Tec激酶区作用蛋白RAI 16的功能和分化调控机理初步研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
背景和目的:肿瘤是一种多基因、多步骤、多因素、多网络的病理机制复杂的疾病,细胞分化障碍是肿瘤形成的本质,利用某些化合物诱导肿瘤细胞分化为正常细胞或近似正常细胞的肿瘤的诱导分化治疗是继手术、放疗、化疗后的新的治疗模式,已成为研究的“热点”。
     Tec(tyrosine kinase expressed in hepatocellular carcinoma)是一种存在于胞质内的非受体型蛋白酪氨酸激酶,是肝干细胞和肝细胞增殖、分化、凋亡信号转导途径中关键的蛋白激酶连接分子。我们在前期工作中以Tec激酶结构域(KD)作为“钓饵”蛋白,利用酵母双杂交技术筛选构建于转录激活结构域(AD)载体的人胎肝cDNA文库,经营养缺陷选择、诱导筛选及初步鉴定,发现并确证了一个新的阳性克隆。筛库所得基因片段经测序和BLAST比对分析,无同源性序列,确定为RAI16(Homo sapiens retinoic acid induced 16)基因,已被GeneBank收录(编号为BC052237),组织来源为脑Ⅳ型星状细胞瘤,其编码蛋白为人视黄酸诱导蛋白16。进而以RAI16蛋白为切入点,利用相关的生物信息学站点及计算机软件,对RAI16进行生物信息学分析,从中获得其编码蛋白质结构方面的重要信息,从而根据结构预测其功能。生物信息学分析提示,RAI16蛋白含6个PKC磷酸化结合位点,8个CK2磷酸化结合位点,3个酪氨酸(Tyr)磷酸化结合位点, 5个豆蔻酰化位点,2个酰胺化位点,1个ATP/GTP-结合位点、无跨膜区结构域,具有一段由31个氨基酸组成的信号肽,且其切割位点位于30~31aa之间,因此推测RAI16蛋白可能为RA信号通路中一类胞浆内重要的“新”信号蛋白分子。
     关于RAI16文献报道很少,功能研究报告更是空白,从其命名知晓,为一种RA诱导蛋白。由于诱导分化剂相关的信号转导过程是揭示其作用机理的关键,结合RA在肿瘤细胞分化中的突出作用和RAI16蛋白的功能的“未知性”,我们选择肝癌细胞为对象,将课题切入点定位于“Tec作用蛋白RAI16的新功能和分化调控机理研究”上,以酵母双杂交系统筛选中Tec酪氨酸激酶与RAI16蛋白相互作用的现象为线索,从肝癌细胞诱导分化和胞内信号转导途径的角度,通过抗体制备、真核表达载体和腺病毒载体的构建及表达检测、免疫组化、激光共聚焦分析、Northern blotting、Western blotting、MTT、流式细胞仪(FCM)检测、划痕实验、Transwell细胞迁移和Metrigel细胞侵袭实验等多种方法明确RAI16的表达谱和亚细胞定位,分析RAI16在肝再生、肝癌形成和ATRA对肝癌细胞诱导分化过程中的作用,探讨RAI16对肝癌细胞诱导分化、细胞周期、细胞增殖与凋亡、迁移与侵袭力等多种生物学行为的影响,探索RAI16蛋白“新”功能,并进一步认识和阐明RAI16在RA诱导肝癌细胞分化的信号转导途径中的关键作用,本研究将为肿瘤细胞诱导分化信号转导分子机理的研究提供新思路和理论依据,为肝癌分子治疗提供新的作用靶点,为进一步研究其作用机制奠定基础。
     研究内容和方法
     1.用纯化的RAI16蛋白合成肽免疫新西兰大白兔,采用一种快速免疫法制备RAI16多克隆抗体,ELISA追踪检测抗体效价,Protein A亲和层析法纯化特异性IgG抗体,Western blot和免疫组化检测抗体的特性,初步检测RAI16在大鼠再生肝组织和肝癌组织中的表达,为进一步研究RAI16的功能提供抗体。
     2.以ATRA诱导人肝癌HepG2细胞后,流式细胞仪检测其对细胞周期、细胞增殖与凋亡的影响;提取总RNA和总蛋白,采用RT-PCR和Western blot检测ATRA诱导后RAI16的表达情况,分析ATRA与RAI16在细胞诱导分化中的关系,为进一步探讨RAI16在诱导分化中的作用机理提供理论基础。
     3.以真核表达质粒pEGFP-C1为载体,从含有全长RAI16基因片段的质粒克隆模板中,利用PCR方法钓取目的基因,将目的基因与目的载体用EcoRI和XhoI分别进行酶切。纯化酶切产物后进行定向连接,构建pEGFP-C1-RAI16,经酶切及测序鉴定。采用激光共聚焦显微镜观察转染pEGFP-C1-RAI16后的亚细胞定位;Western blot分析检测其对HepG2细胞RAI16蛋白水平和诱导分化作用;利用MTT实验检测pEGFP-C1-RAI16质粒转染后对细胞增殖的影响。
     4.以真核表达质粒pDC315-EGFP为载体,从含有全长RAI16基因片段的质粒克隆模板中,利用PCR方法钓取RAI16基因,将其与真核表达载体pDC315-EGFP分别以EcoR I进行酶切。纯化酶切产物后进行定向连接,构建穿梭质粒pDC315-EGFP-RAI16。经测序鉴定后,利用AdMax腺病毒包装系统构建重组腺病毒Ad-RAI16,并进行包装、扩增、纯化和滴度测定;采用激光共聚焦显微镜观察感染Ad-RAI16后的亚细胞定位;通过Western blotting、MTT、流式细胞仪(FCM)检测、划痕实验、Transwell细胞迁移和Metrigel细胞侵袭实验等多种方法分析RAI16对肝癌细胞诱导分化、细胞周期、细胞增殖与凋亡、迁移与侵袭力等多种生物学行为的影响,进一步认识和阐明RAI16在RA诱导肝癌细胞分化的信号转导途径中的关键作用。
     研究结果
     1.成功制备了兔抗人RAI16多克隆抗体。ELISA检测显示该抗体的效价达到1:125 000,相对亲和力常数为10-5~10-6 M-1。Western blot和免疫组化提示该抗体能与RAI16蛋白特异性结合。RAI16在肝、心脏、食管、胃、脑、胰腺等多种组织中均有表达,而且在肝脏再生和肝癌形成过程中其表达呈上调趋势。
     2. ATRA诱导后RAI16的表达明显上调,并且呈时效性关系;MTT检测结果显示:ATRA能够抑制HepG2在体外的增殖,与对照组比较,诱导组细胞生长均2d开始明显受抑(P<0.05),组内不同处理天数之间有显著性差异(P<0.05)。流式细胞仪检测显示:ATRA能够促进肝癌细胞分化和凋亡,抑制细胞DNA合成,使其停滞于G1期。
     3. RAI16真核表达载体的构建及其生物学效应:经测序分析表明成功构建重组表达载体pEGFP-C1-RAI16。转染HepG2细胞发现RAI16主要定位于胞质内内质网等细胞器中,且能显著抑制细胞增殖和分化分子AFP、γ-GT的表达。
     4. RAI16腺病毒表达载体的构建及其生物学效应:经测序分析质粒表达检测表明成功构建重组表达腺病毒Ad-RAI16。感染HepG2细胞发现RAI16主要定位于胞质内内质网等细胞器中;能显著抑制细胞增殖、抑制分化分子AFP、γ-GT的表达和Wnt通路信号传导分子β-Catenin的表达;抑制细胞迁移,削弱细胞侵袭能力。
     结论
     1.成功制备兔抗人RAI16多克隆抗体并进行了特异性亲和纯化,抗体效价和纯度良好;Western blot和免疫组化显示制备抗体特异性良好。RAI16在多种组织中表达,并参与了肝再生和肝癌形成过程的调控。
     2. ATRA能够抑制肝癌细胞增殖,促进其分化和凋亡;能显著增强RAI16的表达。RAI16可能在ATRA的诱导分化过程发挥重要作用。
     3.成功构建了真核表达载体pEGFP-C1-RAI16,RAI16蛋白定位于胞质内内质网等细胞器中;并具有抑制肝癌细胞增殖和促进分化的作用。
     4.成功构建重组腺病毒表达载体Ad-RAI16。进一步研究表明RAI16能够抑制肝癌细胞增殖、侵袭和迁移,促进其分化和凋亡;可直接通过调控Wnt信号转导通路参与肝癌细胞的诱导分化。
Background and objective
     Tumor is a diseas with polygene, multistep, multifactor, many networks and complex pathomechanism. Cell dysdifferentiation is the nature of tumorigeness. The induced differentiation therapy for tumor, which can make tumor cells become mormal cells or similar to the normal cells by using some compounds, is a new therapy pattern following the surgery therapy, radiotherapy and chemotherapy and become the focus of research.
     Tec is a kind of non-receptor tyrosine kinase which is a crucial joint molecule in signal transduction pathway of liver stem cells and liver cell proliferation, differentiation and apoptosis. We use Tec kinase domain as the bait protein in our preliminary work and take advantage of the technique of yeast two-hybrid system to filter the human foetus liver cDNA library that was built from transcription activation domain vector, and through auxotrophic selecting , inducing filtering and preliminary identification, we found and confirmed a new positive clone. After sequencing and making BLAST comparison analysis on the gene fragment got from the filtering result of gene library in this study, we were sure that it was the RAI16 (Homo sapiens retinoic acid induced 16 gene), which has no homology sequence and has been embodied by Genebank and the number is BC052237, and the source of tissue is Brain astrocytoma (grade IV). Its coding protein is the Homo sapiens Retinoic Acid induced protein 16. Afterwards we looked on RAI16 as the cut through point. We mainly used related computer softwares and some bioinformatics websites to analyze RAI16,from which we obtained the significant information of RAI16 protein structure and thus we could predict its unknown new functions according to its structure. The analyses of bioinformatics shows that RAI16 protein contains six protein kinase C phosphorylation sites, nine casein kinase II phosphorylation sites, three tyrosine kinase phosphorylation sites, six N-myristoylation sites, two amidation sites, three tyrosine sulfation sites and one ATP/GTP-binding site. Furthermore, this protein has no transmembrane domain, which contains a section of signal peptide consisted of 31 amino acid and the cleavage site is located between 30-31aa. So we predict it to be a kind of important new decapentaplegic molecule of RA signal path in intracytoplasm.
     At present there is a few of documents reported about RAI16 and its function study is vacuity. While learning from its name it is a kind of inducing protein of RA. Because signal transduction process is the key to reveal its mechanism of action and thinking of its unknowning function and the outstanding effection of RA for cancer cells in induced differentiation, we choose HCC cells which were induced to differentiate by RA or RAI16 to act as our study object and we looked on‘Preliminary Research of function and differentiation regulating mechanism of RAI16 Protein Interacting withTec Tyrosine Kinase’as the breakthrough point. Herein, from the induced differentiation of HCC cells and intracellular signal transduction path, we looked on phenonmenon that RAI16 inacted with Tec in yeast two-hybrid system as clue to identify expression spectrum and subcellular localization of RAI16, to analyze its function in the process of liver regeneration, hepatocarcinogenesis and HCC differentiation induced by ATRA, to investigate RAI16 effect for many kind of biological behavior of HCC cells, such as cell differentiation, cell cycle, cell proliferation or apoptosis , cell migration or invasiveness and etc, and explored the new function by antibody preparation, the construction of eukaryotic expression vector and adenovirus vector, immunohistochemisty, laser confocal microscop, immunoblotting, MTT, FCM, and so forth, which can make us to learn and interpret its crucial role in signal transduction pathway that RA induced HCC cells to differentiate. This study will provide new idea and theoretical evidences for the molecule mechanism of tumorous cell induced differentiation and signal transduction, and provide a new target for molecule targeted therapy of HCC, which can make the found base for the next step of the function mechanism study of RAI16.
     Materials and Methods
     1. Rabbit anti-human RAI16 polyclonal antibody was prepared by using its protein synthetic peptide and a modified rapid immune procedure followed by purification of specific avidity column. The efficacy of this polyclonal antibody was certificated by ELISA, Western blot and immunohistochemistry. The expression of RAI16 was preliminarily detected in regenerating liver tissue and cancer tissue of liver. This can provide antibody for the next step of the function study of RAI16.
     2. To detect the influence of RAI16 for cell cycle, cell proliferation and apoptosis by flow cytometer after HepG2 cells were induced by ATRA, then extracted total RNA and total protein to detect the expression of RAI16 by RT-PCR and Western blot and analyze the relation of ATRA with RAI16 in inducing differentiation, which can provide the theoretical base for the next step of the function mechanism study of RAI16 during the induced differentiation process.
     3. First,the RAI16 genes was amplified by PCR from plasmid cloning templates which contains total length gene fragment of RAI16 then inserted into pEGFP-C1 between EcoRI and XhoI sites. Then, the plasmid vector was sequenced and named as pEGFP-C1-RAI16. Laser confocal microscopy was used to observe the subcellular localization of recombinant RAI16. Western blot was applied to measure the increase level of RAI16 protein and induced differentiation function. Subsequently, cellular survival and proliferation was assayed by MTT.
     4. First,the RAI16 genes was amplified by PCR from plasmid cloning templates which contains total length gene fragment of RAI16 then inserted into pDC315-EGFP at the side of EcoRI sites. The plasmid vector was sequenced and named as pDC315-EGFP-RAI16. Second, AdMax system was used to construct the Ad-RAI16 and to enclose, amplify, depurate and detect titer.Then laser confocal microscopy was used to observe the subcellular localization of recombinant RAI16. Western blot was applied to measure the increase level of RAI16 protein and induced differentiation function. Subsequently, cellular survival and proliferation was assayed by MTT, cellular cycle and apoptosis was analyzed by FCM and the effect for cell biological behaviour was investigated by Transwell cell migration experiment and Metrigel invasion experiment.
     Results
     1. Rabbit anti-human RAI16 polyclonal antibody has been preparated successfully: The titer of the antibody obtained in this experiment determined by ELISA was up to 1∶125 000.The kaff value of the antibody was 10-5~10-6 M-1. Western blot results showed the antibody has high specificity to RAI16 protein. RAI16 can express in many tissues, such as liver, heart, esophagus, stomach, brain, pancreatic and so forth, and its expression was up-regulated during the liver regeneration and hepatocarcinogenesis process.
     2. The induced effect of ATRA: That the expression of RAI16 was up-regulated after induced by ATRA was showed by Western blot. MTT results show that ATRA can inhibition HepG2 cells to proliferate in vitro, cell growth in induced group begin to be restrained obviously after two days comparing with control(P<0.05)and there are significant difference(P<0.05)in different phase after treating. FCM shows that ATRA can promote HCC cells to differentiate and apoptosis, suppress DNA synthesis and make them stagnate in G1 period.
     3. Construction and biological effects of plasmid vector overexpressing RAI16 specifically in eukaryotic: Sequencing analysis show that pEGFP-C1-RAI16 was constructed successfully. After transfected into HepG2 RAI16 was found to mainly localizate in endoplasmic reticulum and intracytoplasm and to significantly suppress cell proliferation and the expression of AFP andγ-GT, which are differentiation molecules.
     4. Construction and biological effects of adenovirus plasmid vector overexpressing RAI16: Sequencing analysis show that Ad-RAI16 was constructed successfully. After infected HepG2 RAI16 was found to mainly localizate in endoplasmic reticulum and intracytoplasm and to significantly suppress cell proliferation and the expression of AFP andγ-GT, which are differentiation molecules, to inhibite cell migration and to weaken invasing ability of tumorous cells.
     Conclusion
     1. Rabbit anti-human RAI16 polyclonal antibody, with high titer and specificity, was successfully produced by a modified rapid immune procedure. This polyclonal antibody can be further applied in ELISA, Western blot and immunohistochemistry to elucidate the roles of RAI16 protein played in many important cellular procedures. The result of initiatory experiment shows that RAI16 genes express in many tissues and regulate liver regeneration and hepatocarcinogenesis.
     2. The proliferation of HepG2 was significantly inhibited,the differentiation and apoptosis was induced and the expression of RAI16 was obviously up-regulated by ATRA. RAI16 may play important role in the process of ATRA induced differentiation.
     3. The eukaryotic overexpressing vector pEGFP-C1-RAI16 was successfully constructed, which may satisfy experiment demand. RAI16 proteins localizate in endoplasmic reticulum and intracytoplasm,which can inhibit the proliferation of HCC cells and make them differentiate.
     4. The recombinant adenovirus overexpressing vectorwas successfully constructed. It was proved that RAI16 can inhibit proliferation, invasion and migration of HCC cells, promote cancer cells to differentiate and apoptosis and play induced differentiating function in HCC through regulating Wnt signal transduction path.
引文
1.王阁,邓婧,杨进,等.视黄酸诱导蛋白16蛋白质结构功能分析[J].世界华人消化杂志,2007, 15(12): 1342-1346.
    2. Li Q, Wang G, Shan JL, et al. MicroRNA-224 is upregulated in HepG2 cells and involved in cellular migration and invasion [J].J Gastroenterol Hepatol. (2009)Sep 27, 1-8.
    3. Zhang ZM, Wang G, Chen C, et al. Rapid induction of PC3/BTG2 gene by hepatopoietin or partial hepatectomy and its mRNA expression in hepatocellular carcinoma [J].Hepatobiliary Pancreat Dis Int, 2009,8(3):288-293
    4. Jijun Zheng, Chuan Chen, Ge Wang, Jing Deng, et al. Effect of MEK1 inhibitor PD98059 on the signal transduction of Tec in hepatoma cells [J]. Chinese-German Journal of Clinical Oncology, 2007, 6(6):542-545.
    5. Chen Chuan, Wang Ge, Suo Jin-you, et al. Hepatic stem cells in different histopathologic types of primary hepatic carcinoma [J].Journal of Clinical Rehabilitative Tissue Engineering Research, 2008, 12, (3):573-577.
    6.张志敏,王阁,杨志祥等,二乙基亚硝胺诱发大鼠肝癌过程中PC3/BTG2基因表达的变化[J].中华肝脏病杂志, 2009,17, (2):107-111.
    7.李琼,王阁,张志敏等,微RNA18与B细胞易位基因在肝癌细胞中差异表达相关性的研究[J].中华肝脏病杂志,2009,17(1):43-36.
    8.许文,王阁等。索拉菲尼诱导大鼠肝细胞癌形成过程中巨噬细胞和自然杀伤细胞的聚集。第三军医大学学报,2009, 31(11):1009-1012
    9.李琼,王阁,单锦露等,MicroRNA 224在HepG2细胞中高表达及其靶基因的生物学预测[J],中华消化外科杂志,2008,7(4):297-299.
    10.李琼,王阁,杨志祥等,PAK4在肝细胞肝癌组织中的表达及其临床意义[J].临床肿瘤学杂志,2008,13(11):961-964.
    11.许文,王阁,邓婧等,Tec激酶区作用蛋白RAI16的原核表达和纯化.世界华人消化杂志[J],2008,16(12):1350-1354.
    12.许文,王阁,杨志祥等,APE1在DEN诱导大鼠肝细胞癌形成过程中作用的研究[J].肿瘤预防与治疗,2008,21(2):127-129.
    13.陈川,王阁,张志敏等,原发性肝癌中Tec表达及磷酸化水平的研究[J].中华肝脏病杂志,2007, 15(12):910-913.
    14.李琼,王阁,王红中等,MicroRNA在HepG2肝癌细胞表达差异谱的研究[J].重庆医学,2007,36(20 ),2024-2025.
    15.张志敏,王阁,陈川等,改进性DEN诱发大鼠肝癌模型的建立与病理形态学研究[J].第三军医大学学报,2007, 29(2):176-17 8.
    16.张志敏,王阁,陈川等,间歇给药对DEN诱发大鼠肝癌模型的影响研究[J].重庆医学,2007,36(20):2035-2037.
    17.陈川,王阁,郑继军等,PP2对人肝癌细胞HepG2中Tec信号转导作用研究[J].重庆医学,2007,36(20):2032-2034.
    18. Higgins GM, Anderson R M. Experimental pathology of the liver : regeneration of the liver by the white rat following partial surgical removal [J ] .Arch Pathol ,1931 ,12 :186-202.
    19. Chomczynski P, Sacchi N. Single step method of RNA isolation by acid guanidinium thiocyanate-phenol-chlorform extraction [J]. Anal Biochem, 1987 ,162(1):156-159.
    20. Liu ZH, Li MY, Cui DF. A novel method for polypeptide design to prepare specific antibody of the peptide and applied to immunoassay[J]. J Immunol Methods. 2003 ,281(1-2):17-25
    21. Merson RR, Franks DG, Karchner SI,et al.Development and characterization of polyclonal antibodies against the aryl hydrocarbon receptor protein family (AHR1, AHR2, and AHR repressor)[J]. Comp Biochem Physiol C Toxicol Pharmacol. 2006,142 (1-2):85-94.
    22. Matsumoto K, Torimaru A,Ishitobi S.Preparation and characterization of a polyclonal antibody from rabbit for detection of trinitrotoluene by a surface plasmon resonance biosensor[J]. Talanta, 2005,68(2): 305-311.
    23. Wang ST, Gui WJ, Guo YR, et al.Preparation of a multi-hapten antigen and broad specificity polyclonal antibodies for a multiple pesticide immunoassay[J]. Anal Chim Acta. 2007,587(2):287-292.
    24. Cao Z, Zhang J, Li Y,et al.Preparation of polyclonal antibody speciWc for AtPLC4, an Arabidopsis phosphatidylinositol-speciWc phospholipase C in rabbits[J].Protein Expr Purif. 2007,52(2):306-312.
    25. Freemantle S J,Dragnev K H,Dmitrovsky E.The Retinoic Acid Paradox inCancer Chemoprevention.J Natl Cancer Inst,2006,98(7):426-427.
    26.董倩,姜达.实体瘤诱导分化治疗研究现状.肿瘤学杂志,2004,10(6):443-446.
    27. Estey E,Koller C,Tsimberidou A M,et al.Potential curability of newlydiagnosed acutepromyelocytic leukemia without use of chemotherapy:Theexample of liposomal all-trans retinoic acid [J].Blood,2005,105(3):1366-1367.
    28. Testi A M,Biondi A,Coco F L,et al.GIMEMA-AIEOP AIDA protocol for thetreatment of newly diagnosed acute promyelocytic leukemia(APL)inchildren[J].Blood,2005,106(2):447-453.
    29. Chung J,Liu C,Smith D E,et al.Restoration of retinoic acid concentrationsuppresses ethanol-enhanced c-Jun expression and hepatocyte proliferation in ratliver [J].Carcinogenesis,2001,22(8):1213-1219.
    30. Rexer B N,Zheng W L,Ong D E.Retinoic acid biosynthesis by normal humanbreast epithelium is via aldehyde dehydrogenase 6,absent in MCF-7cells[J].CancerRes,2001,61(19):7065-7070.
    31.高山,雷道雄,艾中立.肝癌细胞分化诱导作用的研究进展[J].临床外科杂志,2004,12(9):571-573.
    32.张强,范清宇,张殿忠等.全反式维甲酸对人骨肉瘤细胞抑制增殖和诱导分化的实验[J].国临床康复,2005,9(22):150-151.
    33. Zhou HG , Gu GW . Retinoids preventing liver cancer [J] . Shijie Huaren XiaohuaZazhi(World Chin J Digestol),1999,7(1):82-83.
    34. Sin XY,Chen BL,Wang TD.Expression of gap junction protein connexin 43 inHela cell line and its effect on gap junctional intercellular communication[J].Di-siunyi Daxue Xuebao(J Fourth Mil Med Univ),2000,21(3):369-371.
    35.王振义,孙关林.诱导分化疗法应用的现状[J].中华血液学杂志,1994,15(2):105-7.
    36.张雯杰,郑容,林琳等.维甲酸诱导分化在分化型甲状腺癌治疗中的初步应用[J].实用癌症杂志,2005,20(4):396-401.
    37.罗惠霞,王玉炯.组蛋白乙酰化/去乙酰化在真核基因转录调控中的作用[J],中国生物工程杂志,2004,24(2):19-21.
    38.张世国,于雪艳,孔立新,等.9-顺维甲酸对肺鳞癌细胞生长、分化及凋亡的研究[J].解放军医学杂志,2003,28(1):73-75.
    39. Daniele S,Manlio T.Retinoids,Apoptosis and Cancer [J].Current Pharmaceutical Design,2001,7(17):1823-1837.
    40. Yukari M , Akimori W, Kimie N , et al . Antitumoral activity of 13-demethyl or13-substituted analogues of all-trans retinoic acid and 9-cis retinoic acid in the human myeloid leukemia cell line HL-60[J].Biol Pharm Bull,2006,29(9):1803-1809.
    41. Boehm M F,Zhang L,Zhi L,et al.Design and synthesis of potent retinoid X receptor selective ligands that induce apoptosis in leukemia cells[J].J Med Chem,1995,38(16):3146-3155.
    42. Deigner H P,Kinscherf R.Modulating apoptosis:current applications and prospects for future drug development[J].Curr Med Chem,1999,6:399-414.
    43. Pettersson F,Couture M C,Hanna N,et al.Enhanced retinoid-induced apoptosis of MDA-MB-231 breast cancer cells by PKC inhibitors involves activation of ERK [J].Oncogene,2004,23(42):7053-7066.
    44.张传海,许戈良,葛勇胜,等.亚砷酸对人肝癌细胞生长增殖及分化的影响[J].安徽医科大学学报,2005,40(1):10-13.
    45.刘秀英,魏让,阎建文,等.血清酶活性联检在恶性肿瘤诊断中的应用[J].实用医技,2001,8(6):415-416.
    46.张敬一,许顺江,杨慧敏,等.刺五加抗癌作用的实验研究[J].实用肿瘤学杂志,2004,18(6):422-427.
    47.曾小莉,涂植光.人参总皂甙对人肝癌细胞胞浆中某些表型的逆转作用[J].癌症,2000,19(8):776-778.
    48.彭安,陈敏珍,申东兰.唑来膦酸诱导Bel-7402人肝癌细胞分化的研究[J].中国基层医药,2006,13(3):447-449.
    49. Magee TR,Cai Y,El-Houseini ME,et al.Retinoic acid mediates down-regulationof the alpha-fetoprotein gene through decreased expression of hepatocyte nuclearfactors[J].J Bio Chem,1998,273(45):30024-30032.
    50. Wang XW, Xu B.Several new targets of antitumor agents[J].Acta pharmacol Sin,2000,21(5):450-454.
    51.姜圣亮,谭江平,石林祥,等.MTT法检测肝癌细胞化疗敏感性方法学探讨[J].同济大学学报(医学版),2002,23(4):296-298.
    52.邓婧,王阁,杨进等,Tec激酶区作用蛋白RA I16的筛选和结构功能分析[J].第三军医大学学报,2006,28(2):133-135.
    53. Nakai K, Horton P. PSORT: a program for detecting sorting signals in proteins andpredicting their subcellular localization [J]. Trends in Biochemical Sciences, 1999, 24(1):34-35
    54. Peuvel I, Peyret P, Metenier G, et al. The microsporidian polar tube: evidence for a third polar tube protein (PTP3) in Encephalitozoon cuniculi [J]. Molecular and Biochemical Parasitology, 2002, 122(1):69-80
    55. Chen J, Lu Y, Xu J. Identification and characterization of NBEAL1, a novel human neurobeachin-like 1 protein gene from fetal brain, which is up regulated in glioma [J]. Molecular Brain Research, 2004, 125(1-2):147-155
    56. Guo J, Jin G, Meng L, et al. Subcellullar localization of tumor-associated antigen 3H11Ag [J]. Biochemical and Biophysical Research Communications, 2004, 324 (2):922-930
    57. Chalfie M, Tu Y, Euskirchen G, et al. Green fluorescent protein as marker for gene express [J ] . Science, 1994, 263 (5148):802 - 805.
    58. Inouye S, Tsuji FI. Aequorea green fluorescent protein. Expression of the gene and fluorescence characteristics of the recombinant protein [J]. FEBS Letters, 1994, 341 (2-3): 277 - 280.
    59. Valdez BC, Perlaky L, Cai ZJ, et al. Green fluorescent protein tag for studies of drug induced translocation of nucleolar protein RH - IIPGu [J]. Biotechniques, 1998, 24 (6): 1032 - 1036.
    60. Nikles D, Vana K, Gauczynski S, et al. Subcellular localization of prion proteins and the
    37 kD/67 kD laminin receptor fused to fluorescent proteins [J]. Biochim Biophys Acta. 2008, 1782(5):335-340.
    61. Hong SY, Linz JE. Functional expression and subcellular localization of the aflatoxin pathway enzyme Ver-1 fused to enhanced green fluorescent protein [J]. Appl Environ Microbiol. 2008, 74(20):6385-6396.
    62. Altucci L, Leibowitz MD, Ogilvie KM, de Lera AR, Gronemeyer H. RAR and RXR modulation in cancer and metabolic disease [J]. Nat Rev Drug Discov. 2007 ,6(10):793-810.
    63. Wolf G. Retinoic acid as cause of cell proliferation or cell growth inhibition depending on activation of one of two different nuclear receptors [J]. Nutr Rev. 2008, 66(1):55-59.
    64. O'Reilly K, Bailey SJ, Lane MA. Retinoid-mediated regulation of mood: possible cellular mechanisms [J]. Exp Biol Med (Maywood). 2008, 233(3):251-258.
    65. Tao QG, Cheng YJ, Clifford J, et al. Characterization of the murine orphan G-protein-coupled receptor gene Rai3 and its regulation by retinoic acid[J]. Genomics, 2004, 83(2):270-280.
    66. Elsea SH, Girirajan S. Smith-Magenis syndrome [J]. Eur J Hum Genet. 2008, 16(4):412-421.
    67. Walpole SM, Hiriyana KT, Nicolaou A, et al.Identification and characterization of the human homologue (RAI2) of a mouse retinoic acid-induced gene in Xp22[J]. Genomics. 1999, 55(3):275-283.
    68. Nagahata T, Sato T, Tomura A, et al.Identification of RAI3 as a therapeutic target for breast cancer[J]. Endocr Relat Cancer. 2005, 12(1):65-73.
    69. Vendrov AE, Madamanchi NR, Hakim ZS, et al. Thrombin and NAD(P)H oxidase-mediated regulation of CD44 and BMP4-Id pathway in VSMC, restenosis, and atherosclerosis[J]. Circ Res. 2006, 98(10):1254-1263.
    70. Kuhl A, Melberg A, Meinl E, et al. Myofibrillar myopathy with arrhythmogenic right ventricular cardiomyopathy 7: corroboration and narrowing of the critical region on 10q22.3 [J].Eur J Hum Genet. 2008, 16(3):367-373.
    71.雒喜忠,王阁,郑继军等,RAI16基因真核表达载体的构建及在HepG2细胞中的表达[J].第三军医大学学报.2009,31(17):1620-1624.
    72.雒喜忠,王阁,许文等,RAI16蛋白合成肽多克隆抗体的制备及初步应用[J].细胞与分子免疫学杂志.2008,24(9):898-901.
    73. Amarzguioui M, Rossi JJ, Kim D. Approaches for chemically synthesized siRNA and vector-mediated RNAi [J]. FEBS Lett, 2005, 579(26): 5974-5981.
    74. Darling AJ, Boose JA, Spaltro J. Virus assay methods: accuracy and validation. Biologicals [J]. 1998, 26(2):105-109.
    75. Cross D, Burmester JK. Gene therapy for cancer treatment: past, present and future [J]. Clin Med Res, 2006, 4(3): 218-227.
    76. Peng Z. Current status of gendicine in China: recombinant human Ad-p53 agent for treatment of cancers [J]. Hum Gene Ther, 2005, 16(9): 1016-1027.
    77. Wong D, DeMott MS, Demple B. Modulation of the 3'→5'exonuclease activity of human apurinic endonuclease (Ape1) by its 5'-incised Abasic DNA product [J]. J Biol Chem, 2003, 278(38):36242-36249.
    78. Ghosh SS, Gopinath P, Ramesh A. Adenoviral vectors: a promising tool for gene therapy [J]. Appl Biochem Biotechnol, 2006, 133(1): 9-29.
    79. Majhen D, Ambriovic-Ristov A. Adenoviral vectors-How to use them in cancer gene therapy? [J] Virus Res, 2006, 119(2): 121-133.
    80. Relph KL, Harrington KJ, Pandha H. Adenoviral strategies for the gene therapy of cancer [J]. Semin Oncol, 2005, 32(6): 573-582.
    81. Addison, C. L, Hitt, M, Kunsken, D, and Graham, F. L. Comparison of the human versus murine cytomegalovirus immediate early gene promoters for transgene expression by adenoviral vectors [J]. J. Gen. Virol. 1997, 78(17), 1653-1661.
    82. Bett, A. J, Haddara, W, Prevec, L. and Graham, F.L. An efficient and flexible system for construction of adenovirus vectors with insertions or deletions in early regions 1 and 3 [J]. Proc. Natl. Acad. Sci. US.1994, 91: 8802-8806,.
    83. Chartier, C, Degryse, E, Gantzer, M, et al. Efficient generation of recombinant adenovirus vectors by homologous recombination in Escherichia coli [J]. J. Virol. 1996, 70:4805-4810.
    84. Fallaux, F.J, Kranenburg, O, Cramer, S.J, et al. Characterization of 911: a new helper cell line for the titration and propagation of early region 1-deleted adenoviral vectors [J]. Hum Gene Ther, 1996, 7:215-222.
    85. Fallaux, F.J, Bout A, van der Velde, I, et al. New helper cells and matched early region 1-deleted adenovirus vectors prevent generation of replication-competent adenoviruses [J]. Hum Gene Ther, 1998, 9:1909-1917.
    86. Graham, F.L, Smiley, J, Russell, W.C, et al. Characteristics of a human cell line transformed by DNA from human adenovirus type 5 [J]. J. Gen. Virol. 1977, 36:59-72.
    87. Graham, F.L. Covalently closed circles of human adenovirus DNA are infectious [J]. The EMBO J. 1984, 3:2917-2922.
    88. He, T. C, Zhou, S, Da Costa, et al. A simplified system for generating recombinant adenoviruses [J]. Proc. Natl. Acad. Sci. USA. 1998,95: 2509-2514.
    89. Mizuguchi, H, and Kay, M. A. Efficient construction of a recombinant adenovirus vector by an improved in vitro ligation method. Hum [J]. Gene Ther. 1998, 9: 2577-2583.
    90. Ng, P, Parks, R. J, Cummings, et al. A high efficiency Cre/loxP based system for construction of adenoviral vectors [J]. Hum. Gene Ther. 1999,10:2667-2672.
    91. Ng, P, Parks, R. J, Cummings, et al. An enhanced system for construction of adenoviral vectors by the two plasmid rescue method. Hum. Gene Ther.1999, 11:693-699, 2000.
    92. Ng, P, Cummings, D. T, Evelegh, et al. The yeast recombinase FLP functions effectively in human cells for construction of adenovirus vectors. BioTechniques.2000, 29: 524-528.
    93. Wehrli M,Dougan ST,Caldwell K,et a1.Arrow encodes all LDL-receptor-related protein essential for Wingless signalling[J].Nture,2000,407(6803):527-530.
    94. Moon RT,Bowerman B,Boutros M,et a1.The promise and perils of Wnt signaling through beta-catenin[J].Science,2002,296(5573):l644-l646.
    95. Heisenberg CP.Wnt signalling:refocusing on Strabismus[J].Curt Biol,2002。12(19):R657-659.
    96. Kikuchi A,Kishida S,Yamamoto H.Regulation of Wnt signaling by protein-protein interaction and post-translational modifications[J].Exp Mol Med。2006,38(1):1-10.
    97. David J Mulholand,Shoukat Dedhar,GerhardA Coetzee,et a1.Interaction of Nuclear Receptors with the Wnt/β-Catenin/Tcf Signaling Axis:Wnt You Liketo Know? [J]. Endocr Rev,2005,(26):898-915.
    98. DasGupta R,Kaykas A,Moon RT。et a1.Functional genomie analysis of the Wnt-wingless signaling pathway[J].Science,2005,308(5723):826-833.
    99. Zeng L, Fagotto F, Zhang T, et a1.The mouse Fused locus encodes Axin, an inhibitor of the Wnt signaling pathway that regulates embryonic axis formation [J]. Cell 1997, 90:181-192.
    100. Berle H, Bauer A, Stappert J, et a1.Beta-catenin is a target for the ubiquitin-proteasome pathway [J]. EMBO J .1997, 16:3797-3804.
    101. Eresh S, Riese J, Jackson DB, et a1.A CREBbinding site as a target for decapentaplegic signalling during Drosophila endoderm induction [J]. EMBO J.1997, 16:2014-2022.
    102. Takemaru KI, Moon RT. The transcriptional coactivator CBP interacts with beta-catenin to activate gene expression [J]. J Cell Biol. 2000, 149:249-254.
    103. Hecht A, Vleminckx K, Stemmler MP, et a1.The p300/CBP acetyltransferases function as transcriptional coactivators of beta-catenin in vertebrates [J]. EMBO J. 2000, 19:1839-1850.
    104. Tien LT,Ito M,Nakao M,et a1.Expression of beta-catenin in hepatocellular carcinoma [J].World J Gastroenterol. 2005, 11(16):2398-2401.
    105. Cui J,Zhou X,Liu Y,et a1.Wnt signaling in hepatocellular carcinoma:analysis of mutation an dexpression of beta-catenin,T-cell factor-4 and SlYcogen synthase kinase 3-beta genes[J].J Gastroenteml Hepatol,2003,18(3):280-287.
    106. Kimura N, Shimada N, Fukuda M, et al. Regulation of cellular functions by nucleoside diphosphate kinases in mammals[J]. J Bioenerg Biomembr, 2000, 32(3):309-315.
    107. Wang F, Cao Y, Zhao W Z, et al. Taxol inhibits melanoma metastases. Through apoptosis induction, angiogenesis inhibition, and restoration of E-cadherin and nm23 expression[J]. J Pharmacol Sci, 2003,93(2):197-203.
    108. Elagoz S, Egilmez R, Koyuncu A, et al. The intratumoral microvessel density and expression of bFGF and nm23-H1 in colorectal cancer[J]. Pathol Oncol Res,2006, 12(1):21-27.
    109. Liu F, Qi HL, Chen HL. Effects of all-trans retinoic acid and epidermal growth factor on the expression of nm-23-H1 in human hepatocarcinoma cells [J]. J Cancer Res Clin Oncol, 2000,126(2):85-90.
    110. Evans. AR, Limp-Foster. M, Kelley MR. Going APE over ref-1. Mutat Res, 2000,461(9):83-108.
    111. Fan J, Wilson DM. Protein-protein interactions and posttranslational modifications in mammalian base excision repair. Free Radic Biol Med, 2005, 38(9):1121-1138.
    112. Parsons JL, Dianova II, Dianov GL. Ape1 is the major 3′-phosphoglycolate activity in human cell extracts. Nucleic Acids Res, 2004, 32(12):3531-3536.
    113. Chou KM, Cheng YC. An exonucleolytic activity of human apurinic/apyrimidinic endonuclease on 3′mispaired DNA. Nature, 2002, 415(6872):655–659.
    114. Tell G, Quadrifoglio F, Tiribelli C. The many functions of APE1/Ref-1: not only a DNA repair enzyme. Antioxid Redox Signal, 2008, 11(3): ahead of print.
    115. Kelley MR, Moore DH, Cheng L, et al. Altered expression of the multifunctional DNA base excision repair and redox enzyme of the AACR Special Conference on DNA Repair Defects, San Diego, 2000,5(98):210-223.
    116. Moore DH., Michael H, Tritt R, et al. Alterations in the expression of the DNA repair/redox enzyme APE/Ref-1 in epithelial ovarian cancers, Clin. Cancer Res, 2000,6 (4): 602-609.
    117. Fishel ML, Kelley MR. The DNA base excision repair protein Ape1/Ref-1 as atherapeutic and chemopreventive target. Mol Aspects Med, 2007, 28(3-4):375-395.
    118. Bapat A, Fishel M, Kelley MR. Going Ape as an approach to cancer therapeutics. Antioxid Redox Signal, 2008, ahead of print.
    119. Xiang DB, Chen ZT, Wang D, et al. Chimeric adenoviral vector Ad5/F35-mediated APE1 siRNA enhances sensitivity of human colorectal cancer cells to radiotherapy in vitro and in vivo. Cancer Gene Therapy. 2008, 15(10):625-635.
    120. Wang D, Zhong ZY, Xiang DB, et al. APE1 overexpression is associated with cisplatin resistance in non-small cell lung cancer and targeted inhibition of APE1 enhances the activity of cisplatin in A549 cells. Lung Cancer,2009,ahead of print.
    1. Li XD, Wu LM, Xie HY, Xu X, Zhou L, Liang TB, et al. No association exists between E-cadherin gene polymorphism and tumor recurrence in patients with hepatocellular carcinoma after transplantation. Hepatobiliary Pancreat Dis Int 2007;6:254-258.
    2. Bruix J, Hessheimer AJ, Forner A, Boix L, Vilana R, Llovet JM: New aspects of diagnosis and therapy of hepatocellular carcinoma. Oncogene 2006, 25(27):3848-3856.
    3. Avila MA, Berasain C, Sangro B, Prieto J: New therapies for hepatocellular carcinoma. Oncogene 2006, 25(27):3866-3884.
    4. Fontana JA, Rishi AK: Classical and novel retinoids: their targets in cancer therapy. Leukemia 2002, 16(4):463-472.
    5. Clarke N, Germain P, Altucci L, Gronemeyer H: Retinoids: potential in cancer prevention and therapy. Expert Rev Mol Med 2004,6(25):1-23.
    6. Herold C, Ganslmayer M, Ocker M, Hermann M, Hahn EG, Schuppan D: Combined in vitro anti-tumoral action of tamoxifen and retinoic acid derivatives in hepatoma cells. Int J Oncol 2002, 20(1):89-96.
    7. Gerard B, Bleiberg H: Treatment of hepatocarcinoma. Curr Oncol Rep 2004, 6(3):184-191.
    8. You KR, Shin MN, Park RK, Lee SO, Kim DG: Activation of caspase-8 during N-(4-hydroxyphenyl)retinamide-induced apoptosis in Fas-defective hepatoma cells. Hepatology 2001, 34(6):1119-1127.
    9. Kim DG, You KR, Liu MJ, Choi YK, Won YS: GADD153-mediated anticancer effects of N-(4-hydroxyphenyl)retinamide on human hepatoma cells. J Biol Chem 2002, 277(41):38930-38938.
    10. Forsyth N. R., Wright W. E., Shay J. S. Telomerase and differentiation in multicellular organisms: turn it off, turn it on, and turn it off again [J]. Differentiation, 2002, 69: 188-197.
    11. Araki K, Kishihara F, Takahashi K, et al. Hepatocellular carcinoma producing a granulocyte colony-stimulating factor: report of a resected case with a literature review [J]. Liver Int. 2007 , 27(5):716-721.
    12. Musch A, Rabe C, Paik MD, et al. Altered expression of TGF-beta receptors in hepatocellular carcinoma-effects of a constitutively active TGF-beta typeⅠreceptor mutant[J]. Digestion, 2005, 71(2):78- 91.
    13. Forrester E, Ghytil A, Bierie B, et al. Effect of conditional knockout of the typeⅡTGF-beta receptor gene in mammary epithelia on mammary gland development and polyomavirus middle T antigen induced tumor formation and metastasis [J]. Cancer Res, 2005, 65:2296-2302.
    14. Das A, Banik NL, Ray SK. Molecular Mechanisms of the Combination of Retinoid andInterferon-gamma for Inducing Differentiation and Increasing Apoptosis in Human Glioblastoma T98G and U87MG Cells . Neurochem Res. 2008 Mar 27.
    15. Zerbini A, Pilli M, Fagnoni F, et al. Increased immunostimulatory activity conferred to antigen-presenting cells by exposure to antigen extract from hepatocellular carcinoma after radiofrequency thermal ablation [J]. J Immunother. 2008, 31(3):271-282.
    16. Finnberg N, El-Deiry WS. TRAIL death receptors as tumor suppressors and drug targets[J]. Cell Cycle. 2008, 7(11):1525-1528.
    17. Xu DH, Tang J, Li QF, Shi SL, et al. Positional and expressive alteration of prohibitin during the induced differentiation of human hepatocarcinoma SMMC-7721 cells[J]. World J Gastroenterol. 2008 ,14(32):5008-5014.
    18. Tang J, Niu JW, Xu DH, et al. Alteration of nuclear matrix-intermediate filament system and differential expression of nuclear matrix proteins during human hepatocarcinoma cell differentiation[J]. World J Gastroenterol. 2007 ,13(20):2791-2797.
    19. Wu J, Luo RC, Zhang H, Cui YZ. Inhibitory effect of sorafenib combined with arsenic trioxide on hepatocellular carcinoma cells [J]. Nan Fang Yi Ke Da Xue Xue Bao. 2008 ,28(4):639-641.
    20.郑兰东,夏荣龙.丹参对肝脏肿瘤发展的影响[J].医药论坛杂志,2005,26(11):35-39.
    21. Zeng XL,Tu ZG.In vitro induction of differentiation by ginsenoside Rh2 in SMMC-7721 hepatocarcinoma cell line[J].Pharmacol Toxicol,2003,93(6):275-283.
    22.曾小莉,涂植光.端粒酶在人参皂甙Rh2诱导肝癌细胞分化中的作用[J].癌症,2004,23(12):1655-1659.
    23.司维柯,高利宏,刘文武.苦参碱诱导人肝癌细胞分化、凋亡时对GL细胞周期调节因子的调控[J].癌症,2001,20(8):848-851.
    24.黄炜,黄济群,张东方.18β-甘草次酸和甘草酸对人肝癌细胞增殖的抑制和诱导分化作用[J].中国中医药科技,2002,9(2):92-93.
    25.吴晓慧,王顺祥,周少英.甘草酸苷对人肝癌细胞增殖及凋亡的影响[J].中华实用中西医杂志,2005,18(2):267-269.
    26.刘剑波,高学岗,连涛,等.大黄素在体外诱导人肝癌细胞HepG2发生凋亡的初步研究[J].癌症,2003,22(12):1280-1289.
    27.郭昱,郭霞,姚金锋等.黄芩甙对人肝癌细胞BEL-7402增殖、凋亡和分化的影响[J].中药药理与临床,2008,24 (3):21-23.
    28.解方为,欧阳学农,蒋明德.红景天甙对人肝癌细胞的诱导分化作用[J].西南国防医药2005 ,15 (6):613-615.
    29. Zhu WG,Hileman T Key,et al.5-aza-2’-deoxycytidine activates the p53/p21Wafl/Cipl pathway to inhibit cell proliferation.J Biol Chem,2004,279(15):151-161.
    30. Ray A,Shakya A,Kumar D,et al.Overexpression by the induction of cyclin-dependet protein kinase inhibitor p21WAF-1/Cip-1/Sdi-1 expression.J Immunol,2004,172(8):5006
    31. Troncone G,Migliaccio I,Caleo A,et al.p27Kipl expression and grading of liver cancer diagnosed on cytological samples.Diagn Cytopathol,2004,30(6):375
    32. Yokota Y, Mori S. Role of Id family proteins in growth control [J].J Cell Physiol, 2002, 190(1):21-25.
    33. Bu P, Wan YJ. Fenretinide-induced apoptosis of Huh-7 hepatocellular carcinoma is retinoic acid receptorβdependent [J]. BMC Cancer, 2007, 7:236-245.
    34.樊光华,姜浩,欧文胜.人参皂甙Rh2诱导人肝癌细胞BEL-7402细胞凋亡的作用[J].实用癌症杂志,2003,18(1):16-19.
    35. Moon RT, Kohn AD, De Ferrari GV, et al. WNT and beta-catenin signaling: diseases and therapies [J]. Nat Rev Genet, 2004, 5:691-701.
    36. Bengochea A, de Souza MM, Lefran?ois L, et al. Common dysregulation of Wnt/Frizzled receptor elements in human hepatocellular carcinoma [J]. Br J Cancer. 2008, 99(1):143-50.
    37. Lechel A, Rudolph KL. Rho GTP ase and Wnt signaling pathways in hepatocarcinogenesis [J]. Gastroenterology. 2008, 134(3):875-878.
    38. Thompson MD, Monga SP. WNT/beta-catenin signaling in liver health and disease [J]. Hepatology. 2007, 45(5):1298-1305.
    39. Wang Q, Huang S, Yang L, et al. Down-regulation of Sonic hedgehog signaling pathway activity is involved in 5-fluorouracil-induced apoptosis and motility inhibition in Hep3B cells [J]. Acta Biochim Biophys Sin (Shanghai). 2008 , 40(9):819-829.
    40. Kim Y, Yoon JW, Xiao X, et al. Selective down-regulation of glioma-associated oncogene 2 inhibits the proliferation of hepatocellular carcinoma cells [J]. Cancer Res. 2007, 67(8): 3583-3593.
    41. Suwanjunee S, Wongchana W, Palaga T. Inhibition of gamma-secretase affects proliferation of leukemia and hepatoma cell lines through Notch signaling [J]. AnticancerDrugs. 2008, 19(5):477-486.
    42. Gao J, Song Z, Chen Y, et al. Deregulated expression of Notch receptors in human hepatocellular carcinoma [J]. Dig Liver Dis. 2008, 40(2):114-121.
    43. Gramantieri L, Giovannini C, Lanzi A, et al. Aberrant Notch3 and Notch4 expression in human hepatocellular carcinoma [J]. Liver Int. 2007, 27(7):997-1007.
    44. Calvisi DF, Pascale RM, Feo F. Dissection of signal transduction pathways as a tool for the development of targeted therapies of hepatocellular carcinoma [J]. Rev Recent Clin Trials. 2007, 2(3):217-236.
    45. SERANSKI P, HOTF C, RADELOF U, et al. RA I1 is a novel polyglutamine encoding gene that is deleted in Smith-Magenis syndrome patients[ J ]. Gene, 2001, 270 (122) : 69-76.
    46. TOULOUSE A, ROCHEFORT D, ROUSSEL J, et al. Molecular cloning and characterization of human RA I1, a gene associated with schizophrenia [ J ]. Genomics, 2003, 82 (2): 162-171.
    47. WALPOLE SM, HIRIYANA K T, NICOLAOU A, et al. Identification and characterization of the human homologue (RAI2) of amouse retinoic acid-Induced gene in Xp22[ J ]. Genomics, 1999, 55 (3): 275-283.
    48. HUANG KM, GEUNES2BOYER S, WU S, et al. Organization and annotation of the Xcat critical region: elimination of seven positional candidate genes. Genomics, 2004, 83 (5): 893-901.
    49. TAO Q, CHENG Y, CL IFFORD J, et al. Characterization of the murine orphan G-protein-coupled receptor gene Rai3 and its regulation by retinoic acid [ J ]. Genomics, 2004, 83 (2): 270-280.
    50. Kang SH,Song JH,Kang HK,et al.Arsenic trioxide induced apotosis is independent of stress responsive signaling pathways but sensitive to inhibition of inducible nitric oxide synthase in HepG2 cells[J].ExpMol Med,2003,35:83-90.
    51.曾小莉,涂植光.人参皂甙Rh2对人肝癌细胞SMMC-7721信号传导的影响[J].中华肝脏病杂志,2004,12(9):565-566.
    52.李卫东,张明磊,朴云峰.全反式维甲酸与顺铂抑制肝癌细胞的研究[J].中国公共卫生. 2004,20(4):459-460.
    53. Yun-Feng P, Yang S, Pu-Jun G. Inhibitory effect of all-trans retinoic acid on humanhepatocellular carcinoma cell proliferation [J]. World J Gastroenterol, 2003, 9(9):2117-2120.
    54.聂林,余为,何冬梅,等.三氧化二砷对肝癌细胞survivin基因和端粒酶活性的影响[J].肿瘤,2006,26(12):1064-1068.
    55.曾小莉,涂植光.端粒酶在人参皂甙Rh2诱导肝癌细胞分化中的作用[J].癌症,2004,23(12):1655-1659.
    56.左国庆,何松,张燕,等.苦参素改变端粒酶活性对人肝癌细胞株HepG2 QGY体外增殖影响的研究[J].肿瘤学杂志,2005,11(2):126-128.
    57. Yu J, Qiao L, Zimmermann L, et al. Troglitazone inhibits tumor growth in hepatocellular carcinoma in vitro and in vivo [J]. Hepatology, 2006, 43(1):134-143.
    58.罗惠霞,王玉炯.组蛋白乙酰化/去乙酰化在真核基因转录调控中的作用[J],中国生物工程杂志,2004,24(2):19-21.
    59. Blaheta RA, Michaelis M, Driever PH, et al.Evolving anticancer drug valproic acid: insights into the mechanism and clinical studies [J].Med Res Rev,2005,25(4):383-397.
    60. Camacho LH,Olson J,Tong WP,et al.PhaseⅠdose escalation clinical trial of phenylbutyrate sodium administered twice daily to patients with advanced solid tumors [J].Invest New Drugs. 2007, 25(2):131-138.

© 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