用户名: 密码: 验证码:
芪珠方对肝纤维化大鼠TGF-β1/Smad信号传导通路的影响
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
背景
     肝纤维化是多种慢性肝病发展至肝硬化的必经阶段。研究证实肝纤维化是可逆的,但如果不加以治疗,肝纤维化可能会转变为肝硬化。晚期肝硬化病人3年的存活率仅仅为16%,全世界直接死于肝硬化的人数位居人类总死亡数的第6位。肝纤维化的形成原因是由于肝脏受到损害与炎症反应后,肝组织内细胞外基质(ECM)过度增生与异常沉积,导致肝脏内胶原组织的增多,使肝脏结构和肝功能产生异常病变。转化生长因子-β1是肝纤维化的重要靶点,它可以激活肝星状细胞、调节细胞外基质合成和降解导致肝纤维化发生,TGF-β1促纤维化作用主要是通过TGF-β1/Smad信号转导通路来实现的。
     西医对肝纤维化的治疗多为阻断肝纤维化诱因的针对治疗,临床上尚无理想的、公认的抗纤维化特效药物。中医药在治疗肝纤维化方面具有多成分、多环节、多靶点的优势,并已经取得了较好的疗效,拥有广阔的开发和应用前景。芪珠方有清热解毒、益气活血等功效,前期研究表明芪珠方在慢性乙型肝炎的治疗和临床研究中有一定的保肝降酶、抗纤维化作用;对急慢性肝损伤和肝纤维化模型大鼠有一定的保护作用。
     本研究拟运用分子生物学和细胞学技术,观察芪珠方对CC14肝纤维化模型大鼠肝组织TGF-β1/Smad信号通路相关因子及TIMPs/MMPs, ILs等细胞因子的影响,部分阐明抗肝纤维化机制;利用HPLC法检测芪珠方中的有效成分;检测芪珠方有效成分对外源性TGF-β1刺激后的HSC-T6细胞增殖抑制、细胞凋亡、细胞周期以及TGF-β1/Smad信号传导通路的影响,进一步探明芪珠方抗肝纤维化的作用机制。
     方法
     1.体内实验
     SD大鼠70只,随机分为空白组、模型组、芪珠方水提大、中剂量组、芪珠方醇提大、中剂量组,秋水仙碱组,组间大鼠体重差异无显著性。除正常组外,其余各组大鼠皮下注射40%四氯化碳(cC14)橄榄油造模,首次5mL每100g体重,以后每次3mL每100g体重,每周3次,共8周。于造模开始次日给药,芪珠方高、低剂量组16.2,9.72g每100g体重,秋水仙碱组0.5mg每100g体重灌胃,每日1次,连续8周。空白组不做任何处理,模型组大鼠给予同大剂量组灌胃剂量生理盐水灌胃。造模结束后所有大鼠处死,肝脏组织学观察,测定肝脾指数,酶联免疫吸附试验(ELISA)技术检测TNF-a、IL-4、IL-8、IL-10、TIMP-1、MMP-1、MMP-2、MMP-13在各组大鼠肝组织内的表达,TIMP-1、MMP-1、 MMP-2、MMP-13在各组大鼠血清内表达。蛋白免疫印迹法(Western blott)检测Smad2、 Smad3、Smad4、Smad7、TGF-β1蛋白表达:实时荧光定量Real-time PCR)检测Smad2、 Smad3、Smad4、Smad7、TGF-β1基因表达。
     2.芪珠方HPLC实验
     采用高效液相色谱法,色谱条件为Agilent C18柱(250mm×4.6mm,5μm);流动相:A为乙腈,B为水溶液,梯度洗脱,流速0.8mL/min,柱温30℃,检测波长230nm,检测芪珠方与含药血清中有效成分,采用中药色谱指纹图谱相似度评价系统(A版)进行相似度评价。
     3.体外实验
     HSC-T6细胞,DMEM培养基+10%FBS培养(37℃,5%C02),青链霉素作为双抗,0.25%胰蛋白酶消化,细胞生长至75%传代。分组:空白组、叶下珠次素组、虎杖苷组、芍药苷组、黄芪甲苷组,取对数生长期的HSC-T6细胞,每组设6个复孔,加入浓度为5ng/mL外源性TGF-β1作为模型组,空白组与模型组分别加入叶下珠次素、虎杖苷、芍药苷、黄芪甲苷,观察12h、24h、48h。检测各组HSC-T6细胞增殖情况,流式细胞仪检测各组24h细胞周期变化:蛋白免疫印迹法(western blot)检测Bcl-2、Bax、CyclinD1、Smad2、Smad3、 Smad4、Smad7、TGF-pβ,24h蛋白表达;实时荧光定量(real-time PCR)检测Bcl-2、Bax、 CyclinD1、Smad2、Smad3、Smad4、Smad7、TGF-β1,24h基因表达。
     所有结果以均数±标准误差形式,利用SPSS18.0软件进行统计学分析,组间比较使用t检验或方差分析(ANOVA),当p<0.05时,认为处理组和对照组间有统计学差异。
     结果
     第一部分体内实验
     1.与正常组相比,模型组大鼠体质量下降(P<0.01),肝、脾指数上升(P<0.01);模型组与治疗组相比,各治疗组大鼠体重有增加,大剂量组有显著改变(P<0.01):脾脏指数降低(P<0.05);大鼠肝脏大体标本肉眼观察,各治疗组外观接近正常对照组;模型组HE切片可见肝细胞肿胀,广泛气球样变与大量纤维增生,芪珠方组HE切片大鼠肝组织炎性病变程度减轻,肝细胞变性或坏死减少,纤维组织增生减少或逆转。提示肝纤维化模型复制建立成功,芪珠方对肝纤维化有一定的预防作用。
     2.相比空白组,模型组大鼠肝组织内IL4、IL8、TNF-a、TIMP-1、MMP-2表达上调,IL10、MMP-1、MMP-13表达下调(P<0.01);芪珠方能降低肝纤维化大鼠肝组织BL4, IL-8, TNF-a的表达,提高IL-10的表达(P<0.01);同时能降低肝组织TIMP-1、MMP-2的表达,提高MMP-1、MMP-13的表达(P<0.01),血清内的TIMPs/MMPs的表达与肝组织表达一致(P<0.01)。
     3.相比空白组,模型组大鼠肝组织内TGF-β1、Smad2、Smad3、Smad4蛋白和mRNA表达上调,Smad7蛋白和mRNA表达下调(P<0.05);芪珠方干预后,芪珠方治疗组大鼠肝组织内TGF-β1、Smad2、Smad3、Smad4蛋白和mRNA表达不同程度下调(P<0.05),Smad7蛋白和mRNA表达不同程度上调(P<0.05)。
     第二部分芪珠方的HPLC检测
     HPLC结果显示芍药苷、虎杖苷、叶下珠次素是芪珠方中的主要成分;芪珠方醇提法比水提法能更有效的提取方中药材的有效成分。
     第三部分体外实验
     1.叶下珠次素、虎杖苷各剂量组以及芍药苷、黄芪甲苷中剂量组在12h、24h、48h3个时间段对HSC-T6细胞分别有一定的增殖抑制作用,在24h时间段抑制作用最为明显和稳定。
     2.叶下珠次素、虎杖苷、黄芪甲苷组能上调空白组HSC-T6细胞与模型组HSC-T6细胞BaxmRNA和蛋白表达(P<0.05),下调Bcl-2mRNA和蛋白表达(P<0.05),芍药苷能上调空白组HSC-T6细胞与模型组HSC-T6细胞BaxmRNA和蛋白表达(P<0.05),下调Bcl-2蛋白表达(P<0.05)。
     3.叶下珠次素有通过下调模型组HSC-T6细胞周期相关因子CyclinD1蛋白和mRNA表达,该组HSC-T6细胞与模型组相比停留在G1期的数量更多(P<0.05)。
     4.相比空白组,模型组HSC-T6细胞内源性TGF-β1、Smad2,Smad3的蛋白和mRNA表达高于空白组(P<0.05), Smad7的表达低于空白组(P<0.05);叶下珠次素可以下调模型组HSC-T6细胞Smad2蛋白表达、Smad3mRNA表达,上调Smad7蛋白与mRNA表达,下调内源性TGF-β1mRNA与蛋白的表达(P<0.05);虎杖苷可以下调Smad2mRNA与蛋白的表达,下调TGF-β1mRNA与蛋白的表达(P<0.05);芍药苷可以下调Smad2,Smad3蛋白表达,下调Smad3mRNA表达,上调Smad7蛋白与mRNA表达,下调TGF-β1mRNA与蛋白的表达(P<0.05);黄芪甲苷可以上调Smad7蛋白与mRNA表达,下调TGF-β1蛋白的表达(P<0.05)。
     结论
     1.芪珠方能提高肝纤维化模型大鼠体重,减轻肝脏肿大及肝脾湿重,改善肝组织内的纤维化程度,提示芪珠方有保护肝纤维化大鼠和抗肝纤维化作用。
     2.芪珠方能降低肝纤维化大鼠肝组织IL4、IL-8、TNF-a、TIMP-1、MMP-2的表达,提高IL-10、MMP-1、MMP-13的表达,提示芪珠方抗肝纤维化的作用与调节TIMPs/MMPs,ILs和TNF-a,从而减少细胞外基质的分泌,平衡肝内胶原的分泌与降解有关。
     3.芪珠方能通过下调肝纤维化大鼠肝组织内Smad2、Smad3、Smad4mRNA和蛋白的表达,上调Smad7mRNA和蛋白的表达,芪珠方能调节TGF-β1/Smad信号传导通路,从而抑制TGF-β1mRNA和蛋白的表达、抑制肝星状细胞的激活,减少细胞外基质的分泌,降低肝纤维化程度。
     4.叶下珠次素、虎杖苷、芍药苷是芪珠方的有效成分,醇提法在提取上述单体的效果好于水提法,提示芪珠方醇提物调节TIMPs/MMPs、TGF-β1/Smad信号传导通路的作用优于水提物与可以更有效的提取中药中的有效成分有关。
     5.叶下珠次素、虎杖苷各剂量组以及芍药苷、黄芪甲苷中剂量组在12h、24h、48h三个时间段对HSC-T6细胞和外源性TGF-β1刺激模型组HSC-T6细胞有一定的增殖抑制作用,提示芪珠方的抗肝纤维化作用与其有效成分可以抑制肝星状细胞增殖有关。
     6.叶下珠次素、芍药苷、黄芪甲苷、虎杖苷均能调节Bcl-2和Bax的表达从而促进HSC-T6细胞,其抑制HSC-T6细胞增殖作用可能是通过上调Bax和下调Bc1-2表达,促进HSC-T6细胞凋亡而形成的,芪珠方抗肝纤维化的作用可能与其有效成分能促进肝星状细胞凋亡有关。
     7.叶下珠次素有通过下调CyclinD1mRNA与蛋白表达,从而调控细胞周期,使HSC-T6细胞停留在G1期,叶下珠次素抑制HSC-T6细胞增殖作用可能与能调控HSC-T6细胞周期有关。
     8.芪珠方抗肝纤维化的作用与芪珠方中的有效成分叶下珠次素、虎杖苷、芍药苷、黄芪甲苷可以分别调控、共同作用于TGF-β1/Smad信号传导通路,抑制TGF-β1表达,从而抑制肝星状细胞的激活,减少肝组织内ECM沉积有关。
Background
     Hepatic fibrosis is a necessary stage in many chronic liver diseases developing to liver cirrhosis and this process is reversible proved by research. But if Hepatic fibrosis is left untreated, it may be transformed into liver cirrhosis. The survival rate of end-stage liver cirrhosis is only16%in3years, directly death in liver cirrhosis is on the sixth place in human total death of the whole world.
     The mainly formation mechanism of liver fibrosis is that:after the liver damage by inflammation, there are excessive proliferation and abnormal deposition of Extracellular matrix (ECM), leading an abnormal changes of liver structure and function. Transforming growth factor-β1(TGF-β1) is the main reason of Hepatic fibrosis by activate hepatic stellate cells, regulate synthesis and degradation of extracellular matrix, and it is the important target for liver fibrosis. The effect of TGF-β1is mainly realized by TGF-β1/Smad signal transduction pathway. The treatment for Hepatic fibrosis on Modern Medicine is blocking the inducement and there is no specific medicine for Hepatic fibrosis on clinic. In contrast, Traditional Chinese Medicine has various advantages and achieved effect on the treatment of Hepatic fibrosis such as multiple components, multiple links, and multiple targets, has broad prospects for development and application. Compound Qizhu Tablet has many functions such as clearing heat and removing toxicity, benefiting qi for activating blood circulation and it has proved its effect in the treatment of chronic hepatitis B as protection of liver, dropping enzyme and anti-liver fibrosis. Our previous studies have shown that Compound Qizhu Tablet has a protective effect on acute and chronic liver injury and hepatic fibrosis rat model.
     The experiment aims to observe the systematic effect of Compound Qizhu Tablet on Hepatic fibrosis, and make further research on the mechanism. The conclusion of this experiment, providing exploratory research to develop the treatment of Hepatic fibrosis drugs from Traditional Chinese Medicine.
     Objective:
     This study aims to research the effect of TGF-β1/Smad signaling pathway and cell factor as TIMPs/MMPs and ILs influenced by Compound Qizhu Tablet on CC14rats hepatic fibrosis model and verify the mechanism partly on molecular biology and cell biology; To find out the effective components of Compound Qizhu Tablet by HPLC; To observe the influence of effective components of Compound Qizhu Tablet on HSC-T6cell effected by exogenous TGF-β1, by testing cell apoptosis, cell cycle and TGF-β1/Smad signal transduction pathway effects, to provide further experimental evidence for the treatment of liver fibrosis in Traditional Chinese Medicine.
     Methods:
     Experiment1:Treatment of liver fibrosis of Compound Qizhu Tablet in vivo experiments
     Seventy SD rats were randomly divided into normal group, model group, Qizhu high-dose group (water preparation), Qizhu low dose group (water preparation), Qizhu high-dose group (ethanol preparation), Qizhu low dose group (ethanol preparation), and colchicine group. All the rats except normal group received40%CCl4subcutaneous injection twice a week for8weeks, rats was firstly given40%CCl4(5mL per1OOg body weight) and3mL per100g body weight secondly. Rats in both Qizhu high-dose group and small dose group were given16.2,9.72g per100g body weight by ig. Qizhu decoction respectively while rats in colchicine group were fed with0.5mg per100g body weight colchicine. Liver histology and liver and spleen index were observed after the model is done. The expression of TNF-a, IL-4,IL-8,IL-10,TIMP-1,MMP-1,MMP-2,MMP-13in hepatic tissue was detected by enzyme linked immune sorbent assay (ELISA),and as same as the expression of TIMP-1, MMP-1, MMP-2,MMP-13in the blood serum. To detect the protein expression levels of Smad2, Smad3, Smad4, Smad7, and TGF-β1by Western-blot, the mRNA expression levels of Smad2, Smad3, Smad4, Smad7, and TGF-β1by real time quantitative RT-PCR.
     2. Experiment2:HPLC for Compound Qizhu Tablet
     HPLC Method was applied to establish the Compound Qizhu Tablet. The separation was performed on a Agilent C18column (250mm×4.6mm,5μm) with a gradient elution composed of acetonitrile(A) and water (B). The column temperature was set at30℃and the flow rate was0.8Ml/min.The detective wave length was at280nm. HPLC fingerprint was performed on the Similarity evaluation system for chromatographic fingerprint of traditional Chinese medicine (A)
     Experiment3:Treatment of liver fibrosis of the effective components of Compound Qizhu Tablet in vitro experiments
     HSC-T6cells were cultured in DMEM medium supplemented with10%fetal bovine serum,50U penicillin/mL and50μg streptomycin/mL. For sub culturing purposes, cells were detached by treatment with0.25%trypsin/0.02%EDTA at37℃. Cultures were used at75%confluence. Cultures were divided into control group, Polydatin group, Hypophyllanthin group,Astragaloside group, Peoniflorin group, and model group is add exogenous TGF-β1. ALL groups are measured for proliferation and cell cycle change at24h by flow cytometry; The mRNA and protein expression levels of Bcl-2、Bax、CyclinD1、Smad2、Smad3、Smad4、Smad7、TGF-β1were detected by real time quantitative RT-PCR and Western-blot respectively at24h.
     Data are expressed as Mean±SD. The Student's t-test was used to determine the statistical significance of the experimental data.
     Results
     Experiment1:Treatment of liver fibrosis of Compound Qizhu Tablet in vivo experiments
     1.Compared with normal group, body weight of rats in model group decreased (P<0.01), liver index, spleen index increased (P<0.01); Compared with the model group, body weight of rats in treatment group increased, the high dose group were improved significantly (P<0.01); spleen index decreased (P<0.05); On the rat gross anatomy, the treatment group appearance similar to the normal group; According to the pathological condition, the rats in model group were observed that the liver cells swelling, widely balloon degeneration and fibrosis, the rats in treatment group showed Compound Qizhu Tablet can improve liver inflammatory lesions, reduce liver cell degeneration or necrosis, stop or reverse the hyperplasia of fibrous tissue. It showed that liver fibrosis model replication is established successfully, and Compound Qizhu Tablet has a preventive effect on liver fibrosis.
     2. Compared with normal group, the expression of IL4,IL8,TNF-a,TIMP-1,MMP-2in liver fibrosis rats increased, the expression of IL-10,MMP-1,MMP-13decreased (P<0.01);Compound Qizhu Tablet can reduce the expression of IL4, IL-8, TNF-a in liver fibrosis rats, increase the expression of IL-10(P<0.01); At the same time, Compound Qizhu Tablet can reduce the expression of reduce the expression of TIMP-1,MMP-2in liver fibrosis rats, increase the expression of MMP-1,MMP-13(P<0.01), it showed the same TIMPs/MMPs expression in the serum of liver fibrosis rats (P<0.01).
     3.Compared with normal group, protein and gene expression of TGF-β1, Smad2, Smad3, Smad4in model group increased, protein and gene expression of Smad7decreased (P<0.05); Compared with model group, protein and gene expression of TGF-β1, Smad2, Smad3,Smad4in treatment group decreased,and expression of Smad7increased (P<0.05) Experiment2:HPLC for Compound Qizhu Tablet
     Result of HPLC in Compound Qizhu Tablet showed that Polydatin, Hypophyllanthin, and Peoniflorin is the effective components of Compound Qizhu Table; And ethanol preparation is more effective than water preparation.
     Experiment3:Treatment of liver fibrosis of the effective components of Compound Qizhu Tablet in vitro experiments
     1. Hypophyllanthin,Polydatin in each dosage group and dose group of Paeoniflorin, Astragaloside showed effect of proliferation inhibition on HSC-T6cells in12h,24h,48h,and the most obvious performance in on24h.
     2. Hypophyllanthin, Polydatin, Astragaloside can increase protein and gene expression of Bax in normal group and model group HSC-T6cells, decrease the expression of Bcl-2; Paeoniflorin can increase protein and gene expression of Bax in normal group and model group HSC-T6cells, decrease the protein expression of Bcl-2.
     3. Hypophyllanthin can hold the HSC-T6cells of model group in G1phase by decrease the protein and gene expression of cell cycle related factors CyclinD1.
     4. Compared with normal group, HSC-T6in model group protein and gene expression of TGF-β1, Smad2,Smad3increased, protein and gene expression of Smad7decreased; Hypophyllanthin can decrease protein expression of Smad2, gene expression of Smad3, increase protein and gene expression of Smad7and decrease protein and gene expression of endogenous TGF-β1; Polydatin can decrease protein and gene expression of Smad2, decrease protein and gene expression of endogenous TGF-β1; Paeoniflorin can decrease protein and gene expression of Smad3, protein expression of Smad2, increase protein and gene expression of Smad7, and decrease protein and gene expression of endogenous TGF-β1; Astragaloside can increase protein and gene expression of Smad7and decrease protein expression of endogenous TGF-β1.
     Conclusions
     1. Compound Qizhu Tablet can improve the weight of the rats of liver fibrosis model, reduce the enlargement of the liver and spleen wet weigh, and restore the damage of fibrosis in liver tissue.It shows Compound Qizhu Tablet can protect liver fibrosis rats and treat liver fibrosis.
     2.Compound Qizhu Tablet can reduce the expression of IL4, IL-8, TIMP-1, MMP-2,TNF-a in liver fibrosis rats, increase the expression of IL-10, MMP-1, MMP-13.It shows Compound Qizhu Tablet treat hepatic fibrosis though regulate TTMPs/MMPs, ILs and TNF-a in the liver, reduce the expression of extracellular matrix in liver and balance liver collogen.
     3.Compound Qizhu Tablet can decrease protein and gene expression of Smad2, Smad3, Smad4,and increase expression of Smad7, regulat the TGF-β1/Smad Signal Transduction Pathway to decrease the expression of TGF-β1and Inhibiting activation of hepatic stellate cells, reduce the secretion of extracellular matrix, reduce the degree of liver fibrosis.
     4. According to HPLC on Compound Qizhu Tablet, Hypophyllanthin, Phyllanthus, polydatin are the effective component of Compound Qizhu Tablet, ethanol preparation is better than water preparation in the extraction of the monomer, it showed ethanol preparation regulate TIMPs/MMPs, TGF-β1/Smad signal transduction pathway more effective than water preparation With the relevant of in the extraction of the effective component.
     5.Hypophyllanthin,Polydatin in each dosage group and dose group of Paeoniflorin, Astragaloside showed effect of proliferation inhibition on HSC-T6cells in12h,24h,48h,and the most obvious performance in on24h.It showed that treatment of Compound Qizhu Tablet on Hepatic fibrosis is relative to its effective component's effect of proliferation inhibition on HSC-T6cell.
     6. Hypophyllanthin, Polydatin, Astragaloside and Paeoniflorin increase protein and gene expression of Bax in normal group and model group HSC-T6cells; decrease the expression of Bcl-2. Compound Qizhu Tablet's effect on Hepatic fibrosis is relative to proliferation inhibition on HSC-T6cells, as well as the effective component's effect on Promoting HSC-T6apoptosis.
     7. Hypophyllanthin can hold the HSC-T6cells of model group in G1phase by decrease the protein and gene expression of cell cycle related factors CyclinD1and the proliferation inhibition on HSC-T6cells is relative to the control of cell cycle.
     8. Effect on hepatic fibrosis of Compound Qizhu Tablet is relative to its effective component such as Hypophyllanthin, Polydatin, Astragaloside and Paeoniflorin, which can respectively control the TGF-β1/Smad signal transduction pathway, inhibition TGF-β1expression and activation of hepatic stellate cells, reduced ECM deposition in liver tissue.
引文
[1]Albillos A, Nieto M, Ubeda M, et al. The biological response modifier AM3 attenuates the inflammatory cell response and hepatic fibrosis in rats with biliary cirrhosis[C].Gut,2010,59 (7):943.
    [2]孙妩弋,吴成义,魏伟.肝纤维化的中医药治疗现状及现代研究思路[C].2008年安徽中医药继承与创新博士科技论坛论文集,合肥:中国中医药学会,2008,83.
    [3]毛青.抗丙型肝炎病毒新型药物研究进展[C].中华医学会病毒性肝炎慢性化、重症化基础与临床研究进展学术会议论文集,重庆:中华医学会,2009,27.
    [4]程明亮,刘三都.肝纤维化的基础研究与临床[M].北京:人民卫生出版社,1996,106-110.
    [5]Ismail MH, Pinzani M. Reversal of liver fibrosis [J]. Saudi J Gastroenterol,2009,15:72-79.
    [6]Seifert RA, Hart CE, Phillips PE, et al. Two different subunits assoeiated to create isoform specific platelet derived growth factor receptors [J]. J Biol Chem,1989,264:8771.
    [7]Abou Shady M, Friess H, Zimmermann A, et al. Connective tissue growth factor in human liver cirrhosis [J].Liver,2000,2(4):296-304.
    [8]Liu X,Hu H,Yin JQ.Therapeutics strategies against TGF-beta signaling pathway in hepatic fibrosis[J]. Liver Int 2006; 26:8-22.
    [9]Wynn TA. Cellular and molecular mechanisms of fibrosis [J].J Pathol,2008,214:199-210.
    [10]Shek FW, Benyon RC. How can transforming growth factor beta targeted usefully to combat liver fibrosis [J].Eur J Gastroenterology Hepatol, 2004,16:12-36.
    [11]Hemmann S, Graf I, Roderfeld et al. Expression of MMPs and TIMPs in liver fibrosis-a systematic review with special emphasis on anti fibrotic strategies [J].J Hepatol 2007,46:955-975.
    [12]Brew K, Nagase H. The tissue inhibitors of metalloproteinase (TIMPs):an aneient family with structural and functional diversity[J].Bochum Biopsy's Acta 2010,1803:55-71.
    [13]Pan CH, Kn CH, Linc S. Interplay of angiotensin Ⅱ and angiotensin (1-7) in the regulation of metalloproteinase of human cardiocytes [J].Exp Physiol 2008,93:599-612.
    [14]Derynck R, Zhang YE.Smad dependent and Smad-independent pathways in TGF-Beta family signaling [J].Nature,2003,425(6958):577-584.
    [15]Shi Y, Massague J.Mechanisms of TGF-beta signaling from cell membrane to the nucleus [J].Cell,2003, 113:685-700.
    [16]Friedman SL. Mechanisms of hepatic fibro genesis [J]. Gastroenterology,2008,134 (6):1655-1669.
    [17]席文娜,孙水林,李方春.a-2干扰素对大鼠肝组织bcl-2基因表达的影响及意义[J].世界华人消化杂志,2009;17(31):3232-3240.
    [18]Fulda S, Meyer E, Friesen C, et al. Cell type specific involvement of death receptor and mitochondrial pathways in drug-induced apoptosis[J].Oncogene,2001,20 (9):1063-1075.
    [19]贺骏,阮秋蓉,杨秀萍,等.Pinl和CyclinD1在人类5种常见肿瘤中的表达及其意义[J].中国组织化学与细胞化学杂志,2006;15(6):608-612.
    [20]李红雨,徐茜,朱涛,等.宫颈癌细胞株和宫颈上皮组织中Pinl和CyclinDl的表达及临床意义[J].癌症,2006;25(3):367-372.
    [21]王吉耀.重视肝纤维化的基础与临床研究[J].中华医学杂志,2005,85(15):1009-1010.
    [22]Schuppan D, Krebs A, Bauer M, Hahn EG.Hepatitis Cand liver fibrosis[J].Cell Death,2003:10 Suppll: S59-67
    [23]ISON M, SODA M, I NOUE A, et al. Reverse transformation on of hepatic my fibroblast-like cells by TGF-β1/LAP [J]. Bochum Biopsy's Res Commun,2003,311 (4):959-965.
    [24]薛博瑜,顾学兰.肝纤维化的病机认识和辨证论治[J].南京中医药大学学报,2001,17(2):76.
    [25]薛博瑜,殷杰,潘林梅,等.复方芪珠片治疗慢性乙型肝炎100例临床研究[J].南京中医药大学学报,2007,23(1):24.
    [26]谢东浩,薛博瑜,张林松,等.复方芪珠片对大鼠CC14慢性肝损伤及肝纤维化模型的保护作用研究[J].南京中医药大学学报,2011,27(3):250.
    [27]王学东,胡荣听,叶放,等.复方芪珠片对急性肝损伤和肝纤维化的实验研究[J].辽宁中医药大学学 报,2008,10(4):150.
    [28]薛燕.中药复方霰弹理论[M].中国环境科学出版社,北京,1996年.
    [29]谢东浩,马利华,贾晓斌,等.高效液相色谱法测定复方芪珠片中柯里拉京和虎杖苷的含量[J].中国医院药学杂志,2011,31(18):1550-1552.
    [1]Fallowfield JA, Iredale JP.Reversal of liver fibrosis and cirrhosis-an emerging reality [J]. Scott Med J, 2004,49(1):3-61.
    [2]薛博瑜,殷杰,潘林梅,等.复方芪珠片治疗慢性乙型肝炎100例临床研究[J].南京中医药大学学报,2007,23(1):24.
    [3]谢东浩,薛博瑜,张林松,等.复方芪珠片对大鼠CC14慢性肝损伤及肝纤维化模型的保护作用研究[J].南京中医药大学学报,2011,27(3):250.
    [4]王学东,胡荣昕,叶放,等.复方芪珠片对急性肝损伤和肝纤维化的实验研究[J].辽宁中医药大学学报,2008,10(4):150.
    [5]Ohishi T, Saito H, Tsusaka K, et al. Anti-fibro genic effect of an angiotensin converting enzyme inhibitor on chronic carbon tetrachloride-induced hepatic fibrosis in rats[J]. Hepatol Res,2001,21 (2):147-158.
    [6]Kitamura K, Nakamot o Y, Akiyama M, et al. Pathogenic roles of tumor necrosis f actor receptor p55/3 mediated signals in dimethyl 1 nitrosamine-induced murine Liver fibrosis [J]. Laboratory Investigation,2002, 82(5):571-583.
    [7]王宝恩,王志高,殷蔚荑,等.实验性免疫性肝纤维化模型的研究[J]-中华医学杂志,1989,69(8):503-505.
    [8]Keegan A, Martini R, Batey R. Ethanol related liver injury in the rat:a model of statuses, inflammation and percent rat fibrosis [J]. J Hepatol,1995,23 (5):591-600.
    [9]Nan JX, Park EJ, Lee SH, et al. Anti fibrotic effect of Stephanie tetramer on experimental liver fibrosis induced by bile duct Iigation and scission in rats[J]. Arch Pharm Res,2000,23 (5):501-506.
    [10]Cote PJ, Korba BE, Miller RH, et al. Effects of age and viral determinants on chronicity as an outcome of experimental woodchuck hepatitis virus infection [J].Herpetology,2000,31 (1):190-200.
    [11]Jilbert AR, Kotlarski I. Immune responses to duck hepatitis B virus infection [J]. Developmental Comparative Immunology,2000,24 (2-3):285-302.
    [12]Cater EA, MeCarron MJ, Alpent E, et al. Lysol oxidase and collagenase in experiment acute and chronic liver injury [J]. Gastroenterology.1982,82:526-524.
    [13]展玉涛,魏红山,王志荣,等.大黄素抗肝纤维化作用的实验研究[J].中华肝脏病杂志2001,9:235-236.
    [14]韩德五,马学惠,赵元昌.肝硬化动物模型的研究[J].山西医药杂志,1979,8(4):1-3.
    [15]Murie IP, Fernandez-Martinez E, Perez-Alvarez V, et al. Thalidomide ameliorates carbon tetrachloride induced cirrhosis in the rat [J]. Euro, J Gastroenterology Hepatol,2003,15 (9):951-957.
    [16]吕霞,舒建昌,何雅军,秋水仙碱抑制肝星状细胞活化与抗肝纤维化的作用[J].广东医学,2006,27(12):1796-1798.
    [17]Luekey Sw, Peterson DR, Activation of kuppfer cells during the course of carbon tetrachloride induced liver injury and fibrosis in mice [J].exploit patrol,2001,71(3):226-228.
    [1]Henderson NC, Forbes SJ. Hepatic fibro genesis:from within and out with[J].Toxicology,2008,254 (3):130-135.
    [2]孙妩弋,桂双英,吴丽等.芍芪多营对肝纤维化大鼠肝脏星状细胞基质金属蛋白酶13及组织金属蛋白酶抑制因子1表达的影响[J].中国中药杂志,2010,35(11):1447-1451.
    [3]都广礼,陈德兴,张艳等.盯纤维化大鼠肝组织基质金属蛋白酶9及其抑制因子1基因表达的动态变化及下瘀血汤对其影响[J].中国中西医结合消化杂志,2010,18(1):1-4.
    [4]Haas TL, Madri JA. Extracellular matrix-driven matrix metalloproteinase production in endothelial cells: implications for angiogenesis[J].Trends Cardio vase Med,1999,9(3-4):70-77.
    [5]Zhang Xiao-bo, Fan Xian-ming, Wang Wen-jun, et al. The Effect of Inhaled Budesonide on the Expression of MMP-2 and TIMP-2 in Lung Tissue of Pulmonary Fibrosis Rats[J].Journal of Military Surgeon in Southwest,2008,10(4):12-15.
    [6]Zhu GF, Yu CH, Zhang Y, Li YM. Gene expression of interstitial collagens MMP-13 progressive phase of rat liver fibrosis induced by ethanol [J]. Chinese Journal of Hepatology,2003; 11:660-662.
    [7]谢玉梅,聂青和,周永兴,等.肝硬化患者肝组织中TIMP1,TIMP2表达[J].第四军医大学学报,2000,21:790-792.
    [8]Gomez DE, Alonso DF, Yoshiji H, et al. Trusses inhibitors of metalloproteinase structure, regulation and biological functions [J].Eur J Cell Biol,1997,74:111-122.
    [9]陈治新,王小从,张莉娟,等.IL-10对实验性肝纤维化大鼠血小板衍生生长因子-AA、BB表达的影响[J].临床消化病杂志,2003,15(3):99.
    [10]Thompson K, Maltby J, Fallow field J, et al. Interleukin-10 expression and function in experimental murine liver inflammation and fibrosis [J].Herpetology,1998,28(6):1597.
    [11]Louis H, Vanlaethem J L, Wa W, et al. Inerleukin-10controls neutrophil in filtration, hepatocyte proliferation and liver inflammation and fibrosis [J]. Herpetology,1998,28(6):1597.
    [12]Sugmioto R, Enjoji M. Effect of IL-4 and IL-13 oncollagen production in cultured LI90 human hepatic stellate cells[J].Liver Int,2005,25(2):420.
    [13]Tomita K, Tamiya G, Ando S,et al. Tumor necrosis factor alpha signaling through activation of Kuffer cells Plays an essential role in liver fibrosis of non ahoholic stator hepatitis in mice [J]. Gut,2006,55:415-424.
    [14]Migita K, Maeda Y, Abiru S, et al. Immune suppressant FKS06 inhibit matrix metalloproteinase-9 induction in TNF-alpha-stimulated human hepatic stellate cells [J]. Sci,2006,78 (21):2510-2515.
    [1]Inn H. Pathogenesis of fibrosis:role of TGF-beta and CTGF[J].Cropping Rheumatic,2002;14(6):681-685.
    [2]Inagaki Y, Okazaki I. Emerging insights into Transforming growth factor (3/Smad signal in hepatic fibro genesis [J].Gut,2007,56(2):284-292.
    [3]党双锁,李亚萍TGF-β1在肝纤维化研究中的新进展[J].世界华人消化杂志,2010,18(16):1631-1636.
    [4]汤丽霞,杨光,谭家驹。Smad7抑制肝星状细胞胶原蛋白表达[J].南方医学大学学报,2009,29(1):2112-2123.
    [5]Fu R, Wu,J.Ding J,et al. Targeting transforming growth factor beta RⅡ expression inhibits the activation of hepatic stellate cells and reduces collegen synthesis[J].Exp Biol Med,2011,236(3):291-297.
    [6]Hamzavi J,Ehnert S,Godoy P,et al.Disruption of the Smad7 gene enhances CCL4-dependent liver damage and fibrogensis in mice[J].Cell Mol Med,2008,12 (5):2130-2144.
    [7]Miyazawa K, Shinozaki M, Hara T, et al.Two major Smad pathways in TGF-beta super family signaling [J]. Genes Cells,2002,7(12):1191-1204.
    [8]Jun Zhou,De-Wu Zhong,Qun-Wei Wang,et al.Paclitaxel ameliorates fibrosis in hepatic stellate cells via inhibition of TGF-β/Smad activity[J].World J Gastroenterol,2010,16(26):3330-3334.
    [9]Nakao A, Afrakhte M, Moren A, et al. Identification of Smad7, a TGF-beta inducible antagonist of TGF-Beta signaling [J].Nature,1997,389:631-635.
    [10]Jun Zhou,De-Wu Zhong, Qun-Wei Wang, et al. Paclitaxel ameliorate fibrosis in hepatic stellatecell via inhibition of TGF-β/Smad activity[J]. World J Gastronenterol,2010,16 (26):3330-3334.
    [11]阮艺华,刘海林.Smad7与肝纤维化[J].国际消化病杂志,2007,27(3):208-210.
    [12]Miyazawa K, Shinozaki M, Hara T, et al.Two major Smad pathways in TGF-beta super family signaling [J]. Genes Cells,2002,7(12):1191-1204.
    [13]Liu Y, Wen X M, Liu E L, et al. Therapeutic targeting of the PDGF and TGF-β signaling pathways in hepatic stellate cells by PTK787/ZK22258[J].Lab Invest,2009,89(10):1152-1160.
    [14]孙玲.豹皮樟总黄酮血清对TGF-β1干预的大鼠肝星状细胞的作用及部分机制[D].安徽医科大学硕士学位论文,2010.
    [15]杨洁,蔡刁龙,谭献文,等.柴胡、红花、川芎中药单体对大鼠肝纤维化治疗作用的实验研究[J].临床和实验医学杂志,2009,8(7):1-3.
    [16]樊予惠,朱琳,华海婴.川芎嗪对人肝星状细胞TGF-β1信号传导的影响[J].中国当代医药,201 1,18(23):19-21.
    [17]何绮微,杨洁.苦参与茯苓对肝纤维化的作用及机制的研究[J].热带医学杂志,2010,10(8):930-931.
    [18]李志钢,杨晋翔,谢春光,等.中药肝心宁对CC14诱导的肝纤维化大鼠TGF-β1/Smad信号通路的影响[J].成都中医药大学学报,2009,32(2):65-68.
    [19]Tang LX, He RH, Yang G, et al. Asiatic Acid Inhibits Liver Fibrosis by Blocking TGF-beta/Smad Signaling In Vivo and In Vitro[J]. PLoS ONE,2012,7:331-350.
    [20]谈博,张奉学,刘妮,等.川芎嗪和苦参碱对HSC-T6细胞增殖的抑制作用[J].热带医学杂志,2004;4(6):678-681.
    [21]谈博,宋健平,张奉学,等.川芎嗪对HSC-T6细胞Smad蛋白细胞内转位的影响[J].中药新药与临床药理,2006;17(5):320-322.
    [22]董玲,孙剑勇,方国汀,等.甘草酸对转化生长因子β刺激肝星状细胞信号传导的作用[J].中华肝脏病杂志,2005,13(11):828-831.
    [1]袁永兵,张兰珍,郭亚建,等RP-HPLC法测定叶下珠中没食子酸、柯里拉京和鞣花酸的含量[J].北京中医药大学学报,2009,32(1):56-58.
    [2]彭立志,贺劲松,童光东,等.叶下珠提取物抗乙肝病毒及乙肝病毒X基因的研究[J].中西医结合肝病杂志,2006,16(6):340-343.
    [3]谢志春.叶下珠抗乙肝病毒作用的研究现状[J].广西医学,2006,28(2):164-166.
    [4]张芝英.高效液相色谱法测定叶下珠中没食子酸的含量[J].中国药业,2002,11(7):53.
    [5]李家珉.叶下珠中抗PAI-1成分柯里拉京的提取分离[D].广州中医药大学,2009,广州.
    [6]沈志强,陈蓬,段理,等.叶下珠有效部位对凝血系统的影响[J].中草药,2004,35(5):539-542.
    [7]窦志芳,冯前进,闫娟丽,等.苦味叶下珠抗病毒作用的研究进展[J].中华中医药期刊,2007,25(11):2311-2312.
    [8]李兰岚,范适,饶立群,等.叶下珠提取物对体外四氯化碳损伤肝细胞的保护作用[J].中国组织工程与临床康复,2007,11(25):4909-4912.
    [9]冀德富,郭冬艳,裴妙荣.叶下珠总多酚提取工艺的优选[J].山西中医学院学报,2010,11(1):16-18.
    [10]肖凯,宣利江,徐亚明,等.虎杖的化学成分研究[J].中国药学杂志,2003,38(1):12-15.
    [11]莫志贤,邵红霞.白藜芦醇苷体外对过氧化氢导致小鼠肝细胞损伤的保护作用[J].中国药理学通报,2000,16(5):519-521.
    [12]张霖,陈育尧,孙学刚,等.虎杖苷对非酒精性脂肪肝大鼠保护作用及机制研究[J].陕西中医,2010,31(6):756-758.
    [13]杨溪琳.虎杖的现代药用研究[J].中国社区医师,2008,10(1):24.
    [14]陈文霞,谈献和,李伟,等.白花蛇舌草HPLC指纹图谱的研究[J].现代中药研究与实践,2006,20:38-40.
    [15]谭宁华,王双明,杨亚滨,等.白花蛇舌草抗肿瘤活性和初化学研究[J].天然产物研究与开发,2004,14(5):33-36.
    [16]赵浩如,李瑞,林以宁,等.白花蛇舌草不同提取工艺对抗肿瘤活性的影响[J].中国药科大学学报,2002,33(6):510.
    [17]张硕,宋衍芹,倪同尚,等.白花蛇舌草总黄酮抑制人肝癌细胞的靶基因调控[J].世界华人消化杂志,2007,15(10):1060-1066.
    [18]陈秀珍,朱大诚,王艳辉.白花蛇舌草药理作用及临床应用研究进展[J].中药材,2009,32(1):157-160.
    [19]阮金兰,赵钟祥,曾庆忠.赤芍化学成分和药理作用的研究进展[J].中国药理学通报,2003,19(9):965-970.
    [20]陈晓宇,宗云奎.三种中草药及其复方体外对HBsAg、HBeAg分泌的抑制[J].中国药物与临床,2002,2(1):46-48.
    [21]孙丽荣,曹雄,侯凤青,朱心红,高天明等.芍药苷研究进展[J].中国中药杂志,2008,33(18):2028-2029.
    [22]王晓玉,魏伟,唐丽琴,等芍药苷对佐剂性关节炎大鼠腹腔巨噬细胞吞噬功能及其产生细胞因子的影响[J].安徽医科大学学报,2007,42(2):189.
    [23]晏雪生,李瀚曼,彭亚琴,等.芍药苷对人肝癌细胞HepG-2凋亡及其调控基因的影响[J].中华中医药学刊,2007,25(7):1346-1347.
    [24]Ping J, Wang H.The effects and mechanism of indole-3-carbinol oninducing hepatic stellate cell apoptosis [J].Chin Pharmacol,2007,24:48-9.
    [25]胡宗涛,高世乐,秦峰,等.芍药苷对大鼠放射性肝纤维化的保护作用和机制研究[J].解放军药学学报2012,28(4)::283-288.
    [26]黄桢.黄芪多糖的药理研究进展[J].中国临床医药学杂志,2002,11(5):315.
    [27]王淑华,王红霞.浅析黄芪不同剂量的药理作用[J].浙江中医杂志,2002(6):260.
    [28]吕涛,姚希贤,孙泽明.药物血清内黄芪甲苷含量测定及其抑制HSCs活化增殖的实验研究[J].中华中医药学刊,2011,29(12):2770-2773.
    [29]刘浩.前列腺素E2受体信号转导在大鼠免疫性肝纤维化和肝星状细胞胶原合成中的作用及黄芪甲苷对其影响[D].安徽科技大学,2008.
    [30]侯双,杨艳芳,魏建和,等.丹参有效成分合成积累及调控的分子机制研究进展[J].现代中药研究与实践,2010,24(4):82-85.
    [31]刘艾林,李铭源,王一涛,等.丹参药理学活性物质基础研究现状[J].中国药学杂志,2007(9):641-646.
    [32]辜海英,钟森,陈红苹.复方丹参注射液治疗慢性乙型肝炎的荟萃分析[J].现代中西医结合杂志,2009,18(16):1851.
    [33]沙飞,禹志领,王一涛.土茯苓品质与药理研究进展[J].中药材,2006,29(5):516-519.
    [34]李小娜,张兰桐,殷玮.中药复方药效物质基础研究途径与方法[J].中草药,2006,37(6):801-805.
    [35]贾晓斌,陈彦,李霞,等.中药复方物质基础研究新思路和方法[J].中华中医药杂志,2008,23(5):420.
    [36]薛燕等.中药复方霰弹理论[M].中国环境科学出版社,北京,1996年.
    [37]黄熙.方剂体内血清成分谱与靶成分概念的提出及意义[J].第四军医大学学报,1999,10(4):177.
    [38]罗国安,王义明.中药复方的化学研究体系[J].世界科学技术-中药现代化,1999,1(1):11.
    [39]邱峰,姚新生等.中药体内直接物质基础研究的新思路[J].中国药理与临床,1999,15(3):1.
    [40]王本祥,周丽秋.关于中药活性成分的认识及其研究方法[J].中国中药杂志,2001,26(1):10.
    [1]Friedman SL.Liver fibrosis-from bench to bedside[J]. Hepatol 2003:38 Suppl 1:S38-53.
    [2]ISON M, SODA M, I NOUE A, et al. Reverse transformati on of hepaticmyofibroblast-like cells by TGF-β1/LAP[J]. Biochem Biophys Res Commun,2003,311 (4):959-965.
    [3]Safadi R, Friedman SL. Hepatic fibrosis role of hepatic stellate cell activation[J]. Med Gen Med 2002: 4:27.
    [4]Eng FJ, Friedman SL. Fibrogenesis I. New insights into hepatic stellate cell activation:the simple becomes complex [J]. Am I Physiol Gastrointest Liver Physiol 2000:279:G7-G11.
    [5]Rockey DC. Hepatic blood flow regulation by stellate cell in normal and injured liver [Jj.Semin Liver Dis 2001:21:337-349.
    [6]Reeves HL, Friedman SL. Activation of hepatic stellate cell-a key issue in liver fibrosis [J].Front Biosci 2002:7:d808-826.
    [7]李欣,彭小东,张文利,等.四种中药单体的抗肝纤维化作用及其机制[J].中华肝脏病杂志,2008,16(3):193-197.
    [8]王小众.肝纤维化的发病机制及肝纤维化研究进展[J].中西医结合肝病杂志,2001,11:35-38.
    [9]Gressner AM. Transdifferentiation of hepatic stellate cells(Ito cells) to myofibroblasts:a key event in hepatic fibrogenesis [J]. Kidney Int,1996,39-45.
    [10]曾明德,萧树东主编.肝脏与内分泌[M].第一版,人民卫生出版社.1995:168-172.
    [11]刘成海.肝纤维化的基础研究进展[J].中国中西医结合杂志,2006,26(1):11-12.
    [12]Gewalting J, Mangasser-Stephan K, Gartung C, et al. Association of polymorphisms of the transforming growth factor-betal gene with the rate of progression of HCV-induced liver fibrosis[J]. Clin Chim Acta,2002, 316:83-94.
    [13]Bissell DM,Roulot D, George J, et al. Transforming growth factor β and liver[J]. Hepatolgy,2001,34: 859-867.
    [14]Border WA, Noble NA.Transforming growth factor beta in tissuefibrosis [J]. New Engl J Med,1993, 331:1286-1292.
    [15]Nagashio Y,Ueno H, Imamura M, et al.Inhibition of transforming growth factor beta decreases pancreatic fibrosis and protects the pancreas against chronic injury in mice[J].Lab Invest,2004,84:1610-1618.
    [1]Mehal W.Imaeda A.Cell Death and Fibro genesis[J].Semin Liver Dis,2010,30(3):226.
    [2]Harper N,Hughes M, MacFarlane M,et al.Fas-associated death domain protein and caspase-8 are not recruited to the tumor necrosis factor receptor 1 signaling complex during tumor necrosis factor-induced apoptosis[J].J Biol Chem,2003;278:2534-2541.
    [3]Jaeschke H,Gores GJ, Cederbaum AI, et al. Mechanisms of hepatotoxicity [J].Toxicol Sci,2002:65:166-176..
    [4]Baskin-Bey ES, Gores GJ.Death by assoeiation:BH3 domain only proteins and liver injury [J].Am J Physiol Gastrointest Liver Physiol,2005;289:987-990.
    [5]Oltvai ZN, Milliman CL, Korsmeyer SJ.Bcl-2 heterodimerizes in vivo with a conserved homolog, Bax, that accelerates Programmed cell death[J].Cell,1993;74:609-619.
    [6]Rogerio F, Jordao H JR, Vieira AS, et al. Bax and Bcl-2expression and TUNEL labeling in lumbar enlargement of neonatal rats after sciatic axotomy and melaton in treatment [J]. Brain Res,2006,11(12):80-90.
    [7]Higuchi M, Aggarwal BB, Yeh ET. Activation of CPP32-like protease in tumor necrosis faetor induced apoptosis is dependent on mitochondrial function [J].J Clin Invest,1997:99:1751-1758.
    [8]Kotsafti A, Farinati F, Cardin R, el at. Bax inhibitor-1 down-regulation in the progression of chronic liver diseases[J].BMC Gastroenterology.2010,10:35.
    [9]Mitchell C, Mabrouf-Yorgov M, Mayeuf A,et al. Over expression of Bcl-2 in hepatocytes protects against injury but does not attenuate fibrosis in a mouse model of chronic cholestasis liver disease [J]. Lab Invest 2011, 91:273-282.
    [1]席文娜,孙水林,李方春.a-2干扰素对大鼠肝组织bcl-2基因表达的影响及意义[J].世界华人消化杂志,2009;17(31):3232-3240.
    [2]赵宏贤,陈霞,郭勇,等.粉防己碱对大鼠肝星状细胞增殖及细胞周期D1增殖细胞核抗原表达的调控[J].时珍国医国药,2008;19(1):108-110.
    [3]贺骏,阮秋蓉,杨秀萍,等.CyclinD1在人类5种常见肿瘤中的表达及其意义[J].中国组织化学与细胞化学杂志,2006;15(6):608-612.
    [4]Motokura T, Arnold A. CyclinD and oncogenesis [J].CurrPin Genet Dev.1993;3(1):5-10.
    [5]Umekita Y,OHI Y. Overexpression of cyc1 inD1 prediets for poorprognosis in estrogen receptor-negative breast cancer patients [J]. Int J Cancer,2002:98 (3):415-418.
    [6]张伟,杜成友,徐尔侃.肝癌临床发展与cyclinDl表达DNA含量及细胞凋亡发生的关系[J].第三军医大学学报,2005;27(5):46.
    [7]Loyer P, Trembley JH, Katona R,et al. Role of CDK/cyclin complexes in transcription and RNA splicing. Cell Signa [J].2005;17(9):1033-1051.
    [8]Hirsch FR, Witta S, Biomarkers for Prediction of sensitivity to EGFR inhibitiors in non-small Cell lung cancer. Curropin oncol [J].2005;17(2):118-122.
    [9]Chenchen Li, Xinmei Li.The different roles of cyclinDl-CDK4 in STEP and mGluR-LTD during the Postnatal development in mice hippocampus area CA1 [J].BMC Dev Biol,2007:7:57.
    [10]Holley SL, Heighway J, Hoban PR.Induced expression of human CCND1 alterative transcripts in mouse Cyl-1 knockout fibroblasts high lights functional differences [J].Int J Cancer,2005:114(3):364-370.

© 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