靶向肝星状细胞PDGF-β受体小干扰RNA减轻大鼠肝纤维化
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
研究背景及目的
     肝纤维化是肝脏对不同病因所致的慢性损伤共有的过修复反应,其病理特征是肝脏细胞外基质(extracellular matrix,ECM)过度沉积及分布异常。肝星状细胞(hepaticstellate cell,HSC)是纤维化肝脏中产生ECM的最主要细胞,HSC的活化是肝纤维化发生的核心环节,设法抑制HSC激活、增殖与迁移是肝纤维化治疗的重要对策。
     血小板衍生生长因子(platelet-derived growth factor,PDGF)是最强的促进HSC增殖的细胞因子,由A、B、C、D四条高度同源的多肽链组成,其中A、B亚基互相组合可以表现为AA、BB、AB三种分子形式。PDGF受体(platelet-derived growthfactor receptor,PDGFR)由两种亚单位α及β构成,静止的HSC表面只有α亚单位,HSC活化后才表达β亚单位,而且以β亚单位为主,与PDGF-BB具有较强的亲和力。PDGF-BB以及PDGFR-β在肝纤维化过程中发挥重要作用,设法抑制PDGF及其受体表达,阻断PDGF细胞内信号转导是抗肝纤维化的治疗策略之一。
     RNA干扰(RNA interference,RNAi)是一种介导转录后基因沉默的强大工具。它是通过小干扰RNA(small interfering RNA,siRNA)降解生物体或细胞内同源mRNA,导致序列特异性同源基因沉默,使细胞表现出特定基因缺失表型的过程。本研究应用RNAi技术沉默PDGFR-β表达,阻断PDGF的作用,观察在体外对HSC生物学特性及大鼠实验性肝纤维化的影响;并构建胶质纤维酸性蛋白(glial fibrillaryacidic protein,GFAP)基因启动子介导的PDGFR-βsiRNA表达载体,观察其靶向治疗大鼠实验性肝纤维化的疗效。
     实验方法
     1、筛选高效沉默PDGFR-β基因表达的siRNA
     设计3对针对大鼠PDGFR-βmRNA开放阅读框的siRNA分子,用脂质体转染法将siRNA转染HSC-T6细胞,RT-PCR法和Western Blot法检测HSC-T6中PDGFR-βmRNA和蛋白的表达,筛选高效沉默PDGFR-β基因表达的siRNA。
     2、siRNA干扰PDGFR-β表达对HSC-T6生物学特性的影响
     PDGFR-βsiRNA转染HSC-T6后进行PDGF-BB刺激,48小时后收集细胞,用RT-PCR法检测细胞中与纤维化相关的基因表达变化;用BrdU掺入试验检测细胞增殖能力的变化;用Annexin IV/PI染色法检测凋亡细胞数目的变化;用Western Blot检测ERK的磷酸化水平,RT-PCR检测c-fos的表达水平来观察PDGFR-βsiRNA对HSC细胞内MAPK信号转导通路的影响。
     3、构建表达PDGFR-βsiRNA的质粒载体
     siRNA分子在体内具有半衰期短、不稳定等缺点,为了观察PDGFR-βsiRNA在体内对动物实验性肝纤维化的影响,我们将PDGFR-βsiRNA对应的短发夹RNA(shorthairpin RNA,shRNA)插入商品化质粒pCMR30的miR30结构中,构建表达PDGFR-βshRNA的质粒载体(pCMV-shRNA-LacZ),以CMV基因启动子启动下游PDGFR-βshRNA的表达,β-半乳糖苷酶(β-galactosidase,β-gal)基因作为报告基因。另外构建阴性对照质粒(pCMV-NC-LacZ),以CMV基因启动子启动下游对PDGFR-β基因无干扰作用的小RNA的表达。
     4、pCMV-shRNA-LacZ对动物实验性肝纤维化的影响
     构建胆总管结扎(bile duct ligation,BDL)和二甲基亚硝胺(dimethylnitrosamine,DMN)大鼠肝纤维化模型,将pCMV-shRNA-LacZ和pCMV-NC-LacZ质粒在造模前1天通过尾静脉高压注射至大鼠体内。为了保证PDGFR-βshRNA在大鼠体内的稳定表达,以后每5天重新注射一次。造模结束后取血检测血清肝功能,取肝组织用免疫组化法和Western Blot方法检测α平滑肌激动蛋白(α-smooth muscle actin,α-SMA)和PDGFR-β蛋白表达水平,用HE染色和Masson三色染色法观察肝脏的病理变化。
     5、构建含胶质纤维酸性蛋白基因启动子的PDGFR-βshRNA质粒表达载体
     质粒pGfa2-clac包含人的GFAP基因启动子。用PCR克隆的方法将pGfa2-clac中的GFAP基因启动子克隆出来,插入pCMV-shRNA-LacZ的酶切位点之间,取代CMV基因启动子,命名为pGfa-shRNA-LacZ。同理构建阴性对照质粒,命名为pGfa-NC-LacZ。
     6、体内验证pGfa-shRNA-LacZ的HSC靶向性
     由于在正常肝脏中HSC的数量较少,为了增加肝脏中HSC的数目以便于我们观察,我们用四氯化碳(carbon tetrachloride,CCl_4)腹腔内注射制备大鼠急性肝损伤模型,用BDL方法制备大鼠慢性肝损伤模型。将pGfa-shRNA-LacZ和pCMV-shRNA-LacZ分别通过尾静脉高压注射入模型大鼠体内,24小时后处死小鼠,取肝组织用免疫荧光染色和激光共聚焦显微镜技术检测表达β-gal的细胞在两种模型大鼠肝组织中的分布,并观察肝脏中β-gal蛋白和GFAP蛋白、β-gal蛋白和α-SMA蛋白的细胞共定位情况。
     7、pGfa-shRNA-LacZ对大鼠实验性肝纤维化的影响
     构建BDL大鼠肝纤维化模型,将构建好的pGfa-shRNA-LacZ及其相应的阴性对照质粒pGfa-NC-LacZ在造模前1天通过尾静脉高压注射至各组大鼠体内。造模结束后取血检测血清肝功能,取肝组织用免疫组化法检测α-SMA和PDGFR-β蛋白表达水平,用HE染色和Masson三色染色法观察肝脏的病理变化,测定组织羟脯氨酸含量来定量评价PDGFR-βshRNA对大鼠肝纤维化的影响。
     实验结果
     1、针对PDGFR-β基因957位点的siRNA能高效沉默靶基因的表达
     我们合成了针对PDGFR-β基因不同位点的小干扰RNA及其阴性对照RNA,分别命名为PDGFR-βsiRNA_(2597)、PDGFR-βsiRNA_(2687)、PDGFR-βsiRNA_(957)和PDGFR-βsiRNA_(con)。RT-PCR结果显示三对siRNA均能不同程度抑制PDGFR-βmRNA的表达,其中以PDGFR-βsiRNA_(957)的基因沉默效率最高,达到(72±4.20)%,且转染HSC-T6细胞72小时的基因沉默效率明显高于转染24(40±3.14)%和48小时(55±4.67)%,Western Blot检测显示PDGFR-βsiRNA_(957)转染HSC-T6细胞72小时后PDGFR-β蛋白水平降低约(60±7.59)%。以后的实验均用PDGFR-βsiRNA_(957)进行。
     2、PDGFR-βsiRNA能抑制HSC-T6细胞的活化和胶原合成
     RT-PCR和Western Blot结果表明PDGF-BB能提高HSC-T6细胞的活化水平,使α-SMA的基因表达水平升高,而PDGFR-βsiRNA能抑制PDGF-BB的这种效应,使α-SMA的基因表达水平降低约(50±2.20)%;此外,RT-PCR结果还提示PDGF-BB能明显促进HSC-T6 PDGF-B链、结缔组织生长因子(connective tissue growth factor,CTGF)、金属蛋白酶组织抑制因子-1(Tissue Inhibitor of Metalloproteinase-1,TIMP-1)和I型胶原基因表达,而PDGFR-βsiRNA能使PDGF-B、CTGF、TIMP-1和I型胶原基因表达分别下降约(43±3.40)%、(50±5.10)%、(45±1.80)%和(40±2.40)%。但无论是PDGF-BB还是PDGFR-βsiRNA对转化生长因子(Transforming GrowthFactor-β_1,TGF-β_1)和基质金属蛋白酶-2(Matrix Metalloproteinases-2,MMP-2)的基因表达均无明显影响。
     3、PDGFR-βsiRNA抑制HSC-T6细胞的增殖
     BrdU掺入试验表明PDGF-BB明显促进HSC细胞增殖,而PDGFR-βsiRNA能明显抑制PDGF-BB的促增殖效应(流式细胞法显示抑制40%的细胞增殖,细胞免疫荧光法显示抑制35%的细胞增殖)。Western Blot和RT-PCR检测显示PDGF-BB能激活细胞内MAPK信号转导通路,使ERK蛋白磷酸化和c-fos的基因表达明显增加,而PDGF-BB的这种效应被PDGFR-βsiRNA明显抑制,其中c-fos的基因表达减少约35%,ERK蛋白磷酸化减少约50%。Annexin/PI流式细胞检测表明无论PDGF-BB还是PDGFR-βsiRNA对HSC凋亡均无明显影响。
     4、pCMV-shRNA-LacZ能改善肝纤维化大鼠肝功能,减轻其肝纤维化程度
     血清肝功能检测显示BDL模型大鼠的血清丙氨酸氨基转移酶(alanineaminotransferase,ALT)、天冬氨酸氨基转移酶(aspartate aminotransferase,AST)、总胆红素(total bilirubin,TB)和间接胆红素(indirect bilirubin,IDB)水平明显高于假手术组和正常组,而pCMV-shRNA-LacZ处理后转氨酶和胆红素水平均明显降低。DMN模型大鼠的ALT、AST和TB水平明显高于正常组大鼠,而pCMV-shRNA-LacZ处理后转氨酶水平和胆红素均明显下降。HE染色和Masson染色显示pCMV-shRNA-LacZ能明显减轻BDL和DMN引起的大鼠肝损伤和肝纤维化程度。Western Blot检测和免疫组化染色显示BDL模型组和DMN模型组肝脏的PDGFR-β蛋白和α-SMA蛋白水平明显升高,而pCMV-shRNA-LacZ处理后两种蛋白的表达水平均明显降低。
     5、GFAP基因启动子能驱动下游基因在HSC特异表达
     免疫荧光染色显示在CCl_4介导的急性肝损伤和BDL介导的慢性肝损伤模型中,无论是pCMV-shRNA-LacZ处理组还是pGfa-shRNA-LacZ处理组,大鼠肝脏中β-gal蛋白均主要表达在中央静脉周围及汇管区的细胞中。免疫荧光双重染色显示:pCMV-shRNA-LacZ处理组大鼠的β-gal蛋白不仅表达在GFAP蛋白表达阳性的细胞中,还表达在GFAP蛋白表达阴性的细胞中;而pGfa-shRNA-LacZ处理组大鼠肝脏中β-gal蛋白仅表达在GFAP蛋白表达阳性的细胞中。β-gal是我们构建的质粒的报告基因,GFAP是HSC特异表达的标志蛋白,提示GFAP基因启动子能介导下游基因在HSC特异表达。
     6、pGfa-shRNA-LacZ改善BDL大鼠的肝功能,减轻肝纤维化程度
     血清生化检测显示BDL模型组大鼠转氨酶(ALT和AST)和胆红素(TB和IDB)水平明显升高,而pGfa-shRNA-LacZ处理后升高的转氨酶和胆红素水平明显降低。α-SMA和PDGFR-β免疫组化染色以及Masson染色显示单纯BDL大鼠的PDGFR-β蛋白和α-SMA蛋白表达水平明显强于正常大鼠,并出现明显的胶原沉积和胆管增生,而pGfa-shRNA-LacZ处理后PDGFR-β蛋白和α-SMA蛋白表达水平均有明显降低,肝纤维化和胆管增生情况均有改善。BDL大鼠肝组织羟脯氨酸含量较正常大鼠增加约2倍,而pGfa-shRNA-LacZ可使BDL大鼠的肝组织羟脯氨酸含量下降约45%。
     结论
     1、用RNAi技术抑制HSC中PDGFR-β基因表达,阻断PDGFR-β与PDGF-BB的结合,可阻断细胞内MAPK信号转导通路进而抑制细胞的增殖,并可显著抑制HSC的活化和胶原分泌;
     2、用RNAi技术抑制PDGFR-β基因表达,可明显改善BDL和DMN诱导的肝纤维化模型大鼠的肝功能,减轻肝纤维化程度;
     3、GFAP基因启动子可介导下游基因特异性地在HSC中表达;
     4、GFAP基因启动子能介导针对PDGFR-β基因的小干扰RNA特异性地表达在HSC中,并能有效改善BDL大鼠肝功能,减轻肝纤维化程度。
Background and Objective
     Hepatic fibrosis,characterized by the overproduction and accumulation of hepatic extracellular matrix(ECM),was the common and important pathological changes in the chronic liver diseases,through which the chronic hepatitis developed into cirrhosis.It was acknowledged that hepatic cirrhosis couldn't be reversed,while hepatic fibrosis could. Hepatic stellate cell(HSC) was the main resource of hepatic ECM and the cellular basis for hepatic fibrosis.The activation of HSC was recognized as the key event of the pathogenesis of hepatic fibrosis.Inhibiting the activation,proliferation and migration of HSCs is one of the most important strategies to prevent the hepatic fibrogenesis.
     In fibrotic liver,PDGF is the most potent mitogen for HSCs.PDGF binds to structurally related tyrosine kinase receptors that is a homodimer or heterodimer ofαorβsubunits.PDGFR-αis constitutively expressed in quiescent HSCs,whereas PDGFR-βis acquired as the activation of HSCs,the major cell type expressing PDGFR-βin liver. PDGF and PDGFR-βplay important roles in hepatic fibrogenesis,therefore,targeting to PDGF,PDGF receptor and molecules related to PDGF intracellular signaling transduction pathway is attractive for therapeutic intervention in hepatic fibrosis.
     RNAi(RNA interference) is one of the gene-silencing technologies,which is considered as an antisense mechanism of action that utilizes double strand RNase to promote hydrolysis of target RNA.In this study,we inhibited the PDGFR-βexpression by small interference RNA(siRNA) to block the binding of PDGF,and investigated its effect on biological characteristics of HSCs in vitro.Furthermore,we constructed a PDGFR-βshRNA(short hairpin RNA) expression vector and investigated its antifibrogenic effect on experimental hepatic fibrosis.To avoid the non-specific interference of PDGFR-βgene expression in other tissues and cells,we used cell-specific glial fibrillary acidic protein (GFAP) promoter to drive the expression of PDGFR-βshRNA in HSCs of liver,and investigated its antifibrogenic effect in vivo.
     Methods
     1.Screening for the PDGFR-βsiRNA with high gene-silencing efficacy
     The targeted region of siRNA was the coding sequence of PDGFR-βcDNA.We selected 3 pairs 21-nucleotide RNAs and 1 pairs non-specific nucleotide RNA.Transient transfection of siRNA by lipofection method was carried out in the rat HSC-T6 cell line, whose phenotype was activated HSC.Reverse transcription-polymerase chain reaction (RT-PCR) and Western Blot were used to detect the expressions of PDGFR-β.
     2.Investigate the effect of PDGFR-βsiRNA on biological characteristics of HSC
     Cells were transfected with PDGFR-βsiRNA and were stimulated by PDGF-BB,and then were collected.RT-PCR was used to detect the expression of fibrogenesis- related genes.BrdU incorporation was performed to determine cell proliferation.Annexin V/PI staining method was utilized to measure cell apoptosis.Mitogen-activated protein kinases (MAPK) signaling transduction pathway was necessary for the HSC proliferation induced by PDGF,and extracellular-signal- related kinase(ERK) phosphorylation and c-fos expression were the two important processes of MAPK signaling transduction pathway, which were detected by Western Blot and RT-PCR in this study.
     3.Construct plasmid expressing PDGFR-βshRNA
     The disadvantages of chemically synthesized siRNA,such as instability and easy to be degraded by serum nucleases restrict its application in vivo.To investigate the effect of PDGFR-βsiRNA on experimental hepatic fibrosis,we inserted PDGFR-βshRNA into the miR30-based shRNA expression system of pCMR30 Vector to generate pCMV-shRNA-LacZ,in which PDGFR-βshRNA was driven by CMV promoter andβ-galactosidase(β-gal) was the reporter gene.As a negative control,we generated pCMV-NC-LacZ with miR30-based PDGFR-βshRNA in pCMV-shRNA- LacZ replaced by negative control RNA.
     4.Investigate the effect of pCMV-shRNA-LacZ on experimental hepatic fibrosis
     Rats model of hepatic fibrosis were induced by common bile duct ligation(BDL) and dimethylnitrosamine(DMN) treatment,pCMV-shRNA-LacZ solutions were injected once every 5 days with hydrodynamics-based transfection method.to induce stable expression of PDGFR-βshRNA.After the rats models were constructed successfully,rats were sacrificed,serum samples were collected for biochemical tests and liver tissues were taken out for histology and Western Blot analysis.
     5.Construct plasmid expressing PDGFR-βshRNA driven by GFAP promoter
     The GFAP promoter in the plasmid named pGfa2-clac was PCR-cloned into the EcoRI and BamHI site and replaced the CMV promoter of pCMV-shRNA-LacZ to generate pGfa-shRNA-LacZ,pGfa-NC-LacZ was generated by replacement of PDGFR-βshRNA in pGfa-shRNA-LacZ with non-specific nucleotide RNA as a negative control.All the constructed plasmids were verified by PCR and sequencing.
     6.Locate LacZ gene expression in acute and chronic liver injury rats model
     To locate LacZ reporter gene expression driven by GFAP promoter in liver,we injected pGfa-shRNA-LacZ or pCMV-shRNA-LacZ by hydrodynamics-based transfection method into normal rats,acute liver injury rats induced by CCl4 and chronic liver injury rats induced by BDL.Twenty four hours after plasmids injection,all rats were anesthetized and perfused,livers were taken out and postfixed.Immunofluorescence and laser scanning confocal techniques were used to observe the distribution ofβ-gal protein,and the colocalization ofβ-gal andα-SMA orβ-gal and GFAP.
     7.Investigate the effect of pGfa-shRNA-LacZ on experimental hepatic fibrosis
     Hepatic fibrosis was induced by BDL in rats,pGfa-shRNA-LacZ solutions were injected once every 5 days with hydrodynamics-based transfection method.to induce stable expression of PDGFR-βshRNA.After the rats models were constructed successfully,rats were sacrificed,serum samples were collected for biochemical tests and liver tissues were taken out for histology and hydroxyproline content test.
     Results
     1.PDGFR-βsiRNA could downregulate PDGFR-βexpression
     All the 3 pairs of siRNAs could suppress PDGFR-βmRNA expression,especially siRNA_(957),which had the highest gene-silencing efficacy.RT-PCR results showed that siRNA_(957) downregulated the PDGFR-βmRNA expression by about(40±3.14)%, (55±4.67)%,(72±4.20)%and(55±6.19)%at 24h,48h,72h and 96h,respectively.In addition,PDGFR-βprotein expression was downregulated by about(33±4.51)%, (47±5.72)%,(60±7.59)%and(82±7.13)%at 24h,48h,72h and 96h,respectively.
     2.PDGFR-βsiRNA could suppress the activation and proliferation of HSC
     The results of RT-PCR and Western Blot revealed that theα-SMA expression in HSCs was elevated significantly after PDGF-BB stimulation,which was impeded by PDGFR-βsiRNA.Furthermore,the upregulation of PDGF-B,CTGF,TIMP-1 and collagen typeⅠmRNA induced by PDGF-BB was eliminated by siRNA_(957).However,the expressions of TGF-β_1 and MMP-2 mRNA in HSCs were not affected by PDGFR-βsiRNA_(957) and/or PDGF-BB.
     3.PDGFR-βsiRNA inhibited HSC proliferation induced by PDGF-BB,but did not affect the apoptosis of HSC
     To investigate the effect of siRNA_(957) on HSC proliferation,BrdU incorporation was used and analyzed by immunofluorescence and flow cytometry.Results showed that PDGF-BB significantly promoted the proliferation of HSC,which was inhibited by PDGFR-βsiRNA_(957).Additionally,Western blot analysis revealed that the phosphorylation of ERK increased significantly after PDGF-BB stimulation,which was impeded by siRNA_(957),RT-PCR analysis demonstrated that siRNA_(957) suppressed the upregulation of transcription factor c-fos expression induced by PDGF-BB.Annexin V/PI flow cytometry analysis showed that neither PDGF-BB nor siRNA_(957) could affect the percentage of apoptotic HSC.
     4.pCMV-shRNA-LacZ relieved liver injury and ameliorated hepatic fibrosis in rats
     In DMN animal model,results of serum biochemical tests revealed that serum total bilirubin(TB),alanine aminotransferase(ALT) and aspartate aminotransferase(AST) levels were elevated significantly in DMN control rats compared with the normal rats, which were reduced by pCMV-shRNA-LacZ treatment.In BDL animal model,serum TB, direct bilirubin(DB),ALT and AST levels were elevated significantly in BDL control group compared with the sham-operated group,which was eliminated by pCMV-shRNA-LacZ treatment.HE staining and Masson's trichrome staining of liver sections showed that liver injury and hepatic fibrosis in both rats models were ameliorated by pCMV-shRNA-LacZ treatment.Western Blot analysis indicated that both PDGFR-βandα-SMA protein levels were elevated significantly in DMN treatment rats and BDL rats, and remarkably reduced by pCMV-shRNA-LacZ treatment.The results were confirmed by immunohistochemical examination.
     5.GFAP promoter targets LacZ express into HSC
     Immunofluorescence analysis indicated thatβ-gal-positive cells were all distributed in the pericentral and portal area of hepatic lobule in normal rats,acute liver injury rats and chronic liver injury rats for both pCMV-shRNA-LacZ and pGfa-shRNA-LacZ treatment. The results also revealed thatβ-gal proteins were detected in both hepatocytes and HSC, which were positive forα-SMA and/or GFAP staining in the liver treated with pCMV-shRNA-LacZ.While in the liver treated with pGfa-shRNA-LacZ,β-gal proteins were co-expressed with GFAP and/orα-SMA,and were located in the perisinusoidal space, suggesting that GFAP promoter targeted the reporter gene LacZ specifically expressing in HSC and CMV promoter directed LacZ expressing in not only HSCs,but also hepatocytes.
     6.pGfa-shRNA-LacZ relieved liver injury and ameliorated hepatic fibrosis in rats
     Biochemical tests indicated that the elevated serum levels of TB,indirect bilirubin (IDB),ALT and AST induced by BDL could be markedly reduced by pGfa- shRNA-LacZ treatment.Immunohistochemical and Masson's trichrome staining of liver sections showed significant collagen deposition and bile duct hyperplasia in BDL rats,and increased protein expressions of PDGFR-βandα-SMA were also observed in them,while both the hepatic fibrosis and the increased expressions of PDGFR-βandα-SMA protein were significantly relieved by pGfa-shRNA-LacZ treatment.In addition,the hydroxyproline content increased more than 2-fold in BDL rats compared with the sham-operated rats,while pGfa-shRNA-LacZ treatment decreased about half of the hydroxyproline content in BDL rats.
     Conclusion
     1.siRNA against PDGFR-βcould block the binding of PDGF,inhibit the MAPK signaling transduction pathway and suppress HSC proliferation consequently.Furthermore, the activation and ECM production of HSC stimulated by PDGF-BB could also be inhibited by PDGFR-βsiRNA.
     2.PDGFR-βsiRNA is active as an antifibrogenic gene therapeutic method able to reduce the expression of PDGFR-βprotein and ameliorated liver injury and hepatic fibrosis induced by BDL and DMN.
     3.GFAP promoter was able to target genes downstream express into HSC.
     4.PDGFR-βshRNA driven by GFAP promoter could be targeted into HSC,and could inhibit the activation of HSC,relieve liver injury and hepatic fibrosis induced by BDL.
引文
[1] Safadi R, Friedman SL. Hepatic fibrosis-role of hepatic stellate cell activation. Med Gen Med 2002;4:27.
    [2] Kinnman N, Francoz C, Barbu V, et al. The myofibroblastic conversion of peribiliary fibrogenic cells distinct from hepatic stellate cells is stimulated by platelet-derived growth factor during liver fibrogenesis. Lab Invest 2003;83:163-173.
    [3] Pinzani M. PDGF and signal transduction in hepatic stellate cells. Front Biosci 2002;7:d1720-1726.
    [4] Marra F, Arrighi MC, Fazi M, et al. Extracellular signal-regulated kinase activation differentially regulates platelet-derived growth factor's actions in hepatic stellate cells, and is induced by in vivo liver injury in the rat. Hepatology 1999;30:951-958.
    [5] Pinzani M, Milani S, Herbst H, et al. Expression of platelet-derived growth factor and its receptors in normal human liver and during active hepatic fibrogenesis. Am J Pathol 1996;148:785-800.
    [6] Czochra P, Klopcic B, Meyer E, et al. Liver fibrosis induced by hepatic overexpression of PDGF-B in transgenic mice. J Hepatol 2006;45:419-428.
    [7] Breitkopf K, Roeyen C, Sawitza I, et al. Expression patterns of PDGF-A, -B, -C and -D and the PDGF- receptors a and b in activated rat hepatic stellate cells (HSC). Cytokine 2005;31:349-357.
    [8] Wong L, Yamasaki G, Johnson RJ, Friedman SL. Induction of beta-platelet- derived growth factor receptor in rat hepatic lipocytes during cellular activation in vivo and in culture. J Clin Invest 1994;94:1563-1569.
    [9] Vickers TA, Koo S, Bennett CF, et al. Efficient reduction of target RNAs by small interfering RNA and RNase H-dependent antisense agents. A comparative analysis. J Biol Chem 2003;278:7108-7118.
    [10] Qiang H, Lin Y, Zhang X, et al. Differential expression genes analyzed by cDNA array in the regulation of rat hepatic fibrogenesis. Liver Int 2006;26:1126-1137.
    [11] Liu XJ, Yang L, Mao YQ, et al. Effects of the tyrosine kinase inhibitor genistein on the proliferation, activation of cultured rat hepatic stellate cells. World J Gastroenterol 2002;8:739-745. [12] Garcia-Ba(n|~)uelos J, Siller-Lopez F, Miranda A, et al. Cirrhotic rat livers with extensive fibrosis can be safely transduced with clinical-grade adenoviral vectors. Evidence of cirrhosis reversion. Gene Ther 2002;9:127-134.
    [13] Bedossa P, Paradis V. Liver extracellular matrix in health and disease. J Pathol 2003;200:504-515.
    [14]Zhang LJ,Chen YX,Chen ZX,et al.Effect of interleukin-10 and platelet- derived growth factor on expressions of matrix metalloproteinases-2 and tissue inhibitor of metalloproteinases-1 in rat fibrotic liver and cultured hepatic stellate cells.World J Gastroenterol 2004;10:2574-2579.
    [15]Rachfal AW,Brigstock DR.Connective tissue growth factor(CTGF/CCN2) in hepatic fibrosis.Hepatol Res 2003;26:1-9.
    [16]Gao R,Ball DK,Perbal B,Brigstock DR.Connective tissue growth factor induces c-fos gene activation and cell proliferation through p44/42 MAP kinase in primary rat hepatic stellate cells.J Hepato12004;40:431-438.
    [17]Borkham-Kamphorst E,Stoll D,Gressner AM,Weiskirchen R.Inhibitory effect of soluble PDGF-beta receptor in culture-activated hepatic stellate cells.Biochem Biophys Res Commun 2004;317:451-462.
    [1]Borkham-Kamphorst E,Stoll D,Gressner AM,et al.Antisense strategy against PDGF B-chain proves effective in preventing experimental liver fibrogenesis.Biochem Biophys Res Commun 2004;321:413-423.
    [2]Borkham-Kamphorst E,Herrmann J,Stoll D,et al.Dominant-negative soluble PDGF-beta receptor inhibits hepatic stellate cell activation and attenuates liver fibrosis.Lab Invest 2004;84:766-777.
    [3]Iwamoto H,Nakamuta M,Tada S,et al.Platelet-derived growth factor receptor tyrosine kinase inhibitor AG 1295 attenuates rat hepatic stellate cell growth.J Lab Clin Med 2000;135:406-412.
    [4]Di Sario A,Bendia E,Taffetani S,et al.Selective Na+/H+ exchange inhibition by cariporide reduces liver fibrosis in the rat.Hepatology 2003;37:256-266.
    [5]Chen YX,Lu CH,Xie WF,et al.Effects of ribozyme targeting platelet-derived growth factor receptor beta subunit gene on the proliferation and apoptosis of hepatic stellate cells in vitro.Chin Med J(Engl) 2005;118:982-988.
    [6]Akashi H,Matsumoto S,Taira K.Gene discovery by ribozyme and siRNA libraries.Nat Rev Mol Cell Bio12005;6:413-422.
    [7]Novina CD,Sharp PA.The RNAi revolution.Nature 2004;430:161-164.
    [8]Grimm D,Kay MA.Therapeutic short hairpin RNA expression in the liver:viral targets and vectors.Gene Ther 2006;13:563-575.
    [9]Lewis DL,Wolff JA.Delivery of SiRNA and SiRNA expression constructs to adult mammals by hydrodynamic intravascular injection.Methods Enzymol 2005;392:336-350.
    [10]Liu F,Song Y,Liu D.Hydrodynamics-based transfection in animals by systemic administration of plasmid DNA.Gene Ther 1999;6:1258-1266.
    [11]Maruyama H,Higuchi N,Nishikawa Y,et al.High-level expression of naked DNA delivered to rat liver via tail vein injection.J Gene Med 2002;4:333-341.
    [12]Giladi H,Ketzinel-Gilad M,Rivkin L,et al.Small interfering RNA inhibits hepatitis B virus replication,in mice.Mol Ther 2003;8:769-776.
    [13]Zender L,Hutker S,Liedtke C,et al.Caspase 8 small interfering RNA prevents acute liver failure in mice.Proc Natl Acad Sci USA 2003;100:7797-7802.
    [14]Friedman SL.Seminars in medicine of the Beth Israel Hospital,Boston.The cellular basis of hepatic fibrosis.Mechanisms and treatment strategies.N Engl J Med 1993;328:1828-1835.
    [15]Qi Z,Atsuchi N,Ooshima A,et al.Blockade of type beta transforming growth factor signaling prevents liver fibrosis and dysfunction in the rat. Proc Natl Acad Sci U S A 1999;96:2345-2349.
    [1]Yuantai W,Tiancai W,Qiu Z.PD98059 inhibits expression of pERK1 protein and collagen alphal(I)mRNA in rat pancreatic stellate cells activated by platelet-derived growth factor.Indian J Gastroenterol 2005;24:100-103.
    [2]Eitner F,Ostendorf T,Van Roeyen C,et al.Expression of a novel PDGF isoforrn,PDGF-C,in normal and diseased rat kidney.J Am Soc Nephrol 2002;13:910-917.
    [3]Abdollahi A,Li M,Ping G,et al.Inhibition of platelet-derived growth factor signaling attenuates pulmonary fibrosis.J Exp Med 2005;201:925-935.
    [4]Zymek P,Bujak M,Chatila K,et al.The role of platelet-derived growth factor signaling in healing myocardial infarcts.J Am Coll Cardiol 2006;48:2315-2323.
    [5]Zhang Z,Chen J,Jin D.Platelet-derived growth factor(PDGF)-BB stimulates osteoclastic bone resorption directly:the role of receptor beta.Biochem Biophys Res Commun 1998;251:190-194.
    [6]Oreffo RO.Growth factors for skeletal reconstruction and fracture repair.Curr Opin Investig Drugs 2004;5:419-423.
    [7]Beljaars L,Meijer DK,Poelstra K.Targeting hepatic stellate cells for cell-specific treatment of liver fibrosis.Front Biosci 2002;7:e214-222.
    [8]Beljaars L,Molema G,Weert B,et al.Albumin modified with mannose 6-phosphate:A potential carrier for selective delivery of antifibrotic drugs to rat and human hepatic stellate cells.Hepatology 1999;29:1486-1493.
    [9]Beljaars L,Molema G,Schuppan D,et'al.Successful targeting to rat hepatic stellate cells using albumin modified with cyclic peptides that recognize the collagen type Ⅵ receptor.J Biol Chem 2000;275:12743-12751.
    [10]Beljaars L,Weert B,Geerts A,et al.The preferential homing of a platelet derived growth factor receptor-recognizing macromolecule to fibroblast-like cells in fibrotic tissue.Biochem Pharmacol 2003;66:1307-1317.
    [11]张其胜,John ML,张健,等.甘草次酸靶向肝星状细胞治疗肝纤维化的体内研究.中华肝脏病杂志 2005;13:664-667.
    [12]杜施霖,王吉耀,潘弘,等.含有RGD序列环肽介导的干扰素脂质体对大鼠肝纤维化的治疗作用.中华医学杂志 2005;85:1015-1020.
    [13]Elrick LJ,Leel V,Blaylock MG,et al.Generation of a monoclonal human single chain antibody fragment to hepatic stellate cells:a potential mechanism for targeting liver anti-fibrotic therapeutics.J Hepatol 2005;42:888-896.
    [14] Mizuguchi H, Hayakawa T. Targeted adenovirus vectors. Hum Gene Ther 2004;15: 1034-1044.
    [15] Slack JL, Liska DJ, Bornstein P. An upstream regulatory region mediates high-level, tissue-specific expression of the human alpha 1(I) collagen gene in transgenic mice. Mol Cell Biol 1991;11:2066-2074.
    [16] Brenner DA, Veloz L, Jaenisch R, et al. Stimulation of the collagen alpha 1 (I) endogenous gene and transgene in carbon tetrachloride-induced hepatic fibrosis. Hepatology 1993;17:287-292.
    [17] Yata Y, Scanga A, Gillan A, et al. DNase I-hypersensitive sites enhance alphal(I) collagen gene expression in hepatic stellate cells. Hepatology 2003;37:267-276.
    [18] Herrmann J, Arias M, Van De Leur E, et al. CSRP2, TIMP-1, and SM22alpha promoter fragments direct hepatic stellate cell-specific transgene expression in vitro, but not in vivo. Liver Int 2004;24:69-79.
    [19] Bahr MJ, Vincent KJ, Arthur MJ, et al. Control of the tissue inhibitor of metalloproteinases-1 promoter in culture-activated rat hepatic stellate cells: regulation by activator protein-1 DNA binding proteins. Hepatology 1999;29:839-848.
    [20] Inagaki Y, Kushida M, Higashi K, et al. Cell Type-Specific Intervention of Transforming Growth Factor/Smad Signaling Suppresses Collagen Gene Expression and Hepatic Fibrosis in Mice. Gastroenterology 2005; 129:259-268.
    [21] Lee Y, Kim M, Han J, et al. MicroRNA genes are transcribed by RNA polymerase Ⅱ. Embo J 2004;23:4051-4060.
    [22] Xia XG, Zhou H, Samper E, et al. Pol Ⅱ-expresed shRNA knocks down Sod2 gene expression and causes phenotypes of the gene knockout in mice. PLoS Genet 2006;2:e10.
    [23] Huang SL, Wu Y, Yu H, et al. Inhibition of Bcl-2 expression by a novel tumor-specific RNA interference system increases chemosensitivity to 5-fluorouracil in Hela cells. Acta Pharmacol Sin 2006;27:242-248.
    [24] Zeng Y, Wagner EJ, Cullen BR, et al. Both natural and designed microRNAs can inhibit the expression of cognate mRNAs when expressed in human cells. Mol Cell 2002;9:1327-1333.
    [25] Gard AL, White FP, Dutton GR. Extra-neural glial fibrillary acidic protein (GFAP) immunoreactivity in perisinusoidal stellate cells of rat liver. J Neuroimmunol 1985;8:359-375.
    [26] Neubauer K, Knittel T, Aurisch S, et al. Glial fibrillary acidic protein: a cell type specific marker for Ito cells in vivo and in vitro. J Hepatol 1996 ;24:719-730.
    [27] Cassiman D, Libbrecht L, Desmet V, et al. Hepatic stellate cell/myofibroblast subpopulations in fibrotic human and rat livers. J Hepatol 2002;36:200-209.
    [28] Chang D, Ramalho LN, Ramalho FS, et al. Hepatic stellate cells in human schistosomiasis mansoni: a comparative immunohistochemical study with liver cirrhosis. Ada Trop 2006;97:318-323.
    [29] Besnard F, Brenner M, Nakatani Y, et al. Multiple interacting sites regulate astrocyte-specific transcription of the human gene for glial fibrillary acidic protein. J BiolChem 1991;266:18877-18883.
    [30] Wang CY, Wang S. Astrocytic expression of transgene in the rat brain mediated by baculovirus vectors containing an astrocyte-specific promoter. Gene Ther 2006; 13: 1447-1456.
    [31] Maubach G, Lim MC, Zhang CY, et al. GFAP promoter directs lacZ expression specifically in a rat hepatic stellate cell line. World J Gastroenterol 2006;12:723-730.
    [1]Pankhurst QA,Connolly J,Jones SK,et al.Application of magnetic nanoparticles in biomedicine.J Phys D Appl Phys 2003;36:167-181.
    [2]Arias JL,Gallardo V,Gomez-Lopera SA,et al.Synthesis and characterization of poly (ethyl-2-cyanoacrylate) nanoparticles with a magnetic core.J Control Release 2001;77:309-321.
    [3]Dobson J.Magnetic nanoparticles for drug delivery.Drug Dev Res 2006;67:55-60.
    [4]Hu FX,Neoh KG and Kang ET.Synthesis and in vitro anti-cancer evaluation of tamoxifen-loaded magnetite/PLLA composite nanoparticles.Biomaterials 2006;27:5725-5733.
    [5]Yang J,Park SB,Yoon HG,et al.Preparation of poly ε-caprolactone nanoparticles containing magnetite for magnetic drug carrier.Int J Pharm 2006;324:185-190.
    [6]Widder KJ,Senyel AE,and Scarpelli GD.Magnetic microspheres:a model system for site specific drug delivery in vivo.Proc Soc Exp Biol Med 1978;158:141-146.
    [7]Anwer K,Kao G,Proctor B,et al.Ultrasound enhancement of cationic lipid mediated gene transfer to primary tumors following systemic administration.Gene Ther 2000;7:1833-1839.
    [8]Lee RJ and Huang L.Folate-targeted,anionic liposome-entrapped polylysine- condensed DNA for tumor cell-specific gene transfer.J Biol Chem 1996;271:8481-8487.
    [9]Anderson WF.Human gene therapy.Nature 1998;392:25-30.
    [10]Pollok KE,Hanenberg H,Noblitt TW,et al.High-efficiency gene transfer into normal and adenosine deaminase-deficient T lymphocytes is mediated by transduction on recombinant fibronectin fragments.J Virol 1998;72:4882-4892.
    [11]Hou Z,Nguyen Q,Frenkel B,et al.Osteoblast-specific gene expression after transplantation of marrow cells:Implications for skeletal gene therapy.Proc Natl Acad Sci USA 1999;95:7294-7299.
    [12]Chow YH,Brodovich HO,Plumb J,et al.Development of an epithelium-specific expression cassette with human DNA regulatory elements for transgene expression in lung airways.Proc Natl Acad Sci USA 1997;94:14695-14700.
    [13]Binley K,Askham Z,Martin L,et al.Hypoxia-mediated tumour targeting.Gene Ther 2003;10:540-549.
    [14]Lewis DL,Wolff JA.Delivery of SiRNA and SiRNA expression constructs to adult mammals by hydrodynamic intravascular injection.Methods Enzymol 2005;392:336-350.
    [15]Liu F,Song Y,Liu D.Hydrodynamics-based transfection in animals by systemic administration of plasmid DNA.Gene Ther 1999;6:1258-1266.
    [16]Maruyama H,Higuchi N,Nishikawa Y,et al.High-level expression of naked DNA delivered to rat liver via tail vein injection.J Gene Med 2002;4:333-341.
    [17]Gérolami R,Cardoso J,Bralet MP,et al.Enhanced in vivo adenovirus-mediated gene transfer to rat hepatocarcinomas by selective administration into the hepatic artery.Gene Ther 1998;5:896-904.
    [18]Yamashita YI,Shimada M,Hasegawa H,et al.Electroporation-mediated interleukin-12 gene therapy for hepatocellular carcinoma in the mice model.Cancer Res 2001;61:1005-1012.
    [19]Wang JM,Xiao BL,Zheng JW,et al.Effect of targeted magnetic nanoparticles containing 5-FU on expression of bcl-2,bax and caspase 3 in nude mice with transplanted human liver cancer.World J Gastroenterol 2007;13:3171-3175.
    [20]龚连生,张阳德,周少波.磁性化疗纳米粒治疗大鼠移植性肝癌.中国现代医学杂志2001;11:14-16.
    [21]Matsunaga T,Suzuki T,Tanaka M,et al.Molecular analysis of magnetotactic bacteria and development of functional bacterial magnetic particles for nano- biotechnology.Trends Biotechnol 2007;25:182-188.
    [22]解宇,汪浩.磁性细菌的电磁诱导分离及人工培养.杭州师范学院学报(医学版)2007;27:217-221.
    [23]Sun J,Duan J,Dai S,et al.In vitro and in vivo antitumor effects of doxorubicin loaded with bacterial magnetosomes(DBMs) on H22 cells:The magnetic bio- nanoparticles as drug carriers.Cancer Lett 2007;258:109-117.
    [24]Takeyama H,Yamazawa A,Nakamura C,et al.Application of bacterial magnetic particles as novel DNA carriers for ballistic transformation of a marine cyanobacterium.Biotechnol Tech 1995;9:355-360.
    [25]Nakagawa T,Hashimoto R,Maruyama K,et al.Capture and release of DNA using aminosilane-modified bacterial magnetic particles for automated detection system of single nucleotide polymor-phisms.Biotechnol Bioeng 2006;94:862-868.
    [26]Ota H,Lim TK,Tanaka T,et al.Automated DNA extraction from genetically modified maize using aminosilane-modified bacterial magnetic particles.J Biotechnol 2006;125:361-368.
    [27]Sharon N and Lis H.Lectins as cell recognition molecules.Science 1989;246:227-234.
    [28]Han J,Il Yeom Y.Specific gene transfer mediated by galactosylated poly-L-lysine into hepatoma cells.Int J Pharm 2000;202:151-160.
    [29]郑连英,肖玉良.高性能壳聚糖介导基因载体.化学通报 2004;67:W44 1-6.
    [30]Jiang HL,Kwon JT,Kim YK,et al.Galactosylated chitosan-graft- polyethylenimine as a gene carrier for hepatocyte targeting.Gene Ther 2007;14:1389-1398.
    [31]Park JU,Ishihara T,Kano A,et al.Preparation of dendritic graft copolymer consisting of poly-(L-lysine) and arabinogalactan as a hepatocyte specific DNA carrier.Prep Biochem Biotechnol 1999;29:353-70.
    [32]Junbo H,Li Q,Zaide W,et al.Receptor-mediated interlukin-2 gene transfer into human hepatoma cells.Int J Mol Med 1999;3:601-608.
    [33]Ramani K,Hassan Q,Venkaiah B,et al.Site-specific gene delivery in vivo through engineered Sendai viral envelopes.Proc Natl Acad Sci USA 1998;95:11886-11890.
    [34]任常春,田培坤,曲淑敏,等.靶向与胰岛素样生长因子受体的基因导入系统.肿瘤2001;21:431-434。
    [35]Takahashi H,Ozturk M,Wilson B,et al.In vivo expression of two novel tumor-associated antigens and their use in immunolocalization of human hepatocellular carcinoma.Hepatology 1989;9:625-634.
    [36]Leohard M,Julia I S,Raymond H,et al.Targeted gene transfer to hepatocellular carcinoma cells in vitro using a novel monoclonal antibody based gene delivery system.Hepatology 1999;29:82-89.
    [37]Moradpour D,Compagnon B,Wilson BE,et al.Specific targeting of human hepatocellular carcinoma cells by immunoliposomes in vitro.Hepatology 1995;22:1527-1537.
    [38]Sokoloff AV,Wong SC,Ludtke JJ,et al.A new peptide ligand that targets particles and heteroiogous proteins to hepatocytes in vivo.Mol Ther 2003;8:867-872.
    [39]Wong SC,Wakefield D,Klein J,et al.Hepatocyte Targeting of Nucleic Acid Complexes and Liposomes by a T7 Phage p17 Peptide.Mol Pharmaceutics 2006;3:386-397.
    [40]Ruoslahti E.alpha-Fetoprotein in cancer and fetal development.Adv Cancer Res 1979;29:275-346.
    [41]Xu GW,Sun ZT,Forrester K,et al.Tissue-specific growth suppression and chemosensitivity promotion in human hepatocellular carcinoma cells by retroviral- mediated transfer of the wild-type p53gene.Hepatology 1996;24:1264-1268.
    [42]Ido A,Nakata K,Kato Y,et al.Gene therapy for hepatoma cells using a retrovirus vector carrying herpes simplex virus thymidine kinase gene under the control of human α-fetoprotein gene promoter.Cancer Res 1995;55:3105-3109.
    [43]Kanai F,Shiratori Y,Yoshida Y,et al.Gene therapy for alpha-fetoprotein- producing human hepatoma cells by adenovirus-mediated transfer of the herpes simplex virus thymidyne kinase gene.Hepatology 1996;23:1359-1368.
    [44]Su H,Lu R,Chang JC,et al.Tissue-specific expression of herpes simplex virus thymidine kinase gene delivered by adeno-associated virus inhibits thd growth of human hepatocellular carcinoma in athymic mice.Proc Natl Acad Sci USA 1997;94:13891-13896.
    [45]Vollmer CM,Eilber FC,Butterfield LH,et al.Alpha-fetoprotein-specific genetic immunotherapy for hepatocellular carcinoma.Cancer Res 1999;59:3064-3067.
    [46]Grimm CF,Ortmann D,Mohr L,et al.Mouse alpha-fetoprotein-specific DNA- based immunotherapy of hepatocellular carcinoma leads to tumor regression in mice.Gastroenterology 2000;119:1104-1112.
    [47]Gordon EM,Liu PX,Chert ZH,et al.Inhibition of metastatic tumor growth in nude mice by portal vein infusions of matrix-targeted retroviral vectors bearing a cytocidal cyclin G1 construct.Cancer Res 2000;60:3343-3347.
    [48]Beljaars L,Meijer DK,Poelstra K.Targeting hepatic stellate cells for cell-specific treatment of liver fibrosis.Front Biosci 2002;7:e214-222.
    [49]Beljaars L,Molema G,Weert B,et al.Albumin modified with mannose 6-phosphate:A potential carrier for selective delivery of antifibrotic drugs to rat and human hepatic stellate cells.Hepatology 1999;29:1486-1493.
    [50]张其胜,John ML,张健,等.甘草次酸靶向肝星状细胞治疗肝纤维化的体内研究.中华肝脏病杂志 2005;13:664-667.
    [51]Adrian JE,Poelstra K,Scherphof GL,et al.Interaction of targeted liposomes with primary cultured hepatic stellate cells:involvement of multiple receptor systems.J Hepatol 2006;44:560-567.
    [52]Beljaars L,Weert B,Geerts A,et al.The preferential homing of a platelet derived growth factor receptor-recognizing macromolecule to fibroblast-like cells in fibrotic tissue.Biochem Pharmacol 2003;66:1307-1317.
    [53]Beljaars L,Molema G,Schuppan D,et al.Successful targeting to rat hepatic stellate cells using albumin modified with cyclic peptides that recognize the collagen type Ⅵ receptor.J Biol Chem 2000;275:12743-12751.
    [54]杜施霖,王吉耀,潘弘,等.含有RGD序列环肽介导的干扰素脂质体对大鼠肝纤维化的治疗作用.中华医学杂志 2005;85:1015-1020.
    [55]Elrick LJ,Leel V,Blaylock MG,et al.Generation of a monoclonal human single chain antibody fragment to hepatic stellate cells:a potential mechanism for targeting liver anti-fibrotic therapeutics.J Hepatol 2005;42:888-896.
    [56]Herrmann J,Arias M,Van De Leur E,et al.CSRP2,TIMP-1,and SM22alpha promoter fragments direct hepatic stellate cell-specific transgene expression in vitro,but not in vivo.Liver Int 2004;24:69-79.
    [57]Brenner DA,Veloz L,Jaenisch R,et al.Stimulation of the collagen alpha 1(I) endogenous gene and transgene in carbon tetrachloride-induced hepatic fibrosis. Hepatology 1993;17:287-292.
    [58] Slack JL, Liska DJ, Bornstein P. An upstream regulatory region mediates high- level, tissue-specific expression of the human alpha 1(I) collagen gene in transgenic mice. Mol Cell Biol 1991;11:2066-2074.
    [59] Yata Y, Scanga A, Gillan A, et al. DNase I-hypersensitive sites enhance alphal(I) collagen gene expression in hepatic stellate cells. Hepatology 2003;37:267-276.
    [60] Inagaki Y, Kushida M, Higashi K, et al. Cell Type-specific intervention of transforming growth factor/smad signaling suppresses collagen gene expression and hepatic fibrosis in mice. Gastroenterology 2005;129:259-268.
    [61] Gard AL, White FP, Dutton GR. Extra-neural glial fibrillary acidic protein (GFAP) immunoreactivity in perisinusoidal stellate cells of rat liver. J Neuroimmunol 1985;8:359-375.
    [62] Neubauer K, Knittel T, Aurisch S, et al. Glial fibrillary acidic protein: a cell type specific marker for Ito cells in vivo and in vitro. J Hepatol 1996 ;24:719-730.
    [63] Cassiman D, Libbrecht L, Desmet V, et al. Hepatic stellate cell/myofibroblast subpopulations in fibrotic human and rat livers. J Hepatol 2002;36:200-209.
    [64] Chang D, Ramalho LN, Ramalho FS, et al. Hepatic stellate cells in human schistosomiasis mansoni: a comparative immunohistochemical study with liver cirrhosis. Acta Trop 2006;97:318-323.
    [65] Besnard F, Brenner M, Nakatani Y, et al. Multiple interacting sites regulate astrocyte-specific transcription of the human gene for glial fibrillary acidic protein. J Biol Chem 1991 ;266:18877-18883.
    [66] Wang CY, Wang S. Astrocytic expression of transgene in the rat brain mediated by baculovirus vectors containing an astrocyte-specific promoter. Gene Ther 2006; 13: 1447-1456.
    [67] Maubach G, Lim MC, Zhang CY, et al. GFAP promoter directs lacZ expression specifically in a rat hepatic stellate cell line. World J Gastroenterol 2006;12:723-730.
    [68] Miller N, Vile RG. Targeted vectors for gene therapy. FASEB J 1995;9:190-199.

© 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