肝靶向性RNA干扰载体的构建与鉴定
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
RNA干扰(RNA interference,RNAi)是指通过诱导同源mRNA降解导致特定基因发生转录后基因沉默。目前,RNAi技术已广泛应用于基因功能研究和基因治疗等领域。RNAi可以通过siRNA或shRNA来实现,shRNA普遍应用RNA聚合酶Ⅲ启动子来调控其转录,但此类启动子缺乏组织特异性,因此我们选择RNA聚合酶Ⅱ启动子来调控shRNA转录,从而实现RNAi的组织特异性。本研究采用RNA聚合酶Ⅱ启动子中的apoA-I启动子来调控shRNA在肝癌细胞中特异性表达,从而实现肿瘤基因治疗的肝靶向性。
     首先,本实验通过PCR扩增apoA-I基因启动子序列,并将其克隆至pEGFP的报告蛋白EGFP基因上游,构建肝靶向重组载体pAE。将其和阳性对照质粒pEGFP分别转染肝癌细胞和非肝癌细胞,以报告蛋白的表达为标志检测其组织特异性。结果表明apoA-I启动子能够特异性启动绿色荧光蛋白在肝癌细胞中表达,而在食管癌EC9706、乳腺癌MCF7、宫颈癌HeLa细胞中不表达,apoA-I启动子具有较强的肝细胞组织特异性,为肝组织靶向性基因治疗奠定了实验基础。
     其次,我们将肝特异性apoA-I启动子运用到RNAi载体构建中,期望实现肝癌的靶向性基因治疗,试验中用报告蛋白EGFP的表达下调情况来判定载体构建是否成功。由于apoA-I启动子属于PolⅡ启动子,它的转录调控机制有别于PolⅢ启动子,所以我们对shRNA的表达框进行结构修饰,添加了Ribozyme和MAZ结构,与未经修饰的质粒载体做对照,转染肝癌细胞,以报告蛋白的表达为标志检测其活性。实验结果表明,本研究构建的apoA-I启动子调控的RNAi载体不仅能够有效实现RNAi作用,同时可以有效降低脱靶效应,为PolⅡ启动子在RNAi中的应用奠定了实验基础。
     RNAi具有序列特异性的特点,因此碱基突变、缺失等都可能对RNAi效果产生一定的影响。在本研究中,对于PolⅡ启动子调控的shRNA序列中碱基缺失对RNAi效果的影响也进行了初步探索,在上步实验验证有RNAi作用的shRNA序列正义链3'端设计碱基缺失,和未缺失的质粒做对照,转染肝癌细胞,以报告蛋白的表达为标志检测其活性。研究结果发现,在PolⅡ启动子调控的shRNA正义链上进行碱基缺失,shRNA载体也具有一定的RNAi效果,为深入探索RNAi的干扰机制进行了初步的尝试。
     肿瘤的发生和发展是一种复杂的过程,涉及多种基因、多种因素的综合作用,因此联合基因治疗已成为肿瘤基因治疗研究的新方向。为了实现高效、稳定的双基因沉默,并预防RNAi逃逸,在本研究中,我们构建双基因RNA沉默载体pSAD-EGFP和pDAF-EGFP,共转染肝癌细胞,以报告蛋白的表达为标志检测其活性,尝试联合RNAi的可行性。结果显示两种质粒对EGFP都具有沉默效果,为肿瘤的联合RNAi基因治疗奠定了很好的实验基础。
RNA interference (RNAi) is a post-transcriptional gene silencing process by inducing sequence-specific mRNA degradation. Nowadays, RNAi has been widely used in many areas, such as loss-of-function studies and gene therapy. RNAi may be achieved using short hairpin RNA (shRNA) as well as small interfering RNA (siRNA). Although Pol III promoters are commonly used to synthesize shRNA, they lack cell specificity. An alternative way is to use cell-specific Pol II-directed synthesis of shRNA. Here, a liver-specific RNAi system for hepatoma gene therapy was studied using the human apolipoprotein A-I promoter.
     First, the apoA-I promoter containing essential transcriptional regulatory elements was amplified by PCR and then inserted into upstream of the green fluorescent protein gene of pEGFP. The recombinant plasmid with apoA-I promoter and the control plasmid with CMV promoter were transfected into SMMC-7221 and EC9706, MCF7, HeLa, respectively. Cells generated using vector with liver-specific promoter expressed GFP only in the appropriate cell type. By contrast, when transfected with the control vector, cells gave similar levels of expression. The apoA-I promoter could obtain restricted expression of an exogenous gene only in liver cells.
     Second, apoA-I promoter was used to drive shRNA expression in a particular type of cell. It was showed that the apoA-I-driven small hairpin RNAs specifically depress the expression of the exogenous reporter - enhanced green fluorescent protein. To prevent the off-target effects, the gene-specific targeting sequence was cloned between the ribozyme cassette and the MAZ site. The cis-acting hammerhead ribozyme cassette was added to cut off the m~7G cap structure, and the MAZ site was added for Pol II transcriptional pausing. The study provides an efficient way in silencing a gene in a particular type of cell without interfering with other type of cells.
     RNAi has characteristic of sequence-specific. Gene deletion and gene mutation of shRNA will influence RNAi effect. We want to know the influence about a base deletion shRNA regulated by Pol II promoter. A shRNA with a base deletion in its sense strand was designed and inserted into downstream of the Pol II promoter, this vector was subsequently cotransfected with pEGFP into SMMC-7721 accompany with the comparison vector that have no gene deletion. The study demonstrated that the gene deletion shRNA controled by the Pol II promoter could also induce the RNAi effect, this make contribution to the in-depth exploration of RNAi mechanism.
     To achieve efficient, stable, simultaneous gene silencing, and to prevent RNAi escape, a single cell-specific RNAi expression system for silencing double targets is tested in this study. The system was transfected into SMMC-7721 to investigate the influence to RNAi of diffferent direction of the promoters. The silencing efficiency was identified by observing the expression of EGFP. The silencing efficiency of both vectors were comparable to each other. The study demonstrated that the liver-specific vectors construction of combined gene therapy is successful. This method is a promising therapeutic method in which gene silencing of a single cannot account for hepatoma gene therapy.
引文
[1]李广灿,叶召.我国常见恶性肿瘤发病现状.中国肿瘤,1999,8(11):498-499.
    [2]Ruben HA,Bruno S,Jesus P.Gene therapy of liver cancer.World Gastroenterol,2006,12(38):6085-6097.
    [3]Ullu E,Djikeng A,Shi HF,et al.RNA interference:advances and questions.The Royal Society,2002,357:65-70.
    [4]Schubert S,Grunert HP,Zeichhardt H,et al.Maintaining Inhibition:siRNA double expression vectors against coxsackieviral RNAs.Mol.Biol.,2005,346:457-465.
    [5]Lowenstein PR.Clinical trials in gene therapy:ethics of informed consent and the future of experimental medicine.Curr Opin Mol Ther.,2008,10(5):428-430.
    [6]Xia CF,Zhang YF,Zhang Y,et al.Intravenous siRNA of brain cancer with receptor targeting and avidin-biotin technology.Pharmaceutical Research,2007,24(12):2309-2316.
    [7]任雪玲,马玥,徐敏,等.基因治疗中的纳米载体研究进展.第十届中国科协年会论文集,2008,36-37.
    [8]Yang L,Cao Z,Li F,et al.Tumor-specific gene expression using the survivin promoter is further increased by hypoxia.Gene Therapy,2004,11:1215-1223.
    [9]Fukazawa T,Maeda Y,Frances M.Development of a Cancer-Targeted Tissue-Specific Promoter System.Cancer Research,2004,64(1):363-369.
    [10]Selvakumaranm,Bao R,Crijns AP,et al.Ovarian epithelial cell lineage-specific gene expression using the promoter of a retrovirus-like element.Cancer Res,2001,61(4):1291-1295.
    [11]Yu JY,DeRuiter SL,Turner DL.RNA interference by expression of short-interfering RNAs and hairpin RNAs in mammalian cells.PNAS,2002,99:6047-6052.
    [12]Vande WM,Oving I,Munean V,et al.Specific inhibition of gene expression using a stably integrated,inducible small-interfering-RNA vector.Embo reports,2003,4(6):609-615.
    [13]陈青,潘秋卫,蔡荣,等.RNA聚合酶Ⅱ启动子调控RNA干扰在肿瘤治疗中的应用前景.生物化学与生物物理进展,2007,34(8):806-815.
    [14]Becker GJ.Recent developments in cancer therapy and why we need a human cancer genome project.J Am Coll Radiol,2005,2(11):887-888.
    [15]邓婷,纪伟,连霁虹,等.人apoA-I基因启动子转录上调剂筛选系统的建立与应用.天然产物研究与开发,2007,19:776-780.
    [16]Jones DR,Leffak M.A bifunctional regulatory element of the human ApoA-I gene responsive to a distal enhancer.DNA Cell Bio,1999,18(2):107-119.
    [17]Widom RL,Ladias JA,Kouidou S,et al.Synergistic interactions between transcription factors control expression 6f the apolipoprotein A-I gene in liver cells.Mol.Cell.Bio.,1991,11:677-687.
    [18]Higuchi K,Law SW,Hoeg JM,et al.Tissue-specific expression of apolipoprotein A-I (APOA-I) is regulated by the 5'-flanking region of the human ApoA-I gone.Biol.Chem,1988,263(34):18530-18536.
    [19]Matsunaga A,Sasaki J,Han H,et al.Compound heterozygosity for an apolipoprotein A1gene promoter mutation and a structural nonsense mutation with apolipoprotein A1 deficiency.Arterioscler.Thromb.Vasc.Biol.,1999,19:348-355.
    [20]Chalfie M,Tu Y,Euskirchen G,et al.Green fluorescent protein as a marker for gene expression.Science,1994,263:802-805.
    [21]Kain SR,Adams M,Kondepudi A,et al.Green fluorescent protein as a reporter of gene expression and protein localization.Biotechniques,1995,19:650-655.
    [22]程爱娟,毛用敏,崔让庄,等.apoAI启动子-93bp--63bp片段及-75bp位点突变对基因转录调控作用的研究.天津医药,2006,34(9):593-595.
    [23]彭剑虹,周新,刘焰,等.载脂蛋白(a)启动子-418A/G位点和-384C/T位点单核苷酸多态性的研究.中国病理生理杂志,2004,20(8):1368-1372.
    [24]孟葆林,李维琪,毛建平,等.人apoA-I分泌型表达调控细胞模型构建.中国生物工程杂志,2005,25(8):39-44.
    [25]Philippe D,Cristin MG,John EB,et al.Identification of liver receptor homolog-1 as a novel regulator of apolipoprotein A-I gene transcription.Molecular Endocrinology,2004,18(10):2378-2387.
    [26]Panagiota P,Kinya OG,Dipak PR,et al.Promoter elements and factors involved in hepatic transcription of the human apoA-I gene positive and negative regulators bind to overlapping sites.Biological Chemistry,1991,266(9):5790-5797.
    [27]Lungwitz U,Breunig M,Blunk T,et al.Polyethylenimine-based non-viral gene delivery systems.Eur J Pharm Biopharm.,2005,60(2):247-266.
    [28]Kircheis R,Schuller S,Brunner S,et al.Polycation-based DNA complexes for tumor-targeted gene delivery in vivo.Gene Med,1999,1(2):111-120.
    [29]Fire A,Xu S,Montgomery MK,et al.Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans.Nature,1998,391(6669):806-811.
    [30]Tabara H,Grishok A,Mello CC,et al.RNAi in C.elegans:Soaking in the Genome Sequence.Science,1998,282(5388):430-431.
    [31]Raskopf E,Vogt A,Sauerbruch T,et al.siRNA targeting VEGF inhibits hepatocellular carcinoma growth and tumor angiogenesis in vivo.Hepatology,2008,49(6):977-984.
    [32].Hamilton AJ, Baulcombe DC. A Species of Small Antisense RNA in Posttranscriptional Gene Silencing in Plants. Science, 1999,286(5441): 950-952.
    [33] Amarzguioui M, Rossi JJ, Kim D. Approaches for chemically synthesized siRNA and vector-mediated RNAi. FEBS Lett, 2005,579(26): 5974-5981.
    [34] Brummelkamp TR, Bernards R, Agami R. A system for stable expression of short interfering RNAs in mammalian cells. Science, 2002,296(5567): 550-553.
    [35] Paddison PJ, Caudy AA, Bernstein E, et al. Short hairpin RNAs(shRNAs) induce sequence-specific silencing in mammalian cells. GenesDev, 2002,16: 948.
    [36] Bear DG, Fomproix N, Soop T, et al. Nuclear poly(A)-binding protein PABPN1 is associated with RNA polymerase II during transcription and accompanies the released transcript to the nuclear pore. Exp Cell Res, 2003,286(2): 332-344.
    [37] Jackson AL, Bartz SR, Schelter J, et al. Expression profiling reveals off-target gene regulation by RNAi. Nat. Biotechnol., 2003,21: 635-637.
    [38] Fedorov Y, Anderson EM, Birmingham A, et al. Off-target effects by siRNA can induce toxic phenotype. RNA, 2006,12: 1188-1196.
    [39] Matsumoto S, Miyagishi M, Akashi H, et al. Analysis of double-stranded RNA-induced apoptosis pathways using interferon-response noninducible small interfering RNA expression vector library. Biol. Chem., 2005,280: 25687-25696.
    
    [40] Shinagawa T, Ishii S. Generation of Ski-knockdown mice by expressing a long double-strand RNA from an RNA polymerase II promoter. Genes Dev., 2003,17: 1340-1345.
    [41] Cheng T, Xu CY, Wang YB, et al. A rapid and efficient method to express target genes in mammalian cells by baculovirus. World J Gastroenterol, 2004,10(11): 1612-1618.
    [42] Yao K, Lu H, Cheng H, et al. Mutation of siRNA results in thermodynamically unstable duplex which influences knockdown of dmrt1 by RNA interference. Front Biosci., 2006, 11(1): 2623-2630.
    
    [43] Chu CY, Rana TM. Potent RNAi by short RNA triggers. RNA, 2008,14: 1714-1719.
    [44] Reynolds A, Leake D, Boese Q, et al. Rational siRNA design for RNA interence. Nat Biotechnol, 2004,22(3): 326-330.
    [45]Muller OJ, Katus HA, et al. Targeting the heart with gene therapy-optimized gene delivery methods. Cardiovasc Res, 2007, 73:453-462.
    [46] Palu G, Cavaggioni A, Calvi P, et al. Gene therapy of glioblastoma multiforme via combined expression of suicide and cytochine genes: a pilot study in humans. Gene Therapy, 1999, 6(3): 330-337.
    [47] Grill J, Beusechem VW, Van Der VP, et al. Combined targeting of adenoviruses to integrins and epidermal growth factor receptors increases gene transfer into primary glioma cells and spheroids.Clin Cancer Res,2001,7(3):641-650.
    [48]刘清源,施学忠,李来生,等.Rb对HepG2肝癌细胞系增殖的影响.医药论坛杂志,2006,27(22):10-11.
    [49]盖晓东,李国利,黄京子,等.野生型p53基因对肝癌细胞多药耐药的逆转作用及其相关机制.癌症,2006,25(8):954-959.
    [50]Zeng JZ,Wang HY,Wu MC.Molecular cloning,characterization and expression of a novel proline-rich gene associated hepatocellular carcinoma.Gastroenterol Hepatol,2000,15(1):16.
    [51]Nagao K,Tomimatsu M,Endo H,et al.Telomerase reverse transcriptase mRNA expression and telomerase activity in hepatocellular carcinoma.Gastroenterol,1999,34:83-87.
    [52]Lingner J,Hughes TR,Shevchenko A,et al.Reverse transcriptase motifs in the catalytic subunit of telomerase.Science 1997,276(5312):561-567.
    [53]Dufes C,Keith WN,Bilsland A,et al.Synthetic anticancer gene medicine exploits intrinsic antitumor activity of cationic vector to cure established tumors.Cancer Res.,2005,65:8079-8084.
    [54]Zhang PH,Zou L,Tu ZG,RNAi-hTERT Inhibition Hepatocellular Carcinoma Cell Proliferation via Decreasing Telomerase Activity.Journal of Surgical Research,2006,131(1):143-149
    [55]吴江雪,徐本玲,黄文林.血管生成抑制因子的研究进展,癌症,2005,24(3):376-384.
    [56]Miura H,Miyazaki T,Kuroda M.Increased expression of vascular endothelial growth factor in human hepatocellular carcinoma.Journal of Hepatology,1997,27(5):854-861.
    [57]蒋冬贵,夏昆,刘劫,等.RNA体外干扰肝癌Hep-3B细胞VEGF基因的表达.第四军医大学学报,2007,28(15):1349-1351.
    [58]Duda DG.,Batchelor TT,Willett CG.,et al.VEGF-targeted cancer therapy strategies:current progress.Hurdles and future prospects,2007,13(6):223-230.
    [59]Folkman J.Antiangiogenesis in cancer therapy-endostatin and its machanisms of action.Experimental Call Res,2006,312:594-607.
    [60]黄嘉凌,刘燕艳,刘然义,等.肝癌特异性HSV-TK/CD基因表达质粒的构建及其杀伤活性.中山大学学报(医学科学版),2005,26(2):142-145.
    [61]Van Dillen IJ,Mulder NH,Vaalburg W,et al.Influence of the bystander effect on HSV-tk/GCV gene therapy.A review.Curr Gene Ther,2002,2:307-332.
    [62]肖震宇,陈孝平,何松清,等.肿瘤坏死因子相关凋亡诱导配体及其受体在肝癌组织中的表达的研究.中华肝胆外科杂志,2005,11(1):56-58.
    [63]许海苗,王慧芬.肝癌的免疫治疗.国际流行病学传染病学杂志,2007,2(34):29-32.
    [64]姜涛,徐闯,刘国文,等.树突状细胞抗肿瘤疫苗研究进展.动物医学进展,2007, 28(2):62-66.
    [65]陈彦祥,乔卫红,刘栋良,等.靶向性基因治疗载体.大连医科大学学报,2007,29(4):400-403.
    [66]Seeger C,Mason W.Hepatitis B virus biology.Microbiol Mol Biol Rew,2000,64(1):51-68.
    [67]陈锋,胡晓年.非病毒载体在肝癌基因治疗中的应用.国外医学肿瘤学分册,2005,32(4):292-294.
    [68]Chu AC,Tsang SY,Lo EH,et al.Low density lipoprotein as a targeted carrier for doxorubicin in nude mice bearing human hepatoma HepG2 cells.Life Sci.,2001,70(5):591-601.
    [69]海俐,无唾液酸糖蛋白受体介导的肝靶向脂质体配体的设计、合成及靶向性研究.四川大学博士学位论文,2006:3.
    [70]Vorachek WR,Steppan CM,Lima M,et al.Distant Enhancers Stimulate the Albumin Promoter through Complex Proximal Binding Sites.Biol.Chem.,2000,275(37):29031-29041.
    [71]Zhang Y,Huang SZ,Zeng YT.The effect of HSV-t k/GCV on hepatic specific damage driven by murine ALB promoter/enhancer.Acta Genetica Sinica,2004,31(10):1053-1060.
    [72]徐凯成,所剑,赵岳,等.人肝癌细胞N-2ras基因RNA干涉有效靶点的确定.中国实验诊断学,2006,10(1):75-78.
    [73]朱明光,张鑫,袁红艳,等.RNAi沉默HMGA1基因对肝癌细胞凋亡和细胞周期的影响.中国免疫学杂志,2007,23(7):634-636.
    [74]Buckley AF,Burgart LJ,Kakar S.Epidermal growth factor receptor expression and gene copy number in fibrolamellar hepatocellular carcinoma.Hum.Pathol.,2006,37(4):410-414.
    [75]Chen GG,Ho RLK,Wong J,et al.Single nucleotide polymorphism in the promoter region of human alpha-fetoprotein(AFP) gene and its significance in hepatocellular carcinoma (HCC).EJSO,2007,33(7):882-886.
    [76]I do 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 alpha-fetoprotein gene promoter.Cancer Res,1995,55(14):3105.
    [77]魏强,王健伟,郭丽,等.以人甲胎蛋白启动子控制表达HIV-1vpr基因的重组腺病毒诱导肝癌细胞G2期阻滞和细胞凋亡作用的研究.中国实验诊断学,2006,10(7):725-729.

© 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