血管紧张素Ⅱ诱导其前体基因表达及促进血管平滑肌细胞增殖的作用机制研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
血管平滑肌细胞(vascular smooth muscle cells, VSMC)异常增殖是动脉粥样硬化斑块形成、高血压和血管成形术后再狭窄等血管增殖性疾病共同的细胞病理学基础。虽然多种细胞因子和血管活性物质能促进VSMC的增殖,但血管紧张素II(angiotensinII, AngII)在VSMC增殖及高血压、动脉粥样硬化、血管再狭窄等血管重塑性疾病的发生发展过程中处于关键地位。已经证明,在心血管系统中,VSMC不仅是AngII的一个重要来源,而且也是AngII调节血管功能的重要靶细胞。因此,研究AngII调节其前体基因表达及促进VSMC增殖的作用机制,对阐明血管增殖性疾病的分子机制具有重要意义。
     为了阐明AngII调节血管紧张素原基因表达及促进VSMC增殖的分子机制,本研究观察AngII对转录激活蛋白-1(activator protein-1, AP-1)表达活化及其对血管紧张素原基因表达的影响;探讨AP-1与信号转导及转录激活因子5(signal transducer and activator of transcription 5, STAT5)在调节血管紧张素原基因表达中的相互关系;确定AngII诱导VSMC增殖的信号转导途径及roscovitine抗VSMC增殖的作用靶点。
     1 AngII调节其前体基因在VSMC中表达的分子机制
     AP-1是一种激活细胞增殖相关基因表达的转录因子,在AngII诱导其前体基因表达过程中,伴有AP-1(Fos-Jun二聚体)与血管紧张素原基因启动子结合活性的增强。为进一步探讨AngII促进AP-1与其顺式元件结合的分子机制。本部分实验用放线菌酮(cycloheximide, CHX)作为c-Jun的阻断剂,观察AngII对AP-1与血管紧张素原基因启动子相互作用及对血管紧张素原基因表达的影响。实验结果如下:
     1.1不同剂量CHX对VSMC活力的影响
     为了观察CHX对VSMC是否具有的毒性,用不同剂量的CHX处理VSMC后,进行MTT分析。结果显示,在15~45μmol/L浓度范围内,VSMC活力无明显变化。提示在该浓度范围内,CHX对VSMC不产生毒性作用。
     1.2 AngII促进c-Jun蛋白表达与磷酸化
     Western blot结果显示,AngII作用于VSMC 0.5 h后,细胞核内的c-Jun水平即显著升高并在此水平上保持至3 h。而且,免疫细胞化学染色证实,AngII诱导表达的c-Jun主要分布在细胞核内。用抗丝氨酸磷酸化抗体对核提取物进行免疫沉淀后检测磷酸化型c-Jun水平时发现,伴随着c-Jun表达增高,磷酸化型c-Jun水平也平行升高。用CHX预处理VSMC 0.5 h后,再用AngII刺激,c-Jun表达虽然不受影响,但其磷酸化水平明显降低。结果提示,AngII对其前体基因的正反馈调节是通过促进AP-1表达及诱导c-Jun磷酸化活化而实现的,CHX是一种AP-1磷酸化的抑制剂。
     1.3 CHX抑制AngII诱导的血管紧张素原基因表达
     用RT-PCR检测CHX抑制c-Jun磷酸化对血管紧张素原基因表达的影响。结果显示,用AngII处理VSMC 3 h,可显著提高血管紧张素原基因的表达活性。用CHX预处理VSMC可抑制AngII诱导的血管紧张素原基因表达。由此可见,CHX对c-Jun磷酸化的抑制可下调血管紧张素原基因的表达活性,提示AP-1的磷酸化活化是该基因表达所必需的。
     1.4 AngII促进AP-1与血管紧张素原基因启动子结合
     为寻找AngII诱导c-Jun磷酸化与其促进血管紧张素原基因表达之间的关系,用EMSA检测AngII对AP-1与其顺式元件结合活性的影响。结果显示,AngII处理VSMC 0.5 h后,核蛋白与探针的结合活性显著升高,至3 h达高峰。分别加入抗c-Jun和抗STAT5b抗体进行超迁移分析,均可出现抗体-抗原-探针形成的超迁移条带。在CHX预处理的细胞,其核蛋白与探针的结合活性明显下降。为了查明CHX抑制AP-1结合活性的机制,对同样条件下收集的核蛋白进行Western blot分析。结果表明,CHX对AP-1结合活性的抑制效应与AP-1蛋白水平无关,而是CHX抑制AP-1磷酸化的直接结果。上述结果提示,AngII诱导AP-1磷酸化是AngII对血管紧张素原基因正反馈调节的机制之一。
     2 Roscovitine抑制血管平滑肌细胞增殖与c-Jun表达之间的关系
     Roscovitine作为一种细胞周期蛋白依赖激酶(CDKs)的特异性抑制剂,具有诱导肿瘤细胞凋亡和抑制细胞增殖的作用。但roscovitine对VSMC增殖是否具有抑制作用,以及其对VSMC的抑制效应及作用机制与其他细胞是否相同目前尚不清楚。本部分比较roscovitine对VSMC和不同组织来源的细胞系增殖的影响及其作用机制。
     2.1 Roscovitine对细胞增殖的影响
     用细胞计数法检测细胞增殖活力。结果显示,被血清和AngII刺激后,VSMC细胞增殖速率显著加快,分别是对照组(无血清培养组)的2.63和1.68倍。不同浓度的roscovitine(15、30、45μmol/L)预处理VSMC 15 h,均可显著抑制AngII诱导的VSMC增殖。抑制率分别为37.3 %、46.7 %和51.8 %。Roscovitine也可显著抑制血清诱导的VSMC增殖。结果表明,roscovitine可显著抑制体外培养的VSMC增殖。
     Roscovitine也显著抑制血清诱导的HeLa、COS-7、M17细胞的增殖。Roscovitine在30μmol/L时,对HeLa、COS-7、M17细胞的增殖抑制率分别为61.8 %、60.4 %和54.3 % (p<0.001),明显高于对VSMC的抑制作用。结果提示,不同细胞对roscovitine的敏感性有所不同。
     2.2 Roscovitine对c-Jun蛋白表达的影响
     免疫细胞化学染色结果显示,在AngII刺激的VSMC中,c-Jun表达明显增多。Roscovitine预处理VSMC可使c-Jun的表达明显下降。Western blot结果进一步证实,AngII和血清刺激VSMC可显著诱导c-Jun蛋白的表达,roscovitine预处理细胞15 h可显著抑制AngII诱导的c-Jun表达。然而,在HeLa、COS-7和M17细胞中,roscovitine预处理细胞对c-Jun蛋白表达无明显影响。在HeLa、COS-7、M17和VSMC中,roscovitine均不影响STAT5b的表达水平。结果提示,roscovitine抗VSMC增殖的作用与其抑制c-Jun表达有关;roscovitine抑制HeLa、COS-7和M17增殖与抑制c-Jun表达无关。说明roscovitine抗细胞增殖的机理在不同细胞是不同的。
     2.3 Roscovitine在转录水平上抑制c-jun基因在VSMC中的表达
     为进一步探讨roscovitine抑制c-Jun表达的作用环节,用roscovitine处理VSMC、HeLa与M17细胞后,用RT-PCR检测c-jun基因的转录活性。结果显示,roscovitine可下调VSMC中c-jun mRNA水平,但不影响HeLa和M17细胞对c-jun mRNA的表达,与Western blot结果一致。结果提示,roscovitine通过抑制c-jun基因转录而导致VSMC中c-Jun蛋白水平下降。
     3 AngII促进VSMC增殖的信号转导途径及Roscovitine抑制VSMC增殖的作用靶点
     AngII通过与VSMC上的相应受体相互作用而触发细胞增殖信号转导过程,最终引起细胞增殖相关基因表达及VSMC大量增殖。丝裂原活化蛋白激酶(mitogen-activated protein kinase,MAPK)级联反应是转导细胞增殖信号进入细胞核的一条重要信号转导通路。本部分研究观察roscovitine对MAPK信号通路以及对血管紧张素原基因表达的影响,进一步阐明roscovitine抑制VSMC增殖与细胞增殖信号转导之间的关系。
     3.1 Roscovitine对ERK1/2和c-Jun磷酸化的影响
     AngII可诱导ERK1/2快速磷酸化,AngII刺激VSMC 5 min,ERK1/2的磷酸化水平达到高峰,30 min仍维持在较高水平上。给予roscovitine预处理VSMC 15 h后,再用AngII刺激细胞,则磷酸化型ERK1/2几乎检测不到,而各组细胞中ERK1/2的总含量没有变化。结果表明,roscovitine可以完全阻断AngII诱导的ERK1/2磷酸化。为观察roscovitine对AngII诱导c-Jun磷酸化的影响,细胞裂解液用抗丝氨酸磷酸化抗体进行免疫沉淀后,用抗c-Jun抗体检测磷酸化型c-Jun水平。结果显示,roscovitine显著降低c-Jun的磷酸化水平。上述结果表明,在VSMC中,roscovitine不仅抑制AngII诱导的c-Jun表达,而且抑制c-Jun的磷酸化。该抑制作用与其抑制AngII诱导的ERK1/2磷酸化有关。
     3.2 Roscovitine抑制AP-1介导的血管紧张素原基因表达
     本实验进一步探讨roscovitine对c-Jun/AP-1下游基因血管紧张素原基因表达的影响。RT-PCR结果显示,AngII刺激可显著上调血管紧张素原基因的表达。给予roscovitine预处理VSMC,可显著下调AngII刺激引起的血管紧张素原基因的表达水平。
     为进一步证实c-Jun在体内的作用,用c-Jun抗体进行ChIP分析后,用PCR扩增血管紧张素原基因启动子中的AP-1结合序列。实验结果显示,AngII处理细胞可增强c-Jun与血管紧张素原基因启动子的结合活性,给予roscovitine预处理VSMC,可显著抑制c-Jun的结合活性。该结果进一步证实,roscovitine通过抑制c-Jun表达与磷酸化、以及抑制c-Jun与血管紧张素原基因启动子的结合活性,而起到抑制血管紧张素原基因表达的作用。
     4 AP1与STAT5在调节血管紧张素原基因表达中的相互作用
     AngII诱导其前体基因表达与AP-1和STAT5磷酸化活化有关,表明AP-1与STAT5均参与血管紧张素原基因的反式激活过程。本部分实验探讨这两种转录因子在血管紧张素原基因转录激活过程中的相互作用。
     4.1 AngII诱导其前体基因表达与促进c-Jun与STAT5b相互作用有关
     用抗STAT5b抗体对细胞裂解液进行免疫沉淀,抗c-Jun抗体进行Western blot分析。结果显示,AngII刺激前后,均可检测到STAT5b与c-Jun共沉淀,但AngII刺激后,STAT5b与c-Jun的相互作用明显增强。交互式免疫共沉淀进一步证实,STAT5b与c-Jun之间存在物理学上的相互作用,AngII能够诱导二者之间的相互缔合。
     4.2 c-Jun和STAT5b以复合物的形式与血管紧张素原基因调控元件相互作用
     EMSA结果显示,在AngII诱导下,VSMC核蛋白与血管紧张素原基因启动子区AP-1结合位点的结合活性明显增强,加入抗c-Jun或STAT5b抗体均可使核蛋白-DNA复合物发生超迁移。结果表明,核蛋白-DNA复合物中含有c-Jun和STAT5b。由此推测,STAT5b是以直接或间接方式与结合在AP-1结合位点上的AP-1发生相互作用。
     ChIP分析结果显示,用抗STAT5b抗体沉淀富集的DNA-核蛋白复合物中的DNA片断为模板,可以扩增出含AP-1结合位点的血管紧张素原基因调控区片断;用抗c-Jun抗体沉淀富集的DNA-核蛋白复合物中的DNA片断为模板,可以扩增出含SATA5结合位点的血管紧张素原基因调控区片断。AngII刺激后,从抗STAT5b抗体沉淀的复合物中扩增出的含AP-1位点的片断明显增多;经AG490处理后,该基因片断的扩增产物显著减少。这些结果提示,在体内,c-Jun和STAT5b除分别与血管紧张素原基因启动子区相应的顺式元件结合外,同时c-Jun与STAT5b之间也存在相互作用。
     4.3 STAT5b与c-Jun体外结合实验
     GST pull-down结果显示,无论是核蛋白中的STAT5b还是细胞总蛋白中的STAT5b均不能被GST-c-Jun融合蛋白淘选出来。结果表明,c-Jun与STAT5b在体外无直接相互作用。
     4.4 AP-1与STAT5协同激活血管紧张素原基因的表达
     报告基因分析结果显示,c-Jun和STAT5b表达质粒共转染293A细胞时,报告基因的相对活力显著升高,表明AP-1与STAT5在促进血管紧张素原基因表达方面具有正协同作用。在HeLa细胞中,AP-1与STAT5可协同抑制报告基因的表达。提示在不同细胞中,血管紧张素原基因的表达需要不同的转录因子进行组合调控。
     结论
     1. AngII通过促进AP-1表达及磷酸化活化正反馈调节其前体基因表达。
     2. Roscovitine通过特异性抑制c-Jun表达而发挥其对VSMC的抗增殖作用。
     3. Roscovitine抑制VSMC增殖与抑制ERK1/2信号转导通路有关。
     4. STAT5和c-Jun对血管紧张素原基因的转录激活具有正协同作用。
     5. STAT5和AP-1是通过与相应顺式元件结合以及二者之间相互作用实现对血管紧张素原基因表达的组合调控。
The proliferation of vascular smooth muscle cells (VSMC) is the important pathological basis of some vascular proliferative diseases, such as hypertension, atherosclerosis and postangioplasty restenosis. It has been known that a number of the growth factors and vasoactive substances can induce the proliferation of VSMC, AngII play a central role in VSMC proliferation and a variety of pathological processes of vascular remodeling diseases including atherosclerosis, hypertension and restenosis after coronary angioplasty. Not only does VSMC as a source of AngII, but it also serves an important target tissue of AngII-regulating vascular function. Therefore, to investigate mechanisms of AngII-induced VSMC proliferation and regulating its precursor gene expression is more important to clarify the molecular mechanisms of proliferative vascular disease.
     To elucidate the molecular mechanism of angiotensinogen gene expression and VSMC proliferation induced by AngII, we detected the effect of AngII on activator protein-1 (AP-1) expression and activation, and on angiotensinogen gene expression and VSMC proliferation. We also investigated the interaction between AP-1 and STAT5 in the transcriptional activation of angiotensinogen gene. Additionally, signal transduction pathways of AngII-induced VSMC proliferation and the potential targets of the inhibition of VSMC proliferation by roscovitine were studied.
     1 Molecular mechanisms of angiotensinogen gene expression induced by AngII in VSMC
     AP-1 was identified as a transcription factor involved in regulation of the expression of genes responsible for cell proliferation. It has been demonstrated that expression of angiotensinogen gene induced by AngII is related with the increase in the activity of AP-1 binding to angiotensinogen promoter in VSMC. To determine the molecular mechanism of AP-1 binding to its cis-element, we investigated the effect of AngII on expression of angiotensinogen gene and interaction between AP-1 and its cis-element by using cycloheximide (CHX) as an inhibitor of inhibiting c-Jun phosphorylation in this part. The results were as follows:
     1.1 Effects of different concentrations of CHX on viability of the VSMC
     To assess whether CHX has cytotoxic effect on VSMC, the cells were treated with different concentrations of CHX, and then the viability of cells was analyzed by MTT method. The results showed that there was no difference in viability of the VSMC at the range of 15~45μmol/L of CHX. The results suggested that CHX has no cytotoxic effect on VSMC at examined concentration ranges.
     1.2 AngII induces c-Jun proteins expression and phosphorylation
     Western blot results showed that the level of c-Jun in VSMC was significantly increased after the cells were treated with AngII for 0.5 h, and then kept to 3 h at this level. Immunocytochemistry analysis indicated that majority of c-Jun proteins were located in the nucleus. The phosphorylated c-Jun levels were assessed by immunoblotting with anti-c-Jun antibody after the nuclear extracts were immunoprecipitated by anti-phosphoserine antibody. The results confirmed that AngII could induce serine phosphorylation of c-Jun. VSMC was pretreated with CHX, and then stimulated by AngII, the levels of c-Jun protein were not altered, but the phosphorylated c-Jun levels were significantly decreased. These results indicated that the c-Jun expression and phosphorylation induced by AngII is one of the important mechanisms whereby AngII regulates its precursor gene expression in feedback manner. It is found that CHX is an inhibitor of AP-1 phosphorylation.
     1.3 CHX inhibits angiotensinogen gene expression induced by AngII
     The effect of CHX on angiotensinogen gene expression was detected by RT-PCR. The results showed that angiotensinogen gene expression activity was significantly increased after the cells were treated with AngII for 3 h. There was a reduction of angiotensinogen expression induced by AngII in VSMC pretreated with CHX. The results suggested that CHX downregulates the expression of angiotensinogen gene through inhibiting c-Jun phosphorylation. It indicated that the phosphorylation activation of AP-1 is necessary for angiotensinogen gene expression.
     1.4 AngII promotes AP-1 binding to angiotensinogen gene promoter
     To determine the relationship between the c-Jun phosphorylation and angiotensinogen gene expression induced by AngII, the effect of AngII on the binding activity of AP-1 to its cis-acting elements was detected by EMSA. The results showed that the binding activity of AP-1 to the probes was increased significantly after the cells were treated by AngII for 30 min, and reached to preak at 3 h. The supershift analysis using antibody to c-Jun or STAT5b showed that there was appearance of supershifted band. The binding activity of AP-1 to its cis-acting elements was declined significantly in VSMC pretreated with CHX. To explore the mechanism of the inhibition of AP-1 binding activity by CHX, the nuclear protein at same condition was detected by Western blot. The results showed that there was no relationship between c-Jun protein level and AP-1 binding activity. It was results of inhibition of AP-1 phosphorylation by CHX, which suggested that AngII-induced AP-1 phosphorylation was one of the mechanisms of AngII regulating angiotensinogen gene expression.
     2 Roscovitine inhibits VSMC proliferation
     Roscovitine is a potent and specific inhibitor of the CDKs that inhibits cell proliferation and induces apoptosis. However, it was not clear whether roscovitine has same antiproliferative effect in VSMC and other type cells. In this part, we compared the effect of roscovitine on VSMC proliferation with several cell lines from different tissues and the mechanism of inhibition of cell proliferation by roscovitine.
     2.1 Effect of roscovitine on proliferation of different cells
     The cell proliferation activity was determined by cell counting. After the cells were stimulated by 10 % FBS and angiotensin II (10-6 mol/L), the cell number was significantly increased by 2.63 and 1.68 times compared with control, respectively. VSMC was pretreated with different concentration of roscovitine (15, 30, 45μmol/L) for 15 h, the AngII-induced VSMC proliferation was inhibited by roscovitine. The inhibitive rate was 37.3 %, 46.7 % and 51.8 %, respectively. Roscovitine also inhibited FBS- induced VSMC proliferation. The results suggested that roscovitine significantly inhibited VSMC proliferation in vitro.
     Roscovitine also inhibited HeLa, COS-7 and M17 proliferation induced by FBS. The inhibitive rate of roscovitine (30μmol/L) on HeLa, COS-7 and M17 proliferation was 61.8 %, 60.4 % and 54.8 % respectively, which was higher than that of VSMC. These data indicated that the different cell types exhibit variable sensitivity to roscovitine.
     2.2 The effect of roscovitine on c-Jun protein expression
     Immunocytochemistry staining results showed that stimulation with AngII markedly increased expression of c-Jun in VSMC. There was a reduction of AngII-stimulated c-Jun expression in VSMC pretreated with roscovitine. Western blot analysis also confirmed that the levels of c-Jun were significantly increased after the cells were stimulated by AngII or FBS, and the levels of c-Jun were decreased significantly by pretreatment with roscovitine for 15 h. However, in COS-7, HeLa, M17 cell lines, the c-Jun expression was not affected by roscovitine. The STAT5b expression was not affected by roscovitine in HeLa, COS-7, M17 and VSMC. These results suggested that inhibition of c-jun expression was involved in antiproliferative effect of roscovitine and was not related with the inhibition of HeLa, COS-7 and M17. It indicated that there were different mechanisms in roscovitine-inhibiting cell proliferation at different cells.
     2.3 Roscovitine inhibits c-jun gene expression in VSMC at transcriptional level
     The VSMC was pretreated with roscovitine, and then c-jun mRNA level was determined by RT-PCR. We demonstrated that roscovitine caused a down-regulation of c-Jun mRNA in VSMC, but it did not affect c-jun mRNA expression in HeLa and M17 cells. The results indicated that roscovitine down-regulates c-Jun protein expression by inhibiting c-jun gene transcription.
     3 Signal transduction pathways of AngII-induced VSMC proliferation and the targets of the inhibition of VSMC proliferation by roscovitine
     AngII has been shown to induce proliferation of VSMC and through AT1 receptor. Once AngII binds to the AT1R, it activates a series of signaling cascades and this led to the gene expression involved in cell proliferation. The mitogen-activated protein kinase (MAPK) signaling cascades have been shown to play a key role in transducting extracellular signals to nucleus. In this part, we observed the effect of roscovitine on MAPK signaling pathways and angiotensinogen gene expression.
     3.1 Effect of roscovitine on AngII-induced ERK1/2 and c-Jun phosphorylation AngII can rapidly induce ERK1/2 phosphorylation. After stimulation with AngII for 5 min, the phosphorylation of ERK1/2 reached to peak and still kept high level at 30 min. Pretreating the cells with roscovitine for 15 h, the level of ERK1/2 phosphorylation did not detected. The total levels of ERK1/2 were not changed. It showed that roscovitine completely inhibited ERK1/2 phosphorylation induced by AngII. To examine the effect of roscovitine on c-Jun phosphorylation, the cell lysates were immunoprecipitated with anti-phospho-Ser mAb, and immunoblotted using anti-c-Jun antibody, the results showed that phosphorylated c-Jun levels decreased following treatment with roscovitine. The results indicated that roscovitine significantly inhibited the AngII-induced c-Jun expression and phosphorylation, which was associated with the inhibition of ERK1/2 phosphorylation.
     3.2 Roscovitine inhibits AP-1-mediated angiotensinogen gene expression We further examined the effects of roscovitine on expression of
     angiotensinogen, which was one of downstream gene of AP-1. RT-PCR results showed that roscovitine reduced AngII-induced angiotensinogen gene expression.
     To confirm the effect of c-Jun in vivo, ChIP assay was performed. After chromatin was immunoprecipitated with anti-c-Jun antibody, DNA fragments containing AP-1 binding site were subjected to PCR. The ChIP assay demonstrated that treatment with AngII could enhance binding of c-Jun to the angiotensinogen promoter, an effect that could be suppressed by roscovitine, which further demonstrated that roscovitine can block AngII-mediated expression of angiotensinogen by suppressing c-Jun expression and phosphorylation, and then inhibiting the binding of c-Jun to the angiotensinogen gene promoter.
     4 Interaction between AP-1 and STAT5 in transcriptional activation of angiotensinogen gene
     AP-1 and STAT5 phosphorylation activation is involved in AngII-induced its precursor gene expression, which suggested that AP-1 and STAT5 participated in trans-activation of angiotensinogen gene. In the present study, we investigated the interaction between AP-1 and STAT5 in transcriptional activation of angiotensinogen gene.
     4.1 AP-1 and STAT5 interaction is involved in AngII-induced its precursor gene expression
     We carried out co-immunoprecipitation (co-IP) experiments using VSMC lysates treated with or without AngII. The results showed that IP with anti-STAT5b antibody followed by Western blot analysis with anti-c-Jun antibody clearly demonstrated the presence of c-Jun among the immunoprecipitated proteins, and the AngII treatment caused increase in obviously the interaction between c-Jun and STAT5. These results confirmed that the physical interaction between c-Jun and STAT5b occurs in vivo. AngII can induce interaction between c-Jun and STAT5b.
     4.2 The complex formed by c-Jun and STAT5b interacts with cis-elements of angiotensinogen gene
     EMSA results showed that AngII enhanced the binding activity of AP-1 to the promoter of angiotensinogen gene. Supershift analysis was performed with anti-c-Jun or anti-STAT5b antibody. The results indicated that the DNA-protein complex contained both c-Jun and STAT5b. These data indicated that STAT5b and c-Jun can interact directly or indirectly with each other at the AP-1 binding site.
     ChIP assay results showed that DNA sequences contained AP-1 binding site could be amplified when DNA immunoprecipitated using anti-STAT5b antibody were subjected to PCR. DNA immunoprecipitated with anti-c-Jun antibody contained STAT5 binding site. The results demonstrated that there is presence of the interaction between STAT5 and c-Jun when they bind, respectively, to their cis-elements located in the angiotensinogen gene promoter.
     4.3 GST pull-down analysis for interaction between STAT5 and c-Jun in vitro
     GST pull-down results showed that STAT5b in whole cell lysates or nuclear extracts could not be pulled down by using GST-c-Jun fusion proteins. It indicated that c-Jun could not interact directly with STAT5b in vitro.
     4.4 STAT5 synergizes with c-Jun in transactivation of the expression of angiotensinogen gene
     The reporter gene assay results showed that the relative luciferase activity was significantly enhanced, when 293A cells were co-transfected with c-Jun and STAT5b, These results suggested that interaction between AP-1 and STAT5 mediates the synergistic enhancement of angiotensinogen gene expression. Co-transfected c-Jun and STAT5b into HeLa cells could synergistically inhibit angiotensinogen gene expression. These results suggested that in different cells there were distinct transcriptional factors which mediate the angiotensinogen gene expression.
     Conclusion:
     1. AngII positive-feedback regulates its precursor gene expression by inducing AP-1 phosphorylation activation.
     2. Roscovitine exerts an antiproliferative effect on VSMC by specifically inhibiting c-Jun expression.
     3. ERK1/2 signal transduction pathway is involved in inhibition of VSMC proliferation by roscovitine.
     4. STAT5 and c-Jun have a synergistic effect in trans-activation of angiotensinogen gene.
引文
1 聂磊, 韩梅, 温进坤. 血管舒-缩肽在血管平滑肌细胞中的表达和调控. 中国生物化学与分子生物学报, 2005, 21 (1): 107~113
    2 韩梅, 温进坤, 郑斌, 等. 血清饥饿可诱导人血管平滑肌细胞再分化. 中国生物化学与分子生物学报, 2003, 19 (2): 250~255
    3 聂磊,韩梅, 温进坤. JAK-STAT5 参与血管平滑肌血管紧张素原和心钠素基因的转录激活. 中国生物化学与分子生物学报,2006, 22 (1): 70~76
    4 Lee SA, Dritschilo A, Jung M. Impaired ionizing radiation-induced activation of a nuclear signal essential for phosphorylation of c-jun by dually phosphorylated c-jun amino-terminal kinases in ataxia telangiectasia fibroblasts. J Biol Chem, 1998, 273 (49): 32889 ~32894
    5 Franklin CC, Unlap T, Adler V, et al. Multiple signal transduction pathways mediate c-Jun protein phosphorylation. Cell Growth Differ, 1993, 4 (5): 377~385
    6 Bergelson S, KlingmüLler U, Socolovsky M, et al. Tyrosine residues within the intra-cellular domain of the erythropoietin receptor mediate activation of AP-1 transcription factors. J Biol Chem, 1998, 273 (4): 2396~2401
    7 Gronowski AM, Stunff CL, Rotwein P. Acute nuclear actions of growth hormone (GH): Cycloheximide inhibits inducible activator protein-L activity, but does not block GH-regulated signal transducer and activator of transcription activation or gene expression. Endocrinology, 1996, 137 (1): 55~64
    8 Minden A, Lin A, Smeal T, et al. c-Jun N-terminal phosphorylation correlates with activation of the JNK subgroup but not the ERK subgroup of mitogen-activated protein kinase. Mol Cell Biol, 1994, 14 (385): 6683~6688
    9 Smeal T, Binetruy B, Mercola DA, et al. Oncogenic and transcriptionalcooperation with HA-ras requires phosphorylation of c-Jun on serines 63 and 73. Nature, 1991, 354 (6353): 494~496
    10 Karin M. The regulation of AP-1 activity by mitogen-activated protein kinases. J Biol Chem, 1995, 270 (28): 16483~16486
    11 Binetruy B, Smeal T, Karin M. Ha-Ras augments c-Jun activity and stimulates phosphorylation of its activation domain. Nature 1991; 351 (6322): 122~127
    12 Widsom R. AP-1: one switch for many signals. Exp Cell Res, 1999, 253 (1): 180~185
    1 De Azevedo WF, Gaspar RT, Canduri F, et al. Molecular model of cyclin-dependent kinase 5 complexed with roscovitine. Biochem Biophys Res Commun, 2002, 297: 1154~1158
    2 De Azevedo WF, Leclerc S, Meijer L, et al. Inhibition of cyclin-dependent kinases by purine analogues: crystal structure of human cdk2 complexed with roscovitine. Eur J Biochem, 1997, 243: 518~526
    3 Schutte B, Nieland L, Van Engeland M, et al. The effect of the cyclin-dependent kinase inhibitor olomoucine on cell cycle kinetics. Exp Cell Res, 1997, 236: 4~15
    4 Somerville L, Cory JG. Enhanced roscovitine-induced apoptosis is mediated by a caspase-3-like activity in deoxyadenosine-resistant mouse leukemia L1210 cells. Anticancer Res, 2000, 20(5B): 3347~3355
    5 Wesierska-Gadek J, Gueorguieva M, Horky M. Roscovitine-induced up-regulation of p53AIP1 protein precedes the onset of apoptosis in human MCF-7 breast cancer cells. Mol Cancer Ther, 2005, 4: 113~124
    6 Atienza C, Elliott MJ, Dong YB, et al. Adenovirusmediated E2F-1 gene transfer induces an apoptotic response in human gastric carcinoma that is enhanced by cyclin dependent kinase inhibitors. Int J Mol Med, 2000, 6: 55~63
    7 Mihara M, Shintani S, Kiyota A, et al. Cyclin-dependent kinase inhibitor (roscovitine) suppresses growth and induces apoptosis by regulating Bcl-x in head and neck squamous cell carcinoma cells. Int J Oncol, 2002, 21(1): 95~101
    8 Goodyear S, Sharma MC. Roscovitine regulates invasive breast cancer cell (MDA-MB231) proliferation and survival through cell cycle regulatory protein cdk5. Exp Mol Pathol, 2007, 82: 25~32
    9 Pippin JW, Qu Q, Meijer L, et al. Direct in vivo inhibition of the nuclear cell cycle cascade in experimental mesangial proliferativeglomerulonephritis with roscovitine, a novel cyclin-dependent kinase antagonist. J. Clin Invest, 1997, 100: 2512~2520
    10 Atanasova G, Jans R, Zhelev N, et al. Effects of the cyclin-dependent kinase inhibitor CYC202 (R-roscovitine) on the physiology of cultured human keratinocytes. Biochem Pharmacol, 2005, 70: 824~836
    11 Yakisich JS, Siden A, Idoyaga Vargas V, et al. Early inhibition of DNA synthesis in the developing rat cerebral cortex by the purine analogues olomoucine and roscovitine. Biochem Biophys Res Commun, 1998, 243: 674~677
    12 Geisterfer AA, Peach MJ, Owens GK. Angiotensin II induces hypertrophy, not hyperplasia, of cultured rat aortic smooth muscle cells. Circ Res, 1988, 62: 749~758
    13 Ramos K, Cox LR. Primary cultures of rat aortic endothelial and smooth muscle cells. An in vitro model to study xenobiotic-induced vascular cytotoxicity. In Vitro Cell Dev Biol, 1987, 23: 288~296
    14 Bishop-Bailey D, Hla T, Warner TD. Intimal smooth muscle cells as a target for peroxisome proliferator-activated receptor-gamma ligand therapy. Circ Res, 2002, 91: 210~217
    15 姜广建, 温进坤, 韩梅, 等. 高血压相关基因 hrg-1 在血管平滑肌细胞再分化过程中的表达及功能. 中国生物化学与分子生物学报, 2004, 20 (2): 195~199
    16 Wen JK, Han M. Comparative study of induction of iNOS mRNA expression in vascular cells of different species. Biochemistry (Mosc), 2000, 65: 1376~1379
    17 Mgbonyebi OP, Russo J, Russo IH. Roscovitine induces cell death and morphological changes indicative of apoptosis in MDA-MB-231 breast cancer cells. Cancer Res, 1999, 59: 1903~1910
    18 Wesierska-Gadek J, Gueorguieva M, Wojciechowski J, et al. Cell cycle arrest induced in human breast cancer cells by cyclin-dependent kinase inhibitors: a comparison of the effects exerted by roscovitine andolomoucine. Pol J Pharmacol, 2004, 56: 635~641
    19 McClue SJ, Blake D, Clarke R, et al. In vitro and in vivo antitumor properties of the cyclin dependent kinase inhibitor CYC202 (R-roscovitine). Int J Cancer, 2002, 102: 463~468
    20 Alessi F, Quarta S, Savio M, et al. The cyclindependent kinase inhibitors olomoucine and roscovitine arrest human fibroblasts in G1 phase by specific inhibition of CDK2 kinase activity. Exp Cell Res, 1998, 245: 8~18
    21 Wang N, Verna L, Hardy S, et al. Stemerman, c-Jun triggers apoptosis in human vascular endothelial cells. Circ Res, 1999, 85: 387~393
    22 Bossy-Wetzel E, Bakiri L, Yaniv M. Induction of apoptosis by the transcription factor c-Jun. EMBO J, 1997, 16: 1695~1709
    23 Eferl R, Sibilia M, Hilberg F, et al. Functions of c-Jun in liver and heart development. J Cell Biol, 1999, 145: 1049~1061
    24 Zhan Y, Kim S, Yasumoto H, et al. Effects of dominant-negative c-Jun on platelet-derived growth factor–induced vascular smooth muscle cell proliferation. Arterioscler Thromb Vasc Biol, 2002, 22: 82~88
    25 Li BS, Zhang L, Takahashi S, et al. Cyclin-dependent kinase 5 prevents neuronal apoptosis by negative regulation of c-Jun N-terminal kinase 3. EMBO J, 2002, 21: 324~333
    26 Ghahremani MH, Keramaris E, Shree T, et al. Interaction of the c-Jun/JNK pathway and cyclin-dependent kinases in death of embryonic cortical neurons evoked by DNA damage. J Biol Chem, 2002, 277: 35586~35596
    27 Monaco EA, Beaman-Hall CM, Mathur A, et al. Roscovitine, olomoucine, purvalanol: inducers of apoptosis in maturing cerebellar granule neurons. Biochem Pharmacol, 2004, 67: 1947~1964
    28 Du L, Lyle CS, Obey TB, et al. Inhibition of cell proliferation and cell cycle progression by specific inhibition of basal JNK activity: evidence that mitotic Bcl-2 phosphorylation is JNK-independent. J Biol Chem, 2004, 279: 11957~11966
    1 Ross R. The pathogenesis of atherosclerosis: a perspective for the 1990s. Nature, 1993, 362(6423): 801~809
    2 Berk BC, Vekshtein V, Gordon HM, et al. Angiotensin II-stimulated protein synthesis in cultured vascular smooth muscle cells. Hypertension, 1989, 13(4): 305~314
    3 Geisterfer AA, Peach MJ, Owens GK. Angiotensin II induces hypertrophy, not hyperplasia, of cultured rat aortic smooth muscle cells. Circ Res, 1988, 62(4): 749~756
    4 Schorb W, Peeler TC, Madigan NN, et al. Angiotensin II-induced protein tyrosine phosphorylation in neonatal rat cardiac fibroblasts. J Biol Chem, 1994, 269(30): 19626~19632
    5 Chang L, Karin M. Mammalian MAP kinase signalling cascades. Nature, 2001, 410(6824): 37~40
    6 Hommes DW, Peppelenbosch MP, van Deventer SJH. Mitogen activated protein kinase (MAPK) signal transduction pathways and novel anti-inflammatory targets. Gut, 2003, 52: 144~151
    7 Whitmarsh AJ, Davis RJ. Transcription factor AP-1 regulation by mitogen-activated protein kinase signal transduction pathways. J Mol Med, 1996, 74(10): 589~607
    8 Fisher M, Liu B, Glennon PE, et al. Downregulation of the ERK 1 and 2 mitogen activated protein kinases using antisense oligonucleotides inhibits proliferation of porcine vascular smooth muscle cells. Atherosclerosis, 2001, 156(2): 289~295
    9 Kim S, Iwao H. Stress and vascular responses: mitogen-activated protein kinases and activator protein-1 as promising therapeutic targets of vascular remodeling. J Pharmacol Sci, 2003, 91(3): 177~181
    10 Liao B, Monia DF, Dean N, et al. Protein kinase C-ζ mediates angiotensin II activation of ERK1/2 in vascular smooth muscle cells. J Biol Chem.1997, 272(10): 6146~6150
    11 Viedt C, Soto U, Krieger-Brauer HI, et al. Differential activation of mitogen-activated protein kinases in smooth muscle cells by angiotensin II: involvement of p22phox and reactive oxygen species. Arterioscler Thromb Vasc Biol, 2000, 20(4): 940~948
    12 Shaulian E, Karin M. AP-1 as a regulator of cell life and death. Nat Cell Biol, 2002, 4(5): E131~E136
    13 Angel P, Karin M. The role of Jun, Fos and the AP-1 complex in cell-proliferation and transformation. Biochim Biophys Acta, 1991, 1072(2-3): 129~157
    14 Han TH, Lamph WW, Prywes R. Mapping of epidermal growth factor-, serum-, and phorbol ester-responsive sequence elements in the c-jun promoter. Mol Cell Biol, 1992, 12(10): 4472~4477
    15 Angel P, Hattori K, Smeal T, et al. The jun proto-oncogene is positively autoregulated by its product, Jun/AP-1. Cell, 1988, 55(5):875~885
    16 Meijer L, Borgne A, Mulner O, et al. Biochemical and cellular effects of roscovitine, a potent and selective inhibitor of the cyclin-dependent kinases cdc2, cdk2 and cdk5. Eur J Biochem, 1997, 243: 527~536.
    17 Zhao YL, Liu J, Li L, et al. Role of Ras/PKC~/MEK/ERK1/2 signaling pathway in angiotensinII-induced vascular smooth muscle cell proliferation. Regul Pepti, 2005, 128(1): 43~50
    18 李爱英, 温进坤, 韩梅. AP-1 在 AngII 正反馈调节其前体基因表达中的作用. 生物化学与生物物理学进展,2006, 33(8): 775~780
    19 聂磊, 韩梅, 温进坤. 血管舒-缩肽在血管平滑肌细胞中的表达和调控. 中国生物化学与分子生物学报,2005, 21(1): 94~100
    20 Khachigian LM, Fahmy RG, Zhang G, et al. c-Jun Regulates Vascular Smooth muscle cell growth and neointima formation after arterial injury: inhibition by a novel DNA enzyme targeting c-Jun. J Biol Chem, 2002, 277(25): 22985~22991
    21 Ross R. The pathogenesis of atherosclerosis--an update. N Engl J Med,1986, 314: 488~500
    22 Newby AC, George SJ. Proposed roles for growth factors in mediating smooth muscle proliferation in vascular pathologies. Cardiovasc Res, 1993, 27(7): 1173~118
    23 McClue SJ, Blake D, Clarke R, et al. In vitro and in vivo antitumor properties of the cyclin-dependent kinase inhibitor CYC202 (R-roscovitine). Int J Cancer, 2002, 102(5): 463~468
    24 Alessi F, Quarta S, Savio M, et al. The cyclin-dependent kinase inhibitors olomoucine and roscovitine arrest human fibroblasts in G1 phase by specific inhibition of CDK2 kinase activity. Exp Cell Res, 1998, 245(1): 8~18
    25 Mgbonyebi OP, Russo J, Russo IH. Roscovitine induces cell death and morphological changes indicative of apoptosis in MDA-MB-231 breast cancer cells. Cancer Res, 1999, 59(8): 1903~1910
    26 O’Sullivana M, Scotta SD, McCarthya N, et al. Differential cyclin E expression in human in-stent stenosis smooth muscle cells identifies targets for selective anti-restenosis therapy. Cardiovasc Res, 2003, 60(3): 673~ 683
    27 Kyaw M, Yoshizumi M, Tsuchiya K, et al. Antioxidants inhibit JNK and p38 MAPK activation but not ERK1/2 activation by angiotensin II in rat aortic smooth muscle cells. Hypertens Res, 2001, 24(3): 251~261
    28 Wilkie N, Morton C, Ng LL, et al. Stimulated mitogen activated protein kinase is necessary but not sufficient for the mitogenic response to angiotensin II. A role for phospholipase D. J Biol Chem, 1996, 271(50): 32447~32453
    29 Zhou CH, Qian ZY, Zheng SG, et al. ERK1/2 pathway is involved in the inhibitory effect of crocetin on angiotensin II-induced vascular smooth muscle cell proliferation. Eur J Pharmacol, 2006, 535(1-3): 61~68
    30 Atanasova G, Jans R, Zhelev N, et al. Effects of the cyclin-dependent kinase inhibitor CYC202 (R-roscovitine) on the physiology of culturedhuman keratinocytes. Biochem Pharmacol, 2005, 70(6): 824~836
    31 Whittaker SR, Walton MI, Garrett MD, et al. The cyclindependent kinase inhibitor CYC202 (R-roscovitine) inhibits retinoblastoma protein phosphorylation, causes loss of cyclin D1, and activates the mitogen-activated protein kinase pathway. Cancer Res, 2004, 64(1): 262~272
    32 Jin N, Hatton ND, Harrington MA, et al. H2O2-induced egr-1, fra-1, and c-jun gene expression is mediated by tyrosine kinase in aortic smooth muscle cells. Free Radic Biol Med, 2000, 29(8): 36~746
    33 Eguchi S, Inagami T. Signal transduction of angiotensin II type 1 receptor through receptor tyrosine kinase. Regul Pept, 2000, 91(1-3): 13~20
    34 Curran T, Franza Jr BR. Fos and Jun: the AP-1 connection. Cell, 1988, 55(3): 395~397
    35 Hirai S, Bourachot B, Yaniv M. Both Jun and Fos contribute to transcription activation by the heterodimer. Oncogene, 1990, 5(1): 39~46
    36 Vogt PK, Bos TJ. Jun: oncogene and transcription factor. Adv Cancer Res, 1990, 55: 1~35
    1 Marrcro MB, Schicffer B, Paxton WG, et al. Direct stimulation of Jak/STAT pathway by the angiotensin II AT1 receptor. Nature, 1995, 375:247~250
    2 聂磊, 韩梅, 温进坤. JAK-STAT 参与血管平滑肌细胞血管紧张素原和心钠素基因的转录激活. 中国生物化学与分子生物学报, 2006, 22 (1): 70~76
    3 Guo YL, Mascareno E, Siddiqui MAQ. Distinct components of janus kinase/signal transducer and activator of transcription signaling pathway mediate the regulation of systemic and tissue localized renin-angiotensin system. Mol Endocrinol, 2004, 18 (4): 1033~1041
    4 Brasier AR, Li JY. Mechanisms for inducible control of angiotensinogen gene transcription. Hypertension, 1996, 27 (3): 465~475
    5 Orlinick JR, Chao MV. Interactions of cellular polypeptides with the cytoplasmic domain of the mouse Fas antigen. J Biol Chem, 1996, 271 (15): 8627~8632
    6 李爱英, 温进坤, 韩梅. AP-1 在 AngII 正反馈调节其前体基因表达中的作用. 生物化学与生物物理学进展, 2006, 33 (8): 775~780
    7 Marrero MB, Schieffer B, Li B, et al. Role of jansus kinase/signal transducer and activator of transcription and mitogen-activated protein kinase cascades in angiotensin II and platelet derived growth factor inducedvascular smooth muscle cell proliferaction. J Boil chem, 1997, 272 (39): 24684~24690
    8 Mascareno E, Dhar M, Siddiqui MAQ. Signal transduction and activator of transcription (STAT) protein-dependent activation of angiotensinogen promoter: A cellular signal for hypertrophy in cardiac muscle. Proc Natl Acad Sci, 1998, 95 (10): 5590~5594
    9 聂磊, 韩梅, 温进坤. 血管舒-缩肽在血管平滑肌细胞中的表达和调控. 中国生物化学与分子生物学报, 2005, 21 (1): 107~113
    10 Lee PJ, Camhi SL, Chin BY, et al. AP-1 and STAT mediate hyperoxia- induced gene transcription of heme oxygenase-1. Am J Physiol Lung Cell Mol Physiol, 2000, 279 (1): 175~182
    11 Xu WL, Comhair SAA, Zheng S, et al. STAT-1 and c-Fos interaction in nitric oxide synthase-2 gene activation. Am J Physiol Lung Cell Mol Physiol, 2003, 285 (1): 137~148
    12 Nakajima H, Brindle PK, Handa M, et al. Functional interaction ofSTAT5 and nuclear receptor co-repressor SMRT: implications in negative regulation of STAT5-dependent transcription. EMBO J, 2001, 20 (23): 6836~6844
    13 Shuai K: Modulation of STAT signaling by STAT-interacting proteins. Oncogene, 2000, 19: 2638~2644
    14 Zhang X, Wreszczynska MH, Horvath CM, et al. Interacting regions in Stat3 and c-Jun that participate in cooperative transcriptional activation. Mol Cell Biol, 1999, 19: 7138~714
    15 Horvai AE, Xu L, Korzus E, et al. Nuclear integration of JAK/STAT and Ras/AP-1 signaling by CBP and p300. Proc Natl Acad Sci, 1997, 94: 1074~1079
    1 Geisterfer AA, Peach MJ, Owens GK. Angiotensin II induces hypertrophy, not hyperplasia, of cultured rat aortic smooth muscle cells. Circ Res, 1988, 62: 749~756
    2 Xi XP, Graf K, Goetze S, et al. Central role of the MAPK pathway in ang II-mediated DNA synthesis and migration in rat vascular smooth muscle cells. Arterioscler Thromb Vasc Biol, 1999, 19: 73~82
    3 Henriksen EJ, Jacob S, Kinnick TR, et al. Selective angiotensin II receptor receptor antagonism reduces insulin resistance in obese Zucker rats. Hypertension, 2001, 38: 884~890
    4 Igarashi M, Hirata A, Yamaguchi H, et al. Candesartan inhibits carotid intimal thickening and ameliorates insulin resistance in balloon-injured diabetic rats. Hypertension, 2001, 38: 1255~1259
    5 Schiffrin EL, Park JB, Intengan HD, et al. Correction of arterial structure and endothelial dysfunction in human essential hypertension by the angiotensin receptor antagonist losartan. Circulation, 2000, 101: 1653~1659
    6 Griendling KK, Sorescu D, Ushio-Fukai M. NAD(P)H oxidase: role incardiovascular biology and disease. Circ Res, 2000, 86: 494~501
    7 Hunyady L, Catt KJ. Pleiotropic AT1 receptor signaling pathways mediating physiological and pathogenic actions of angiotensin II. Mol Endocrinol, 2006, 20: 953~970
    8 Suzuki H, Motley ED, Frank GD, et al. Recent progress in signal transduction research of the angiotensin II type-1 receptor: protein kinases, vascular dysfunction and structural requirement. Curr Med Chem Cardiovasc Hematol Agents, 2005, 3: 305~322
    9 Marrero MB, Fulton D, Stepp D, et al. Angiotensin II-induced insulin resistance and protein tyrosine phosphatases. Arterioscler Thromb Vasc Biol, 2004, 24: 2009~2013
    10 Schmitz U, Ishida T, Ishida M, et al. Angiotensin II stimulates p21-activated kinase in vascular smooth muscle cells: role in activation of JNK. Circ Res,1998, 82: 1272~1278
    11 Ushio-Fukai M, Griendling KK, Akers M, et al. Temporal dispersion of activation of phospholipase C-beta1 and -gamma isoforms by angiotensin II in vascular smooth muscle cells. Role of alphaq/11, alpha12, and beta gamma G protein subunits. J Biol Chem, 1998, 273: 19772~19777
    12 Ushio-Fukai M, Alexander RW, Akers M, et al. Angiotensin II receptor coupling to phospholipase D is mediated by the betagamma subunits of heterotrimeric G proteins in vascular smooth muscle cells. Mol Pharmacol, 1999, 55: 142~149
    13 Yan C, Kim D, Aizawa T, et al. Functional interplay between angiotensin II and nitric oxide: cyclic GMP as a key mediator. Arterioscler Thromb Vasc Biol, 2003, 23: 26~36
    14 Jin L, Ying Z, Hilgers RH, et al. Increased RhoA/Rho-kinase signaling mediates spontaneous tone in aorta from angiotensin II-induced hypertensive rats. J Pharmacol Exp Ther, 2006, 318: 288~295
    15 Uehata M, Ishizaki T, Satoh H, et al. Calcium sensitization of smooth muscle mediated by a Rho-associated protein kinase in hypertension.Nature, 1997, 389: 990~994
    16 Srivastava AK. High glucose-induced activation of protein kinase signaling pathways in vascular smooth muscle cells: a potential role in the pathogenesis of vascular dysfunction in diabetes (review). Int J Mol Med, 2002, 9: 85~89
    17 Giugliano D, Ceriello A, Paolisso G. Oxidative stress and diabetic vascular complications. Diabetes Care, 1996, 19: 257~267
    18 Yasunari K, Kohno M, Kano H, et al. Antioxidants improve impaired insulin-mediated glucose uptake and prevent migration and proliferation of cultured rabbit coronary smooth muscle cells induced by high glucose. Circulation, 1999, 99: 1370~378
    19 Touyz RM, Schiffrin EL. Signal transduction mechanisms mediating the physiological and pathophysiological actions of angiotensin II in vascular smooth muscle cells. Pharmacol Rev, 2000, 52: 639~672
    20 Griendling KK, Sorescu D, Ushio-Fukai M. NAD(P)H oxidase: role in cardiovascular biology and disease. Circ Res, 2000, 86: 494~501
    21 Campbell WB, Gebremedhin D, Pratt PF, et al. Identification of epoxyeicosatrienoic acids as endothelium-derived hyperpolarizing factors. Circ Res, 1996, 78: 415~423
    22 Grant SL, Lassegue B, Griendling KK, et al. Specific regulation of RGS2 messenger RNA by angiotensin II in cultured vascular smooth muscle cells. Mol Pharmacol, 2000, 57: 460~467
    23 Heximer SP, Knutsen RH, Sun X, et al. Hypertension and prolonged vasoconstrictor signaling in RGS2-deficient mice. J Clin Invest, 2003, 111:1259~1270
    24 Adams LD, Geary RL, McManus B, et al. A comparison of aorta and vena cava medial message expression by cDNA array analysis identifies a set of
    68 consistently differentially expressed genes, all in aortic media. Circ Res, 2000, 87: 623~631
    25 Cho H, Harrison K, Schwartz O, et al. The aorta and heart differentiallyexpress RGS (regulators of G-protein signalling) proteins that selectively regulate sphingosine 1-phosphate, angiotensin II and endothelin-1 signalling. Biochem J, 2003, 371: 973~980
    26 Touyz RM. Reactive oxygen species and angiotensin II signaling in vascular cells—implications in cardiovascular disease. Braz J Med Biol Res, 2004, 37: 1263~1273
    27 Taylor WR, Griendling KK. Role of NADH/NADPH oxidase-derived H2O2 in angiotensin II-induced vascular hypertrophy. Hypertension, 1998, 32: 488~495
    28 Ushio-Fukai M, Zafari AM, Fukui T, et al. p22phox is a critical component of the superoxide-generating NADH/NADPH oxidase system and regulates angiotensin II-induced hypertrophy in vascular smooth muscle cells. J Biol Chem, 1996, 271: 23317~23321
    29 Touyz RM, Yao G, Quinn MT, et al. p47phox associates with the cytoskeleton through cortactin in human vascular smooth muscle cells: role in NAD(P)H oxidase regulation by angiotensin II. Arterioscler Thromb Vasc Biol, 2005, 25: 512~518
    30 Ohtsu H, Frank GD, Utsunomiya H, et al. Redox-dependent protein kinase regulation by angiotensin II: mechanistic insights and its pathophysiology. Antioxid Redox Signal, 2005, 7: 1315~1326
    31 Salmeen A, Barford D. Functions and mechanisms of redox regulation of cysteine-based phosphatases. Antioxid Redox Signal, 2005, 7: 560~577
    32 Arredondo M, Nunez MT. Iron and copper metabolism. Mol Aspects Med, 2005, 26: 313~327
    33 Chen XL, Dodd G, Thomas S, et al. Activation of Nrf2/ARE pathway protects endothelial cells from oxidant injury and inhibits inflammatory gene expression. Am J Physiol Heart Circ Physiol, 2006, 290: H1862~H1870
    34 Papaiahgari S, Zhang Q, Kleeberger SR, et al. Hyperoxia stimulates an Nrf2-ARE transcriptional response via ROSEGFR-PI3K-Akt/ERK MAPkinase signaling in pulmonary epithelial cells. Antioxid Redox Signal, 2006, 8: 43~52
    35 Wu S, Gao J, Ohlemeyer C, Roos D, et al. Activation of AP-1 through reactive oxygen species by angiotensin II in rat cardiomyocytes. Free Radic Biol Med, 2005, 39: 1601~1610
    36 Gryglewski RJ, Palmer RM, Moncada S. Superoxide anion is involved in the breakdown of endothelium-derived vascular relaxing factor. Nature, 1986, 320: 454~456
    37 Taniyama Y, Ushio-Fukai M, Hitomi H, et al. Role of p38MAPK and MAPKAPK-2 in angiotensin II-induced Akt activation in vascular smooth muscle cells. Am J Physiol Cell Physiol, 2004, 287: C494~C499
    38 Sugden PH, Clerk A. Regulation of the ERK subgroup of MAP kinase cascades through G protein-coupled receptors. Cell Signal, 1997, 9: 337~351
    39 Eguchi S, Matsumoto T, Motley ED, et al. Identification of an essential signaling cascade for mitogen-activated protein kinase activation by angiotensin II in cultured rat vascular smooth muscle cells. Possible requirement of Gq-mediated p21ras activation coupled to a Ca2+/calmodulin-sensitive tyrosine kinase. J Biol Chem, 1996, 271: 14169~14175
    40 Liao DF, Monia B, Dean N,et al. Protein kinase C-zeta mediates angiotensin II activation of ERK1/2 in vascular smooth muscle cells. J Biol Chem, 1997, 272: 6146~6150
    41 Bokemeyer D, Lindemann M, Kramer HJ. Regulation of mitogenactivated protein kinase phosphatase-1 in vascular smooth muscle cells. Hypertension, 1998, 32: 661~667
    42 Cui T, Nakagami H, Iwai M, et al. Pivotal role of tyrosine phosphatase SHP-1 in AT2 receptor-mediated apoptosis in rat fetal vascular smooth muscle cell. Cardiovasc Res, 2001, 49: 863~871
    43 Touyz RM, He G, Deng LY, et al. Role of extracellular signal-regulatedkinases in angiotensin II-stimulated contraction of smooth muscle cells from human resistance arteries. Circulation, 1999, 99: 392~399
    44 Allen RT, Krueger KD, Dhume A, et al. Sustained Akt/PKB activation and transient attenuation of c-jun N-terminal kinase in the inhibition of apoptosis by IGF-1 in vascular smooth muscle cells. Apoptosis, 2005, 10: 525~535
    45 Nishimura K, Li W, Hoshino Y, et al. Role of AKT in cyclic strain-induced endothelial cell proliferation and survival. Am J Physiol Cell Physiol, 2006, 290: C812~C821
    46 Rocic P, Jo H, Lucchesi PA. A role for PYK2 in ANG II-dependent regulation of the PHAS-1-eIF4E complex by multiple signaling cascades in vascular smooth muscle. Am J Physiol Cell Physiol, 2003, 285: C1437~C1444
    47 Ishida M, Ishida T, Thomas SM, et al. Activation of extracellular signal-regulated kinases (ERK1/2) by angiotensin II is dependent on c-Src in vascular smooth muscle cells. Circ Res, 1998, 82: 7~12
    48 Ishida T, Ishida M, Suero J, et al. Agoniststimulated cytoskeletal reorganization and signal transduction at focal adhesions in vascular smooth muscle cells require c-Src. J Clin Invest, 1999, 103: 789~797
    49 Ichijo H, Nishida E, Irie K, et al. Induction of apoptosis by ASK1, a mammalian MAPKKK that activates SAPK/JNK and p38 signaling pathways. Science, 1997, 275: 90~94
    50 Tobiume K, Matsuzawa A, Takahashi T, et al. ASK1 is required for sustained activations of JNK/p38 MAP kinases and apoptosis. EMBO, 2001, 2: 222~228
    51 Kudoh S, Komuro I, Mizuno T, et al. Angiotensin II stimulates c-Jun NH2-terminal kinase in cultured cardiac myocytes of neonatal rats. Circ Res, 1997, 80: 139~146
    52 Force T, Pombo CM, Avruch JA, et al. Stress-activated protein kinases in cardiovascular disease. Circ Res, 1996, 78: 947~953
    53 Kim S, Murakami T, Izumi Y, et al. Extracellular signal-regulated kinase and c-Jun NH2-terminal kinase activities are continuously and differentially increased in aorta of hypertensive rats. Biochem Biophys Res Commun, 1997, 236: 199~204
    54 Eguchi S, Dempsey PJ, Frank GD,et al. Activation of MAPKs by angiotensin II in vascular smooth muscle cells. Metalloprotease-dependent EGF receptor activation is required for activation of ERK and p38 MAPK but not for JNK. J Biol Chem , 2001, 276: 7957~7962
    55 Nishida M, Tanabe S, Maruyama Y, et al. G alpha 12/13-and reactive oxygen species-dependent activation of c-Jun NH2-terminal kinase and p38 mitogen-activated protein kinase by angiotensin receptor stimulation in rat neonatal cardiomyocytes. J Biol Chem, 2005, 280: 18434~18441
    56 Ohtsu H, Mifune M, Frank GD, et al. Signal-crosstalk between Rho/ROCK and c-Jun NH2-terminal kinase mediates migration of vascular smooth muscle cells stimulated by angiotensin II. Arterioscler ThrombVasc Biol, 2005, 25: 1831~1836
    57 Touyz RM, Yao G, Viel E, et al. Angiotensin II and endothelin-1 regulate MAP kinases through different redox-dependent mechanisms in human vascular smooth muscle cells. J Hypertens, 2004, 22: 1141~1149
    58 Taniyama Y, Weber DS, Rocic P, et al. Pyk2- and Srcdependent tyrosine phosphorylation of PDK1 regulates focal adhesions. Mol Cell Biol, 2003, 23: 8019~8029
    59 Berk BC, Corson MA. Angiotensin II signal transduction in vascular smooth muscle: role of tyrosine kinases. Circ Res, 1997, 80: 607~616
    60 Madamanchi NR, Li S, Patterson C, et al. Reactive oxygen species regulate heat-shock protein 70 via the JAK/STAT pathway. Arterioscler Thromb Vasc Biol, 2001, 21: 321~326
    61 Frank GD, Saito S, Motley ED, et al. Requirement of Ca2+ and PKC delta for Janus kinase 2 activation by angiotensin II: involvement of PYK2. Mol Endocrinol, 2002,16: 367~377
    62 Marrero MB, Venema VJ, Ju H, et al. Regulation of angiotensin II-induced JAK2 tyrosine phosphorylation: roles of SHP-1 and SHP-2. Am J Physiol Cell Physiol, 1998, 275: C1216~C1223
    63 Okuda M, Kawahara Y, Nakayama I,et al. Angiotensin II transduces its signal to focal adhesions via angiotensin II type 1 receptors in vascular smooth muscle cells. FEBS Lett, 1995, 368: 343~347
    64 Rocic P, Govindarajan G, Sabri A, et al. A role for PYK2 in regulation of ERK1/2 MAP kinases and PI 3-kinase by ANG II in vascular smooth muscle. Am J Physiol Cell Physiol, 2001, 280: C90~C99
    65 Heeneman S, Haendeler J, Saito Y, et al. Angiotensin II induces transactivation of two different populations of the plateletderived growth factor beta receptor. Key role for the p66 adaptor protein Shc. J Biol Chem , 2000, 275: 15926~15932
    66 Linseman DA, Benjamin CW, Jones DA. Convergence of angiotensin II and platelet-derived growth factor receptor signaling cascades in vascular smooth muscle cells. J Biol Chem, 1995, 270: 12563~12568
    67 Gratton JP, Bernatchez P, Sessa WC. Caveolae and caveolins in the cardiovascular system. Circ Res, 2004, 94: 1408~1417
    68 203. Ushio-Fukai M, Griendling KK, Becker PL, et al. Epidermal growth factor receptor transactivation by angiotensin II requires reactive oxygen species in vascular smooth muscle cells. Arterioscler Thromb Vasc Biol, 2001, 21: 489~495
    69 Prenzel N, Zwick E, Daub H, et al. EGF receptor transactivation by G-protein-coupled receptors requires metalloproteinase cleavage of proHB-EGF. Nature, 1999, 402: 884~888
    70 Ogihara T, Asano T, Ando K, et al. Angiotensin IIinduced insulin resistance is associated with enhanced insulin signaling. Hypertension , 2002, 40: 872~879
    71 Nosadini R, Tonolo G. The role of the renin angiotensin hormonal system in the metabolic syndrome and type 2 diabetes. Nutr Metab CardiovascDis, 2004,14: 88~93
    72 Ottensmeyer FP, Beniac DR, Luo RZ, et al. Mechanism of transmembrane signaling: insulin binding and the insulin receptor. Biochemistry, 2000, 39: 12103~12112
    73 Taniyama Y, Hitomi H, Shah A, et al. Mechanisms of reactive oxygen species-dependent downregulation of insulin receptor substrate-1 by angiotensin II. Arterioscler Thromb Vasc Biol, 2005, 25: 1142~1147
    74 Andreozzi F, Laratta E, Sciacqua A, et al. Angiotensin II impairs the insulin signaling pathway promoting production of nitric oxide by inducing phosphorylation of insulin receptor substrate-1 on Ser312 and Ser616 in human umbilical vein endothelial cells. Circ Res, 2004, 94: 1211~1218
    75 Schieffer B, Schieffer E, Hilfiker-Kleiner D, et al. Expression of angiotensin II and interleukin 6 in human coronary atherosclerotic plaques: potential implications for inflammation and plaque instability. Circulation, 2000, 101: 1372~1378
    76 Schena M, Mulatero P, Schiavone D, et al. Vasoactive hormones induce nitric oxide synthase mRNA expression and nitric oxide production in human endothelial cells and monocytes. Am J Hypertens, 1999, 12: 388~397
    77 Rajagopalan S, Kurz S, Munzel T, et al. Angiotensin II-mediated hypertension in the rat increases vascular superoxide production via membrane NADH/NADPH oxidase activation. Contribution to alterations of vasomotor tone. J Clin Invest, 1996, 97: 1916~1923
    78 Pueyo ME, Gonzalez W, Nicoletti A, et al. Angiotensin II stimulates endothelial vascular cell adhesion molecule-1 via nuclear factor-kappaB activation induced by intracellular oxidative stress. Arterioscler Thromb Vasc Biol, 2000, 20: 645~651
    79 Kalra D, Sivasubramanian N, Mann DL. Angiotensin II induces tumor necrosis factor biosynthesis in the adult mammalian heart through aprotein kinase C-dependent pathway. Circulation, 2002, 105: 2198~2205
    80 Li DY, Zhang YC, Philips MI, et al. Upregulation of endothelial receptor for oxidized low-density lipoprotein (LOX-1) in cultured human coronary artery endothelial cells by angiotensin II type 1 receptor activation. Circ Res, 1999, 84: 1043~1049
    81 Weiss D, Kools JJ, Taylor WR. Angiotensin II-induced hypertension accelerates the development of atherosclerosis in apoE-deficient mice. Circulation, 2001, 103: 448~454
    82 Ruiz-Ortega M, Lorenzo O, Ruperez M, et al. Angiotensin II activates nuclear transcription factor kappaB through AT1 and AT2 in vascular smooth muscle cells: molecular mechanisms. Circ Res, 2000, 86: 1266~1272
    83 Kranzhofer R, Schmidt J, Pfeiffer CA, et al. Angiotensin induces inflammatory activation of human vascular smooth muscle cells. Arterioscler Thromb Vasc Biol, 1999, 19: 1623~1629
    84 Braun-Dullaeus RC, Mann MJ, Ziegler A, et al. A novel role for the cyclin-dependent kinase inhibitor p27(Kip1) in angiotensin II-stimulated vascular smooth muscle cell hypertrophy. J Clin Invest, 1999, 104: 815~823
    85 Kim S, Ohta K, Hamaguchi A, Yukimura T, Miura K, Iwao H. Angiotensin II induces cardiac phenotypic modulation and remodeling in vivo in rats. Hypertension, 1995, 25: 1252~1259
    86 Mifune M, Sasamura H, Shimizu-Hirota R, et al. Angiotensin II type 2 receptors stimulate collagen synthesis in cultured vascular smooth muscle cells. Hypertension, 2000, 36: 845~850
    87 Matsubara H, Moriguchi Y, Mori Y, et al. Transactivation of EGF receptor induced by angiotensin II regulates fibronectin and TGF-beta gene expression via transcriptional and post-transcriptional mechanisms. Mol Cell Biochem, 2000, 212: 187~201
    88 Touyz RM, He G, El Mabrouk M, et al. p38 Map kinase regulates vascularsmooth muscle cell collagen synthesis by angiotensin II in SHR but not in WKY. Hypertension, 2001, 37: 574~580
    89 Iozzo RV. Matrix proteoglycans: from molecular design to cellular function. Annu Rev Biochem, 1998, 67: 609~652
    90 Sasamura H, Shimizu-Hirota R, Nakaya H, et al. Effects of AT1 receptor antagonist on proteoglycan gene expression in hypertensive rats. Hypertens Res, 2001 24: 165~172
    91 Shimizu-Hirota R, Sasamura H, Mifune M, et al. Regulation of vascular proteoglycan synthesis by angiotensin II type 1 and type 2 receptors. J Am Soc Nephrol, 2001, 12: 2609~2615
    92 Yusuf S, Sleight P, Pogue J, Bosch J, et al. Effects of an angiotensin-converting-enzyme inhibitor, ramipril, on cardiovascular events in high-risk patients. The Heart Outcomes Prevention Evaluation Study Investigators. N Engl J Med, 2000, 342: 145~153
    93 Daugherty A, Rateri DL, Lu H, et al. Hypercholesterolemia stimulates angiotensin peptide synthesis and contributes to atherosclerosis through the AT1A receptor. Circulation, 2004. 110: 3849~3857
    94 Daugherty A, Manning MW, Cassis LA. Angiotensin II promotes atherosclerotic lesions and aneurysms in apolipoprotein E-deficient mice. J Clin Invest, 2000, 105: 1605~1612
    95 Wassmann S, Czech T, van Eickels M, et al. Inhibition of diet-induced atherosclerosis and endothelial dysfunction in apolipoprotein E/angiotensin II type 1A receptor double-knockout mice. Circulation, 2004, 110: 3062~3067
    96 Yang BC, Phillips MI, Mohuczy D, et al. Increased angiotensin II type 1 receptor expression in hypercholesterolemic atherosclerosis in rabbits. Arterioscler Thromb Vasc Biol, 1998, 18: 1433~1439
    97 Diep QN, Amiri F, Touyz RM, et al. PPARalpha activator effects on Ang II-induced vascular oxidative stress and inflammation. Hypertension, 2002, 40: 866~871
     98 Ryan MJ, Didion SP, Mathur S, Faraci FM, Sigmund CD. PPAR-(gamma) agonist rosiglitazone improves vascular function and lowers blood pressure in hypertensive transgenic mice. Hypertension, 2004, 43: 661~666
    1 Ambros V. The functions of animal microRNAs. Nature, 2004, 431: 350~355
    2 Kummerfeld SK and Teichmann SA. DBD: A transcription factor prediction database. Nucleic Acids Res, 2006, 34: D74–D81
    3 Reece-Hoyes JS, Deplancke B, Shingles J. et al. A compendium of C. elegans regulatory transcription factors: A resource for mapping transcription regulatory networks. Genome Biol, 2005, 6: R110
    4 Deplancke B, Mukhopadhyay A Ao W, Elewa AM, et al. A gene-centered C. elegans protein–DNA interaction network. Cell, 2006, 125: 1193–1205
    5 Harbison CT, Gordon DB, Lee TI, et al. Transcriptional regulatory code of a eukaryotic genome. Nature, 2004, 431: 99~104
    6 Levine M and Tjian R. Transcription regulation and animal diversity. Nature, 2003, 424: 147~151
    7 Maston, GA, Evans, SK, and Green, MR. Transcriptional regulatory elements in the human genome. Annu Rev Genomics Hum Genet, 2006, 7: 29~59
    8 Imanishi T, Itoh T, Suzuki, YO’Donovan C, et al. Integrative annotation of
    21,037 human genes validated by full-length cDNA clones. PLoS Biol, 2004, 2 (6): 856~875
    9 Carninci P, Kasukawa T, Katayama S, et al. The transcriptional landscape of the mammalian genome. Science, 2005, 309: 1559~1563
    10 Carninci P, Sandelin A, Lenhard B, et al. Genome-wide analysis of mammalian promoter architecture and evolution. Nat Genet, 2006, 38: 626~635
    11 Kim TH, Barrera LO, Qu C, et al. Direct isolation and identification of promoters in the human genome. Genome Res, 2005a, 15: 830~839
    12 Cooper SJ, Trinklein ND, Anton ED, et al. Comprehensive analysis of transcriptional promoter structure and function in 1% of the human genome. Genome Res, 2006, 16: 1~10
    13 Hallikas O, Palin K, Sinjushina N, et al. Genome-wide prediction of mammalian enhancers based on analysis of transcription-factor binding affinity. Cell, 2006, 124: 47~59
    14 Gupta M and Liu JS. De novo cis-regulatory module elicitation for eukaryotic genomes. Proc. Natl. Acad. Sci, 2005, 102: 7079~7084
    15 Elnitski L, Jin VX, Farnham PJ, et al. Locating mammalian transcription factor binding sites: A survey of computational and experimental techniques. Genome Res, 2006, 16 (12): 1455~1464
    16 Sabo PJ, Kuehn MS, Thurman R, et al. Genome-scale mapping of DNaseI sensitivity in vivo using tiling DNA microarrays. Nat. Methods, 2006, 3: 511~518
    17 Tompa M, Li N, Bailey TL, et al. Assessing computational tools for the discovery of transcription factor binding sites. Nat. Biotechnol, 2005, 23: 137~144
    18 Kellis M, Patterson N, Endrizzi M, et al. Sequencing and comparison of yeast species to identify genes and regulatory elements. Nature, 2003, 423: 241~254
    19 Xie X, Lu J, Kulkobas EJ, et al. Systematic discovery of regulatory motifs in human promoters and 3 UTRs by comparison of several mammals. Nature, 2005, 434: 338~345
    20 Rual JF, Venkatesan K, Hao, T, et al. Towards a proteome-scale map of the human protein–protein interaction network. Nature, 2005, 437: 1173~1178
    21 Newman JRS. and Keating AE. Comprehensive identification of human bZip interactions with coiled-coil arrays. Science, 2003, 300: 2097~2101
    22 Gavin AC, Aloy P, Grandi P, et al. Proteome survey reveals modularity of the yeast cell machinery. Nature, 2006, 440: 631~636
    23 Krogan NJ, Cagney G, Yu H, et al. Global landscape of protein complexes in the yeast Saccharomyces cerevisiae. Nature, 2006, 440: 637~643
    24 Li S, Armstrong CM, Bertin N, Ge H, et al. A map of the interactome network of the metazoan C. elegans. Science, 2004, 303: 540~543
    25 Blais A and Dynlacht BD. Constructing transcriptional regulatory networks.Genes & Dev, 2005, 19: 1499~1511
    26 Workman CT, Mak HC, McCuine S, et al. A systems approach to mapping DNA damage response pathways. Science, 2006, 312: 1054~1059
    27 Bieda M, Xu X, Singer MA, et al. Unbiased location analysis of E2F1-binding sites suggests a widespread role for E2F1 in the human genome. Genome Res, 2006, 16: 595~605
    28 Odom DT, Zizlsperger, N, Gordon DB, et al. Control of pancreas and liver gene expression by HNF transcription factors. Science, 2004, 303: 1378~1381
    29 Zhang X, Odom DT, Koo SH, et al. Genome-wide analysis of cAMP-response element binding protein occupancy, phosphorylation, and target gene activation in human tissues. Proc. Natl Acad Sci, 2005, 102: 4459~4464
    30 Boyer LA, Lee TI, Cole MF, et al. Core transcriptional regulatory circuitry in human embryonic stem cells. Cell, 2005, 122: 947~956
    31 Oh SW, Mukhopadhyay A, Dixit BL, et al. Identification of direct targets of DAF-16 controlling longevity, metabolism and diapause by chromatin immunoprecipitation. Nat Genet, 2005, 38: 251~257
    32 van Steensel, B and Henikoff S. Identification of in vivo DNA targets of chromatin proteins using tethered Dam methyltransferase. Nat Biotechnol, 2000, 18: 424~428
    33 Orian A, van Steensel B, Delrow J, et al. Genomic binding by the Drosophila Myc, Max, Mad/Mnt transcription factor network. Genes & Dev, 2003, 17: 1101~1114
    34 Mukherjee S, Berger MF, Jona G, et al. Rapid analysis of the DNA-binding specificities of transcription factors with DNA microarrays. Nat Genet, 2004, 36: 1331~1339
    35 Liu X, Noll DM, Lieb JD, et al. DIP-chip: Rapid and accurate determination of DNA-binding specificity. Genome Res, 2005, 15: 421~427
    36 Meng X, Brodsky MH, and Wolfe SA. A bacterial one-hybrid system fordetermining the DNA-binding specificity of transcription factors. Nat Biotechnol, 2005, 23: 988~994
    37 Shannon P, Markiel A, Ozier O, et al. Cytoscape: A software environment for integrated models of biomolecular interaction networks. Genome Res, 2003, 13: 2498~2504
    38 Lall S, Grun D, Krek A, et al. A genome-wide map of conserved microRNA targets in C. elegans. Curr Biol, 2006, 16: 460~471

© 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