用户名: 密码: 验证码:
A20基因对大鼠颈动脉球囊损伤后内膜增生的影响及其分子机制的研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
目的:经皮冠状动脉介入治疗(percutaneous coronary intervention, PCI)是治疗冠心病的有效手段,但是再狭窄(restenosis, RS)仍是制约这一技术应用的主要问题。大量研究证实,再狭窄的机制主要由于球囊扩张和支架置入时导致血管壁损伤,引起炎症反应,进而导致平滑肌细胞的增殖和移行。由于血管壁的机械性损伤所致的炎性和增殖效应是造成再狭窄的主要原因,多种生长因子、细胞因子、粘附分子等参与其病理生理过程,单纯抑制某种特定的因子并不能完全抑制病灶形成。因此,阻断由炎症损伤引起的平滑肌细胞增殖、分化、迁移等最终共同通路的关键基因成为有效防止再狭窄的研究方向。
     基于导管将基因准确地送至狭窄血管的局部进行基因治疗是预防血管再狭窄较有效的方法之一,而选择合适的基因和载体是基因治疗再狭窄的关键之一。
     近年来在细胞内源性保护机制的研究中发现了一类新的蛋白质,即锌脂蛋白A20(zinc finger protein A20),简称A20。A20是NF-κB信号系统重要的负反馈调节因子,也是防止体内炎症反应失控的重要调节蛋白。A20的表达能够被LPS和TNF等所诱导,是NF-κB信号系统活化的产物,同时A20通过抑制TNF和LPS刺激TNFR-1和TLR4诱导的NF-κB的活化而对细胞损伤起保护作用。已有研究证实NF-κB的激活是球囊损伤动脉后血管狭窄发生的重要机制之一。结构研究显示,A20既有去泛素活性,又有泛素连接酶活性,是一种重要的泛素调节酶,在A20等泛素调节酶的作用下泛素调节参与了NF-κB前体的处理、NF-κB抑制性结合蛋白IκB的降解和IκB激酶IKK的活化等主要环节,反馈抑制了NF-κB的活化。
     一方面由于再狭窄的炎症机制及A20调控炎症的机制,另一方面由于NF-κB的激活导致球囊损伤后血管狭窄及A20对NF-κB活化的负反馈抑制作用,由此我们选择A20基因作为再狭窄发生发展过程中的干预靶点,运用基因治疗方法对体内A20的表达进行调控,深入探讨球囊损伤术后血管内膜增生过程中A20在损伤局部组织中的表达变化及局部过表达A20对损伤后内膜增生、平滑肌细胞增殖和表型改变,内皮再生及TLR4/NF-κB信号通路的影响,为进一步明确A20对再狭窄过程中炎症反应的调节作用以及揭示机体内源性保护机制提供必要的理论依据。
     方法:
     1. pCAGGS-GFP/A20重组质粒的鉴定和大量提取:将获得的pCAGGS-GFP/A20质粒菌保涂Amp抗性的平板后,过夜培养。挑单菌落于3ml Amp抗性的LB培养基中37℃摇床中过夜培养,集菌后,进行小量质粒提取,并使用EcoRⅤ/SmaⅠ进行双酶切鉴定,酶切产物1%琼脂糖凝胶电泳。将鉴定好的质粒菌保进行无内毒素质粒的大量提取,将质粒DNA提取物按适当比例稀释,并测定浓度。
     2.动物模型制备和实验分组,大鼠颈总动脉内膜球囊剥脱术用来建立再狭窄模型:健康雄性Sprague-Dawley(SD)大鼠体重350~400g,104只随机分为4组(n=26):假手术组;模型组(单纯损伤组);对照组(pCAGGS-GFP+转染试剂组);治疗组(pCAGGS-GFP/A20+转染试剂组)。10%水合氯醛(3ml/kg)腹腔注射麻醉后,取颈正中切口,钝性分离左颈总动脉,暴露左颈总动脉及其分叉处,用显微血管夹暂时夹闭左颈总动脉近心端及颈内动脉,将2F Fogarty球囊导管从颈外动脉切口插入至左颈总动脉,用50μl生理盐水充盈球囊,来回抽动3次以剥脱颈总动脉内膜。退出球囊,治疗组向损伤血管节段内缓慢注入灌注液(Lipofectamine 2000转染试剂40μl + pCAGGS-GFP/A20质粒20μl),对照组向损伤血管节段内缓慢注入灌注液(Lipofectamine 2000转染试剂40μl+ pCAGGS-GFP质粒20μl)局部作用30分钟。模型组只进行球囊损伤术,假手术组只进行左颈外动脉结扎,不进行球囊损伤。术后不同时间点根据不同要求分别处死动物。转染后24h取大鼠局部血管进行冰冻切片荧光显微镜下观察绿色荧光蛋白的表达籍以评估A20在损伤动脉壁的转染效率;术后14d病理组织学方法进行内膜增生的形态学分析;术后10d溴脱氧尿嘧啶核苷(BrdU)标记技术评估血管平滑肌细胞(VSMC)的增殖指数;术后10d免疫组织化学染色观察A20、核因子-κB p65(NF-κB p65)、Toll样受体4(TLR4)在各组大鼠颈总动脉中的表达情况;术后3d、7d、14d、28d通过逆转录-聚合酶链式反应(RT-PCR)、Western-blotting法分别检测A20、NF-κB p65、TLR4在颈动脉组织的mRNA和蛋白表达。
     3.健康雄性Sprague-Dawley(SD)大鼠体重350~400g,36只随机分为3组(n=12):假手术组;对照组(pCAGGS-GFP+转染试剂组);治疗组(pCAGGS-GFP/A20+转染试剂组)。球囊损伤后2周伊文思蓝(Evans blue)染色法评估内皮再生情况;术后3d、7d透射电镜观察血管超微结构改变。
     4.体外培养大鼠主动脉平滑肌细胞并鉴定,以LPS(20mg/l)作为刺激因素,以不同浓度大黄素(Emodin, EMD)作为干预因素,通过MTT和BrdU掺入方法评估各组VSMC增殖情况,以RT-PCR检测A20 mRNA表达,以Western-blotting法检测A20、NF-κB p65蛋白的表达。
     结果:
     1.将提取的重组真核表达质粒pCAGGS-GFP/A20使用EcoRⅤ/SmaⅠ进行双酶切鉴定,酶切产物1%琼脂糖凝胶电泳,质粒大小及酶切结果正确。
     2.将pCAGGS-GFP/A20于活体转染至大鼠球囊损伤的颈动脉局部,24h后荧光显微镜下可见治疗组残存血管内膜及中膜有大量绿色荧光分布,证明转染成功。
     3.大鼠颈总动脉球囊损伤术后14d模型组和对照组血管内膜显著增生。A20基因转染可显著抑制新生内膜面积的增加(减少47.8%,P<0.05)和内/中膜比值的增加(减少42.9%,P<0.05)。术后10d治疗组VSMC体内增殖指数(BrdU指数)(9.6±2.3%)显著少于对照组(26.7±5.1%,P<0.01)。术后10d治疗组血管壁A20免疫组化染色阳性细胞率显著高于对照组(P<0.05),NF-κB p65、TLR4免疫组化染色阳性细胞率显著低于对照组(P<0.05)。术后3d、7d、14d、28d治疗组血管组织A20的mRNA和蛋白表达量显著高于对照组(P<0.05),治疗组血管组织NF-κB p65、TLR4的mRNA和蛋白表达量显著低于对照组(P<0.05)。
     4.球囊损伤后2周,pCAGGS-GFP/A20质粒转染治疗组与对照组比较,显示更大面积的内皮修复,由伊文思蓝染色证明。治疗组再内皮化的面积占最初损伤面积的百分数( 73.1±3.7% )显著大于对照组(55.6±3.3%,P<0.05)。说明A20转染治疗组与对照组比较显示更大面积的内皮修复。
     5.假手术组大鼠VSMC电镜下呈典型的“收缩表型”,球囊损伤后7d对照组VSMC表型发生明显改变,呈典型的“合成表型”,损伤后3d对照组可见合成型细胞及过渡型细胞,治疗组术后7d可见接近收缩型的VSMC。
     6.体外培养的大鼠主动脉VSMC经LPS刺激后,MTT OD值与BrdU掺入OD值均显著高于空白组。经不同浓度EMD干预结果显示随EMD浓度增大,MTT OD值及BrdU掺入OD值均逐渐变小,其中低剂量组与对照组相比无显著性差异(P>0.05),高剂量组与对照组相比存在显著性差异(P<0.05)。RT-PCR和Western-blot结果显示LPS刺激可使A20mRNA和蛋白以及NF-κB p65蛋白表达均增加,经不同浓度EMD干预后A20mRNA和蛋白表达随EMD浓度递增而进一步增加,NF-κB p65蛋白表达随EMD浓度递增而逐渐减少。
     结论:
     1.大鼠颈动脉球囊损伤后内源性锌指蛋白A20在局部表达增加,并具有明显的时效性,说明损伤情况下,A20表达增加是机体的一种重要的内源性抗炎保护效应机制,可能对于调控损伤后适度的炎症反应具有重要意义。
     2.大鼠颈动脉球囊损伤后局部转染A20基因可获得锌指蛋白A20mRNA和蛋白在局部组织细胞内高表达。
     3.大鼠颈动脉球囊损伤后局部转染A20基因可抑制损伤血管内膜增生及平滑肌细胞的增殖,其分子机制可能通过其负反馈抑制了TLR4/NF-κB介导的胞内信号转导途径。
     4.大鼠颈动脉球囊损伤后局部转染A20基因可促进损伤血管内皮再生,抑制损伤处VSMC表型转化,其分子机制可能通过其负反馈抑制了TLR4/NF-κB介导的胞内信号转导途径。
     5. EMD对LPS诱导的大鼠主动脉VSMC增殖具有抑制作用,其机制可能通过增加体内炎症反应调节蛋白A20的表达并进一步负反馈抑制了NF-κB的激活,进一步证实了A20对VSMC增殖的影响。
Object:Percutaneous coronary intervention (PCI) is now one of the most effective methods to treat coronary artery disease, but restenosis (RS) following PCI is a primary problem to limit its long-term benefits. Many investigates confirmed that restenosis was the arterial wall’s healing response to mechanical injury and comprised two main processes- neointimal hyperplasia (smooth muscle proliferation/migration, extracellular matrix deposition) and vessel remodeling. Many growth factors, cytokine and adhesion molecule participate this pathophysiological procedure, and just inhibiting certain factor can not suppress lesion formation completely. Therefore, it becomes an effective approach of inhibiting restenosis to suppress the critical gene which regulates cell proliferation, migration and differentiation and so on the final common path.
     Catheter-based gene therapy has emerged as one of the most promising approaches to prevent coronary artery restenosis, because it appears capable of delivering therapeutic gene specifically to the location of the disease, at a precise site in the arterial wall. The choice of efficacious therapeutic gene and appropriate vector are the crux of the matter for a successful gene therapy.
     A kind of new protein molecule, zinc finger protein A20 (ZFPA20) has been found in recent-years’researches of cellular endogenous protective mechanism. It is confirmed that zinc finger protein A20 is not only important negative feedback factor to regulate signaling pathway of NF-κB but also essential regulating proteinum to suppress inflammatory responses in vivo. It is product of activated NF-κB sgnaling system induced by LPS or TNF. A20 has a protective effect on damaged cells through terminating the activation of NF-κB induced by TLR4, which is activated by LPS. Previous researches have shown that the activation of NF-κB initiate lesion formation and inflammatory response after vascular injury. A20 has both ubiquitinating and deubiquitinating activities. As an important ubiquitinating modification enzyme, A20 is involed in dealing with NF-κB prosoma, IκBαdegradation and IKK (IκB-kinase) activation. A20 leads to a negative-feedback response that terminates activation of NF-κB though above main mechanisms.
     One side because of inflammation mechanism of restenosis and regulation the inflammation mechanism of A20, on the other hand, bacause the activation of NF-κB initiates restenosis after vascular injury and A20 negative-feedback inhibits activation of NF-κB, A20 was served as target of happening and development process of restenos. A20 expression in vivo was regulated by gene therapy. The present study was designed to evaluate the transfection efficiency of A20 gene in injured arterial wall and to investigate the effect of in vivo local transfection of zinc finger protein A20 gene on intimal hyperplasia, proliferation and the phenotype transformation of vascular smooth muscle cells and reendothelialization of balloon-injured rat carotid arteries and its possible molecular mechanism.
     Methods:
     1. Identification and extraction of recombinant plasmid DNA in large quantity: Smearing the obtained E.coli carrying pCAGGS-GFP/A20 plasmid on ampicillin resistance plate, overnight cultured. puting single colony from plate into 3ml ampicillin resistance LB medium and overnight cultured on 37℃shaking table. After collecting bacteria, extracting recombinant plasmid DNA in small quantity and identificating by using EcoRⅤ/SmaⅠdouble restriction enzyme digestion. The production 1% agarose gel electrophoresis was performed. Endo-free plasmid DNA was extracted in large quantity from E.coli carrying plasmid. Plasmid DNA extraction was diluted in suitable proportion and detected the concentration.
     2. Establishment of rat carotid injury model and grouping. Balloon catheter denudation of the endothelium of rat common carotid artery was routinely used as a model of restenosis. One hundred and four male Sprague-Dawley rats weighing 350-400g were randomly divided into 4 groups (n=26): the sham group (no injury), the model group (simple injury), the control group (vacant vector and transfection regent group) and the therapeutic group (A20 gene and transfection regent group). Rats were anesthetized with 10% chloraldurat (3ml/kg intraperitoneally, i.p.). After a midcervial incision, the left common carotid artery and its bifurcation were exposed. Two vascular clips were placed at the distal end and in the middle of the injured arterial segment. A cannula was introduced into the common carotid artery via the external carotid artery. The left common carotid artery was balloon-injured three times with a 2F balloon catheter (2F Fogarty, Edwards Lifesciences Corporation), then pulling out Balloon catheter. pCAGGS-GFP/A20 20μl or pCAGGS-GFP 20μl with 40μl Lipofectamine 2000 (Invitrogen Corporation) was gradually instilled into the lumen for 30 min. Just balloon-injured operation in the model group. The sham group was only tied with a ligature at the external carotid artery without the balloon- injured operation. Rat were sacrificed according to various requirement on the different day after operation respectively. The transfection efficiency of plasmid in injured arterial wall was evaluated by using fluorescence microscope 24 hours after transfection. Quantification of intimal hyperplasia was determined by histopathologic examination 14 days after operation. Proliferation index of VSMCs in vivo was assessed by thymidine analogue bromodeoxyuridine (BrdU) labeling technique 10 days after operation. The expression of A20, nuclear factor-kappaB p65 (NF-κBp65) and toll like receptor 4 (TLR4) of carotid arteries in different groups was confirmed by immunohistochemical staining, reverse transcription-polymerase chain reaction (RT-PCR) and Western blot analysis at different time point.
     3. Thirty six male Sprague-Dawley(SD) rats were randomly divided into three groups (n=12): the sham group(no injury), the control group (vacant vector and transfection regent group) and the therapeutic group (A20 gene and transfection regent group). Rats were sacrificed two weeks after operation to evaluate reendothelialization by Evans blue dye staining. Ultrastructural change of injured artery was observed with transmission electron microscope at day 3 and 7 after balloon injury.
     4. Cultured rat aorta smooth muscle cells (SMCs) were identified by specificα-actin antibody. VSMCs were pretreated by emodin (5~50μg/l) for 30min and then were stimulated by LPS (20mg/l) for 24h. VSMCs proliferation was assessed by MTT and BrdU corporation. The quantification of A20 mRNA expression was measured by RT-PCR. The quantification of A20, NF-κBp65 protein expression was measured by Western blot analysis.
     Results:
     1. After the pCAGGS-GFP/A20 plasmid was digested by EcoRⅤ/SmaⅠdouble restriction enzyme, the production was identified by 1% agarose gel electrophoresis, plasmid size and enzyme digested result was right.
     2. Much green fluorescence distribution of zinc finger protein A20 gene existed in the remnant intima and media In vivo 24 hours after transfection by using fluorescence microscope, which confirmed the transgene efficiency in injured vascular wall.
     3. Significant intimal hyperplasia was observed in the model and control group at day 14 after injury. A20 gene transfection markedly reduced the neointimal area (47.8% reduction; P< 0.05) and intimal to media area ratio (42.9% reduction; P< 0.05) in the therapeutic group. Proliferation index of VSMCs (BrdU index) at day 10 after operation decreased more significantly in the therapeutic group (9.6±2.3%) than in the control group (26.7±5.1%,P<0.01). The A20 positive cells ratio of the therapeutic group was much more than that of the control group (P<0.05), whereas, the NF-κBp65 and TLR4 positive cells ratio of the therapeutic group was much less than that of the control group at day 10 after injury (P<0.05). A significantly higher level of A20 mRNA, protein expression was measured in the therapeutic group than in control group at every time point after operation (P<0.05). A significantly lower level of NF-κBp65 and TLR4 mRNA, protein expression was observed in the therapeutic group than in control group at every time point after operation (P<0.05).
     4. At 2 weeks after balloon injury, compared with control group ,the arteries of animals treated with A20 gene transfection showed a greater area of recovered endothelium, as evidenced by Evans blue staining. Quantification of Re-endothelialization, based on the percentage of re-endothelialization area in the originally injured area re-endothelialized (control 55.6±3.3% vs. therapeutic 79.1±3.7%, P<0.05) revealed a significant acceleration of endothelial recovery in the A20 gene therapeutic group compared with the control group.
     5. Representative contractile phenotype of VSMC was shown in the sham group and synthetic phenotype was shown at day 7 after balloon injury in the control group. Synthetic phenotype and transitional phenotype was shown at day 3 after balloon injury in the control group. At day 7 VSMC showed relatively more myofilament and less organelles and looked like contractile phenotype in the therapeutic group.
     6. Compered with the vacant group, the control group showed significantly higher MTT and BrdU corporating OD value. We found that emodin induced a concentration-dependent decrease in the level of MTT and BrdU corporating OD value. Compered with the control group, low-dose groups have not significant difference, high-dose group showed significant difference. The expression of A20 mRNA and protein,NF-κBp65 protein were up-reglulated after VSMCs were stimulated by LPS. The expression of A20 mRNA and protein were more increased in a concentration- dependent manner after VSMCs were pretreated by emodin on different concentration. While the expression of NF-κBp65 protein were down-regulated.
     Conclusion:
     1. Local endogenous zinc finger protein A20 expression was increased in balloon-injured rat carotid artery, and possesed evident chronergy. It is confirmed that zinc finger protein A20 is critical for the regulation of inflammatory responses. It is the important regulation protein for inhibiting inflammatory responses in vivo.
     2. Local transfection of A20 gene resulted in the high expression of zinc finger protein A20 mRNA and protein overexpression in balloon- injured rat carotid artery,
     3. Local transfection of A20 gene suppressed intimal hyperplasia and VSMC proliferation of balloon-injured rat carotid artery. Its possible molecular mechanism is that A20 is critical for the regulation of inflammatory responses through terminating the activation of NF-κB induced by injury.
     4. Local transfection of A20 gene accelerated reendothelialization and inhibited the phenotype transformation of VSMC of balloon-injured rat carotid artery. Its possible molecular mechanism is that A20 is critical for the regulation of inflammatory responses through terminating the activation of NF-κB induced by injury.
     5. EMD can inhibit LPS-mediated rat aorta VSMC proliferation..the possible mechanism is that EMD can suppress activation of NF-kB via increasing the expression of inflammatory reaction regulatory protein A20 in vivo ,which has confirmed further the effect of A20 on the VSMC proliferation.
引文
[1] Costa MA, Simon DI. Molecular basis of restenosis and drug-eluting stents. Circulation. 2005;111(17):2257-73.
    [2] Welt FG, Rogers C. Inflammation and restenosis in the stent era. Arterioscl Thromb Vasc Biol. 2002;22:1769-76.
    [3] Boone DL, Turer EE, Lee EG, et al. The ubiquitin-modifying enzyme A20 is required for termination of Toll-like receptor responses. Nat Immunol. 2004; 5 (10):1052-60.
    [4] Lee EG, Boone DL, Chai S, et al. Failure to regulate TNF-induced NF -κB and cell death responses in A20 - deficient mice. Science. 2000;289:2350-4.
    [5] Cercek B, Yamashita M, Dimayuga P, et al. Nuclear factor-κB activity and arterial response to balloon injury. Atherosclerosis. 1997;131:59-66.
    [6] Heyninck K, Beyaert R. A20 inhibits NF-kappaB activation by dual ubiquitin- editing functions. Trends Biochem Sci. 2005;30:1-4.
    [7] Ueno H, Haruno A, Morisaki N, et al. Local expression of C-type natriuretic peptide markedly suppresses neointimal formation in rat injured arteries through an autocrine/paracrine loop. Circulation. 1997;96:2272-9.
    [8] Park KW, Yang HM, Youn SW, et al. Constitutively active glycogen synthase kinase-3beta gene transfer sustains apoptosis, inhibits proliferation of vascular smooth muscle cells, and reduces neointima formation after balloon injury in rats. Arterioscler Thromb Vasc Biol. 2003;23:1364-9.
    [9] Zoldhelyi P, McNatt J, Xu XM, et al. Prevention of arterial thrombosis by adenovirus-mediated transfer of cyclooxygenase gene. Circulation. 1996;93: 10-17.
    [10] Gennaro G, Menard C, Giasson E, et al. Role of p44/p42 MAP kinase in the age-dependent increase in vascular smooth muscle cell proliferation and neointimal formation. Arterioscler Thromb Vasc Biol. 2003;23:204-10.
    [11] Okano HJ, Pfaff DW, Gibbs RB. Expression of EGFR-, p75NGFR-, and PSTAIR (cdc2)-like immunoreactivity by proliferating cells in the adult rat hippocampal formation and forebrain. Dev Neurosci. 1996;18:199-209.
    [12] Schartl M, Bocksch W, Koschyk DH,et al. Use of intravascular ultrasound to compare effects of different strategies of lipid-lowering therapy on plaquevolume and composition in patients with coronary artery disease. Circulation. 2001;104(4):387-92.
    [13] Kimmel SE, Localio AR, Krone RJ, et al. The effects of contemporary use of coronary stents on in-hospital mortality. Registry Committee of the Society for Cardiac Angiography and Interventions. J Am Coll Cardiol. 2001;37(2): 499-504.
    [14] Singh M, Rihal CS, Berger PB, et al. Improving outcome over time of percutaneous coronary interventions in unstable angina. J Am Coll Cardiol. 2000;36(3):674-8.
    [15] Ritchie JL, Maynard C, Every NR, et al. Coronary artery stent outcomes in a Medicare population: less emergency bypass surgery and lower mortality rates in patients with stents. Am Heart J. 1999;138(3 Pt 1):437-40.
    [16] Serruys PW, de Jaegere P, Kiemeneij F, et al. A comparison of balloon- expandable-stent implantation with balloon angioplasty in patients with coronary artery disease. Benestent Study Group. N Engl J Med. 1994;331(8): 489-95.
    [17] Fischman DL, Leon MB, Baim DS, et al. A randomized comparison of coronary- stent placement and balloon angioplasty in the treatment of coronary artery disease. Stent Restenosis Study Investigators. N Engl J Med. 1994;331(8): 496-501.
    [18] Mintz GS, Popma JJ, Pichard AD, et al. Arterial remodeling after coronary angioplasty: a serial intravascular ultrasound study. Circulation. 1996;94(1): 35-43.
    [19] Post MJ, Borst C, Kuntz RE. The relative importance of arterial remodeling compared with intimal hyperplasia in lumen renarrowing after balloon angioplasty. A study in the normal rabbit and the hypercholesterolemic Yucatan micropig. Circulation. 1994;89(6):2816-21.
    [20] Karas SP, Gravanis MB, Santoian EC, et al. Coronary intimal proliferation after balloon injury and stenting in swine: an animal model of restenosis. J Am Coll Cardiol. 1992 Aug;20(2):467-74.
    [21] Rogers C, Karnovsky MJ, Edelman ER. Inhibition of experimental neointimal hyperplasia and thrombosis depends on the type of vascular injury and the site of drug administration. Circulation. 1993 Sep;88(3):1215-21.
    [22] Libby P, Schwartz D, Brogi E, et al. A cascade model for restenosis. A special case of atherosclerosis progression. Circulation. 1992 Dec;86(6 Suppl):III47-52.
    [23] Tanaka H, Sukhova GK, Swanson SJ,et al. Sustained activation of vascular cells and leukocytes in the rabbit aorta after balloon injury. Circulation. 1993 Oct;88(4 Pt 1):1788-803.
    [24] Tanaka H, Sukhova G, Schwartz D,et al. Proliferating arterial smooth muscle cells after balloon injury express TNF-alpha but not interleukin-1 or basic fibroblast growth factor. Arterioscler Thromb Vasc Biol. 1996 Jan;16(1):12-8.
    [25] Gaspardone A, Crea F, Versaci F, et al. Predictive value of C-reactive protein after successful coronary-artery stenting in patients with stable angina. Am J Cardiol. 1998 Aug;82(4):515-8.
    [26] Versaci F, Gaspardone A, Tomai F, et al. Predictive value of C-reactive protein in patients with unstable angina pectoris undergoing coronary artery stent implantation. Am J Cardiol. 2000 Jan;85(1):92-5.
    [27] Van Belle E, Tio FO, Chen D, et al. Passivation of metallic stents after arterial gene transfer of phVEGF165 inhibits thrombus formation and intimal thickening. J Am Coll Cardiol. 1997 May;29(6):1371-9.
    [28] French BA, Mazur W, Ali NM, et al. Percutaneous transluminal in vivo gene transfer by recombinant adenovirus in normal porcine coronary arteries, atherosclerotic arteries, and two models of coronary restenosis. Circulation. 1994 Nov;90(5):2402-13.
    [29] Wivel NA, Wilson JM. Methods of gene delivery. Hematol Oncol Clin North Am. 1998 Jun;12(3):483-501.
    [30] Verma IM, Somia N. Gene therapy-promises, problems and prospects. Nature. 1997 Sep 18;389(6648):239-42.
    [31] Mahato RI, Rolland A, Tomlinson E. Cationic lipid-based gene delivery systems: pharmaceutical perspectives. Pharm Res. 1997 Jul;14(7):853-9.
    [32] Ruponen M, Herrala Y, Urtti A. Interaction of polymeric and liposomal gene delivery systems with extracelluar glycosaminoglycans: physicochemiacal and transfection studies. Biochem Biophys Acta. 1999,1415: 331-41.
    [33] Ross PC, Hui SW. Polythylene glycol enhancers lipoplex-cell association and lipofection. Biochim Biophys Acta. 1999;1421:273-83.
    [34] Choi YH, Liu F, Kim JS, et al. Polyethylene glycol-grafted poly-L-lysine as polymeric gene carrier. J Controlled Release. 1998;54:39-48.
    [35] Veit K, Boissel JP, Buerke M, et al. Highly efficient liposome-mediated gene transfer of inducible nitric oxide synthase in vivo and in vitro in vascularsmooth muscle cells. Cardiovasc Res. 1999;43(3):808-22.
    [36] Wertz IE, O'Rourke KM, Zhou H, et al. De-ubiquitination and ubiquitin ligase domains of A20 downregulate NF-kappaB signalling. Nature. 2004;430(7000): 694-9.
    [37] Beyaert R, Heyninck K, Van Huffel S. A20 and A20-binding proteins as cellular inhibitors of nuclear factor-kappa B-dependent gene expression and apoptosis. Biochem Pharmacol. 2000; 60(8):1143-51.
    [38] Makarova KS, Aravind L, Koonin EV. A novel superfamily of predicted cysteine proteases from eukaryotes, viruses and Chlamydia pneumoniae. Trends Biochem Sci. 2000;25(2):50-2.
    [39] Opipari AW Jr, Boguski MS, Dixit VM. The A20 cDNA induced by tumor necrosis factor alpha encodes a novel type of zinc finger protein. J Biol Chem. 1990;265(25):14705-8.
    [40] Evans PC, Ovaa H, Hamon M, et al. Zinc-finger protein A20, a regulator of inflammation and cell survival, has de-ubiquitinating activity. Biochem J. 2004;378(Pt 3):727-34.
    [41] Silverman N, Fitzgerald K. DUBbing down innate immunity. Nat Immunol. 2004 Oct;5(10):1010-2.
    [42] Ben-Neriah Y. Regulatory functions of ubiquitination in the immune system. Nat Immunol. 2002 Jan;3(1):20-6.
    [43] Wilkinson KD, Ventii KH, Friedrich KL, et al. The ubiquitin signal: assembly, recognition and termination. Symposium on ubiquitin and signaling. EMBO Rep. 2005 Sep;6(9):815-20.
    [44] Hu M, Li P, Li M, et al. Crystal structure of a UBP-family deubiquitinating enzyme in isolation and in complex with ubiquitin aldehyde. Cell. 2002 Dec 27;111(7):1041-54.
    [45] Zempo N, Kenagy RD, Au YP, et al. Matrix metalloproteinases of vascular wall cells are increased in balloon-injured rat carotid artery. J Vasc Surg. 1994 Aug;20(2):209-17.
    [46] Fingerle J, Au YP, Clowes AW, et al. Intimal lesion formation in rat carotid arteries after endothelial denudation in absence of medial injury. Arteriosclerosis. 1990;10(6):1082-7.
    [47] Signore PE, Machan LS, Jackson JK, et al. Complete inhibition of intimal hyperplasia by perivascular delivery of paclitaxel in balloon-injured rat carotidarteries. J Vasc Interv Radiol. 2001 Jan;12(1):79-88.
    [48] Lamfers ML, Lardenoye JH, de Vries MR, et al. In vivo suppression of restenosis in balloon-injured rat carotid artery by adenovirus-mediated gene transfer of the cell surface-directed plasmin inhibitor ATF.BPTI. Gene Ther. 2001 Apr;8(7):534-41.
    [49] Ascher E, Scheinman M, Hingorani A, et al. Effect of p53 gene therapy combined with CTLA4Ig selective immunosuppression on prolonged neointima formation reduction in a rat model. Ann Vasc Surg. 2000 Jul;14(4):385-92.
    [50] Clowes AW, Clowes MM, Vergel SC, et al. Heparin and cilazapril together inhibit injury-induced intimal hyperplasia. Hypertension. 1991 Oct;18(4 Suppl):II65-9.
    [51] Hanke H, Strohschneider T, Oberhoff M, et al. Time course of smooth muscle cell proliferation in the intima and media of arteries following experimental angioplasty. Circ Res. 1990 Sep;67(3):651-9.
    [52] Nakamura S, Takeda Y, Kano M, et al. Application of bromodeoxyuridine (BrdU) and anti-BrdU monoclonal antibody for the in vivo analysis of proliferative characteristics of human leukemia cells in bone marrows. Oncology. 1991; 48(4): 285-9.
    [53] Sen R, Baltimore D. Multiple nuclear factors interact with the immunoglobulin enhancer sequences. Cell. 1986 Aug 29;46(5):705-16.
    [54] Vink A, Schoneveld AH, van der Meer JJ, et al. In vivo evidence for a role of toll-like receptor 4 in the development of intimal lesions. Circulation. 2002;106:1985-90.
    [55] Ginn-Pease ME, Whisler RL. Redox signals and NF-kappaB activation in T cells. Free Radic Biol Med. 1998 Aug;25(3):346-61.
    [56] Maffia P, Ianaro A, Pisano B, et al. Beneficial effects of caffeic acid phenethyl ester in a rat model of vascular injury. Br J Pharmacol. 2002 Jun;136(3):353-60.
    [57] Ialenti A, Ianaro A, Maffia P, et al. Role of nuclear factor-kappaB in a rat model of vascular injury. Naunyn Schmiedebergs Arch Pharmacol. 2001 Oct;364(4): 343-50.
    [58] Autieri MV. Expression of anaphase promoting complex 5 in ballon angioplasty injured rat carotid arteries and nutogeu stimulated human vascular smooth muscle cells. Biochem Biophs Res Commun. 2001;282(3):723-8.
    [59] Sasu S, LaVerda D, Qureshi N, et al. Chlamydia pneumoniae and chlamydialheat shock protein 60 stimulate proliferation of human vascularsmooth muscle cells via toll-likereceptor4 and p44/p42 mitogen- activated protein kinase activation. Circ Res. 2001;89(3):244-50.
    [60] Hollestelle SC, De Vries MR, Van Keulen JK, et al. Toll-like receptor 4 is involved in outward arterial remodeling. Circulation. 2004 Jan 27;109(3):393-8.
    [61] Okamura Y, Watari M, Jerud ES, et al. The extra domain A of fibronectin activates Toll-like receptor 4. J Biol Chem. 2001 Mar 30;276(13):10229-33.
    [62] Yamazaki S, Muta T, Takeshige K. A novel IkappaB protein, IkappaB-zeta, induced by proinflammatory stimuli, negatively regulates nuclear factor-kappaB in the nuclei. J Biol Chem. 2001;276:27657-62.
    [63] Patel VI, Daniel S, Longo CR, et al. A20, a modulator of smooth muscle cell proliferation and apoptosis, prevents and induces regression of neointimal hyperplasia. FASEB J. 2006 Jul;20(9):1418-30.
    [64] O’Reilly SM, Moynagh PN. Regulation of Toll-like receptor 4 signalling by A20 zinc finger protein. Biochem Biophys Res Commun. 2003;303(2):586-93.
    [65] Wang AB, Li HL, Zhang R, et al. A20 attenuates vascular smooth muscle cell proliferation and migration through blocking PI3k/Akt singling in vitro and in vivo. J Biomed Sci. 2007 May;14(3):357-71.
    [1] Asahara J, Bauters C, Pastore C, et al. Local delivery of vascular endothelial growth factor accelerates reendothelialization and attenuates intimal hyperplasia in balloon injured rat carotid artery. Circulation. 1995; 91: 2793-801.
    [2] Meurice T, Bauters C, Auffray JL, et al. Basic fibroblast growth factor restores endothelium dependent responses after balloon injury of rabbit arteries. Circulation. 1996;93:18-22.
    [3] Losordo DW, Isner JM, Diaz-Sandoval LJ. Endothelial recovery: the next target in restenosis prevention. Circulation. 2003 Jun 3;107(21):2635-7.
    [4] Boone DL, Turer EE, Lee EG, et al. The ubiquitin-modifying enzyme A20 is required for termination of Toll-like receptor responses. Nat Immunol. 2004 Oct;5(10):1052-60.
    [5] Lee EG, Boone DL, Chai S, et al. Failure to regulate TNF-induced NF-kappaB and cell death responses in A20-deficient mice. Science. 2000 Sep 29;289 (5488): 2350-4.
    [6] Cercek B, Yamashita M, Dimayuga P, et al. Nuclear factor-kappaB activity and arterial response to balloon injury. Atherosclerosis. 1997 May;131(1):59-66.
    [7] Ialenti A, Ianaro A, Maffia P, et al. Role of nuclear factor-kappaB in a rat model of vascular injury. Naunyn Schmiedebergs Arch Pharmacol. 2001;364:343-50.
    [8] Ueno H, Haruno A, Morisaki N, et al. Local expression of C-type natriuretic peptide markedly suppresses neointimal formation in rat injured arteries through an autocrine/paracrine loop. Circulation. 1997;96:2272-9.
    [9] Park KW, Yang HM, Youn SW, et al. Constitutively active glycogen synthase kinase-3beta gene transfer sustains apoptosis, inhibits proliferation of vascular smooth muscle cells, and reduces neointima formation after balloon injury in rats. Arterioscler Thromb Vasc Biol. 2003;23: 1364-9.
    [10] Zoldhelyi P, McNatt J, Xu XM, et al. Prevention of arterial thrombosis by adenovirus-mediated transfer of cyclooxygenase gene. Circulation. 1996 Jan 1; 93(1):10-7.
    [11]崔连群,王晓军,刘继东,等.粒细胞集落刺激因子对大鼠颈动脉PCI后内膜修复和增生的影响.介入放射学杂志.2005;14:90-2.
    [12] Zhang C, Yang J, Jennings LK. Attenuation of neointima formation through the inhibition of DNA repair enzyme PARP-1 in balloon-injured rat carotid artery.Am J Physiol Heart Circ Physiol. 2004 Aug;287(2):H659-66.
    [13]汤健,朝枢主编.心肺内分泌学.北京:北京科学出版社.1991; 42-5.
    [14] Lowe HC, Oesterle SN, Khachigian LM. Coronary in-stent restenosis: current status and future strategies. J Am Coll Cardiol. 2002;39(2):183-93.
    [15] Asahara T, Chen D, Tsurumi Y, et al. Accelerated restitution of endothelial integrity and endothelium2dependent function following phVEGF165 gene transfer. Circulation. 1996; 94:3291-302.
    [16] Losordo DW, Isner JM, Diaz-Sandoval LJ. Endothelial recovery: the next target in restenosis prevention. Circulation. 2003;107(21):2635-7.
    [17] Faries PL, Rohan DI, Wyers MC, et al. Vascular smooth muscle cells derived from atherosclerotic human arteries exhibit greater adhesion, migration, and proliferation than venous cells. J Surg Res. 2002;104(1):22-8.
    [18] Doi K, Ikeda T, Itoh H, et al. C-type natriuretic peptide induces redifferentiation of vascular smooth muscle cells with accelerated reendothelialization. Arterioscler Thromb Vasc Biol. 2001;21(6):930-6.
    [19] Noiseux N, Boucher CH, Cartier R, et al. Bolus endovascular PDGFR-beta antisense treatment suppressed intimal hyperplasia in a rat carotid injury model. Circulation. 2000;102(11):1330-6.
    [20] Haudenschild CC, Schwartz SM. Endothelial regeneration. II. Restitution of endothelial continuity. Lab Invest. 1979;41(5):407-18.
    [21] Oberhoff M, Novak S, Herdeg C, et al. Local and systemic delivery of low molecular weight heparin stimulates the reendothelialization after balloon angioplasty. Cardiovasc Res. 1998 Jun; 38(3):751-62.
    [1] Topol EJ, Serruys PW. Frontiers in interventional cardiology. Circulation. 1998;98:1802-20.
    [2] Welt FG, Rogers C. Inflammation and restenosis in the stent era. Arterioscl Thromb Vasc Biol. 2002;22:1769-76.
    [3]郭丹杰,棣成斌,陈源源.大黄素对血管平滑肌细胞增殖影响的实验研究.中华内科杂志.1996;35(3):157-9
    [4]王翔飞,葛均波,孙爱军,等.大黄素通过p53途径抑制血管平滑肌细胞增殖的实验研究.中华心血管病杂志.2006;34(1):44-9.
    [5]任立群,张秀云,孙波,等.大鼠胸主动脉平滑肌细胞的组织贴块法培养及鉴定.吉林大学学报(医学版).2002;28(2):135-7.
    [6] Nozawa Y, Matsuura N, Miyake H, et al. Effects of TH-142177 on angiotensin II-induced proliferation, migration and intracellular signaling in vascular smooth muscle cells and on neointimal thickening after balloon injury. Life Sci. 1999;64(22):2061-70.
    [7]西冈五夫.大黄的生物活性及其有效成分.国外医学·中医中药分册.1986;8:155.
    [8]陈琼华.大黄的实验研究和临床应用.新医学杂志.l974;5:228.
    [9]刘志江.胡伟新,藜磊石,等.大黄素对肾脏系膜细胞增殖细胞核抗原的影响.肾脏病透析与肾移植.1993;2:45.
    [10] Zhu F, Liu XG, Liang NC. Effect of emodin and apigenin on invasion of human ovarian carcinoma HO-8910PM cells in vitro. Ai Zheng. 2003;22(4):358-62.
    [11] Kuo YC, Sun CM, Ou JC, et al. A tumor cell growth inhibitor from Polygonum hypoleucum Ohwi. Life Sci. 1997;61(23):2335-44.
    [12]王翔飞,葛均波,孙爱军,等.大黄素自身抑制血管平滑肌细胞迁移和增殖.中国介入心脏病学杂志.2006;14(4):241-4.
    [13] Müller SO, Eckert I, Lutz WK, et al. Genotoxicity of the laxative drug components emodin, aloe-emodin and danthron in mammalian cells: topoisomerase II mediated? Mutat Res. 1996;371(3-4):165-73.
    [14] Jayasuriya H, Koonchanok NM, Geahlen RL, et al. Emodin, a protein tyrosine kinase inhibitor from Polygonum cuspidatum. J Nat Prod. 1992;55(5):696-8.
    [15] Yim H, Lee YH, Lee CH, et al. Emodin, an anthraquinone derivative isolated from the rhizomes of Rheum palmatum, selectively inhibits the activity ofcasein kinase II as a competitive inhibitor. Planta Med. 1999;65(1):9-13.
    [16] Iglesias V, Alguacil LF, Alamo C, et al. Effects of yohimbine on morphine analgesia and physical dependence in the rat. Eur J Pharmacol. 1992; 211(1): 35-8.
    [17]苏静怡,唐朝枢.心血管疾病的病理生理基础和发病机制.北京:北京医科大学中国协和医科大学联合出版社.1994;46-7,266-77.
    [18]汤健,周爱儒.原癌基因与心血管疾病.北京:北京医科大学协和医科大学联合出版社.1990;86-7.
    [19] Wu CC, Hong HJ, Chou TC, et al. Evidence for inducible nitric oxide synthase in spontaneously hypertensive rats. Biochem Biophys Res Commun. 1996; 228 (2):459-66.
    [20]李剑,林树新,李莹,等.脂多糖对离体培养大鼠血管平滑肌细胞增殖的影响.生理学报.1999;51(1):14-8.
    [1] Dixit VM, Green S, Sarma V, et al. Tumor necrosis factor-alpha induction of novel gene products in human endothelial cells including a macrophage-specific chemotaxin. J Biol Chem. 1990;265(5):2973-8.
    [2] Opipari AW Jr, Boguski MS, Dixit VM. The A20 cDNA induced by tumor necrosis factor alpha encodes a novel type of zinc finger protein. J Biol Chem. 1990; 265(25):14705-8.
    [3] Krikos A, Laherty CD, Dixit VM. Transcriptional activation of the tumor necrosis factor alpha-inducible zinc finger protein, A20, is mediated by kappa B elements. J Biol Chem. 1992;267(25):17971-6.
    [4] Heyninck K, Beyaert R. A20 inhibits NF-kB activation by dual ubiquitin-editing Functions. TRENDS in Biochemical Sciences. 2005;30(1)1-4.
    [5] Klinkenberg M, Van Huffel S, Heyninck K, et al. Functional redundancy of the zinc fingers of A20 for inhibition of NF-kappaB activation and protein-protein interactions. FEBS Lett. 2001;498(1):93-7.
    [6] Beyaert R, Heyninck K, Van Huffel S. A20 and A20-binding proteins as cellular inhibitors of nuclear factor-kappa B-dependent gene expression and apoptosis. Biochem Pharmacol. 2000;60(8):1143-51.
    [7] Lee EG, Boone DL, Chai S, et al. Failure to regulate TNF-induced NF-kappaB and cell death responses in A20-deficient mice. Science. 2000;289(5488):2350-4.
    [8] Arvelo MB, Cooper JT, Longo C, et al. A20 protects mice from D-galactosamine/ lipopoly- saccharide acute toxic lethal hepatitis. Hepatology. 2002;35(3):535-43.
    [9] He KL, Ting AT. A20 inhibits tumor necrosis factor (TNF) alpha-induced apoptosis by disrupting recruitment of TRADD and RIP to the TNF receptor 1 complex in Jurkat T cells. Mol Cell Biol. 2002;22(17):6034-45.
    [10] Li Q, Verma IM. NF-kappaB regulation in the immune system. Nat Rev Immunol. 2002;2(10): 725-34.
    [11] Gerondakis S, Grossmann M, Nakamura Y, et al. Genetic approaches in mice to understand Rel/NF-kappaB and IkappaB function: transgenics and knockouts. Oncogene. 1999;18(49): 6888-95.
    [12] Wertz IE, O'Rourke KM, Zhou H, et al. De-ubiquitination and ubiquitin ligasedomains of A20 downregulate NF-kappaB signalling. Nature. 2004;430(7000): 694-9.
    [13] Boone DL, Turer EE, Lee EG, et al. The ubiquitin-modifying enzyme A20 is required for termination of Toll-like receptor responses. Nat Immunol. 2004;5 (10):1052-60.
    [14] Chen G, Goeddel DV. TNF-R1 signaling: a beautiful pathway. Science. 2002; 296(5573):1634-5.
    [15] Lee TH, Shank J, Cusson N, et al. The kinase activity of Rip1 is not required for tumor necrosis factor-alpha-induced IkappaB kinase or p38 MAP kinase activation or for the ubiquitination of Rip1 by Traf2. J Biol Chem. 2004;279(32): 33185-91.
    [16] Kanayama A, Seth RB, Sun L, et al. TAB2 and TAB3 activate the NF-kappaB pathway through binding to polyubiquitin chains. Mol Cell. 2004;15(4):535-48.
    [17] O'Reilly SM, Moynagh PN. Regulation of Toll-like receptor 4 signalling by A20 zinc finger protein. Biochem Biophys Res Commun. 2003;303(2):586-93.
    [18] Vogel SN, Fitzgerald KA, Fenton MJ. TLRs: differential adapter utilization by toll-like receptors mediates TLR-specific patterns of gene expression. Mol Interv. 2003;3(8):466-77.
    [19] Silverman N, Maniatis T. NF-kappaB signaling pathways in mammalian and insect innate immunity. Genes Dev. 2001;15(18):2321-42.
    [20] Yamamoto M, Sato S, Hemmi H, et al. Role of adaptor TRIF in the MyD88- independent toll-like receptor signaling pathway. Science. 2003;301(5633): 640-3.
    [21] Oshiumi H, Matsumoto M, Funami K, et al. TICAM-1, an adaptor molecule that participates in Toll-like receptor 3-mediated interferon-beta induction.Nat Immunol. 2003;4(2):161-7.
    [22] Yamamoto M, Sato S, Mori K, et al. Cutting edge: a novel Toll/IL-1 receptor domain- containing adapter that preferentially activates the IFN-beta promoter in the Toll-like receptor signaling. J Immunol. 2002;169(12):6668-72.
    [23] Meylan E, Burns K, Hofmann K, et al. RIP1 is an essential mediator of Toll-like receptor 3-induced NF-kappa B activation. Nat Immunol. 2004;5(5):503-7.
    [24] Hofmann RM, Pickart CM. Noncanonical MMS2-encoded ubiquitin-conjugating enzyme functions in assembly of novel polyubiquitin chains for DNA repair. Cell. 1999;96(5):645-53.
    [25] Sun L, Chen ZJ. The novel functions of ubiquitination in signaling. Curr Opin Cell Biol. 2004;16(2):119-26.
    [26] Wang C, Deng L, Hong M, et al. TAK1 is a ubiquitin-dependent kinase of MKK and IKK. Nature. 2001;412(6844):346-51.
    [27] Evans PC, Ovaa H, Hamon M, et al. Zinc-finger protein A20, a regulator of inflammation and cell survival, has de-ubiquitinating activity. Biochem J. 2004; 378(Pt 3):727-34.
    [28] Prince MR, Choyke PL, Knopp MV. Putting the brakes on cylindromatosis? N Engl J Med. 2004;350(2):187-8.
    [29] Deng L, Wang C, Spencer E, et al. Activation of the IkappaB kinase complex by TRAF6 requires a dimeric ubiquitin-conjugating enzyme complex and a unique polyubiquitin chain Cell. 2000;103(2):351-61.
    [30] Pickart CM. Back to the future with ubiquitin. Cell. 2004;116(2):181-90.
    [31] Peng J, Schwartz D, Elias JE, et al. A proteomics approach to understanding protein ubiquitination. Nat Biotechnol. 2003;21(8):921-6.
    [32] Mauro C, Pacifico F, Lavorgna A, et al. ABIN-1 binds to NEMO/IKKgamma and co-operates with A20 in inhibiting NF-kappaB. J Biol Chem. 2006;281(27): 18482-8.
    [33] Hutti JE, Turk BE, Asara JM, et al. IkappaB kinase beta phosphorylates the K63 deubiquitinase A20 to cause feedback inhibition of the NF-kappaB pathway. Mol Cell Biol. 2007;27(21):7451- 61.
    [34] Wu CJ, Conze DB, Li T, et al. Sensing of Lys 63-linked polyubiquitination by NEMO is a key event in NF-kappaB activation. Nat Cell Biol. 2006;8(4):398-406.
    [35] Heyninck K, De Valck D, Vanden Berghe W, et al. The zinc finger protein A20 inhibits TNF-induced NF-kappaB-dependent gene expression by interfering with an RIP- or TRAF2- mediated transactivation signal and directly binds to a novel NF-kappaB-inhibiting protein ABIN. J Cell Biol. 1999;145(7):1471-82.
    [36] Heyninck K, Kreike MM, Beyaert R. Structure-function analysis of the A20-binding inhibitor of NF-kappa B activation, ABIN-1. FEBS Lett. 2003; 536 (1-3):135-40.
    [37] Hughes DP, Marron MB, Brindle NP. The antiinflammatory endothelial tyrosine kinase Tie2 interacts with a novel nuclear factor-kappaB inhibitor ABIN-2. Circ Res. 2003;92(6):630-6.
    [38] Zhang SQ, Kovalenko A, Cantarella G, et al. Recruitment of the IKKsignalosome to the p55 TNF receptor: RIP and A20 bind to NEMO (IKKgamma) upon receptor stimulation. Immunity. 2000;12(3):301-11.
    [39] Van Huffel S, Delaei F, Heyninck K, et al. Identification of a novel A20-binding inhibitor of nuclear factor-kappa B activation termed ABIN-2. J Biol Chem. 2001;276(32):30216-23.

© 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