑肌细胞增殖及对HSP70和p53表达的影响[J].中国应用心理学杂志,1998,14(2):109-113.
    [10] 李佃贵,李俊峡,李振斌,等.Tet对血管内膜剥脱后再狭窄的预防作用及其分子机制研究[J].河北医科大学学报,2002,23(2):68-70.
    [11] Yu XC, Wu S, Chen CF, et al. Antihypertensive and anti-arrhythmic effects of an extract of Radix Stephaniae Tetrandrae in the rat. J Pharm Pharmacol. 2004, 56(1): 115-122.
    [12] Fang DC, Jiang MX. A new calcium antagonist of chinese medicinal origin: Tetrandrine[J]. J Hypertension, 1986, 4(supply 6): 150-152.
    [13] Liu QY, Karpinski E, Pang PK. Tetrandrine inhibits both T and L calcium channel currents in ventricular cells[J]. J Cardiovasc Pharmacol, 1992, 20(4): 513-519.
    [14] Wu SN, Jan CR, Li HF, et al. Stimulation of large-conductance Ca~(2+)-activated K~+ channels by Evans blue in cultured endothelial cells of human umbilical veins[J]. Biochem Biophys Res Commun, 1999, 254: 666-674.
    [15] Qiu Y, Li YY, Li SG, et al. Effect of Qingyitang on activity of intracellular Ca2+-Mg2+- ATPase in rats with acute pancreatitis. World J Gastroenterol, 2004, 10(1): 100-104.
    [16] 王中峰,开丽,肖欣荣.粉防己碱对肺动脉平滑肌细胞钙激活钾通道的双重作用[J].中国药理学报,1999,20(3):253-256.
    [17] Shengnan Wu, Huifang Li, Yighing Lo. Characterization of tetrandrine-induced inhibition of large-conductance calcium-activated potassium channels in a human endothelial cell line(HUV-EC-C)[J]. Pharmacology and Experimental Therapeutics, 2000, 292(1): 188-195.
    [18] 敖行,何华英,张乐之.粉防己碱对心血管的药理作用研究进展[J].中国药业,2000,9(7),63-65.
    [19] 李风前,陆彬.Tet制剂研究进展[J].中草药,1999,6(30):475-477.
    [20] Berridge MJ. Inositol triphosphate and calcium signaling[J]. Nature, 1993, 361(6410): 315-325.
    [21] Yusufi ANK, Jingfei C, Thompson MA, et al. Differential mechanisms of Ca~(2+) release from vascular smooth muscle cell microsomes[J]. Exp Biol Med, 2002, 227(1): 36-44.
    [22] King VF, Garcia ML, Himmel D, et al. Interaction of tetrandrine with slowly inactivating calcium channels. Characterization of calcium channel modulation by an alkaloid of Chinese medicinal herb origin[J]. J Biol Chem, 1988, 263(5): 2238-2244.
    [23] Liu QY, Li B, Gang JM, et al. Tetrandrine, a Ca~(2+) antagonist: effects and mechanisms of action in vascular smooth muscle cells[J]. J Pharmacol Exp Ther, 1995, 273(1): 32-39.
    [24] Kwan CY, Ma FM, Hui SC. Inhibition of endothelium-dependent vascular relaxation by tetrandrine[J]. Life Sci, 1999, 64(25): 2391-2400.
    [25] Brondello JM, Brunet A, Pouyssegur J, et al. The dual specificity mitogen-activated protein kinase phosphatase-1 and-2 are induced by the P42/P44 MAPK cascade[J]. J Biol Chem, 1997, 272(2): 1368-1376.
    [26] Nishizuka Y. Protein kinase C and lipid signaling for sustained cellular response[J]. FASEB, 1995, 9(7): 484-496.
    [27] Bowling N, Walsh RA, Song G, et al. Increased protein kinase C activity and expression of Ca~(2+)-sensitive isoforms in the failing human heart[J]. Circulation, 1999, 99(3): 384-391.
    [28] Fang LH, Zhang YH, Ma JJ, et al. Inhibitory effects of tetrandrine on the serum-and platelet-derived growth factor-BB-induced proliferation of rat aortic smooth muscle cells through inhibition of cell cycle progression, DNA synthesis, ERK1/2 activation and c-fos expression[J]. Atherosclerosis, 2004, 174(2): 215-223.
    [1] Casscells W, Engler D, Willerson JT, et al. Mechanism of restenosis[J]. Tex Heart Inst. 1994, 21(1): 68-77
    [2] 吴永全,汪丽蕙.基质金属蛋白酶与血管成形术后再狭窄[J].中国介人心脏病学杂志.1997,5(2):92-93
    [3] 方达超,江明性.粉防己碱抗钙作用研究[J].中华医学杂志,1983,63(12):772-774
    [4] Liu QY, Karpinski E, Pang KT. Tetrandrine inhibits both T and L calcium channel currents in ventricular cells[J]. J Vardiovasc Pharmacol, 1992, 20(4): 513-519
    [5] Qiu Y, Li YY, Li SG, et al. Effect of Qingyitang on activity of intracellular Ca2+-Mg2+- ATPase in rats with acute pancreatitis. World J Gastroenterol, 2004, 10(1): 100-104.
    [6] 王中峰,开丽,肖欣荣.粉防己碱对大鼠肺动脉平滑肌细胞小电导钙激活钾通道的作用[J].中国药理学报,2000,16(2):199-201
    [7] 熊一力,张露,姚伟星.粉防己碱抑制血管平滑肌细胞增殖及对HSP70和p53表达的影响[J].中国应用生理学杂志,1998,14(2):109-112
    [8] Yao Xue Xue Bao. Depressive effect of tetrandrine on collagen synthesis in vascular smooth muscle cells. 2001, 36(7): 481-484.
    [9] Feldman LJ, Mazighi M, Scheuble A, et al. Differential expression of matrix metalloproteinases after stent implantation and balloon angioplasty in the hypercholesterolemic rabbit. Circulation, 2001, 103: 3117-3122.
    [10] Dollery CM, McEwan JR, Henney AM. Matrix metalloproteinases and cardiovascular disease[J]. Circ Res, 1995, 77(5): 863-868.
    [11] 吴永全,汪丽蕙.MMP2和MMP9在损伤血管中的原位表达[J].中国介人心脏病学杂志,1998,6(2):89-91
    [12] 周秀霞,温进坤,韩梅等.血管再狭窄发生过程中MMP2基因与胶原转换的动态变化[J].中国生物化学与分子生物学学报,1999,15(6):997-1001
    [13] Vallee BL, Auld DS. Zinc coordination, function and structure of zinc enzymes and other proteins[J]. Biochemistry, 1990, 29(24): 5647-5659
    [14] 李玉瑞主编 细胞间质的生物化学及研究方法.第一版,北京:人民卫生出版社,1988,26-39
    1. Leon MB, Baim DS, Popma JJ, et al. A clinical trial comparing three antithrombotic drug regimens after coronary artery stenting: Stent Anticoagulation Restenosis Study Investigators. N Engl J Med, 1998, 339: 1665-1671
    2. Heidland UE, Klimek WJ, Michel CJ, et al. Einfluss einer intrakoronaren DipyridamolApplikation auf die Inzidenz der Restenose nach PTCA. Eine prospektiv randomisierte. Untersuchung. Med Klin, 1998, 93: 579-584
    3. Kunishima T, Musha H, Eto, F, et al. A randomized trial of aspirin versus cilostazol therapy after successful coronary stent implantation. Clin Ther 1997, 19: 1058-1066
    4. Tsuchikane E, Fukuhara A, Kobayashi T, et al. Impact of cilostazol on restenosis after percutaneous coronary balloon angioplasty. Circulation, 1999, 100: 21-26
    5. Osinski MT, Schror K. Inhibition of platelet-derived mitogenesis by phosphodiesterase 3 inhibitors: role of protein kinase A in smooth muscle cell mitogenesis. Biochemical Pharmacology, 2000, 60: 381-387
    6. Savage MP, Goldberg S, Bove, AA, et al. Effect of thromboxane A2 blockade on clinical outcome and restenosis after successful coronary angioplasty: Multi-Hospital Eastern Atlantic Restenosis Trial(M-HEART Ⅱ). Circulation, 1995, 92: 3194-3200
    7. Gershlick AH, Spriggins D, Davies SW, et al. Failure of epoprostenol(prostacyclin, PGI2) to inhibit platelet aggregation and to prevent restenosis after coronary angioplasty: results of a randomised placebo controlled trial. Br Heart J, 1994, 71: 7-15
    8. Darius H, Nixdorff U, Zander J, et al. Effects of ciprostene on restenosis rate during therapeutic transluminal coronary angioplasty. Agents Actions Suppl, 1992, 37: 305-311.
    9. Samer MG, Philip H, Neal AS. Overview of therapies for prevention of restenosis after coronary interventions. Pharmacol Ther, 2001, 92: 165-178
    10. Lablanche JM, McFadden EP, Meneveau N, et al. Effect of nadroparin, a low-molecular-weight heparin, on clinical and angiographic restenosis after coronary balloon angioplasty: the FACT study. Circulation, 1997, 96: 3396-3402
    11. Serruys PW, Herrman JP, Simon R, et al. A comparison of hirudin with hcparin in the prevention of restenosis after coronary angioplasty. Helvetica Investigators. N Engl J Med, 1995, 333: 757-763
    12. Xing C, Song R, Miranda GM, ct al. Hirulog-like peptide reduces restenosis and expression of tissue factor and transforming growth factor-β in carotid artery of athcrosclerotic rabbits. Atherosclerosis, 2003, 169: 31-40.
    13. Berg JM, Kelder JC, Suttorp MJ, et al. Effect of coumarins started before coronary angioplasty on acute complications and long-term follow-up: a randomized trial. Circulation, 2000, 102: 386-391
    14. Hillegass WB, Ohman EM, Leimberger JD, et al. A meta-analysis of randomized trials of calcium antagonists to reduce restenosis after coronary angioplasty. Am J Cardiol, 1994, 73: 835-839
    15. Faxon DP. Effect of high dose angiotensin-converting enzyme inhibition on restenosis: final results of the MARCATOR Study, a multicenter, double-blind, placebo-controlled trial of cilazapril. The Multicenter American Research Trial With Cilazapril ARer Angioplasty to Prevent Transluminal Coronary Obstruction and Restenosis(MAR-CATOR) Study Group. J Am Coll Cardiol, 1995, 25: 362-369
    16. Jinno T, Iwai M, Li Z, et al. Calcium channel blocker azelnidipine enhances vascular protective effects of AT1 receptor blocker Olmesartan. Hypertension, 2004, 43(2): 263
    17. Bartune kL. Endoteling a rechemicka choroba srdecni. Vnitr Lek, 2000: 46: 49-53
    18. Lablanche JM, Grollier G, Lusson JR, et al. Effect of the direct nitric oxide donors linsidomine and molsidomine on angiographic restenosis after coronary balloon angioplasty. The ACCORD Study. Angioplastic Coronaire Corvasal Diltiazem. Circulation, 1997, 95: 83-89
    19. Serruys PW, Klein W, Tijssen JP, et al. Evaluation of ketanserin in the prevention of restenosis after percutaneous transluminal coronary angioplasty. A multicenter randomized double-blind placebo-controlled trial. Circulation, 1993, 88: 1588-1601
    20. Currier JW, Faxon DP. Restenosis after percutaneous transluminal coronary angioplasty: have we been aiming at the wrong target? J Am Coll Cardi-1, 1997, 25(2): 516
    21. Tamai H, Katoh O, Suzuki S, et al. Impact of tranilast on restenosis after coronary angioplasty: tranilast restenosis following angioplasty trial(TREAT). Am Heart J, 1999, 138: 968-975
    22. Ishiwata S, Verheye S, Robinson KA, et al. Inhibition of neointima formation by tranilast in pig coronary arteries after balloon angioplasty and stent implantation. J Am Coll Cardiol, 2000, 35: 1331-1337
    23. Walter DH, Schachinger V, Eisner M, et al. Effect of statin therapy on restenosis after coronary stent implantation. Am J Cardiol, 2000, 85: 962-968
    24. Gallo R, Padureen A, Jayamman T, et al. Inhibition of intimal thickening after balloon angioplasty in porcine coronary arteries by targeting regulators of the cell cycle. Circulation, 1999, 16: 2164-2170
    25. Axel DL, Ktmert W, Goggelmann C, et al. Paclitaxe linhibits arterial smooth muscle cell proliferation and migration in vitro and in vivo using local drug delivery. Circulation, 1997, 96: 636-645
    26. Sierevogel MJ, Pasterkamp G, Velema E, et al. Oral matrix metalloproteinases inhibition and arterial remodeling after balloon dilation. Circulation, 2001, 102: 302-307
    27. Tardif JC, Cote G, Lesperanee J, et al. Probueol and multi-vitamins in the prevention of restenosis after coronary angioplasty. Multivitamins and Probueol Study Group. N Engl J Med, 1997, 337: 365-372
    28. Yokoi H, Daida H, Kuwabara Y, et al. Effectiveness of an antioxidant in preventing restenosis after percutaneous transluminal coronary angioplasty: the Probucol Angioplasty Restenosis Trial. J Am Coll Cardiol, 1997, 30: 855-862
    29. Arakawa E. Cardiovascular Disease: L-ascorbic acid may ameliorate vascular dysfunction. Health and Medicine Week, 2004, 5: 148
    30. Serruys PW, Foley DP, Hofling B, et al. Carvedilol for prevention of restenosis after directional coronary atherectomy: final results of the European Carvedilol Atherectomy Restenosis (EUROCARE) trial. Circulation, 2000, 101: 1512-1518
    31. Neumann FJ, Kastrati A, Meithke T, et al. Effect of antibiotic treatment against Chlamydia pneumoniae with roxithromycin on neointimal proliferation after coronary stent placement. J Am Coll Cardiol, 2001, 37(suppl. 1): 9A
    32. Leaf A, Jorgensen MB, Jacobs AK, et al. Do fish oils prevent restenosis after coronary angioplasty? Circulation, 1994, 90: 2248-2257.
    33. Cairns JA, Gill J, Morton B, et al. Fish oils and low-molecular-weight heparin for the reduction of restenosis after percutaneous transluminal coronary angioplasty. The EMPAR Study. Circulation, 1996, 94: 1553-1560
    34. Emanuelsson H, Beatt KJ, Bagger JP, et al. Long-term effects of angiopeptin treatment in coronary angioplasty. Reduction of clinical events but not angiographic restenosis. European Angiopeptin Study Group. Circulation, 1995, 91: 1689-1696
    35. George J, Sack J, Barshack I, et al. Inhibition of intimal thickening in the rat carotid artery injury model by a nontoxic Ras inhibitor. Arterioscler Thromb Vase Biol, 2004, 24(2): 363
    36. Yue TL, Vickery-Clark L, Louden CS, et al. Selective estrogen receptor modulator idoxifene inhibits smooth muscle cell proliferation, enhances reendothelialization, and inhibits neointimal formation in vivo after vascular injury. Circulation, 2000, 102(19): Ⅲ281
    1. Steg PG, Tahlil O, Aubailly N, et al. Reduction of restenosis after angioplasty in an atheromatous rabbit model by suicide gene therapy. Circulation. 1997, 96: 408-411.
    2. Harrell RL, Rajanayagam S, Doanes AM, et al. Inhibition of vascular smooth muscle cell proliferation and neointimal accumulation by adenovirus-mediated gene transfer of cytosine deaminase. Circulation. 1997, 96: 621-627.
    3. Smith RC, Wills KN, Antelman D, et al. Adenoviral constructs encoding phosphorylationcompetent full-length and truncated forms of the human retinoblastoma protein inhibit myocyte proliferation and neointima formation. Circulation. 1997, 96: 1899-1905.
    4. Claudio PP, Fratta L, Farina F, et al. Adenoviral RB2/p130 gene transfer inhibits smooth muscle cell proliferation and prevents restenosis after angioplasty. Circ Res. 1999, 85: 1032-1039.
    5. Luo Z, Sata M, Nguyen T, et al. Adenovirus-mediated delivery of fas ligand inhibits intimal hyperplasia after balloon injury in immunologically primed animals. Circulation. 1999, 99: 1776-1779.
    6. Chen D, Krasinski K, Sylvester A, et al. Downregulation of cyclin-dependent kinase 2 activity and cyclin A promoter activity in vascular smooth muscle cells by p27(KIP1), an inhibitor of neointima formation in the rat carotid artery. J Clin Invest. 1997, 99: 2334-2341.
    7. Scheinman M, Ascher E, Levi GS, et al. p53 gene transfer to the injured rat carotid artery decreases neointimal formation. J Vasc Surg. 1999, 29: 360-369.
    8. Kim SC, Rho MC, Lee HS, et al. Caspase-3-dependent apoptosis in vascular smooth muscle cell by proteasome inhibition. J Cardio phar, 2003, 42(4): 554-560
    9. Pollman MJ, Hall JL, Mann M J, et al. Inhibition of neointimal cell bcl-x expression induces apoptosis and regression of vascular disease. Nature Med, 1998, 4: 222-227.
    10. Izumi Y, Kim S, Yoshiyama Y, et al. Activation of apoptosis signal-regulating kinase 1 in injured artery and its critical role in neointimal hyperplasia. Circulation, 2003, 108(22): 2812.
    11. Scheinman M, Ascher E, Levi GS, et al. p53 gene transfer to the injured rat carotid artery decreases neointimal formation. J Vasc Surg. 1999, 29:360-369.
    
    12. Shi Y, Fard A, Galeo A. Transcatheter delivery of c-myc antisense oligomers reduces neointimal formation in a porcine model of coronary artery balloon injury. Circulation. 1994, 90: 944-951.
    
    13. Cioffi CL, Garay M, Johnston JF, et al. Selective inhibition of A-Raf and C-Raf mRNA expression by antisense oligodeoxynucleotides in rat vascular smooth muscle cells: role of A-Raf and C-Raf in serum-induced proliferation. Mol Pharmacol. 1997, 51:383-389.
    14. Ueno H, Yamamoto H, Ito S, et al. Adenovirus-mediated transfer of a dominant-negative H-ras suppresses neointimal formation in balloon-injured arteries in vivo. Arterioscler Thromb Vasc Bio. 1997, 17:898-904.
    15. Iaccarino G, Smithwick LA, Lefkowitz RJ, et al. Targeting G[beta][gamma] signaling in arterial vascular smooth muscle proliferation: a novel strategy to limit restenosis. Proc Natl Acad Sci USA. 1999, 96:3945-3950.
    16. Sarjeant JM, Lawrie A, Kinnear C, et al. Apolipoprotein D inhibits platelet derived growth factor-BB-induced vascular smooth muscle cell proliferated by preventing translocation of phosphorylated extracellular signal regulated kinase 1/2 to the nucleus. Arteriosclerosis Thrombosis Vascular Biology.2003, 23(12): 2172-2177.
    17. Autieri MV, Yue TL, Ferstein GZ, et al. Antisense oligonucleotides to the p65 subunit of NF-[kappa]B inhibit human vascular smooth muscle cell adherence and proliferation and prevent neointima formation in rat carotid arteries. Biochem Biophys Res Commun. 1995, 213: 827-836
    18. Morishita R, Gibbons GH, Horiuchi M, et al. A gene therapy strategy using a transcription factor decoy of the E2F binding site inhibits smooth muscle proliferation in vivo. Proc Natl Acad Sci U S A. 1995, 92:5855-5859.
    19. Goukassian DA, Kishore R, Krasinski K, et al. Engineering the response to vascular injury: divergent effects of deregulated E2F1 expression on vascular smooth muscle cells and endothelial cells result in endothelial recovery and inhibition of neointimal growth. Circulation Res. 2003, 93(2):162
    20. Smith RC, Branellec D, Gorski DH, et al. p21CIP1-mediated inhibition of cell proliferation by over expression of the gax homeodomain gene. Genes Dev. 1997, 11:1674-1689
    21.Mano T, Luo Z, Malendowicz SL, et al. Reversal of GATA-6 downregulation promotes smooth muscle differentiation and inhibits intimal hyperplasia in balloon-injured rat carotid artery. Circ Res. 1999, 84: 647-654.
    22. Hudziak RM, Summerton J, Weller DD, et al. Antiproliferative effects of steric blocking phosphorodiamidate morpholino antisense agents directed against c-myc. Antisense Nucleuc Acid Drug Dev. 2000, 10(3): 163-176
    23. Stephan D, San H, Yang ZY, et al. Inhibition of vascular smooth muscle cell proliferation and intimal hyperplasia by gene transfer of[beta]-interferon. Mol Med. 1997, 3: 593-599
    24. Van Belle E, Maillard L, Tio FO, et al. Accelerated endothelialization by local delivery of recombinant human vascular endothelial growth factor reduces in-stent intimal formation. Biochem Biophys Res Commun. 1997, 235: 311-316
    25. Tallant EA, Clark MA. Molecular mechanisms of inhibition of vascular growth by angiotensin(1-7). Hypertension, 2003, 42(4): 574-579
    26. Gousseva N, Kugathasan K, Chesteman CN, et al. Early growth response factor-1 mediates insulin-inducible vascular endothelial cell proliferation and regrowth after injury. J Cell Biochem. 2001, 81(3): 532-534
    27. Sclzman CH, Shames BD, Whitehill TA, et al. Class Ⅱ cytokine receptor ligands inhibit human vascular smooth muscle proliferation. Surgery 1998, 124(2): 318-326
    28. Girard N, Maingonnat C, Bertrand P, et al. Hyaluoectinse secretion by monocytes: downregulation by IL-13, upregulation by IL-10. Cytokine. 1999, 11(8): 579-584
    29. Lijinen HR, Soloway P, Collen. Tissure inhibitor of matric metalloproteinase-1 impairs arterial neointima formation after vascular injure in mice. Circ Res.1999, 85: 1186-1191
    30. Hanna AK, Fox JC, Neschis DG, et al. Antisense basic fibroblast growth factor gene transfer reduces neointimal thickening after arterial injury. J Vasc Surg. 1997, 25: 320-325
    31. Sirois MG, Michael S, Edelman ER. Antisense oligonucleotide inhibition of PDGFR-beta receptor subunit expression directs suppression of intimal thickening. Circulation, 1997, 95: 669-676
    32. Deguchi J, Namba T, Hamada H, et al. Targeting endogenous platelet-derived growth factor B-chain by adenovirus-mediated gene transfer potently inhibits in vivo smooth muscle proliferation after arterial injury. Gene Ther. 1999, 6: 956-965
    33. Motoko K, Noboru F, Hideyuki A, et al. Chimeric DNA-RNA hammerhead ribozyme targeting PDGF A-chain mRNA specifically inhibits neointima formation in rat carotid artery after balloon injury. Cardiovascular Research. 2003(57) 265-276
    34. Chen L, Daum G, Forough R, et al. Overexpression of human endothelial nitric oxide synthase in rat vascular smooth muscle cells and in balloon-injured carotid artery. Circ Res. 1998, 82: 862-870
    35. Shears LL, Kibbe MR, Murdock AD, et al. Efficient inhibition of intimal hyperplasia by adenovirus-mediated inducible nitric oxide synthase gene transfer to rats and pigs in vivo. J Am Coll Surg. 1998, 187: 295-306
    36. Guo K, Andres V, Walsh K. Nitric oxide-induced dowuregulation of cdk2 activity and cyclin A gene transcription in vascular smooth cells. Circulation. 1998, 97(20): 2066
    37. Fiona MD, Rodney LS, Huiying C, et al. Mechanism of eNOS gene transfer inhibition of vascular smooth muscle cell proliferation. Cell physiology. Bethesda: Jap 2003, 53(1): C191
    1. Massague J, Blain SW, Lo RS. TGFbeta signaling in growth control, cancer, andheritable disorders. Cell. 2000, 103(2): 295-309.
    2. Hajnal A. Fine-tuning the RAS signaling pathway: Zn~(2+) makes the difference. Mol Cell. 2002, 9(5): 927-928.
    3. Mann DJ, Higgins T, Jones NC, et al. Differential control of cyclins D1 and D3 and CDK inhibitor p27 by diverse signaling pathways in Swiss 3T3 cells. Oncogene. 1997; 14: 1759-1766.
    4. Ortega S, Malumbres M, Barbacid M. Cyclin D-dependent kinases, INK4 inhibitors and cancer. Biochim Biophys Acta. 2002, 1602(1): 73-87.
    5. Xiaotang Hu, Kenneth S, Zuckerman. Transforming growth factor: signal transduction pathways, cell cycle mediation, and effects on hematopoiesis[J]. J of Hematotherapy and Stem Cell. Research. 2001, 10: 67-74.
    6. Sumudra P. Repression of transforming growth factor β receptor type 1 promoter expression by 3p1 deficiency[J]. Oncogene. 2000, 19: 4660-4667.
    7. Wakefield LM, Roberts AB. TGF-Beta signaling: positive and negative effects on tumorigenesis. current opinion in genetics and development. 2002, 12(1): 22-29.
    8. Seoane J, Pouponnot C, Staller P. TGFβ influences myc, Miz-1 and smad to control the CDK inhibitor p15ink4b[J]. Nature cell biology. 2001, 3(4): 400-408.
    9. Majesky MW, Lindner V, Twardzik DR, et al. Production of transforming growth factor betal during repair of arterial injury. J Clin Invest. 1991, 88(3): 904-910
    10. Nikol S, Isner JW, Pickering JG, et al. Expression of J transforming growth factor betal is increased in human vascular restenosis lesions. J Clin Invest. 1992, 90(4): 1582-1592
    11. Smith JD, Bryant SR, Couper LL, et al. Soluble transforming growth factor-β type Ⅱ receptor inhibits negative remodeling, fibroblast transdifferentiation, and intimal lesion formation but not endothelial growth. Circ Res. 1999, 84: 1212-1222.
    12. Schulick AH, Taylor AJ, Zuo W, et al. Overexpression of transforming growth factor-β1 in artedal endothelium causes hyperplasia, apoptosis and cartilaginous metaplsia. Proc Natl Acad Sci USA. 1998, 95: 6983-6988.
    13. Chamberlain J, Gunn J, Francis SE, et al. TGFbeta is active, and correlates with activators of TGFbeta, following porcine coronary angioplasty. Cardiovasc Res. 2001, 50(1): 125-136.
    14. Kingston PA, Sinha S, David A, et al. Adenovirus-mediated gene transfer of a secreted transforming growth factor-beta type Ⅱ receptor inhibits luminal loss and constrictive remodeling after coronary angioplasty and enhances adventitial collagen deposition. Circulation. 2001, 20; 104(21): 2595-2601.
    15. McCaffrey TA, ConsigLi S, Du B, et al. Decreased type Ⅱ/type Ⅰ TGF-beta receptor ratio in cells derived from human atherosclerotic lesions. Conversion from an antiproliferative to profibrotic response to TGF-betal. J Clin Invest. 1995, 96(6): 2667-2675.
    16. Nakajima H, Nakajima HO, Salcher O, et al. Atrial but not ventricular fibrosis in mice expressing a mutant transforming growth factor-β1 transgene in the heart. Circ Res. 2000, 17: 571-579.
    17. Stouffer GA, Owens GK, TGF-βPromotes proliferation of cultured SMC via both PDGFAA-dependent and PDGF-AA-independent and mechanisms. J Clin Invest. 1994, 93: 2048-2055
    18. Goodman LV, Majack RA. Vascular smooth muscle cells express distinct orming growth factor-β receptor phenotypes as function of cell density in culture. J Biol Chem. 1989, 264: 5241-5244
    19. Xin-Hua Feng, Ellen HF, Rik Derynck. Transforming Growth Factor-β induced downregulation of Cyclin A expression requires a functional TGF-β receptor complex. The American Society for Biochemistry and Molecular Biology, 1995, 270(41): 24237-24245.
    20. Shiyou Chen, Magdalena Kulk, Robert J, et al. Smad proteins regulate transcriptional induction of the SM22[alpha] gene by TGF-[beta]. Nucleic Acids Research. 2003, 31(4): 1302-1310.
    [1] Obaya AJ, Sedivy JM. Regulation of cyclin-CDK activity in mammalian cells[J]. Cell Mol Life Sci, 2002, 59: 126-142.
    [2] Hulleman E, Boonsha J. Regulation of G1 phase progression by growth factors and the extracellular matrix[J]. Cell Mol Life Sci, 2001, 58(1): 80-93.
    [3] Kadis P. The cdk-activating kinase(CAK): from yeast to mammals[J]. Cell Mol Life Sci, 1999, 55: 184-296.
    [4] Hagopian JC, Kirtley MP, Stevenson LM, et al. Kinetic basis for activation of cdk/cyclin A by phosphorylation[J]. Bion Chem, 2001, 276: 275-280.
    [5] Parker LL, Piwnica-Worms H. Inactivation of the p34cdk2-cyclin B complex by the human WEE1 tyrosine kinasis[J]. Science, 1992, 257: 1955-1957.
    [6] Abrieu A, Brassac T, Galas S, et al. The polo-like kinasis Pix1 is a component of the MPF amplification loop at the G2/M-phase transition of the cell cycle in Xenopus eggs[J]. J Cell Sci, 1998, 111: 1751-1757.
    [7] Ryo A, Liou YC, Lu KP, et al. Prolyl isomerase Pin1: a catalyst for oncogenesis and a potential[J]. Cell Sci, 2003, 116(5): 773-783
    [8] Serrano M, Hannon GJ, Beach D. A new regulatory motif in cell-cycle control causing specific of cyclinD/CDK4[J]. Nature, 1993, 366(6456): 704-707.
    [9] Chen J, Willinghan T, Shford M, et al. Tumor suppression and inhibition if aneuploid cell accumulation in human brain tumor cells by actopic overexpression if the cyclin-dependent kinasis inhibitor 27~(kip1)[J]. J Clin Invest, 1996, 97(4): 1983-1988.
    [10] Grana X, Reddy EP. Cell cycle control in mammalian cells: role of cyclins cyclin dependent kinases(CDKs) growth suppressor genes and cyclin dependent kinase inhibitors(CDKIs) growth suppressor gene and cyclin dependent kinase inhibitors(CKIs)[J]. Oncogene, 1995, 11 (2): 211-219.
    [11] Gulbis JM, Kelman Z, Hurwitz J, et al. Structure of the C-terminal region of p21~(WAF1/CIP2): complexed with human PCNA[J]. Cell, 1996, 87: 297-300.
    [12] Lee MH, Reynisdottir I, Massague J, et al. Cloning of p57~(KIP2) a cyclin-dependent kinase inhibitor with unique domain structure and tissue distribution[J]. Genes Dev, 1995, 9(6): 639-649.
    [13] Nakayama KI, Hatakeyama S, Nakayama K. Regulation by proteolysis of cyclin E and p27~(kip1)[J]. Biochem Biophys Res Commun, 2001, 282(4); 853-860.
    [14] Moberg KH, Daphne WB, Doke CR. Wahrer Archipelago regulates mutated in human cancer cell lines[J]. Nature, 2001, 413: 311-316.
    [15] Strohmaier H, Charles HS, Peter K. Human F-box protein hCdc4 targets cyclin E for proteolysis and is murated in a breast cancer cell lines[J]. Nature, 2001, 413: 316-322.
    [16] Koepp DM, Schaefer LR, Xin Y. Phosphorylation-dependent ubiquitination of cyclin E by the SCF fbw7 ubiquitin ligase[J]. Science, 2001, 294: 173-177.
    [17] Horowitz JM, Park SH, Bogenrnaun E, et al. Frequent inactivation of the retinoblastoma anti-oncogene is restricted to a subset of human tumor cells[J]. Proc Natl Acad Sci USA, 1990, 87(7): 2775-2779.
    [18] Speir E, Epstein SE. Inhibition of smooth muscle cell proliferation by an antisense oligodeoxynucleotide targeting the messenger RNA proliferating cell nuclear antigen[J]. Circulation, 1992, 86(2): 538 547.
    [19] 汤雪明,王一飞,朱平.医学细胞生物学[M].北京,科学出版社,2004.396-398.
    [20] Pilz RB, Casted DE. Regulation of gene expression by cyclic GMP[J]. Circulation, 2003, 93(11): 1034-1046.
NGLC 2004-2010.National Geological Library of China All Rights Reserved.
Add:29 Xueyuan Rd,Haidian District,Beijing,PRC. Mail Add: 8324 mailbox 100083
For exchange or info please contact us via email.