用户名: 密码: 验证码:
预防移植静脉内膜增生和再狭窄的实验研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
自1898年Gluck等首次将自体静脉用于动脉重建以来,自体静脉移植越来越多地被用于临床,迄今仍是修复肢体动脉缺损和冠状动脉旁路手术的主要方法,但常因内膜增生、再狭窄等原因影响远期疗效,Raja报道术后10年通畅率不足50%,因此有效预防移植静脉移植后内膜增生和再狭窄一直是大量的骨外科、血管外科、心脏外科学者研究的热点。移植静脉发生内膜增生是为了适应环境改变而发生的结构性重塑形,病理过程涉及到血栓形成,内皮细胞的免疫激活,白细胞的粘附、浸润,血管平滑肌细胞的迁移、增殖和凋亡,细胞外基质的形成和迁移,各种因子的激活和信号传导等机制,是一个受多种因素影响、多因子和多基因参与、多种机制调控的连续变化的病理过程,机制非常复杂,因此从根本上彻底阻止移植静脉再狭窄的发生至今仍是一个亟待解决的难题。
     内膜增生是移植静脉发生再狭窄最根本和最主要的病理变化,因此减轻移植静脉内膜的过度增生是预防移植静脉再狭窄的关键,基于此我们设计本课题,通过实验研究探讨以下三个问题:1、局部应用5-氟尿嘧啶对移植静脉内膜增生的影响;2、单层和双层自体静脉外支架对移植静脉内膜增生的影响;3、抑制外周组织粘连对移植静脉内膜增生的影响。
     第一部分局部应用5-氟尿嘧啶抑制移植静脉内膜增生的实验研究
     平滑肌细胞的过度增殖、迁移是移植静脉发生内膜增生最关键的机制,因此抑制平滑肌细胞的过度增殖、迁移,诱导平滑肌细胞凋亡,合理地调控血管的重构,是预防移植静脉内膜增生和再狭窄最直接有效的途径。5-氟尿嘧啶(5-FU)是一种嘧啶类的抗代谢药物,可以通过干扰DNA合成的途径抑制细胞增殖,诱导细胞凋亡,局部应用安全性高。Cragg研究发现5-氟尿嘧啶对体外培养的平滑肌细胞增殖有明显抑制作用。基于此,我们建立家兔自体颈外静脉-颈总动脉模型,通过实验动物验证局部应用5-氟尿嘧啶是否能够抑制在体移植静脉平滑肌细胞增殖和诱导平滑肌细胞凋亡,从而调控血管的重构,预防移植静脉再狭窄的发生。
     目的:探讨局部应用5-氟尿嘧啶(5-fluorouracil, 5-FU)对动脉缺损静脉移植后内膜增生的影响。
     方法:选用64只5月龄雄性新西兰大白兔,体重2.8~3.0kg,随机分为A、B、C、D四组(n=16)。切取右侧颈外静脉3cm,游离左侧颈总动脉,中间切除长约1cm,建立动脉缺损模型,将切取静脉远近端倒置后用9-0无损伤缝线端端吻合于动脉缺损,吻合结束后A组、B组、C组动物分别用于50mg/ml、25mg/ml、12.5mg/ml的5-氟尿嘧啶中浸泡的脑棉(大小为12mm×30mm×1mm)完全包裹移植静脉外膜方式给药,共5次,每次1min,D组用生理盐水取代5-氟尿嘧啶同样方法处理。分别于术后1、2、4、6周切取移植静脉,行HE染色、Masson染色观察移植静脉管壁组织学变化,PCNA免疫组化染色、TUNEL标记染色观察移植静脉平滑肌细胞增殖和凋亡情况,透射电镜观察细胞超微结构变化。
     结果:HE染色、Masson染色观察到:术后1、2、4、6周A组、B组移植静脉内膜增厚均明显小于C组和D组,PCNA免疫组化染色和TUNEL法标记染色观察到:术后1、2、4周A组、B组增殖细胞少于C组和D组,术后1、2周内膜、中膜凋亡细胞A组和B组明显多于C组和D组。内膜厚度、内膜增生程度、管腔狭窄程度结果分别为:1周A组12.69±1.68μm,0.73±0.05,0.025±0.003;B组17.52±2.01μm,0.86±0.06,0.027±0.004;C组21.92±1.85μm,1.06±0.09,0.036±0.006;D组26.45±3.86μm,1.18±0.08,0.041±0.005。2周A组24.61±2.91μm,0.86±0.06,0.047±0.003;B组37.28±2.78μm,1.17±0.09,0.060±0.004;C组46.52±2.25μm,1.41±0.08,0.073±0.003;D组52.07±3.29μm,1.45±0.05,0.081±0.006。4周A组61.09±6.84μm,1.38±0.08,0.106±0.007;B组63.61±8.25μm,1.40±0.07,0.107±0.010;C组80.04±7.65μm,1.64±0.07,0.129±0.011;D组84.45±9.39μm,1.68±0.10,0.138±0.014。6周A组65.27±5.25μm,1.46±0.07,0.113±0.005;B组65.82±7.12μm,1.45±0.05,0.112±0.011;C组84.45±6.39μm,1.69±0.09,0.135±0.007;D组87.27±8.96μm,1.76±0.05,0.140±0.012。均为A组、B组小于C组和D组(P<0.05)。内膜的增殖指数、凋亡指数和外膜的增殖指数、凋亡指数结果分别为:1周A组4.83±1.96,30.51±4.01,14.61±2.17,9.23±1.40;B组8.01±2.18,29.27±5.18,20.25±3.18,6.93±1.05;C组9.09±1.27,19.21±3.16,26.80±3.13,3.89±1.11;D组12.25±2.13,28.71±4.17,19.01±4.12,3.83±1.34。2周A组18.92±3.73,36.16±2.27,22.31±3.94,10.72±2.32;B组19.16±3.26,32.41±3.74 , 30.51±4.01 , 5.13±2.22; C组25.97±2.17 , 29.24±2.11 ,37.81±4.50,5.94±1.79;D组27.06±2.91,27.15±2.51,42.71±3.60,5.21±1.91。4周A组10.17±1.21,11.73±3.21,20.00±3.16,5.19±1.83;B组11.18±2.01,11.23±2.15,21.13±2.62,4.99±1.41;C组15.01±2.03,11.04±2.84,24.64±2.98,4.15±2.31;D组15.96±2.44,12.21±3.68,28.70±2.08,4.86±1.69。6周A组6.67±2.10,5.69±2.31,2.51±1.53,3.84±1.41;B组6.67±1.81,5.04±1.71,2.47±1.51,3.53±1.34;C组6.88±2.01,5.99±2.13,3.40±0.98,3.74±1.03;D组8.21±2.27,6.01±2.03,3.28±1.09,3.64±1.08。术后1、2、4周的细胞增殖指数均为A组、B组小于C组和D组(P<0.05),术后1、2周内膜、中膜细胞凋亡指数A组和B组均大于C组和D组(P<0.05)。透射电镜观察到A组、B组相对于C组和D组粗面内质网、高尔基体、核糖体等合成细胞器含量明显减少。
     结论:局部应用5-FU可抑制移植静脉平滑肌细胞增殖,诱导术后早期平滑肌细胞凋亡,有效地减轻内膜增生,降低移植静脉管腔再狭窄程度。
     第二部分自体静脉外支架对移植静脉内膜增生影响的实验研究
     1963年,Parsonne等首先报道了于移植静脉周围应用外支架(external stent)能够减轻移植静脉的内膜增生,有效地预防移植静脉狭窄,之后许多学者通过实验研究证实应用外支架是一种很有前景的预防移植静脉狭窄的措施。外支架的作用是对抗移植静脉移植后的早期扩张,通过刺激新生外膜形成或直接对抗作用使移植静脉管腔内的血流动力学特性更加接近动脉,从而减少因血流动力学改变所引发的一系列病理改变。目前应用的外支架多为人工合成材料,植入体内可引发慢性炎症、局部感染、静脉外膜纤维化等不良反应,而且其弹性和顺应性与动脉差异很大,因此并不能充分发挥减少移植静脉血流动力学改变的作用。我们选用自体相容性好、弹性和顺应性与动脉相近的自体静脉作为移植静脉的外支架,在移植静脉外周放置单层和双层自体静脉外支架,通过动物实验观察自体静脉外支架能否对抗移植静脉的早期扩张,抑制平滑肌细胞增生,预防移植静脉再狭窄的发生。
     目的:探讨自体静脉外支架对动脉缺损静脉移植后内膜增生的影响。
     方法:选用36只5月龄雄性新西兰大白兔,体重2.8~3.0kg,随机分为A组、B组和C组(n=12)。切取右侧颈外静脉6cm,等分为三段,远心段均用作移植静脉,B组以中段作为自体外支架,A组以近心段套叠中段做成双层自体外支架。游离左侧颈总动脉,中间切除长约0.5cm,建立动脉缺损模型,将移植静脉远近端倒置后吻合于动脉缺损,通血后行移植静脉外径测量。分别于术后2、4周切取移植静脉,行HE染色、Masson染色观察移植静脉管壁组织学变化,PCNA免疫组化染色观察移植静脉平滑肌细胞增殖情况。
     结果:通血后移植静脉直径A组1.62±0.31mm,B组2.21±0.40mm,C组2.65±0.58mm,A组     结论:双层自体静脉外支架可以有效地抑制移植静脉内膜增生,减轻移植静脉管腔再狭窄程度。
     第三部分抑制外周组织粘连对移植静脉内膜增生影响的实验研究
     移植静脉植入后均与外周组织间存在明显的粘连,粘连机制是移植静脉外膜滋养血管长入、从外周组织获得血运的途径。如果采取措施阻止外周粘连,阻止外膜滋养血管长入,屏蔽移植静脉从周围组织获得血运,可能导致内膜缺血,而内膜缺血促进促有丝分裂原的产生,是内膜增生的重要原因。Fogelstrand和Zhang等学者通过动物实验研究发现外周组织来源的平滑肌细胞也是导致移植静脉内膜增生的重要细胞来源,外周组织源性平滑肌细胞通过外周组织粘连迁移至移植静脉,参与内膜增生的形成,阻断外周组织的粘连可以抑制内膜增生。因此,阻止外周组织粘连对移植静脉内膜增生可以产生两种相反的作用机制,究竟哪一种机制占主导地位,抑制周围组织粘连将对内膜增生产生怎样的影响是一个需要探究的课题。透明质酸钠局部应用可以抑制粘连的发生,而且透明质酸钠为液性,局部应用后不通过改变移植静脉的生物力学机制影响内膜增生。因此,我们设计采用移植静脉周围应用透明质酸钠的方法抑制移植静脉周围组织粘连,与常规移植静脉对照,揭示其抑制外周组织粘连对移植静脉平滑肌细胞增殖、细胞因子(PDGF)的表达和分布以及对内膜增生的影响。
     目的:探讨抑制外周组织粘连对移植静脉平滑肌细胞增殖、细胞因子(PDFG)的表达和分布以及对内膜增生的影响。
     方法:选用24只5月龄雄性新西兰大白兔,体重2.8~3.0kg,随机分为A组、B组(n=12)。切取右侧颈外静脉3cm,游离左侧颈总动脉,中间切除长约1cm,建立动脉缺损模型,将切取静脉远近端倒置后用9-0无损伤缝线端端吻合于动脉缺损,吻合结束后A组动物用透明质酸钠凝胶0.2ml涂布于移植静脉外膜及两个吻合口。分别于术后1、2、4周行移植静脉切取前肉眼观察,切取移植静脉后行HE染色、Masson染色观察移植静脉管壁组织学变化,PCNA免疫组化染色、PDGF免疫组织化学染色观察移植静脉平滑肌细胞增殖和PDFG的表达、分布情况。
     结果:术后1、2、4周肉眼观察B组移植静脉周围均有明显粘连,A组术后1、2周无粘连,术后4周有轻度粘连。HE染色、Masson染色、PCNA免疫组化染色和PDGF免疫组化染色观察到:术后2、4周A组移植静脉内膜厚度小于B组,术后1、2、4周A组增殖细胞均少于B组,术后1周A组外膜无PDGF阳性细胞,B组外膜内见阳性的炎性细胞;术后2、4周A组内膜、中膜和外膜PDGF阳性细胞均少于B组。内膜厚度、内膜增生程度、管腔狭窄程度结果分别为:1周A组25.51±3.93μm,1.18±0.07,0.041±0.005;B组26.18±4.16μm,1.19±0.09,0.042±0.006。2周A组44.27±2.53μm,1.22±0.06,0.062±0.003;B组50.99±3.76μm,1.40±0.03,0.078±0.004。4周A组69.85±6.76μm,1.49±0.07,0.114±0.009;B组84.43±6.41μm,1.69±0.09,0.137±0.007。术后2、4周A组的内膜厚度、内膜增生程度、管腔狭窄程度均小于B组(P<0.05)。内膜、中膜的增殖指数结果分别为:1周A组7.43±2.21,21.53±3.24;B组11.41±2.01,28.63±4.48。2周A组20.01±3.21,35.81±3.41;B组26.78±4.14,42.63±4.15。4周A组11.41±2.01,22.09±2.65;B组15.52±2.37,28.64±3.90。术后2、4周A组的内膜和中膜增殖指数均小于B组(P<0.05)。内膜、中膜外膜的PDGF表达率结果分别为:1周A组7.67±1.61,19.59±3.66,2.46±1.53;B组7.60±2.42,20.58±4.39,10.25±2.31。2周A组11.37±2.55,19.81±3.09,12.90±3.26;B组19.45±3.48,30.63±5.16,30.47±5.84。4周A组6.15±1.94,11.09±2.83,10.19±2.44;B组10.51±2.03,18.64±3.21,26.51±3.84。术后1周A组外膜PDGF表达率低于B组(P<0.05),术后2、4周A组的内膜、中膜和外膜PDGF表达率均小于B组(P<0.05)。
     结论:局部应用透明质酸钠可以有效地抑制移植静脉外周粘连的形成,并在一定程度上减轻内膜增生;抑制外周组织粘连并不加重移植静脉内膜增生和管腔狭窄。
Autogenous vein graft has been more and more common in clinical practice since autogenous vein was first used for artery reconstruction by Gluck in 1898. Heretofore autogenous vein graft continues to be the primary method for the reconstruction of the artery defect and coronary artery bypass graft. However, the long-term therapeutic efficacy is always limited by the intimal hyperplasia and stenosis of vein graft. Raja reported that patency rate of vein graft was no more than 50% 10 years after surgery. So it is always a focus of study for the researcher of orthopaedics, vascular surgery and cardiac surgery to prevent intimal hyperplasia and stenosis of vein graft. Intimal hyperplasia is the result of remodeling for vein graft to adapt itself to a charged environment, the pathologic process comprised thrombogenesis, activating of endothelial cell, leukocyte adhesion, Leukocytic infiltrate,smooth muscle cells migration, proliferation and apoptosis, extracellular matrix formation and migration, activating cytokine and signal transducting, it is a complex and continuously changing pathogenesis influenced by multiple factor and interfered by multiple cytokine and gene. Accordingly, it has been being a pressing difficult problem to prevent the occurrence of stenosis of vein graft.
     It is the critical way of preventing stenosis to inhibit the intimal hyperplasia of vein graft because intimal hyperplasia is the primary and leading pathological change of stenosis of vein graft. For this reason, we designed a project to answer 3 questions: First, to assess the effect of 5-Fluorouracil(5-FU) applied topically on intimal hyperplasia of the vein grafts; Second, to assess the effect of the autologous single-layer and double-layer venous external stents on intimal hyperplasia; Third, to assess the effect of preventing peripheral adhesion of vein graft on intimal hyperplasia.
     Part 1 Inhibitory effects of topical application of 5-fluorouracil on intimal hyperplasia of the vein grafts in rabbits
     The excessive proliferation and migration of vascular smooth muscle cell are the critical pathogenesis of intimal hyperplasia, so it is a immediate way of preventing intimal hyperplasia and stenosis to inhibit excessive proliferation and migration of vascular smooth muscle cell and to induce apoptosis and to regulate the remodeling of vein graft. 5-Fluorouracil(5-FU) is a antimetabolite belonging to miazines, it can inhibit cell proliferation and induce cell apoptosis, and it is very safe applied locally. Cragg reported that 5-Fluorouracil can effectively inhibit proliferation of vascular smooth muscle cell cultured in vitro. So, external jugular vein grafts was interposed into the carotid artery of rabbits and applied 5-Fluorouracil to test the effect of 5-Fluorouracil in inhibiting proliferation of vascular smooth muscle cell and inducing apoptosis of vascular smooth muscle cell in vivo and regulating the remodeling and preventing stenosis of vein graft.
     Objective To assess the effect of 5-Fluorouracil(5-FU) applied topically on intimal hyperplasia of the vein grafts in rabbits.
     Methods Sixty-four male New Zealand white rabbits, aged 5 months and weighing 2.8 to 3.0kg, were randomly divided into 4 groups: Group A, B, C and D (n=16 rabbits per group). Artery defect model was established by cutting about 1 cm artery from the middle part of the dissociated left common carotid artery. A section about 3 cm was cut from the right external jugular vein, and the harvested vein was inverted and end-to-end anastomosed to the artery defect with 9-0 non-traumatic suture. After anastomosis, the extima of the grafted veins in group A, B, and C was completely wrapped with cotton sheet (12mm×30mm×1mm in size) immersed by 5-FU at a concentration of 50.0, 25.0, and 12.5 mg/mL, respectively, and each vein was treated 5 times (1 minute at a time). In group D, the extima of the graft veins was treated with normal sal ine instead of 5-FU. The grafted veins were obtained 1, 2, 4, and 6 weeks after operation, HE staining and Masson staining were preformed for histological changes of grafted vein wall, proliferating cell nuclear antigen (PCNA) immunohistochemistry staining and terminal deoxymudeotityl transferase mediated dUTP nick end labeling (TUNEL) staining were conducted for proliferation and apoptosis of smooth muscle cell of the grafted vein, and transmission electron microscope observation was performed for cellular ultrastructure.
     Result The HE staining, Masson staining showed that the thickness of intima in group A and B was obviously less than that in group C and D at 1, 2, 4, and 6 weeks after operation. PCNA immunohistochemistry staining and TUNEL labeling staining showed that the proliferation cells in group A and B were less than that in group C and D at 1, 2, and 4 weeks after operation and the apoptotic cells in group A and B were more than that in group C and D at 1 and 2 weeks after operation. The thickness of the intima, the degree of intima hyperplasia, the degree of vessel lumen stenosis of four groups at different time points were as follows: at 1 week after operation, group A [(12.69±1.68)μm, 0.73±0.05, 0.025±0.003], group B [(17.52±2.01)μm, 0.86±0.06, 0.027±0.004], group C [(21.92±1.85)μm, 1.06±0.09, 0.036±0.006] and group D [(26.45±3.86)μm, 1.18±0.08, 0.041±0.005]; at 2 weeks after operation, group A [(24.61±2.91)μm, 0.86±0.06, 0.047±0.003], group B [(37.28±2.78)μm, 1.17±0.09, 0.060±0.004], group C [(46.52±2.25)μm, 1.44±0.08, 0.073±0.003], and group D [(52.07±3.29)μm, 1.45±0.05, 0.081±0.006]; at 4 weeks after operation, group A [(61.09±6.84)μm, 1.38±0.08, 0.106±0.007], group B [(63.61±8.25)μm, 1.40±0.07, 0.107±0.010], group C [(80.04±7.65)μm, 1.64±0.07, 0.129±0.011], and group D [(84.45±9.39)μm, 1.68±0.10, 0.138±0.014]; at 6 weeks after operation, group A [(65.27±5.25)μm, 1.46±0.07, 0.113±0.005], group B [(65.82±7.12)μm, 1.45±0.05, 0.112±0.011], group C [(84.45±6.39)μm, 1.69±0.09, 0.135±0.007], and group D [(87.27±8.96)μm, 1.76±0.05, 0.140±0.012]. Group A and B were inferior to group C and D in terms of the above three parameters 1, 2, 4 and 6weeks after operation (P<0.05). Cell proliferation index and cell apoptosis index of intima and that of media were as follows: at 1 week after operation, group A (4.83±1.96, 30.51±4.01, 14.61±2.17, 9.23±1.40), group B (8.01±2.18, 29.27±5.18, 20.25±3.18, 6.93±1.05), group C (9.09±1.27, 19.21±3.16, 26.80±3.13, 3.89±1.11) and group D (12.25±2.13, 28.71±4.17, 19.01±4.12, 3.83±1.34); at 2 weeks after operation, group A (18.92±3.73, 36.16±2.27, 22.31±3.94, 10.72±2.32), group B (19.16±3.26, 32.41±3.74, 30.51±4.01, 5.13±2.22), group C (25.97±2.17, 29.24±2.11, 37.81±4.50, 5.94±1.79), and group D (27.06±2.91, 27.15±2.51, 42.71±3.60, 5.21±1.91); at 4 weeks after operation, group A (10.17±1.21, 11.73±3.21, 20.00±3.16, 5.19±1.83), group B (11.18±2.01, 11.23±2.15, 21.13±2.62, 4.99±1.41), group C (15.01±2.03, 11.04±2.84, 24.64±2.98, 4.15±2.31), and group D (15.96±2.44, 12.21±3.68, 28.70±2.08, 4.86±1.69); at 6 weeks after operation, group A (6.67±2.10, 5.69±2.31, 2.51±1.53, 3.84±1.41), group B (6.67±1.81, 5.04±1.71, 2.47±1.51, 3.53±1.34), group C (6.88±2.01, 5.99±2.13, 3.40±0.98, 3.74±1.03), and group D (8.21±2.27, 6.01±2.03, 3.28±1.09, 3.64±1.08). Group A and B were inferior to group C and D in terms of cell proliferation index of intima and media 1, 2 and 4 weeks after operation (P<0.05). Group A and B were superior to group C and D in terms of cell apoptosis index of intima and media 1 and 2 weeks after operation (P<0.05). Transmission electron microscope observation showed that the synthetic cell organelles such as rough endoplasmic reticulum, golgi apparatus, and ribosome in group A and B were obviously less than those in group C and D.
     Conclusion Topical application of 5-FU can inhibits proliferation of vascular smooth muscle cell and induces cell apoptosis of vascular smooth muscle cell, so that it can effectively inhibit intima hyperplasia and stenosis of the vein grafts.
     Part 2 The effects of the autologous venous external stents on intimal hyperplasia of the vein grafts in rabbits
     In 1963, Parsonne first reported that external stent can inhibit intimal hyperplasia and stenosis of vein graft. After that, many researcher proved that the application of external stent was a promising and effective means of preventing stenosis of vein graft. External stent can counteract the early distension of vein grafted, and it made hemodynamic index of vein graft approaching that of the host artery by the means of prevoking neoadventitia forming and its counteraction, so that the pathologic process induced by the change of hemodynamic index was inhibited. At present the material of most external stent is artificial synthetics, which was likely to evoke inflammation, local infection and adventitial fibrosis of vein graft if implanted. In addition, the mechanical properties of artificial synthetics were far from artery, so this kind of external stents cannot enough inhibit the change of hemodynamic index of vein grafted. Accordingly, the autologous venous external stents, whose mechanical properties were approaching the artery and unlikely to trigger reject reaction, were selected and tested. Single-layer and double-layer venous external stents were applied to vein graft in rabbits to test the effect of autologous venous external stents in counteracting the early distension of vein
     grafted and preventing intimal hyperplasia and stenosis of vein graft. Objective To assess the effect of the single-layer and double-layer autologous venous external stents on intimal hyperplasia of the vein grafts in rabbits.
     Methods Thirty-six male New Zealand white rabbits, aged 5 months and weighing 2.8 to 3.0kg, were randomly divided into 3 groups: Group A, B and C, with 12 rabbits in each group. First, a section about 6cm long of vein was cut from the right external jugular vein of each rabbit and severed to have 3 equal-length segments. Next, each distal segment prepared for anastomosis. The proximal segment invaginating middle segment in group A and only middle segment in group B were used for the external stent. Later, the left common carotid artery was separated from surrounding tissue, from it a section about 0.5 cm long was cut away. Finally, the vein graft was inverted and end-to-end anastomosed to the two ends of the artery with a 9-0 suture. After bloodstream re-established, the diameter of each vein graft was measured. At 2, 4 weeks postoperative,the graft veins were cut off and histologically examined by the means of HE staining and Masson staining. The smooth muscle cells(SMC) proliferation was studied by the immunohistochemical detection of proliferating cell nuclear antigen(PCNA).
     Result After bloodstream re-established, the diameters of vein graft of group A, group B and group C were 1.62±0.31mm, 2.21±0.40mm and 2.65±0.58mm respectively. The diameter of vein graft of group A was smaller than of group B and group C, and that of group B was smaller than group C. (P<0.05). At 2 and 4 weeks after operation, HE staining, Masson staining and PCNA immunohistochemistry staining showed that the thickness of intima in group A and group B was obviously less than that in group C and the proliferation cells of intimal and media in group A and group B were less than that in group C. The thickness of the intima, the degree of intima hyperplasia, the degree of vessel lumen stenosis of 3 groups at different time points were as follows: at 2 week after operation, group A [(35.13±3.24)μm, 0.88±0.03, 0.046±0.004], group B [(44.53±3.55)μm, 1.08±0.02, 0.058±0.005] and group C [(51.62±4.72)μm, 1.21±0.04, 0.068±0.007]; at 4 weeks after operation, group A [(51.73±6.08)μm, 1.01±0.07, 0.076±0.008], group B [(69.29±6.33μm), 1.32±0.08, 0.101±0.009] and group C [(85.00±7.36)μm, 1.55±0.03, 0.134±0.003]. Group A was inferior to group B and C and Group B was inferior to group C in terms of the above three parameters 2 and 4weeks after operation (P<0.05). Cell proliferation index of intima and that of media were as follows: at 2 weeks after operation, group A (16.98±3.96, 28.39±6.02), group B (24.07±4.13, 36.41±6.93) and group C (29.90±4.68, 48.45±5.60); at 4 weeks after operation, group A (6.84±1.98, 15.65±5.12), group B (11.01±2.61, 23.31±4.30) and group C (14.96±4.14, 29.64±4.15). Group A and B were inferior to group C and Group B was inferior to group C in terms of cell proliferation index of intima and media 2 and 4 weeks after operation (P<0.05).
     Conclusion The autologous venous two-layer external stents inhibit intimal hyperplasia and relieve the stenosis of the vein gratfs effectively. Part 3 The effects of inhibit perivascular adhesion on intimal hyperplasia of the vein grafts in rabbits
     There was apparent adhesion between vein grafted and surrounding tissue. Perivascular adhesion was the way which vasa vasorum enter through to and hyperplasia if to prevent perivascular adhesion and vasa vasorum going in. However, Fogelstrand and Zhang et al reported that vascular smooth muscle cell derived from surrounding tissue played a very important role in intimal hyperplasia, which was proved by animal experiment. Vascular smooth muscle cell derived from surrounding tissue migrated to vein grafted through perivascular adhesion, so it can inhibit intimal hyperplasia to prevent perivascular adhesion. Accordingly, opposite actions on intimal hyperplasia were caused by preventing adhesion. It should be studied which action was dominant and what the effect of preventing adhesion on intimal hyperplasia was. Local application of sodium hyaluronate can prevent adhesion, but also it didn’t inflenced intimal hyperplasia of vein graft by biomechanical mechanism because it was liquid. For these reasons, sodium hyaluronate was applied locally to adventitia of vein graft in rabbits to test the effect of sodium hyaluronate on cell proliferation, expression and distribution of PDGF and intimal hyperplasia of vein graft.
     Objective To assess the effect of sodium hyaluronate applied topically on cell proliferation, expression and distribution of PDGF and intimal hyperplasia of vein graft in rabbits.
     Methods twenty-four male New Zealand white rabbits, aged 5 months and weighing 2.8 to 3.0kg, were randomly divided into 3 groups: Group A and B (n=12 rabbits per group). Artery defect model was established by cutting about 1 cm artery from the middle part of the dissociated left common carotid artery. A section about 3 cm was cut from the right external jugular vein, and the harvested vein was inverted and end-to-end anastomosed to the artery defect with 9-0 non-traumatic suture. After anastomosis, the adventitia and two anastomosis of the grafted veins in group A was applied 0.2ml sodium hyaluronate locally to. The grafted veins were obtained 1, 2 and 4 weeks after operation, HE staining and Masson staining were preformed for histological changes of grafted vein wall, proliferating cell nuclear antigen (PCNA) and platelet-derived growth factor (PDGF) immunohistochemistry staining were conducted for proliferation and expression and distribution of PDGF of the grafted vein.
     Result The macroscopic observation showed that there was not apparent adhesion in group B at 1 and 2 weeks and was slight adhesion at 4 weeks after operation in group A and there was apparent adhesion in group B. HE staining, Masson staining showed that the thickness of intima in group A was obviously less than that in group B at 2 and 4 weeks after operation. PCNA and PDGF immunohistochemistry staining showed that the proliferation cells in group A was less than that in group B at 1, 2, and 4 weeks after operation and the positive cells of PDGF immunohistochemistry staining of intimal, media and adventitia in group A was less than that in group B at 1, 2 and 4 weeks after operation. The thickness of the intima, the degree of intima hyperplasia, the degree of vessel lumen stenosis of 2 groups at different time points were as follows: at 1 week after operation, group A [(25.51±3.93)μm, 1.18±0.07, 0.041±0.005] and group B [(26.18±4.16)μm, 1.19±0.09, 0.042±0.006]; at 2 weeks after operation, group A [(44.27±2.53)μm, 1.22±0.06, 0.062±0.003] and group B [(50.99±3.76)μm, 1.40±0.03, 0.078±0.004]; at 4 weeks after operation, group A [(69.85±6.76)μm, 1.49±0.07, 0.114±0.009] and group B [(84.43±6.41)μm, 1.69±0.09, 0.137±0.007]. Group A was inferior to group B in terms of the above three parameters 2 and 4weeks after operation (P<0.05). Cell proliferation index and cell apoptosis index of intima and that of media were as follows: at 1 week after operation, group A (7.43±2.21, 21.53±3.24) and group B (11.41±2.01, 28.63±4.48); at 2 weeks after operation, group A (20.01±3.21, 35.81±3.41) and group B (26.78±4.14, 42.63±4.15); at 4 weeks after operation, group A (11.41±2.01, 22.09±2.65) and group B (15.52±2.37, 28.64±3.90). Group A was inferior to group B in terms of cell proliferation index of intima and media 2 and 4 weeks after operation (P<0.05). The percentage of PDGF-positive cells of intima, media and adventitia was as follows: at 1 week after operation, group A (7.67±1.61, 19.59±3.66, 2.46±1.53) and group B (7.60±2.42, 20.58±4.39, 10.25±2.31); at 2 weeks after operation, group A (11.37±2.55, 19.81±3.09, 12.90±3.26) and group B (19.45±3.48, 30.63±5.16, 30.47±5.84); at 4 weeks after operation, group A (6.15±1.94, 11.09±2.83, 10.19±2.44) and group B (10.51±2.03, 18.64±3.21, 26.51±3.84). Group A was inferior to group B in terms of the percentage of PDGF-positive cells of intima, media and adventitia 2 and 4 weeks after operation (P<0.05) and Group A was inferior to group B that of adventitia 1 week after operation (P<0.05).
     Conclusion Topical application of sodium hyaluronate prevents perivascular adhesion and inhibits intima hyperplasia. It doesn’t promote intima hyperplasia and stenosis of the vein graft to prevent perivascular adhesion.
引文
1 Moses JW, Leon MB, Popma JJ, et al. Sirolimus-eluting stents versusstandard stents in patients with stenosis in a native coronary artery. N Engl J Med, 2003,349(14):1315-1323.
    2 Raja SG. Composite arterial grafting. Expert Rev Cardiovasc Ther, 2006, 4(4):523-533.
    3 Fogelstrand P, Feral CC, Zargham R, et al. Dependence of proliferative vascular smooth muscle cells on CD98hc (4F2hc, SLC3A2). J Exp Med, 2009,206(11):2397-2406.
    4 Yucel S, Bahcivan M, Gol MK, et al. Reduced intimal hyperplasia in rabbits via medical therapy after carotid venous bypass. Tex Heart Inst J, 2009,36(5):387-392.
    5刘继红,何学志,谷天祥,等.局部应用紫杉醇预防兔移植静脉再狭窄.中国胸心血管外科临床杂志, 2008,15(002):126-129.
    6程宏. 5-氟尿嘧啶诱导细胞凋亡的分子机制.中国医学文摘:肿瘤学, 2003,17(001):75-76.
    7崔韶晖,王艳颖,陈曦,等. 5-氟尿嘧啶诱导肿瘤细胞凋亡的研究.大连民族学院学报, 2008,10(003):207-210.
    8 Cragg AH, Stoll LL, Smith TP, et al. Effect of antineoplastic agents on smooth muscle cell proliferation in vitro: implications for prevention of restenosis after transluminal angioplasty. J Vasc Interv Radiol, 1992,3 (2):273-277.
    9郭明珂,张经歧,田德虎,等. 5-氟尿嘧啶预防屈肌腱粘连的临床研究.中国修复重建外科杂志, 2008,22(007):794-796.
    10郭明珂,张经歧,田德虎,等. 5-氟尿嘧啶防治肌腱粘连的实验研究.中国修复重建外科杂志, 2007,21(008):842-846.
    11张鲁锋,肖锋,李简,等.局部缓释雷帕霉素抑制静脉移植血管内膜增生.中华医学杂志, 2006,86(024):1706-1709.
    12 Albrecht, T, Speck, U, Baier, C, et al. Reduction of Stenosis Due to Intimal Hyperplasia After Stent Supported Angioplasty of Peripheral Arteries by Local Administration of Paclitaxel in Swine. Investigative Radiology, 2007,42(8):579.
    13 Ishiwata, S, Tukada, T, Nakanishi, S, et al. Postangioplasty restenosis:platelet activation and the coagulation-fibrinolysis system as possible factors in the pathogenesis of restenosis. American Heart Journal, 1997, 133(4):387-392.
    14 Verrier ED, Morgan EN. Endothelial response to cardiopulmonary bypass surgery. Ann Thorac Surg, 1998,66(5 Suppl):S17-19; discussion S25-28.
    15 Stark VK, Hoch JR, Warner TF, et al. Monocyte chemotactic protein-1 expression is associated with the development of vein graft intimal hyperplasia. Arterioscler Thromb Vasc Biol, 1997,17(8):1614-1621.
    16 Eefting D, de Vries MR, Grimbergen JM, et al. In vivo suppression of vein graft disease by nonviral, electroporation-mediated, gene transfer of tissue inhibitor of metalloproteinase-1 linked to the amino terminal fragment of urokinase (TIMP-1.ATF), a cell-surface directed matrix metalloproteinase inhibitor. J Vasc Surg, 2010,51(2):429-437.
    17 Allaire E, Clowes AW. Endothelial cell injury in cardiovascular surgery: the intimal hyperplastic response. Ann Thorac Surg, 1997,63(2):582-591.
    18 Lardenoye JH, de Vries MR, Deckers M, et al. Inhibition of intimal hyperplasia by the tetracycline derived mmp inhibitor doxycycline in vein graft disease in vitro and in vivo. EuroIntervention, 2005,1(2):236-243.
    19 Zhang J, Fu M, Zhu X, et al. Peroxisome proliferator-activated receptor delta is up-regulated during vascular lesion formation and promotes post-confluent cell proliferation in vascular smooth muscle cells. J Biol Chem, 2002,277(13):11505-11512.
    20 Lozano E, Segarra M, Garcia-Martinez A, et al. Imatinib mesylate inhibits in vitro and ex vivo biological responses related to vascular occlusion in giant cell arteritis. Ann Rheum Dis, 2008,67(11):1581-1588.
    21 Wu L, Zhang W, Li H, et al. Inhibition of aortic intimal hyperplasia and cell cycle protein and extracellular matrix protein expressions by BuYang HuanWu Decoction. J Ethnopharmacol, 2009,125(3):423-435.
    22 Wu J, Zhang C. Neointimal hyperplasia, vein graft remodeling, and long-term patency. Am J Physiol Heart Circ Physiol, 2009,297 (4) :H1194-1195.
    23 Berceli SA, Tran-Son-Tay R, Garbey M, et al. Hemodynamically driven vein graft remodeling: a systems biology approach. Vascular, 2009,17 Suppl 1:S2-9.
    24 Cai X. Regulation of smooth muscle cells in development and vascular disease: current therapeutic strategies. Expert Rev Cardiovasc Ther, 2006, 4(6):789-800.
    25 Liu HC, Chen GG, Vlantis AC, et al. 5-fluorouracil mediates apoptosis and G1/S arrest in laryngeal squamous cell carcinoma via a p53-independent pathway. Cancer J, 2006,12(6):482-493.
    26万昕,高山,余勤,等.高效液相色谱法测定鼠脑组织中5-氟尿嘧啶浓度.四川医学, 2004,25(003):355-356.
    27 Moran SL, Ryan CK, Orlando GS, et al. Effects of 5-fluorouracil on flexor tendon repair. J Hand Surg [Am], 2000,25(2):242-251.
    28 Daniels JT, Occleston NL, Crowston JG, et al. Effects of antimetabolite induced cellular growth arrest on fibroblast-fibroblast interactions. Exp Eye Res, 1999,69(1):117-127.
    29 Huehns TY, Gonschior P,Hofling B. Adventitia as a target for intravascular local drug delivery. Heart, 1996,75(6):537-538.
    30 Zhang L, Freedman NJ, Brian L, et al. Graft-extrinsic cells predominate in vein graft arterialization. Arterioscler Thromb Vasc Biol, 2004,24 (3):470-476.
    31 Fogelstrand P, Osterberg K,Mattsson E. Reduced neointima in vein grafts following a blockage of cell recruitment from the vein and the surrounding tissue. Cardiovasc Res, 2005,67(2):326-332.
    32 Shan J, Nguyen TB, Totary-Jain H, et al. Leptin-enhanced neointimal hyperplasia is reduced by mTOR and PI3K inhibitors. Proceedings of the National Academy of Sciences, 2008,105(48):19006-19011.
    1段亮,肖明第,游庆军,等.大孔隙非限制性血管外支架抑制移植静脉内膜增生的实验研究.中国胸心血管外科临床杂志, 2007,14(5):358-362.
    2高长青,田晓东.非限制性外支架防治移植静脉狭窄的早期形态学观察.中华胸心血管外科杂志, 2006,22(003):186-189.
    3 Vijayan V, Shukla N, Johnson JL, et al. Long-term reduction of medial and intimal thickening in porcine saphenous vein grafts with a polyglactin biodegradable external sheath. Journal of Vascular Surgery, 2004,40 (5):1011-1019.
    4张鲁锋,肖锋,李简,等.局部缓释雷帕霉素抑制静脉移植血管内膜增生.中华医学杂志, 2006,86(024):1706-1709.
    5 Moses JW, Leon MB, Popma JJ, et al. Sirolimus-eluting stents versus standard stents in patients with stenosis in a native coronary artery. N Engl J Med, 2003,349(14):1315-1323.
    6 Raja SG. Composite arterial grafting. Expert Rev Cardiovasc Ther, 2006, 4(4):523-533.
    7 Khan UA, Krishnamoorthy B, Najam O, et al. A comparative analysis of saphenous vein conduit harvesting techniques for coronary artery bypass grafting--standard bridging versus the open technique. Interact Cardiovasc Thorac Surg, 2010,10(1):27-31.
    8 Siani A, Accrocca F, Antonelli R, et al. Prejudices and realities in the use of 'unsuitable' saphenous vein graft for infrapopliteal revascularization. G Chir, 2008,29(6-7):261-264.
    9 Fonseca FA, Izar MC, Fuster V, et al. Chronic endothelial dysfunction afteroversized coronary balloon angioplasty in pigs: a 12-week follow-up of coronary vasoreactivity in vivo and in vitro. Atherosclerosis, 2001, 154(1):61-69.
    10 Kachlik D, Lametschwandtner A, Rejmontova J, et al. Vasa vasorum of the human great saphenous vein. Surg Radiol Anat, 2003,24(6):377-381.
    11 Kachlik D, Baca V, Stingl J, et al. Architectonic arrangement of the vasa vasorum of the human great saphenous vein. J Vasc Res, 2007,44 (2):157-166.
    12姚双权,张英泽,田德虎,等.移植损伤对移植静脉影响的实验研究.中国修复重建外科杂志, 2006,20(010):1021-1025.
    13 Skalak TC, Price RJ. The role of mechanical stresses in microvascular remodeling. Microcirculation, 1996,3(2):143-165.
    14 Grus T, Lindner J, Vidim T, et al. The anastomosis angle is a key to improved long-term patency of proximal femoropopliteal bypass. Ann Vasc Surg, 2009,23(5):598-605.
    15 Coppola G, Caro C. Arterial geometry, flow pattern, wall shear and mass transport: potential physiological significance. J R Soc Interface, 2009, 6(35):519-528.
    16 Dangaria JH, Butler PJ. Macrorheology and adaptive microrheology of endothelial cells subjected to fluid shear stress. Am J Physiol Cell Physiol, 2007,293(5):C1568-1575.
    17 Langille BL. Morphologic responses of endothelium to shear stress: reorganization of the adherens junction. Microcirculation, 2001,8 (3):195-206.
    18 Yamamoto K, Ando J. Shear-stress sensing via P2 purinoceptors in vascular endothelial cells. Nippon Yakurigaku Zasshi, 2004,124 (5):319-328.
    19 Maniatis NA, Brovkovych V, Allen SE, et al. Novel mechanism of endothelial nitric oxide synthase activation mediated by caveolae internalization in endothelial cells. Circ Res, 2006,99(8):870-877.
    20 Ohno M, Cooke JP, Dzau VJ, et al. Fluid shear stress induces endothelialtransforming growth factor beta-1 transcription and production. Modulation by potassium channel blockade. J Clin Invest, 1995,95 (3):1363-1369.
    21 Cotter EJ, von Offenberg Sweeney N, Coen PM, et al. Regulation of endopeptidases EC3.4.24.15 and EC3.4.24.16 in vascular endothelial cells by cyclic strain: role of Gi protein signaling. Arterioscler Thromb Vasc Biol, 2004,24(3):457-463.
    22 Morinaga K, Okadome K, Kuroki M, et al. Effect of wall shear stress on intimal thickening of arterially transplanted autogenous veins in dogs. J Vasc Surg, 1985,2(3):430-433.
    23 Dobrin PB. Mechanical factors associated with the development of intimal and medial thickening in vein grafts subjected to arterial pressure. A model of arteries exposed to hypertension. Hypertension, 1995,26(1):38-43.
    24 Berceli SA, Showalter DP, Sheppeck RA, et al. Biomechanics of the venous wall under simulated arterial conditions. J Biomech, 1990, 23(10):985-989.
    25 Owens CD, Wake N, Jacot JG, et al. Early biomechanical changes in lower extremity vein grafts--distinct temporal phases of remodeling and wall stiffness. J Vasc Surg, 2006,44(4):740-746.
    26 Stooker W, Gok M, Sipkema P, et al. Pressure-diameter relationship in the human greater saphenous vein. Ann Thorac Surg, 2003,76(5):1533-1538.
    27 Asada H, Sasajima T, Inaba M, et al. Venous blood flow through vein grafts before harvesting minimizes endothelial cell desquamation immediately after implantation. J Cardiovasc Surg (Torino), 2004,45 (2):139-142.
    28 Stooker W, Niessen HWM, Jansen EK, et al. Surgical sealant in the prevention of early vein graft injury in an ex vivo model. Cardiovascular Pathology, 2003,12(4):202-206.
    29廖东华,赵黎.自体静脉移植血管的应力应变关系及其相关组织形态学研究.中国生物医学工程学报, 2000,19(003):261-265.
    30 Sho M, Sho E, Singh TM, et al. Subnormal shear stress-induced intimalthickening requires medial smooth muscle cell proliferation and migration. Exp Mol Pathol, 2002,72(2):150-160.
    31 Kraiss LW, Geary RL, Mattsson EJ, et al. Acute reductions in blood flow and shear stress induce platelet-derived growth factor-A expression in baboon prosthetic grafts. Circ Res, 1996,79(1):45-53.
    32 Song RH, Kocharyan HK, Fortunato JE, et al. Increased flow and shear stress enhance in vivo transforming growth factor-beta1 after experimental arterial injury. Arterioscler Thromb Vasc Biol, 2000,20(4):923-930.
    33 Meguro T, Nakashima H, Kawada S, et al. Effect of external stenting and systemic hypertension on intimal hyperplasia in rat vein grafts. Neurosurgery, 2000,46(4):963-969; discussion 969-970.
    34 Barra JA, Volant A, Leroy JP, et al. Constrictive perivenous mesh prosthesis for preservation of vein integrity. Experimental results and application for coronary bypass grafting. J Thorac Cardiovasc Surg, 1986, 92(3 Pt 1):330-336.
    35 Powell JT, Gosling M. Molecular and cellular changes in vein grafts: influence of pulsatile stretch. Curr Opin Cardiol, 1998,13(6):453-458.
    36 Angelini GD, Lloyd C, Bush R, et al. An external, oversized, porous polyester stent reduces vein graft neointima formation, cholesterol concentration, and vascular cell adhesion molecule 1 expression in cholesterol-fed pigs. J Thorac Cardiovasc Surg, 2002,124(5):950-956.
    37 Izzat MB, Mehta D, Bryan AJ, et al. Influence of external stent size on early medial and neointimal thickening in a pig model of saphenous vein bypass grafting. Circulation, 1996,94(7):1741-1745.
    38秦任甲.动脉粥样硬化好发部位血液流态和参数分析.中国医学物理学杂志, 2009,26(002):1102-1104.
    39 Teng ZZ, Ji GY, Chu HJ, et al. Does PGA external stenting reduce compliance mismatch in venous grafts?. Biomed Eng Online, 2007,6:12.
    40廖东华,赵黎.自体静脉移植血管的应力应变关系及其相关组织形态学研究.中国生物医学工程学报, 2000,19(3):261-265.
    41 George SJ, Izzat MB, Gadsdon P, et al. Macro-porosity is necessary for thereduction of neointimal and medial thickening by external stenting of porcine saphenous vein bypass grafts. Atherosclerosis, 2001,155 (2):329-336.
    1段亮,肖明第,游庆军,等.大孔隙非限制性血管外支架抑制移植静脉内膜增生的实验研究.中国胸心血管外科临床杂志, 2007,14(005):358-362.
    2 Barker, SGE, Tilling, LC, Miller, GC, et al. The adventitia and atherogenesis: removal initiates intimal proliferation in the rabbit which regresseson generation of a'neoadventitia'. Atherosclerosis, 1994,105(2): 131-144.
    3 George SJ, Izzat MB, Gadsdon P, et al. Macro-porosity is necessary for the reduction of neointimal and medial thickening by external stenting of porcine saphenous vein bypass grafts. Atherosclerosis, 2001,155(2): 329-336.
    4 Fogelstrand P, Osterberg K,Mattsson E. Reduced neointima in vein grafts following a blockage of cell recruitment from the vein and the surrounding tissue. Cardiovasc Res, 2005,67(2):326-332.
    5 Zhang L, Freedman NJ, Brian L, et al. Graft-extrinsic cells predominate in vein graft arterialization. Arterioscler Thromb Vasc Biol, 2004,24 (3):470-476.
    6 Tuncay I, Ozbek H, Atik B, et al. Effects of hyaluronic acid on postoperative adhesion of tendo calcaneus surgery: an experimental studyin rats. J Foot Ankle Surg, 2002,41(2):104-108.
    7 Menderes A, Mola F, Tayfur V, et al. Prevention of peritendinous adhesions following flexor tendon injury with seprafilm. Ann Plast Surg, 2004,53(6):560-564.
    8 Nishida J, Araki S, Akasaka T, et al. Effect of hyaluronic acid on the excursion resistance of tendon grafts. A biomechanical study in a canine model in vitro. J Bone Joint Surg Br, 2004,86(6):918-924.
    9张鲁锋,肖锋,李简,等.局部缓释雷帕霉素抑制静脉移植血管内膜增生.中华医学杂志, 2006,86(024):1706-1709.
    10 Sharma P, Maffulli N. Tendon injury and tendinopathy: healing and repair. J Bone Joint Surg Am, 2005,87(1):187-202.
    11 Halici M, Karaoglu S, Canoz O, et al. Sodium hyaluronate regulating angiogenesis during Achilles tendon healing. Knee Surg Sports Traumatol Arthrosc, 2004,12(6):562-567.
    12 Nehez L, Vodros D, Axelsson J, et al. Prevention of postoperative peritoneal adhesions: effects of lysozyme, polylysine and polyglutamate versus hyaluronic acid. Scand J Gastroenterol, 2005,40(9):1118-1123.
    13 Angelini GD, Izzat MB, Bryan AJ, et al. External stenting reduces early medial and neointimal thickening in a pig model of arteriovenous bypass grafting. J Thorac Cardiovasc Surg, 1996,112(1):79-84.
    14 Izzat MB, Mehta D, Bryan AJ, et al. Influence of external stent size on early medial and neointimal thickening in a pig model of saphenous vein bypass grafting. Circulation, 1996,94(7):1741-1745.
    15王韶进,戴国锋,李昕,等.透明质酸钠预防屈肌腱粘连的临床研究.中国修复重建外科杂志, 2002,16(001):28-30.
    16 Estes, JM, Adzick, NS, Harrison, MR, et al. Hyaluronate metabolism undergoes an ontogenic transition during fetal development: implications for scar-free wound healing. Discussion. Journal of pediatric surgery, 1993, 28(10):1227-1231.
    17 Buwitt U, Koch C, Tatje D, et al. Platelet-derived growth factor isoforms AA, AB, and BB differentially activate poly r(I):r(C)-induced genes inhuman fibroblast FS4 cells. DNA Cell Biol, 1992,11(9):641-650.
    18 Kenagy RD, Fukai N, Min SK, et al. Proliferative capacity of vein graft smooth muscle cells and fibroblasts in vitro correlates with graft stenosis. J Vasc Surg, 2009,49(5):1282-1288.
    19 Zhang J, Fu M, Zhu X, et al. Peroxisome proliferator-activated receptor delta is up-regulated during vascular lesion formation and promotes post-confluent cell proliferation in vascular smooth muscle cells. J Biol Chem, 2002,277(13):11505-11512.
    20 Lozano E, Segarra M, Garcia-Martinez A, et al. Imatinib mesylate inhibits in vitro and ex vivo biological responses related to vascular occlusion in giant cell arteritis. Ann Rheum Dis, 2008,67(11):1581-1588.
    21 Willis AI, Pierre-Paul D, Sumpio BE, et al. Vascular smooth muscle cell migration: current research and clinical implications. Vasc Endovascular Surg, 2004,38(1):11-23.
    22 Mehta, D, George, SJ, Jeremy, JY, et al. External stenting reduces long-term medial and neointimal thickening and platelet derived growth factor expression in a pig model of arteriovenous bypass grafting. Nature medicine, 1998,4(2):235-239.
    23 Ishiwata, S, Tukada, T, Nakanishi, S, et al. Postangioplasty restenosis: platelet activation and the coagulation-fibrinolysis system as possible factors in the pathogenesis of restenosis. American Heart Journal, 1997, 133(4):387-392.
    24 Fox PL, DiCorleto PE. Regulation of production of a platelet-derived growth factor-like protein by cultured bovine aortic endothelial cells. J Cell Physiol, 1984,121(2):298-308.
    25 Sj lund, M, Rahm, M, Claesson-welsh, L, et al. Expression of PDGFα-andβ-receptors in rat arterial smooth muscle cells is phenotype and growth state dependent. Growth Factors, 1990,3(3):191-203.
    26 Roy-Chaudhury P, Kelly BS, Miller MA, et al. Venous neointimal hyperplasia in polytetrafluoroethylene dialysis grafts. Kidney Int, 2001,59 (6):2325-2334.
    27 Bowen-Pope, DF, Hart, CE,Seifert, RA. Sera and conditioned media contain different isoforms of platelet-derived growth factor (PDGF) which bind to different classes of PDGF receptor. J Biol Chem, 1989,264 (5):2502-2508.
    28 Mehta D, George SJ, Jeremy JY, et al. External stenting reduces long-term medial and neointimal thickening and platelet derived growth factor expression in a pig model of arteriovenous bypass grafting. Nat Med, 1998, 4(2):235-239.
    29 Kachlik D, Lametschwandtner A, Rejmontova J, et al. Vasa vasorum of the human great saphenous vein. Surg Radiol Anat, 2003,24(6):377-381.
    30 Kachlik D, Baca V, Stingl J, et al. Architectonic arrangement of the vasa vasorum of the human great saphenous vein. J Vasc Res, 2007,44 (2):157-166.
    31 Aviad, AD, Houpt, JB. The molecular weight of therapeutic hyaluronan (sodium hyaluronate): how significant is it? Journal of rheumatology, 1994,21(2):297-301.
    1 Moses JW, Leon MB, Popma JJ, et al. Sirolimus-eluting stents versus standard stents in patients with stenosis in a native coronary artery. N Engl J Med, 2003,349(14):1315-1323.
    2 Raja SG. Composite arterial grafting. Expert Rev Cardiovasc Ther, 2006, 4(4):523-533.
    3 Fogelstrand P, Feral CC, Zargham R, et al. Dependence of proliferative vascular smooth muscle cells on CD98hc (4F2hc, SLC3A2). J Exp Med, 2009,206(11):2397-2406.
    4 Yucel S, Bahcivan M, Gol MK, et al. Reduced intimal hyperplasia in rabbits via medical therapy after carotid venous bypass. Tex Heart Inst J, 2009,36(5):387-392.
    5刘继红,何学志,谷天祥,等.局部应用紫杉醇预防兔移植静脉再狭窄.中国胸心血管外科临床杂志, 2008,15(002):126-129.
    6 Souza DS. A new no-touch preparation technique: technical notes. Scandinavian Cardiovascular Journal, 1996,30(1):41-44.
    7 Souza DS, Christofferson RH, Bomfim V, et al. " No-touch" technique using saphenous vein harvested with its surrounding tissue for coronary artery bypass grafting maintains an intact endothelium. Scandinavian Cardiovascular Journal, 1999,33(6):323-329.
    8 Muzaffar, S, Shukla, N, Angelini, GD, et al. Hypoxia and the expression of gp91 phox and endothelial nitric oxide synthase in the pulmonary artery. Thorax, 2005,60:305-313.
    9 Siow RC, Churchman AT. Adventitial growth factor signalling and vascular remodelling: potential of perivascular gene transfer from the outside-in. Cardiovasc Res, 2007,75(4):659-668.
    10 Parang P, Arora R. Coronary vein graft disease: pathogenesis and prevention. Can J Cardiol, 2009,25(2):e57-62.
    11 Souza DS, Dashwood MR, Tsui J, et al. Improved patency in vein grafts harvested with surrounding tissue: results of a randomized study using three harvesting techniques. The Annals of thoracic surgery, 2002, 73(4):1189.
    12 Souza DS, Dashwood MR, Tsui JC, et al. Improved patency in vein grafts harvested with surrounding tissue: results of a randomized study using three harvesting techniques. Ann Thorac Surg, 2002,73(4):1189-1195.
    13 Dashwood MR, Savage K, Tsui JC, et al. Retaining perivascular tissue of human saphenous vein grafts protects against surgical and distension-induced damage and preserves endothelial nitric oxide synthase and nitric oxide synthase activity. J Thorac Cardiovasc Surg, 2009,138 (2):334-340.
    14 Khan UA, Krishnamoorthy B, Najam O, et al. A comparative analysis of saphenous vein conduit harvesting techniques for coronary artery bypass grafting--standard bridging versus the open technique. Interact Cardiovasc Thorac Surg, 2010,10(1):27-31.
    15 Lopes RD, Hafley GE, Allen KB, et al. Endoscopic versus open vein-graft harvesting in coronary-artery bypass surgery. N Engl J Med, 2009,361 (3):235-244.
    16 Ramakrishna H, Fassl J, Sinha A, et al. The year in cardiothoracic and vascular anesthesia: selected highlights from 2009. J Cardiothorac Vasc Anesth, 2010,24(1):7-17.
    17 Adcock GD, Adcock OT Jr, Wheeler JR, et al. Arterialization of reversed autogenous vein grafts: quantitative light and electron microscopy of canine jugular vein grafts harvested and implanted by standard or improved techniques. J Vasc Surg, 1987,6(3):283-295.
    18 Metcalfe MJ, Lip GY,Dargie HJ. Factors influencing coronary artery bypass graft patency. Cardiovasc Surg, 1994,2(6):679-685.
    19 Schaeffer U, Tanner B, Strohschneider T, et al. Damage to arterial and venous endothelial cells in bypass grafts induced by several solutions used in bypass surgery. Thorac Cardiovasc Surg, 1997,45(4):168-171.
    20 Rosenfeldt FL, He GW, Buxton BF, et al. Pharmacology of coronary artery bypass grafts. Ann Thorac Surg, 1999,67(3):878-888.
    21 Jesuthasan LS, Angus JA,Rosenfeldt FL. In vitro comparison of glyceryl trinitrate-verapamil with other dilators of human saphenous vein. ANZ JSurg, 2003,73(5):313-320.
    22 Thatte HS, Biswas KS, Najjar SF, et al. Multi-photon microscopic evaluation of saphenous vein endothelium and its preservation with a new solution, GALA. Ann Thorac Surg, 2003,75(4):1145-52, 1152.
    23 He W, Yong T, Teo WE, et al. Fabrication and endothelialization of collagen-blended biodegradable polymer nanofibers: potential vascular graft for blood vessel tissue engineering. Tissue Eng, 2005,11 (9-10):1574-1588.
    24王明松,邹良建,黄盛东,等.自体移植静脉内皮细胞的缺血安全时限.中国组织工程研究与临床康复, 2007,11(008):1401-1404.
    25 Ishiwata, S, Tukada, T, Nakanishi, S, et al. Postangioplasty restenosis: platelet activation and the coagulation-fibrinolysis system as possible factors in the pathogenesis of restenosis. American Heart Journal, 1997, 133(4):387-392.
    26 Zimmermann N, Gams E,Hohlfeld T. Aspirin in coronary artery bypass surgery: new aspects of and alternatives for an old antithrombotic agent. Eur J Cardiothorac Surg, 2008,34(1):93-108.
    27 Kawasome H, Papst P, Webb S, et al. Targeted disruption of p70(s6k) defines its role in protein synthesis and rapamycin sensitivity. Proc Natl Acad Sci U S A, 1998,95(9):5033-5038.
    28 Sugo S, Minamino N, Kangawa K, et al. Endothelial cells actively synthesize and secrete adrenomedullin. Biochem Biophys Res Commun, 1994,201(3):1160-1166.
    29 Norman PE, House AK. Heparin reduces the intimal hyperplasia seen in microvascular vein grafts. Aust N Z J Surg, 1991,61(12):942-948.
    30 Cambria RP, Ivarsson BL, Fallon JT, et al. Heparin fails to suppress intimal proliferation in experimental vein grafts. Surgery, 1992,111 (4):424-429.
    31 Wilson NV, Salisbury JR,Kakkar VV. The effect of low molecular weight heparin on intimal hyperplasia in vein grafts. Eur J Vasc Surg, 1994, 8(1):60-64.
    32 Stohr S, Meyer T, Smolenski A, et al. Effects of heparin on aortic versus venous smooth muscle cells: similar binding with different rates of [3H]thymidine incorporation. J Cardiovasc Pharmacol, 1995,25 (5):782-788.
    33 Varty K, Allen KE, Jones L, et al. The influence of low molecular weight heparin on neointimal proliferation in cultured human saphenous vein. Eur J Vasc Surg, 1994,8(2):174-178.
    34 Yamazaki T, Taniguchi I, Kurusu T, et al. Effect of amlodipine on vascular responses after coronary stenting compared with an angiotensin-converting enzyme inhibitor. Circ J, 2004,68(4):328-333.
    35 Muller DW, Golomb G, Gordon D, et al. Site-specific dexamethasone delivery for the prevention of neointimal thickening after vascular stent implantation. Coron Artery Dis, 1994,5(5):435-442.
    36王剑,刘宏宇,李君权,等.左旋精氨酸对大鼠自体移植静脉内膜增生的影响.哈尔滨医科大学学报, 2004,28(006):515-517.
    37王春喜,吴咸中.内皮素, NO与自体静脉移植再狭窄的关系.中华实验外科杂志, 1999,16(005):400-401.
    38 Nunes GL, Hanson SR, King SB 3rd, et al. Local delivery of a synthetic antithrombin with a hydrogel-coated angioplasty balloon catheter inhibits platelet-dependent thrombosis. J Am Coll Cardiol, 1994,23(7):1578-1583.
    39 Porter KE, Turner NA. Statins for the prevention of vein graft stenosis: a role for inhibition of matrix metalloproteinase-9. Biochem Soc Trans, 2002,30(2):120-126.
    40杨云波,廖端芳,朱炳阳.普罗布可抗兔动脉成形术后再狭窄与调节血管重塑的关系[J].中国病理生理杂志, 2003,19(9):1174-1177.
    41宋自芳,李伟,郑启昌,等.阿托伐他汀对大鼠自体移植静脉内膜增生的影响.中国病理生理杂志, 2009,25(004):631-635.
    42 Fulton GJ, Davies MG, Barber L, et al. Localized versus systemic angiotensin II receptor inhibition of intimal hyperplasia in experimental vein grafts by the specific angiotensin II receptor inhibitor L158,809. Surgery, 1998,123(2):218-227.
    43 Okumura K, Sone T, Kondo J, et al. Quinapril prevents restenosis after coronary stenting in patients with angiotensin-converting enzyme D allele. Circ J, 2002,66(4):311-316.
    44 Yoshida O, Hirayama H, Nanasato M, et al. The angiotensin II receptor blocker candesartan cilexetil reduces neointima proliferation after coronary stent implantation: a prospective randomized study under intravascular ultrasound guidance. Am Heart J, 2005,149(1):e1-e6.
    45 Ujiie, Y, Hirosaka, A, Mitsugi, M, et al. Effects of angiotensin-converting enzyme inhibitors or an angiotensin receptor blocker in combination with aspirin and cilostazol on in-stent restenosis. International Heart Journal, 2006,47(2):173-184.
    46 Song QB, Wei MJ, Duan ZQ, et al. Batroxobin reduces intracellular calcium concentration and inhibits proliferation of vascular smooth muscle cells. Chin Med J (Engl), 2004,117(6):917-921.
    47 Margolin L, Fishbein I, Banai S, et al. Metalloproteinase inhibitor attenuates neointima formation and constrictive remodeling after angioplasty in rats: augmentative effect ofαvβ3 receptor blockade. Atherosclerosis, 2002,163(2):269-277.
    48 Bendeck MP, Conte M, Zhang M, et al. Doxycycline modulates smooth muscle cell growth, migration, and matrix remodeling after arterial injury. Am J Pathol, 2002,160(3):1089-1095.
    49 Gallo R, Padurean A, Jayaraman T, et al. Inhibition of intimal thickening after balloon angioplasty in porcine coronary arteries by targeting regulators of the cell cycle. Circulation, 1999,99(16):2164-2170.
    50 Suzuki T, Kopia G, Hayashi S, et al. Stent-based delivery of sirolimus reduces neointimal formation in a porcine coronary model. Circulation, 2001,104(10):1188-1193.
    51 Doostzadeh J, Clark LN, Bezenek S, et al. Recent progress in percutaneous coronary intervention: evolution of the drug-eluting stents, focus on the XIENCE V drug-eluting stent. Coron Artery Dis, 2010,21(1):46-56.
    52 Wang XT, Venkatraman S, Boey F, et al. Effects of controlled-releasedsirolimus from polymer matrices on human coronary artery smooth muscle cells. Journal of Biomaterials Science, Polymer Edition, 2007,18 (11):1401-1414.
    53陈宏林,尤庆生,沈卫东,等.雷帕霉素抑制兔自体移植静脉内膜过度增殖.江苏医药, 2007,33(005):500-502.
    54 Berk BC, Gordon JB,Alexander RW. Pharmacologic roles of heparin and glucocorticoids to prevent restenosis after coronary angioplasty. J Am Coll Cardiol, 1991,17(6 Suppl B):111-117.
    55武国,时德.局部缓释雷公藤内酯醇对移植静脉平滑肌细胞增殖和凋亡的影响.重庆医科大学学报, 2008,33(006):683-685.
    56 Leon MB, Mauri L, Popma JJ, et al. A Randomized Comparison of the Endeavor Zotarolimus-Eluting Stent Versus the TAXUS Paclitaxel-Eluting Stent in De Novo Native Coronary Lesions 12-Month Outcomes From the ENDEAVOR IV Trial. J Am Coll Cardiol, 2010,55(6):543-554.
    57郭明珂,张英泽,田德虎,等.局部应用5-氟尿嘧啶抑制移植静脉内膜增生的实验研究.中国修复重建外科杂志, 2009,23(8):940-946.
    58王庆涛,张强,啜东宇.甲硫氨酸脑啡肽对移植静脉再狭窄的作用用机制.实用新医学, 2007,8(004):299-302.
    59龚裕兴,王伟,傅玉才,等.大鼠主动脉内膜损伤后新生内膜各种生长因子表达与表没食子儿茶素没食子酸酯的干预.中国组织工程研究与临床康复, 2007,11(023):4473-4479.
    60胡佳心,阮新民,石国亮,等.金钠多注射液预防静脉桥再狭窄的实验研究.辽宁中医药大学学报, 2008,10(012):156-158.
    61翁书和,赵军礼,陈镜合,等.莪术油对动脉损伤大鼠血管成形术后再狭窄的抑制作用.广州中医药大学学报, 2003,20(004):282-284.
    62李琦,温进坤,韩梅.黄芪和当归对血管平滑肌细胞表型标志基因表达和细胞增殖的影响.中国动脉硬化杂志, 2004,12(002):147-150.
    63 Celletti FL, Waugh JM, Amabile PG, et al. Vascular endothelial growth factor enhances atherosclerotic plaque progression. Nat Med, 2001,7 (4):425-429.
    64 Bond M, Murphy G, Bennett MR, et al. Localization of the death domainof tissue inhibitor of metalloproteinase-3 to the N terminus. Metalloproteinase inhibition is associated with proapoptotic activity. J Biol Chem, 2000,275(52):41358-41363.
    65 Sugimoto M, Yamanouchi D,Komori K. Therapeutic approach against intimal hyperplasia of vein grafts through endothelial nitric oxide synthase/nitric oxide (eNOS/NO) and the Rho/Rho-kinase pathway. Surg Today, 2009,39(6):459-465.
    66 O'Connor DM, O'Brien T. Nitric oxide synthase gene therapy: progress and prospects. Expert Opin Biol Ther, 2009,9(7):867-878.
    67 Wu S, Wang X, Guo L, et al. Adenovirus mediated endothelial nitric oxide synthase gene transfer prevents restenosis of vein grafts. ASAIO J, 2004, 50(3):272-277.
    68 Sharif F, Hynes SO, Cooney R, et al. Gene-eluting stents: adenovirus-mediated delivery of eNOS to the blood vessel wall accelerates re-endothelialization and inhibits restenosis. Mol Ther, 2008,16 10):1674-1680.
    69 Fetalvero KM, Martin KA,Hwa J. Cardioprotective prostacyclin signaling in vascular smooth muscle. Prostaglandins Other Lipid Mediat, 2007,82 (1-4):109-118.
    70 Eefting D, de Vries MR, Grimbergen JM, et al. In vivo suppression of vein graft disease by nonviral, electroporation-mediated, gene transfer of tissue inhibitor of metalloproteinase-1 linked to the amino terminal fragment of urokinase (TIMP-1.ATF), a cell-surface directed matrix metalloproteinase inhibitor. J Vasc Surg, 2010,51(2):429-437.
    71 Yang J, Zeng Y, Li Y, et al. Intravascular site-specific delivery of a therapeutic antisense for the inhibition of restenosis. Eur J Pharm Sci, 2008,35(5):427-434.
    72 Johnson JL, van Eys GJ, Angelini GD, et al. Injury induces dedifferentiation of smooth muscle cells and increased matrix-degrading metalloproteinase activity in human saphenous vein. Arterioscler Thromb Vasc Biol, 2001,21(7):1146-1151.
    73 Ehsan A, Mann MJ, Dell'Acqua G, et al. Long-term stabilization of vein graft wall architecture and prolonged resistance to experimental atherosclerosis after E2F decoy oligonucleotide gene therapy. J Thorac Cardiovasc Surg, 2001,121(4):714-722.
    74 Wolff RA, Malinowski RL, Heaton NS, et al. Transforming growth factor-beta1 antisense treatment of rat vein grafts reduces the accumulation of collagen and increases the accumulation of h-caldesmon. J Vasc Surg, 2006,43(5):1028-1036.
    75 Kipshidze NN, Iversen P, Kim HS, et al. Advanced c-myc antisense (AVI-4126)-eluting phosphorylcholine-coated stent implantation is associated with complete vascular healing and reduced neointimal formation in the porcine coronary restenosis model. Catheter Cardiovasc Interv, 2004,61(4):518-527.
    76 George SJ, Angelini GD, Capogrossi MC, et al. Wild-type p53 gene transfer inhibits neointima formation in human saphenous vein by modulation of smooth muscle cell migration and induction of apoptosis. Gene Ther, 2001,8(9):668-676.
    77 Owens CD, Wake N, Jacot JG, et al. Early biomechanical changes in lower extremity vein grafts--distinct temporal phases of remodeling and wall stiffness. J Vasc Surg, 2006,44(4):740-746.
    78 Stooker W, Gok M, Sipkema P, et al. Pressure-diameter relationship in the human greater saphenous vein. Ann Thorac Surg, 2003,76(5):1533-1538.
    79 Dobrin PB. Mechanical factors associated with the development of intimal and medial thickening in vein grafts subjected to arterial pressure. A model of arteries exposed to hypertension. Hypertension, 1995,26(1):38-43.
    80 Sho M, Sho E, Singh TM, et al. Subnormal shear stress-induced intimal thickening requires medial smooth muscle cell proliferation and migration. Exp Mol Pathol, 2002,72(2):150-160.
    81 Kraiss LW, Geary RL, Mattsson EJ, et al. Acute reductions in blood flow and shear stress induce platelet-derived growth factor-A expression in baboon prosthetic grafts. Circ Res, 1996,79(1):45-53.
    82 Song RH, Kocharyan HK, Fortunato JE, et al. Increased flow and shear stress enhance in vivo transforming growth factor-beta1 after experimental arterial injury. Arterioscler Thromb Vasc Biol, 2000,20(4):923-930.
    83 Meguro T, Nakashima H, Kawada S, et al. Effect of external stenting and systemic hypertension on intimal hyperplasia in rat vein grafts. Neurosurgery, 2000,46(4):963-969; discussion 969-970.
    84 Barra JA, Volant A, Leroy JP, et al. Constrictive perivenous mesh prosthesis for preservation of vein integrity. Experimental results and application for coronary bypass grafting. J Thorac Cardiovasc Surg, 1986,92(3 Pt 1):330-336.
    85 Powell JT, Gosling M. Molecular and cellular changes in vein grafts: influence of pulsatile stretch. Curr Opin Cardiol, 1998,13(6):453-458.
    86 Angelini GD, Lloyd C, Bush R, et al. An external, oversized, porous polyester stent reduces vein graft neointima formation, cholesterol concentration, and vascular cell adhesion molecule 1 expression in cholesterol-fed pigs. J Thorac Cardiovasc Surg, 2002,124(5):950-956.
    87 Izzat MB, Mehta D, Bryan AJ, et al. Influence of external stent size on early medial and neointimal thickening in a pig model of saphenous vein bypass grafting. Circulation, 1996,94(7):1741-1745.
    88 Zweep HP, Satoh S, van der Lei B, et al. Degradation of a supporting prosthesis can optimize arterialization of autologous veins. Ann Thorac Surg, 1993,56(5):1117-1122.
    89 Jeremy JY, Bulbulia R, Johnson JL, et al. A bioabsorbable (polyglactin), nonrestrictive, external sheath inhibits porcine saphenous vein graft thickening. J Thorac Cardiovasc Surg, 2004,127(6):1766-1772.
    90杨斌,吴清玉,李巅远,等.可降解壳聚糖血管外周支持与静脉移植物早期结构的变化.中华外科杂志, 2003,41(009):688-690.
    91 Huynh, TTT, Davies, MG, Trovato, MJ, et al. Alterations in wall tension and shear stress modulate tyrosine kinase signaling and wall remodeling in experimental vein grafts. Journal of Vascular Surgery, 1999,29(2):334-344.
    92 Krejca, M, Skarysz, J, Szmagala, P, et al. A new outside stent-does itprevent vein graft intimal proliferation? European Journal of Cardio-Thoracic Surgery, 2002,22(6):898-903.
    93 Violaris AG, Newby AC,Angelini GD. Effects of external stenting on wall thickening in arteriovenous bypass grafts. Ann Thorac Surg, 1993,55 (3):667-671.
    94 George, SJ, Izzat, MB, Gadsdon, P, et al. Macro-porosity is necessary for the reduction of neointimal and medial thickening by external stenting of porcine saphenous vein bypass grafts. Atherosclerosis, 2001,155 (2):329-336.
    95 Stooker, W, Niessen, HWM, Wildevuur, WR, et al. Perivenous application of fibrin glue reduces early injury to the human saphenous vein graft wall in an ex vivo model. European Journal of Cardio-Thoracic Surgery, 2002, 21(2):212-217.
    96 George SJ, Izzat MB, Gadsdon P, et al. Macro-porosity is necessary for the reduction of neointimal and medial thickening by external stenting of porcine saphenous vein bypass grafts. Atherosclerosis, 2001,155 (2):329-336.
    97 Karayannacos PE, Hostetler JR, Bond MG, et al. Late failure in vein grafts: mediating factors in subendothelial fibromuscular hyperplasia. Ann Surg, 1978,187(2):183-188.
    98 Sarac TP, Carnevale K, Smedira N, et al. In vivo and mechanical properties of peritoneum/fascia as a novel arterial substitute. J Vasc Surg, 2005,41(3):490-497.
    99郭明珂,张奇,陈占法,等.自体静脉外支架对移植静脉内膜增生影响的实验研究.中华外科杂志, 2010,48(2):138-141.
    100 Crom R, Wulf P, van Nimwegen H, et al. Irradiated versus nonirradiated endothelial cells: effect on proliferation of vascular smooth muscle cells. J Vasc Interv Radiol, 2001,12(7):855-861.
    101 Scott NA, Crocker IR, Yin Q, et al. Inhibition of vascular cell growth by X-ray irradiation: comparison with gamma radiation and mechanism of action. Int J Radiat Oncol Biol Phys, 2001,50(2):485-493.
    102 Waksman, R, Raizner, AE, Yeung, AC, et al. Use of localised intracoronaryβradiation in treatment of in-stent restenosis: the inhibit randomised controlled trial. The Lancet, 2002,359 (9306):551-557.
    103郎晓讴,李军,曾克,等. 32P局部照射对移植静脉内膜平滑肌细胞增殖与凋亡的影响.中华外科杂志, 2002,40(002):133-135.
    104 Powers, BE, Thames, HD,Gillette, EL. Long-term adverse effects of radiation inhibition of restenosis: radiation injury to the aorta and branch arteries in a canine model. International Journal of Radiation Oncology Biology Physics, 1999,45(3):753-759.
    1 Bacchetta MD, Salemi A, Milla F, et al. Low-dose spironolactone: effects on artery-to-artery vein grafts and percutaneous coronary intervention sites. Am J Ther, 2009,16(3):204-214.
    2 Raja SG. Composite arterial grafting. Expert Rev Cardiovasc Ther, 2006, 4(4):523-533.
    3 Khan UA, Krishnamoorthy B, Najam O, et al. A comparative analysis of saphenous vein conduit harvesting techniques for coronary artery bypass grafting--standard bridging versus the open technique. Interact Cardiovasc Thorac Surg, 2010,10(1):27-31.
    4 Siani A, Accrocca F, Antonelli R, et al. Prejudices and realities in the use of 'unsuitable' saphenous vein graft for infrapopliteal revascularization. G Chir, 2008,29(6-7):261-264.
    5 Ballermann BJ, Marsden PA. Endothelium-derived vasoactive mediators and renal glomerular function. Clin Invest Med, 1991,14(6):508-517.
    6 Botting R, Vane JR. Vasoactive mediators derived from the endothelium. Arch Mal Coeur Vaiss, 1989,82 Spec No 4:11-14.
    7 Fonseca FA, Izar MC, Fuster V, et al. Chronic endothelial dysfunction after oversized coronary balloon angioplasty in pigs: a 12-week follow-up of coronary vasoreactivity in vivo and in vitro. Atherosclerosis, 2001,154 (1):61-69.
    8 Kachlik D, Lametschwandtner A, Rejmontova J, et al. Vasa vasorum of the human great saphenous vein. Surg Radiol Anat, 2003,24(6):377-381.
    9 Kachlik D, Baca V, Stingl J, et al. Architectonic arrangement of the vasa vasorum of the human great saphenous vein. J Vasc Res, 2007,44 (2):157-166.
    10 Ishiwata, S, Tukada, T, Nakanishi, S, et al. Postangioplasty restenosis: platelet activation and the coagulation-fibrinolysis system as possible factors in the pathogenesis of restenosis. American Heart Journal, 1997, 133(4):387-392.
    11 Schwarz JB, Langwieser N, Langwieser NN, et al. Novel role of the CXC chemokine receptor 3 in inflammatory response to arterial injury: involvement of mTORC1. Circ Res, 2009,104(2):189-200, 8p following 200.
    12 Verrier ED, Morgan EN. Endothelial response to cardiopulmonary bypass surgery. Ann Thorac Surg, 1998,66(5 Suppl):S17-19; discussion S25-28.
    13 Stark VK, Hoch JR, Warner TF, et al. Monocyte chemotactic protein-1 expression is associated with the development of vein graft intimal hyperplasia. Arterioscler Thromb Vasc Biol, 1997,17(8):1614-1621.
    14 Eefting D, de Vries MR, Grimbergen JM, et al. In vivo suppression of vein graft disease by nonviral, electroporation-mediated, gene transfer of tissue inhibitor of metalloproteinase-1 linked to the amino terminal fragment of urokinase (TIMP-1.ATF), a cell-surface directed matrix metalloproteinase inhibitor. J Vasc Surg, 2010,51(2):429-437.
    15 Allaire E, Clowes AW. Endothelial cell injury in cardiovascular surgery: the intimal hyperplastic response. Ann Thorac Surg, 1997,63(2):582-591.
    16 Lardenoye JH, de Vries MR, Deckers M, et al. Inhibition of intimal hyperplasia by the tetracycline derived mmp inhibitor doxycycline in vein graft disease in vitro and in vivo. EuroIntervention, 2005,1(2):236-243.
    17 Zhang J, Fu M, Zhu X, et al. Peroxisome proliferator-activated receptor delta is up-regulated during vascular lesion formation and promotes post-confluent cell proliferation in vascular smooth muscle cells. J Biol Chem, 2002,277(13):11505-11512.
    18 Lozano E, Segarra M, Garcia-Martinez A, et al. Imatinib mesylate inhibitsin vitro and ex vivo biological responses related to vascular occlusion in giant cell arteritis. Ann Rheum Dis, 2008,67(11):1581-1588.
    19 Willis AI, Pierre-Paul D, Sumpio BE, et al. Vascular smooth muscle cell migration: current research and clinical implications. Vasc Endovascular Surg, 2004,38(1):11-23.
    20 Patel VA, Zhang QJ, Siddle K, et al. Defect in insulin-like growth factor-1 survival mechanism in atherosclerotic plaque-derived vascular smooth muscle cells is mediated by reduced surface binding and signaling. Circ Res, 2001,88(9):895-902.
    21 Camozzi M, Zacchigna S, Rusnati M, et al. Pentraxin 3 inhibits fibroblast growth factor 2-dependent activation of smooth muscle cells in vitro and neointima formation in vivo. Arterioscler Thromb Vasc Biol, 2005,25 (9):1837-1842.
    22 Andersen HR, Maeng M, Thorwest M, et al. Remodeling rather than neointimal formation explains luminal narrowing after deep vessel wall injury: insights from a porcine coronary (re)stenosis model. Circulation, 1996,93(9):1716-1724.
    23 Siow RC, Mallawaarachchi CM,Weissberg PL. Migration of adventitial myofibroblasts following vascular balloon injury: insights from in vivo gene transfer to rat carotid arteries. Cardiovasc Res, 2003,59(1):212-221.
    24 Li G, Chen SJ, Oparil S, et al. Direct in vivo evidence demonstrating neointimal migration of adventitial fibroblasts after balloon injury of rat carotid arteries. Circulation, 2000,101(12):1362-1365.
    25 Stary HC, Blankenhorn DH, Chandler AB, et al. A definition of the intima of human arteries and of its atherosclerosis-prone regions. A report from the Committee on Vascular Lesions of the Council on Arteriosclerosis, American Heart Association. Circulation, 1992,85(1):391-405.
    26 Wu L, Zhang W, Li H, et al. Inhibition of aortic intimal hyperplasia and cell cycle protein and extracellular matrix protein expressions by BuYang HuanWu Decoction. J Ethnopharmacol, 2009,125(3):423-435.
    27 Wu J, Zhang C. Neointimal hyperplasia, vein graft remodeling, andlong-term patency. Am J Physiol Heart Circ Physiol, 2009,297 (4):H1194-1195.
    28 Berceli SA, Tran-Son-Tay R, Garbey M, et al. Hemodynamically driven vein graft remodeling: a systems biology approach. Vascular, 2009,17 Suppl 1:S2-9.
    29 Bush HL Jr, McCabe ME,Nabseth DC. Functional injury of vein graft endothelium. Role of hypothermia and distention. Arch Surg, 1984,119 (7):770-774.
    30 Menashi S, Lu H, Soria C, et al. Endothelial cell proteases: physiological role and regulation. Baillieres Clin Haematol, 1993,6(3):559-576.
    31 Pye DA, Kumar S. Endothelial and fibroblast cell-derived heparan sulphate bind with differing affinity to basic fibroblast growth factor. Biochem Biophys Res Commun, 1998,248(3):889-895.
    32 Levin EG. Latent tissue plasminogen activator produced by human endothelial cells in culture: evidence for an enzyme-inhibitor complex. Proc Natl Acad Sci U S A, 1983,80(22):6804-6808.
    33 Fox PL, DiCorleto PE. Regulation of production of a platelet-derived growth factor-like protein by cultured bovine aortic endothelial cells. J Cell Physiol, 1984,121(2):298-308.
    34 Vlodavsky I, Folkman J, Sullivan R, et al. Endothelial cell-derived basic fibroblast growth factor: synthesis and deposition into subendothelial extracellular matrix. Proc Natl Acad Sci U S A, 1987,84(8):2292-2296.
    35 Hansson HA, Jennische E,Skottner A. Regenerating endothelial cells express insulin-like growth factor-I immunoreactivity after arterial injury. Cell Tissue Res, 1987,250(3):499-505.
    36 Owens GK, Geisterfer AA, Yang YW, et al. Transforming growth factor-beta-induced growth inhibition and cellular hypertrophy in cultured vascular smooth muscle cells. J Cell Biol, 1988,107(2):771-780.
    37王明松,邹良建,黄盛东,等.自体移植静脉内皮细胞的缺血安全时限.中国组织工程研究与临床康复, 2007,11(008):1401-1404.
    38 Bienvenu, JG, Tanguay, JF, Chauvet, P, et al. Relationship betweenplatelets and neutrophil adhesion and neointimal growth after repeated arterial wall injury induced by angioplasty in pigs. Journal of vascular research, 2000,38(2):153-162.
    39 Nilius B. Signaltransduction in vascular endothelium: the role of intracellular calcium and ion channels. Verh K Acad Geneeskd Belg, 1998, 60(3):215-250.
    40 Ohno N, Itoh H, Ikeda T, et al. Accelerated reendothelialization with suppressed thrombogenic property and neointimal hyperplasia of rabbit jugular vein grafts by adenovirus-mediated gene transfer of C-type natriuretic peptide. Circulation, 2002,105(14):1623-1626.
    41 Shimokawa H, Aarhus LL,Vanhoutte PM. Porcine coronary arteries with regenerated endothelium have a reduced endothelium-dependent responsiveness to aggregating platelets and serotonin. Circ Res, 1987, 61(2):256-270.
    42 Skalak TC, Price RJ. The role of mechanical stresses in microvascular remodeling. Microcirculation, 1996,3(2):143-165.
    43 Grus T, Lindner J, Vidim T, et al. The anastomosis angle is a key to improved long-term patency of proximal femoropopliteal bypass. Ann Vasc Surg, 2009,23(5):598-605.
    44 Coppola G, Caro C. Arterial geometry, flow pattern, wall shear and mass transport: potential physiological significance. J R Soc Interface, 2009,6 (35):519-528.
    45 Dangaria JH, Butler PJ. Macrorheology and adaptive microrheology of endothelial cells subjected to fluid shear stress. Am J Physiol Cell Physiol, 2007,293(5):C1568-1575.
    46 Langille BL. Morphologic responses of endothelium to shear stress: reorganization of the adherens junction. Microcirculation, 2001,8 (3):195-206.
    47 Yamamoto K, Ando J. [Shear-stress sensing via P2 purinoceptors in vascular endothelial cells]. Nippon Yakurigaku Zasshi, 2004,124 (5):319-328.
    48 Maniatis NA, Brovkovych V, Allen SE, et al. Novel mechanism of endothelial nitric oxide synthase activation mediated by caveolae internalization in endothelial cells. Circ Res, 2006,99(8):870-877.
    49 Ohno M, Cooke JP, Dzau VJ, et al. Fluid shear stress induces endothelial transforming growth factor beta-1 transcription and production. Modulation by potassium channel blockade. J Clin Invest, 1995,95 (3):1363-1369.
    50 Cotter EJ, von Offenberg Sweeney N, Coen PM, et al. Regulation of endopeptidases EC3.4.24.15 and EC3.4.24.16 in vascular endothelial cells by cyclic strain: role of Gi protein signaling. Arterioscler Thromb Vasc Biol, 2004,24(3):457-463.
    51 Morinaga K, Okadome K, Kuroki M, et al. Effect of wall shear stress on intimal thickening of arterially transplanted autogenous veins in dogs. J Vasc Surg, 1985,2(3):430-433.
    52 Dobrin PB. Mechanical factors associated with the development of intimal and medial thickening in vein grafts subjected to arterial pressure. A model of arteries exposed to hypertension. Hypertension, 1995,26(1):38-43.
    53 Berceli SA, Showalter DP, Sheppeck RA, et al. Biomechanics of the venous wall under simulated arterial conditions. J Biomech, 1990,23 (10):985-989.
    54 Ishida H, Lee YH. Study of hydrogen bonding and thermal properties of polybenzoxazine and poly-(-caprolactone) blends. Journal of Polymer Science Part A: Polymer Chemistry,2001,39(7):736-749.
    55 Lee T, Esemuede N, Sumpio BE, et al. Thrombospondin-1 induces matrix metalloproteinase-2 activation in vascular smooth muscle cells. J Vasc Surg, 2003,38(1):147-154.
    56 Prewitt RL. Autoregulation of blood flow, endothelial nitric oxide synthase and microvascular rarefaction. J Hypertens, 2002,20(2):177-178.
    57 Muzaffar S, Shukla N, Angelini GD, et al. Hypoxia and the expression of gp91 phox and endothelial nitric oxide synthase in the pulmonary artery. Thorax, 2005,60:305-313.
    58 Muzaffar, S, Shukla, N, Angelini, GD, et al. Hypoxia and the expression of gp91 phox and endothelial nitric oxide synthase in the pulmonary artery. Thorax, 2005,60:305-313.
    59 Ibrahim AI, Obeid MT, Jouma MJ, et al. Detection of herpes simplex virus, cytomegalovirus and Epstein-Barr virus DNA in atherosclerotic plaques and in unaffected bypass grafts. J Clin Virol, 2005,32(1):29-32.
    60 Egashira K, Zhao Q, Kataoka C, et al. Importance of monocyte chemoattractant protein-1 pathway in neointimal hyperplasia after periarterial injury in mice and monkeys. Circulation research, 2002,90 (11):1167-1172.
    61 Fogelstrand P, Feral CC, Zargham R, et al. Dependence of proliferative vascular smooth muscle cells on CD98hc (4F2hc, SLC3A2). J Exp Med, 2009,206(11):2397-2406.
    62郭明珂,张英泽,田德虎,等.局部应用5-氟尿嘧啶抑制移植静脉内膜增生的实验研究.中国修复重建外科杂志, 2009,23(8):940-946.
    63 Hutter R, Carrick FE, Valdiviezo C, et al. Vascular endothelial growth factor regulates reendothelialization and neointima formation in a mouse model of arterial injury. Circulation, 2004,110(16):2430-2435.
    64 Wildgruber M, Weiss W, Berger H, et al. Early endothelial and haematological response to cryoplasty compared with balloon angioplasty of the superficial femoral artery--a pilot study. Br J Radiol, 2007,80 (954):430-436.
    65 Tang YD, Pandey A, Kolmakova A, et al. Use of a novel anti-proliferative compound coated on a biopolymer to mitigate platelet-derived growth factor-induced proliferation in human aortic smooth muscle cells: comparison with sirolimus. Glycoconj J, 2009,26(6):721-732.
    66 Zhang Q, Lu L, Pu L, et al. Neointimal hyperplasia persists at six months after sirolimus-eluting stent implantation in diabetic porcine. Cardiovasc Diabetol, 2007,6:16.
    67 Erl W, Hansson GK, de Martin R, et al. Nuclear factor-kappa B regulates induction of apoptosis and inhibitor of apoptosis protein-1 expression invascular smooth muscle cells. Circ Res, 1999,84(6):668-677.

© 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