RNA干扰抑制人结肠癌HT-29细胞COX-2基因表达的实验研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
第一部分人COX-2小分子干扰RNA的设计、合成及转染条件的优化
     目的设计并化学合成人COX-2基因的小分子干扰RNA,并进一步优化转染人结肠癌HT-29细胞的条件。方法遵循RNA干扰目标序列的选取原则,利用互联网资源对人COX-2基因mRNA序列设计4条小干扰RNA(siRNA),经退火合成。以阴性荧光素特异性siRNA为报告基因,通过脂质体lipofectamineTM2000转染人结肠癌HT-29细胞,利用荧光显微镜计数观察转染效率,MTT法检测转染后对HT-29细胞的毒性。结果成功设计并合成4对人COX-2基因的小分子干扰RNA,在24孔板中筛选出最优化的转染条件为1.5ul lipofectamineTM2000和40pmol siRNA。结论成功设计并合成了人COX-2基因的小分子干扰RNA,同时优化的最佳转染条件为进一步研究人COX-2小干扰RNA功能提供了条件。
     第二部分COX-2小干扰RNA对人结肠癌HT-29细胞COX-2表达及细胞生长影响的研究
     目的研究COX-2 siRNA对环氧合酶-2(COX-2)高表达的人结肠癌HT-29细胞增殖和凋亡的影响。方法从化学合成的4对小干扰RNA中筛选出最佳的靶向COX-2 siRNA转染人结肠癌HT-29细胞,采用逆转录-聚合酶链反应(RT-PCR)和Western印迹分别从mRNA和蛋白水平检测COX-2表达,四甲基偶氮唑蓝(MTT)和Hoechst染色检测COX-2表达被抑制后细胞增殖和凋亡的情况。结果COX-2 siRNA能在mRNA和蛋白水平上下调人结肠癌HT-29细胞中COX-2基因的表达(P<0.05),以转染72h时显著。COX-2表达被抑制后,HT-29细胞生长受到明显抑制(P<0.05),凋亡细胞显著增加。结论COX-2与结肠癌细胞生长和凋亡密切相关,抑制结肠癌细胞中COX-2表达可以抑制结肠癌细胞生长,促进结肠癌细胞凋亡。
     第三部分人COX-2shRNA真核表达载体的构建、鉴定和转染条件的优化
     目的构建环氧合酶-2(COX-2)基因靶向性的shRNA真核表达载体,并进一步优化在转染人结肠癌HT-29的转染条件,以观察转染结肠癌细胞株HT-29细胞后,其对COX-2活性的抑制效率。方法设计短发夹结构的COX-2 siRNA对应模版DNA序列,退火处理后克隆至pGPH1-GFP-Neo质粒,构建重组质粒pshCOX-2。酶切并测序鉴定。优化转染条件后,将重组质粒转染人结肠癌HT-29细胞,采用逆转录-聚合酶链反应(RT-PCR)、Western印迹分别从mRNA和蛋白水平检测COX-2表达。结果pshCOX-2经鉴定与设计目的序列完全一致。转染重组质粒72h后可以显著抑制HT-29细胞COX-2 mRNA表达与对照组比较有显著性差异(F=349.58,P=0.0001),对照组之间则无差异(P>0.05),蛋白水平的抑制效率与mRNA水平一致。结论成功构建无内毒素活性的COX-2 shRNA真核表达质粒pshCOX-2,可稳定抑制人结肠癌HT-29细胞COX-2基因表达,为利用RNAi技术在体内和体外水平研究COX-2在结肠癌中的作用机制打下了基础。
     第四部分抑制COX-2蛋白表达对人结肠癌HT-29细胞生物学活性的影响
     目的体外观察pshCOX-2抑制COX-2活性对HT-29细胞生物学活性的影响,并探讨可能的作用机制。方法应用pshCOX-2瞬时转染HT-29细胞,MTT法检测细胞的生长曲线,放射免疫法(RIA)和ELISA法分别检测转染后细胞培养上清中PGE2和VEGF的表达;优化G418浓度、筛选和鉴定稳定表达pshCOX-2的HT-29细胞株,应用过河实验观察细胞迁徙能力的改变,集落形成实验观察细胞增殖能力的改变。免疫组化法观察稳定细胞株中COX-2和EGFR的表达,采用RT-PCR检测稳定细胞株中VEGF、MMP-2、EGFR mRNA的表达。结果pshCOX-2转染HT-29后72h,细胞增长明显受抑,细胞培养上清中PGE2和VEGF的表达明显下降。稳定表达pshCOX-2细胞株内COX-2蛋白和mRNA均下调,细胞越过划痕的时间为(7.67±1.53)d,与对照组有显著性差异(F=9.50,P=0.0138);细胞集落密度为(257.67±39.53)个,明显低于其平行对照组(F=37.07,P=0.0004)。免疫组化提示pshCOX-2细胞株内COX-2和EGFR表达均有下调,RT-PCR显示pshCOX-2组VEGF、MMP-2、EGFR mRNA表达下调。结论抑制COX-2活性能抑制HT-29细胞增殖和迁徙能力,同时可以抑制VEGF、EGFR、MMP-2表达和PGE2产生,有潜在的抑制肿瘤血管形成作用。
     第五部分抑制COX-2蛋白对结肠癌细胞凋亡影响的实验研究
     目的观察pshCOX-2抑制COX-2活性对HT-29细胞凋亡和对于化疗药物敏感性的影响,并探讨可能的作用机制。方法应用pshCOX-2瞬时转染HT-29细胞,联合使用5-Fu处理HT-29细胞,MTT法检测细胞的生长曲线,流式细胞仪分析凋亡率。应用TUNEL法观察稳定表达pshCOX-2的HT-29细胞凋亡形态,应用RT-PCR和Western blot法从转录水平和蛋白水平检测Bcl-2/Bax和Fas/FasL表达的变化。
     结果pshCOX-2瞬时转染3d后可以明显抑制HT-29细胞生长,与空白和阴性对照相比(P<0.05),联合使用5-Fu后在7d时细胞生长受抑作用,且呈协同作用,差异有显著性(F=231.94,P=0.0001)。流式细胞仪检测细胞DNA含量,发现转染组凋亡指数(7.07%)明显高于对照组的0.30%和0.27%(F=24.71,P=0.0013),低于联合处理组13.57%,差异有显著性(P<0.05)。TUNEL法原位凋亡指数结果与流式细胞仪结果类似,RT-PCR和Western blot结果提示稳定表达pshCOX-2细胞株Bax和Fas表达上调,Bcl-2表达呈下调趋势,而FasL表达与对照组相无显著性差异(P<0.05)。结论COX-2shRNA可以抑制细胞增殖,促进凋亡,增强肿瘤细胞对5-Fu的敏感性,COX-2shRNA促进凋亡的机制可能与上调Bax和Fas表达,下调Bcl-2表达有关。
     第六部分荷瘤鼠模型建立及COX-2shRNA对结肠癌细胞致瘤性的影响
     目的通过裸鼠体内实验,进一步验证抑制COX-2表达对结肠癌致瘤的影响并探讨可能的作用机制。方法选用BALB/C裸鼠进行体内实验。裸鼠25只。随机分为5组:A.空白对照组5只;B.阴性质粒组5只;C.转染pshCOX-2组5只;D.5-Fu处理组5只;E.5-Fu+转染pshCOX-2联合组5只。于每只裸鼠右胁腹前肢皮下分别接种等量的不同转染细胞(5×106个细胞/100ul/只),接种42d后处死老鼠。观察肿瘤生长状况、成瘤潜伏期,计算抑瘤率。免疫组化法检测瘤组织内COX-2和EGFR蛋白表达情况,测定瘤块中MVD;TUNEL法检测肿瘤的凋亡指数,透视电镜观察瘤块中肿瘤细胞凋亡情况。结果转染pshRNA组成瘤潜伏期延长、生长曲线、瘤体积和瘤重与对照组比较(P均<0.05),联合治疗组抑瘤作用更强,与转染pshRNA组相比较(P均<0.05);pshRNA组原位凋亡明显增多,透视电镜观察pshRNA组出现较多明显凋亡形态的细胞。免疫组化提示pshRNA组COX-2和EGFR表达均较对照组下调(P<0.05),瘤组织中MVD,对照组明显高于pshRNA组(P<0.05)。结论体内实验表明抑制肿瘤细胞COX-2表达可以有效抑制肿瘤的生长,促进凋亡,抑制肿瘤血管形成,同时可以增加肿瘤细胞对化疗药物敏感性,以此基因为靶点进行的小干扰RNA基因治疗,将有可能成为人类结肠癌治疗提供有效的手段。
The First Section: Study on designing and synthesis of COX-2 siRNA and optimizing transfection conditions
     Objective To design and chemically synthesize siRNA targeted on human COX-2 and optimize transfection conditions. Methods To follow the principle of selecting RNA interference target sequence, to utilize the web resource,design four potential small interference RNA (siRNA) of COX-2,and then were annealed. The FAM- negative-siRNA was transfected to HT-29 cells by lipofectamineTM2000. To optimize transfecting conditions, the fluorescence microscope was used for deciding the efficiency of transfection and the activity of cells were measured by MTT after transfecting. Results The four siRNA targeted on COX-2 were successfully constructed. The best transfection conditions was the combination with 1.5ul lipofectamineTM2000 and 40pmol siRNA in the 24 wells board. Conclusions The four siRNA targeted on COX-2 were successfully designed and constructed. The study on effects of COX-2 siRNA on HT-29 cells will be performed in the most optimal transfection conditions.
     The Second Section: Study on effects of COX-2 siRNA-mediated gene silencing on the expression of COX-2 and proliferation of human colon cancer cells
     Objective To investigate the effect of synthesized COX-2 specific siRNA on the cell proliferation and apoptosis of COX-2 overexpressing human colon cancer cells.
     Methods Human colon cancer cells of the line HT-29 were cultured and transfected with the most optimal COX-2 siRNA by screening from four siRNA. RT-PCR and Western blot were used to detect the expression of COX-2 mRNA and protein. The method of MTT was used to examine the proliferation of the cells. The apoptosis of the cells was detected by Hoechest staining. Results The expression of COX-2 mRNA and protein in the COX-2 siRNA group decreased remarkably(P<0.05),especially 72 hours after transfecting. The proliferation of HT-29 cells transfected with COX-2 siRNA did not changed significantly 24 hours after transfecting, however, decreased 48 hours after and especially 72 hours and 1 week after(P<0.05), Meanwhile, the apoptosis of the HT-29 cells transfected with COX-2 siRNA was significantly higher than that of the cells transfected with the negative COX-2 siRNA and that of the untransfected cells. Conclusions COX-2 is closely related to the proliferation and apoptosis of tumor cells, transfection of the specific siRNA targeting on COX-2 helps inhibiting the expression of COX-2, thus inhibiting the growth and enhancing the apoptosis of the tumor cells.
     The Third Section: Study on construction and identification of the eukaryotic expression plasmid for cyclooxygenase-2 specific shRNA and optimizing transfection conditions
     Objective To clone the recombinant eukaryotic expression plasmid of specific small interfering RNA (siRNA)against COX-2 gene and optimize transfection conditions. Methods The COX-2 siRNA template DNA sequence for short hairpin RNA (shRNA)was designed and synthesized. The annealed siRNA template was inserted into pGPH1-GFP-Neo plasmid. The recombinant plasmid (pshCOX-2) was transformed into DH5αstrain and identified by restrictive enzyme digestion and sequence analysis. The effect of the recombinant plasmid on the COX-2 expression of human colon cancer HT-29 cells was detected by RT-PCR and Western blot after optimizing transfection conditions. Results It was confirmed by restrictive enzyme digestion and sequence analysis that the recombinant plasmid was cloned and the aim sequence was obtained. The COX-2 expression of HT-29 cells was inhibited at mRNA and protein levels 72 hours after transfected with the recombinant plasmid. The difference is significant between the group transfected with the recombinant plasmid and the control groups (F=349.58,P=0.0001), but the difference between the control groups is not significant(P>0.05). Conclusion COX-2 shRNA expression plasmid pshCOX-2 successfully constructed can lastingly inhibit the expression of COX-2 gene in human colon cancer HT-29 cells. It may be a tool to study the relations between COX-2 and colon carcinoma in vitro and in vivo.
     The Fourth Section: Study on effects of gene silencing of COX-2 on the biological activity of HT-29 cells
     Objective To observe the effect on proliferation of HT-29 cells with short hairpin RNA targeted on COX-2 gene in vitro, and to explore the potential mechanisms. Methods After HT-29 cells were transient transfected with pshCOX-2 plasmid, growth curves were detected by MTT methods, and the methods of RIA and ELISA were respectively used to estimate the content of PGE2 and VEGF in supernatant. Cell clones which stably expressed pshCOX-2 were obtained in the medium which contained G418 after 3-4 weeks. The time of healing the wound was utilized to observe the effect on cell migration, invasion and locomotion of HT-29-pshCOX-2 cells. The numbers of clone formation were used to observe the ability of proliferation of HT-29-pshCOX-2 cells.
     The expressions of COX-2 and EGFR of HT-29- pshCOX-2 cells was judged by immunohistochemistry and the expression of VEGF, MMP-2 and EGFR mRNA was detected by RT-PCR. Results The growth of HT-29 cells was inhibited at 72h after transient transfected with pshCOX-2 and the content of PGE2 and VEGF in supernatant decreased. The expression of COX-2 of HT-29 cells which stably expressed pshCOX-2 down-regulated. Time when cells healed the wound was (7.67±1.53) d, which is longer than control groups (F=9.50 , P=0.0138). The number of clone formation was 257.67±39.53, which is few than control groups(F=37.07,P=0.0004)The expression of COX-2 and EGFR of HT-29-pshCOX-2 decreased and VEGF, MMP-2 and EGFR mRNA down-regulated significantly. Conclusions Inhibiting COX-2 expression can decrease the proliferation and invasion in HT-29 cells, downregulate the expression of VEGF,MMP-2 and EGFR mRNA and decrease the synthesis of PGE2; It may potentially inhibit angiopoiesis of tumor, which is foundation to the next in vivo experiment.
     The Fifth Section: Study on blocking COX-2 protein expression to promote HT-29 apoptosis
     Objective To observe the effect on chemotherapy- sensibility and apoptosis of HT-29 cells with short hair-pin RNA targeted on COX-2 gene in vitro, and to explore the potential mechanisms. Methods After HT-29 cells were transient transfected with pshCOX-2 plasmid, 5-Fu was sequently added or singly used, growth curves were detected by MTT methods, apoptosis rate were analyzed by flow cytometry. Morphocytology of apoptotic HT-29-pshCOX-2 cells were observed by TUNEL methods, and the expression of Bcl-2/Bax and Fas/FasL of these cells was measured by RT-PCR and Western blot. Results The growth of HT-29 cells was inhibited at 72h after transient transfected with pshCOX-2 in vitro, and it is significant compared with control groups(P>0.05). The synergistic inhibition can be found in the HT-29-pshCOX-2 cells 7d after 5-Fu used(F=231.94,P=0.0001). FCM show the apoptosis ratio is (7.07%) in pshCOX-2 group, it is statistically significance between pshCOX-2 group and control groups(0.3 % and 0.27%)(F=24.71,P=0.0013), but lower than combination group(13.57%). The results of TUNEL were the same as the FCM. The results of RT-PCR and Western blot showed that the expression of Bax and Fas up-regulated, Bcl-2 down-regulated and FasL had no changes in pshCOX-2 group. Conclusions: The stably expression of COX-2shRNA in HT-29 cells can inhibit the proliferation of HT-29 cells and promote the apoptosis of HT-29 cells. Down-regulating the expression of COX-2 by pshCOX can increase the chemotherapy- sensibility of 5-Fu.
     The Sixth Section: The therapeutic effect of COX-2 shRNA on colon carcinoma HT-29 cells in vivo
     Objective To investigate the effect of COXsiRNA gene therepy in vivo, and to explore the potential mechanisms. Methods We carried out vivo study with BALB/C/nu/nu nude mice. 25 nude mice were randomly divided into five groups, and 5×106 cells per mouse was subcutaneously injected on the right HS abdomen of mice(A)control group: 5 mice injected with HT-29 cells;(B)negative control group: 5 mice injected with HT-29pshNC cells;(C) pshCOX-2 group: 5 mice injected with HT-29pshCOX-2cells;(D)5-Fu group:5 mice injected in the abdomen with 5-Fu 50ug/kg/d×5d;(E)5-Fu+HT-29pshCOX-2 group(combination group):C+D. All mice were sacrificed 42 days later and the changes of weight of all nude mice were observed. The volume and weight of tumor were measured and the rates of tumor inhibition were evaluated. The growth curves and stage of latency of tumor were observed, and morphocytology of apoptotic cells were detected by TUNEL and TEM. The expression of COX-2, EGFR and CD34 were stained by immunohistochemistry. The microvessel density (MVD) of tumor tissue was evaluated. Results Stage of latency lengthen and growth of tumor in group C was inhibited. The volume and weight of tumors were significantly different between group C and control groups(P<0.05). The effect of inhibition of tumor in group C was weaker than combination group (P<0.05).The apoptosis in situ of tumors in group C increased, it is statistically significance compared with the control groups (P<0.05). The expression of COX-2 and EGFR decreased in group C,and the MVD of group C were scarce and punctiform. But in control group, MVD are intensive and annuliform, it is statistically significance between group C and control groups (P<0.05). Under TEM,more apoptosic cells in groupC can be found. Conclusions: By inhibiting the expression of COX-2 of mice, the growth of tumor can be inhibited; more apoptosic cells can be found in tumor, Inhibiting COX-2 expression can decrease vascularization of tumor and improve the sensibility of chemotherapy. The therapeutic effect of COXsiRNA in vivo suggested that COX-2 gene be a good target for siRNA gene therapy of colon carcinoma. We obtain a useful theoretical basis and measure for gene therapy of colon carcinoma.
引文
[1] Jemal A,Siegel R, Ward E, et al. Cancer statistics,2007. CA Cancer J Clin.2007,57:43-66
    [1]张振书,马强.结肠癌的流行病学,胃肠病学,2001,6:231-232
    [2] Marnett LJ, DuBois RN. COX-2:A Target for Cancer Prevention. Annu Rev Pharmacol Toxicol,2002,42:55-80
    [3] Appleby SB, Ristim?ki A, Neilson K, et al. Structure of the human cyclo-oxygenase-2 gene. Biochem J,1994,302:723-727
    [4] Chandrasekharan NV, Dai H, Roos KL, et al. Cox-3,a cyclooxygenase-1 variant inhibited by acetaminophen and other analgesic/antipyretic drugs:Cloning,structure,and expression. PNAS,2002,99:13926-13931
    [5] Warner TD, Mitchell JA. Cyclooxygenase-3(COX-3):Filling in the gaps toward a COX continuum? PNAS,2002,99:133371-13373
    [6] Masferrer JL,Leahy KM,Koki AT,et al. Antiangiogenic and antitumor activities of cyclooxygenase-2 inhibitors. Cancer Res,2000,60:1306-1311
    [7] Gallo O,Franchi A,Magnelli L,et al. Cyclooxygenase-2 pathway correlates with VEGF expression in head and neck cancer. Implications for tumor angiogenesis and metastasis. Neoplasia,2001,3:53-61
    [8] Gwyn K , Sinicrope FA. Chemoprevention of colorectal cancer. Am J Gastroenterol,2002,97:13-21
    [9]王磊,陈卫昌,谢学顺等,塞来昔布对实验性结肠癌原位移植瘤生长及血管形成的影响,中华肿瘤防治杂志,2006,13:1295-1300
    [10] Gr?sch S, Tegeder I, Niederberger E, et al. COX-2 independent induction of cell cycle arrest and apoptosis in colon cancer cells by the selective COX-2 inhibitor celecoxib. FASEB Letters,2001,15:2742-2744
    [11] Brummelkamp TR, Bernards R. New tools for functional mammalian cancer genetics. Nat Rev Cancer,2003, 10:781-789
    [12] Barańska M, Skretkowicz J. Prospects of gene therapy. Wiad Lek. 2007, 60:305-311
    [13] Fire A,Xu S,Montgomery MK,et al. Potent and specific interference by double- stranded RNA in Caenorhabditis elegans. Nature, 1998, 391:806-811
    [14] Hannon GJ. RNA interference.Nature, 2000, 418: 244-251
    [15] Arenz C, Schepers U. RNA interference: An ancient mechanism or a state of the art therapeutic application. Naturwissenschaften, 2003, 90:345-359
    [16] Elabashir SM, Harborth J, Lendeckel W, et al. Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature , 2001, 411:494-498
    [17] Pai SI, Lin YY, Macaes B, Meneshian A, et al. Prospects of RNA interference therapy for cancer.Gene Therapy, 2006, 13, 464–477
    [18]陈杰,白春学,张敏等,RNA干扰技术在哺乳动物中的应用。生物化学与生物物理进展,2003;30:650-654
    [19] Aoki Y, Cioca DP, Oidaira H, et al.RNA interference may be more potent than antisense RNA in human cancer cell lines. Clin Exp Pharmacol Physiol, 2003, 30:96-102
    [20] Wei Lv, Chao Zhang, Jia Hao. RNAi technology: A Revolutionary tool for the colorectal cancer therapeutics. World J Gastroenterol 2006,12: 4636-4639
    [1] Vermeulen A, Robertson B, Dalby AB, et al. Double-stranded regions are essential design components ofpotent inhibitors of RISC function. RNA, 2007,13: 723-730
    [2] Ui-Tei K, Naito Y, Takahashi F, et al. Guidelines for the selection of highly effective siRNA sequences for mammalian and chick RNA interference. Nucleic Acids Res., 2004, 32:936-948
    [3] Elabashir SM, Harborth J, Lendeckel W, et al. Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature , 2001, 411:494-498
    [4] Hannon GJ. RNA interference.Nature, 2000, 418: 244-251
    [5] Zamore PD, Tuschl T, Sharp PA, et al. RNAi: double-stranded RNA directs the ATP-dependent cleavage of mRNA at 21 to 23 nucleotide intervals. Cell, 2000, 101:25-33
    [6] Bosher JM, Labouess M. RNA interference: genetic wand and genetic watchdog. Nature Cell boil, 2000, 2:E31-E36
    [7] Clemens MJ, Elia A. PKR-a protein kinase regulated by double-stranded RNA. Int J Biochem Cell Biol. 1997, 29:945-949
    [8] Tuschl T, Zamore PD, Lehmann R, et al. Targeted mRNA degradation by double-stranded RNA in vitro. Genes Dev, 1999, 13:3191-3197
    [9] Reynolds A, Leake D, Boese Q, et al. Rational siRNA design for RNA interference. Nat Biotechnol, 2004,3:326-330
    [10] Rolland AP. Form genes to gene medicines:recent advances in nonviral gene delivery. Crit Rev Ther Grug Carrier Syst. 1998, 15:143-198
    [11] Spencer SC. Electroporation technique of DNA transfection. Applied Biochemistry and Biotechnology, 1993, 42:75-82
    [12] Walters DK, Jelinek DF. The effectiveness of double-stranded short inhibitory RNAs(siRNAs) may depend on the method of transfection. Antisense Nucleic Acid Drug Dev, 2002, 12:411-418
    [13] Wang D, Jing NH, Lin QS. Strearylamine liposome as a new efficient reagentfor DNA transfection of eukaryotic cells. Biochem Biophy Res Commu, 1996, 226:450-455
    [14] Zhang S, Zhao B, Jiang H, et al. Cationic lipids and polymers mediated vectors for delivery of siRNA. J Control Release. 2007, 123:1-10
    [15] Gilmore IR, Fox SP, Hollins AJ,et al. Delivery strategies for siRNA-mediated gene silencing.Curr Drug Deliv. 2006, 3:147-5
    [16] Byrnes AP, Rusby JE, Wood MJA, et al. Adenovirus gene transfer causes inflammation in the brain. Neuroscience, 1995, 66: 1015-1021
    [17] Liu CM, Liu DP, Dong WJ, et al. Retrovirus vector-mediated stable gene silencing in human cell. Biochemical and Biophysical Research Communications, 2004, 313: 716-720
    [18] Li S,Ma Z. Nonviral gene therapy. Curr Gene Ther, 2001, 1:201-226
    [19] Dalby B, Cates S, Harris A, et al. Advanced transfection with Lipofectamine 2000 reagent: Primary neurons, siRNA, and high-througuput applications. Methods, 2004, 33: 95-103
    [20] Djurovic S,Iversen N,Jeansson S, et al. Comparison of nonviral transfection and adeno-associated viral transduction on cardiomyocytes. Mol Biotechnol, 2004, 28:21-32
    [1] Fire A,Xu SQ,Montgomery MK,et al. Potent and specific interference by double- stranded RNA in Caenorhabditis elegans. Nature, 1998, 391:806-811
    [2] Hannon GJ. RNA interference.Nature, 2000, 418: 244-251
    [3] Arenz C, Schepers U. RNA interference: An ancient mechanism or a state of the art therapeutic application. Naturwissenschaften, 2003, 90:345-359
    [4] Elabashir SM, Harborth J, Lendeckel W, et al. Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature , 2001, 411:494-498
    [5]陈杰,白春学,张敏等,RNA干扰技术在哺乳动物中的应用。生物化学与生物物理进展,2003;30:650-654
    [6] Aoki Y, Cioca DP, Oidaira H, et al.RNA interference may be more potent than antisense RNA in human cancer cell lines. Clin Exp Pharmacol Physiol, 2003, 30:96-102
    [7] Reynolds A, Leake D, Boese Q, et al. Rational siRNA design for RNA interference. Nat Biotechnol, 2004, 3:326-330
    [8] Hammond SM. Dicing and slicing: The core machinery of the RNA interference pathway. FEBS Letters. 2005, 579 :5822–5829
    [9] He DM, Zhang Y, Liu GX. Bcl-2 small hairpin RNAs enhance radiation-induced apoptosis in A549 cells. Cell Biol Int. 2007,31:1442-1445
    [10] Brummelkamp TR, Bernards R, Agami R, Stable suppression of tumorigenicity by virus mediated RNA interference. Cancer Cell, 2002, 2: 243-247
    [11] Izquierdo M.Short interfering RNAs as a tool for cancer gene therapy. Cancer Gene Ther,2005, 3:217-227
    [12] Barańska M, Skretkowicz J. Prospects of gene therapy. Wiad Lek. 2007, 60:305-311
    [13] Appleby SB, Ristim?ki A, Neilson K, et al. Structure of the human cyclo-oxygenase-2 gene. Biochem J,1994,302:723-727
    [14] Marnett LJ, DuBois RN. COX-2:A Target for Cancer Prevention. Annu Rev Pharmacol Toxicol,2002,42:55-80
    [15] Gwyn K.,Sinicrope FA. Chemoprevention of colorectal cancer. Am J Gastroenterol,2002,97:13-21
    [16] Masferrer JL,Leahy KM,Koki AT,et al. Antiangiogenic and antitumor activities of cyclooxygenase-2 inhibitors. Cancer Res,2000,60:1306-1311
    [17] Folkman J. Angiogenesis and apoptosis. Cancer Biology,2003,13: 159–167
    [18] Tuynman JB,Peppelenbosch MP, Richel DJ. COX-2 inhibition as tool to treat and prevent colorectal cancer. Crit Rev Oncol Hematol,2004, 52:81-101
    [19] Husain SS, Szabo IL, Tamawski AS. NSAIDs inhibition of GI cancer growth: clinical implications and molecular mechanisms of action. Am J Gastroentcrol,2002,97:542-553
    [20] Hawk ET, Levin B.Colorectal Cancer Prevention. J.clin.oncology,2005,23:379-391
    [21] Gr?sch S, Tegeder I, Niederberger E,et al. COX-2 independent induction of cell cycle arrest and apoptosis in colon cancer cells by the selective COX-2 inhibitor celecoxib. FASEB Letters,2001,15:2742-2744
    [22] Aagaard L, Rossi JJ. RNAi therapeutics: principles, prospects and challenges. Adv Drug Deliv Rev. 2007, 59:75-86
    [1] Brummelkamp TR, Bernards R. New tools for functional mammalian cancer genetics. Nat Rev Cancer,2003, 10:781-789
    [2] Ui-Tei K, Naito Y, Takahashi F, Haraguchi T, et al. Guidelines for the selection of highly effective siRNA sequences for mammalian and chick RNA interference. Nucleic Acids Research,2004,32:936-948
    [3] Levenkova N, Gu Q, Rux JJ. Gene specific siRNA selector. Bioinformatics, 2004, 20:430-432
    [4] Birmingham A, Anderson E, Sullivan K, et al. A protocol for designing siRNAs with high functionality and specificity. Nat Protoc,2007, 2:2068-2078
    [5] Marnett LJ, DuBois RN. COX-2:A Target for Cancer Prevention. Annu Rev Pharmacol Toxicol,2002,42:55-80
    [6] Hannon GJ. RNA interference.Nature, 2000, 418: 244-251
    [7] Couzin J. Breakthrough of the year.Small RNAs make big splash. Science,2002,298:2296-2297
    [8] Hong J, Wei N, Chalk A, et al.Focusing on RISC assembly in mammalian cells. Biochem Biophys Res Commun. 2008, 368:703-708
    [9] Bernstein E, Caudy AA, Hammond SM, et al. Role for a bidentate ribonuclease in the initation step of RNA interference. Nature, 2001, 409:363-366
    [10] Baulcombe DC, MacFarlane SA. Gene silencing: RNA makes no protein. Curr Biol, 1999, 9: R599-R601
    [11] Elabashir SM, Martinez J, Patkaniowska A, et al. Functional anatomy of siRNAs for mediating efficient RNAi in Drosophila melanogaster embryo lysate. EMBO J, 2001c, 20:6877-6888
    [12] Martinez J, Patkaniowska A, Urlaub H, et al. Single-stranded antisense siRNAs guide target RNA cleavage in RNAi. Cell, 2002, 110:563-574
    [13] Plasterk RH, Ketting RF. The silence of the genes. Curr Opin Genet Dev, 2000, 10:562-567
    [14] Table H, Sarkissian M, Kelly WG, et al. The rde-1 gene, RNA interference,and transposon silencing in C.elegans. Cell, 1999, 99:123-132
    [15] Ketting RF, Haverkamp THA, Luenen HG, et al. Mut-7 of C.elegans, is a homolog of Werner syndrome helicase and Rnase D. Cell, 1999, 99:133-141
    [16] Dalmay T, Hamilton A, Rudd S, et al. An RNA-dependent RNA polymerase gene in Arabidopsis is required for posttranscriptional gene silencing mediated by a transgene but not by a virus. Cell, 2000, 101:543-553
    [17] Sijen T, Fleenor J, Simmer F, et al. On the role of RNA amplification in dsRNA-triggered gene silencing. Cell, 2001, 107:465-476
    [18] Stein P, Svoboda P, Anger M, et al. RNAi: Mammalian oocytes do it without RNA-dependent RNA polymerase. Rn. a A Publication of the Rn.a Society. 2003, 9:187-192
    [19] Hammond SM. Dicing and slicing: The core machinery of the RNA interference pathway. FEBS Lett. 2005, 579 :5822–5829
    [20] Aoki Y, Cioca DP, Oidaira H, et al.RNA interference may be more potent than antisense RNA in human cancer cell lines. Clin Exp Pharmacol Physiol, 2003, 30:96-102
    [21]Byrnes AP.,Rusby JE.,Wood MJ, et al. Adenovirus gene transfer causes inflammation in the brain. Neuroscience, 1995, 66: 1015-1024
    [22] Liu CM, Liu DP, Dong WJ, et al. Retrovirus vector-mediated stable gene silencing in human cell. Biochemical and Biophysical Research Communications, 2004, 313: 716-720
    [23] Misteli T, Spector DL.Applications of the green fluorescent protein in cell biology and biotechnology. Nat Biotechnol, 1997, 15: 961-964
    [1] Marnett LJ, DuBois RN. COX-2:A Target for Cancer Prevention. Annu Rev Pharmacol Toxicol,2002,42:55-80
    [2] Appleby SB, Ristim?ki A, Neilson K, et al. Structure of the human cyclo-oxygenase-2 gene. Biochem J,1994,302:723-727
    [3] Noriyuki M, Sumi T, Zhi X, et al.Vascular endothelial growth factor, matrix metalloproteinases, and cyclooxygenase-2 influence prognosis of uterine cervical cancer in young women.Int J Oncol. 2007,31:531-536
    [4] Sivula A, Talvensaari-Mattila A, Lundin J, et al. Association of cyclooxygenase-2 and matrix metalloproteinase-2 expression in human breast cancer.Breast Cancer Res Treat. 2005, 89:215-220
    [5] Masferrer JL,Leahy KM,Koki AT,et al. Antiangiogenic and antitumor activities of cyclooxygenase-2 inhibitors. Cancer Res,2000,60:1306-1311
    [6] Gallo O,Franchi A,Magnelli L,et al. Cyclooxygenase-2 pathway correlates with VEGF expression in head and neck cancer. Implications for tumor angiogenesis and metastasis. Neoplasia,2001,3:53-61
    [7] Gr?sch S, Tegeder I, Niederberger E, et al. COX-2 independent induction of cell cycle arrest and apoptosis in colon cancer cells by the selective COX-2 inhibitor celecoxib. FASEB Letters,2001,15:2742-2744
    [8] Brummelkamp TR, Bernards R. New tools for functional mammalian cancer genetics. Nat Rev Cancer,2003, 10:781-789
    [9] Barańska M, Skretkowicz J. Prospects of gene therapy. Wiad Lek. 2007, 60:305-311
    [10] Cheng TL, Chang WT.Construction of simple and efficient DNA vector-based short hairpin RNA expression systems for specific gene silencing in mammaliancells.Methods Mol Biol. 2007,408:223-241
    [11] Birmingham A, Anderson E, Sullivan K, et al.A protocol for designing siRNAs with high functionality and specificity. Nat Protoc. 2007, 2:2068-2078
    [12] Tjandrawinata RR, Dahiya R, Hughes-Fulford M. Induction of cyclooxygenase-2 mRNA by prostaglandin E2 in human prostatic cancinoma cells. Br J Cancer,1997,75:1111-1118
    [13] Sheng H,Shao J,Washington MK,et al. Prostaglandin E2 increases growth and motility of colorectal carcinoma cells. Biol. Chem,2001,276:18075-18081
    [14] Uefuji K, Ichikura T, Mochizuki H. Cyclooxygenase-2 expression is related to prostaglandin biosynthesis and angiogenesis in human gastric cancer. Clin Cancer Res,2000,6:135-138
    [15] Husain SS, Szabo IL, Tamawski AS. NSAID inhibition of GI cancer growth :clinical implications and molecular mechanisms of action. Am J Gastroentcrol,2002,97:542-553
    [16] Bisacchi D, Benelli R, Vanzetto C, et al. Anti-angiogenesis and angioprevention:mechanisms,problems and perspectives. Cancer Detection and Prevention,2003,27:229-238
    [17] Hicklin DJ,Ellis LM. Role of the Vascular Endothelial Growth Factor Pathway in Tumor Growth and Angiogenesis. J.Clin. Onco. 2005,23:1011-1027
    [18] Fang J, Shing Y, Wiederschain D, et al. Matrix metalloproteinase-2 is required for the switch to the angiogenic phenotype in a tumor model. PNAS,2000,97(8):3884-3889
    [19] Takahashi Y, Kawahara F, Noguchi M, et al. Activation of matrix metalloproteinase-2 in human breast cancer cells overexpressing cyclooxygenase-1 or–2. FEBS Letters,1999,460:145-148
    [20]Noriyuki M, Sumi T, Zhi X, et al.Vascular endothelial growth factor, matrix metalloproteinases, and cyclooxygenase-2 influence prognosis of uterine cervical cancer in young women.Int J Oncol. 2007,31:531-536
    [21] Lee EJ, Choi EM, Kim SR,et al. Cyclooxygenase-2 promotes cell proliferation, migration and invasion in U2OS human osteosarcoma cells.Exp Mol Med.2007,39:469-476
    [22] Ito H, Duxbury M, Benoit E. et al. Fibronectin-induced COX-2 mediates MMP-2 expression and invasiveness of rhabdomyosarcoma. Biochem Biophys Res Commun. 2004,318:594-600
    [23] Leigh C. Endothelial cells and VEGF in vascular development. Nature. 2005,438: 937-945
    [24]严辉,陈卫昌,蔡衍郎.胃癌及癌旁组织EGFR和KI - 67抗原表达的免疫组化研究.苏州大学学报, 2004, 24:855-860
    [25] Blanchot-Jossic F, Jarry A, Masson D, et al. Up-regulated expression of ADAM17 in human colon carcinoma: co-expression with EGFR in neoplastic and endothelial cells.J Pathol, 2005,207: 156-163
    [26]Prenzel N, Zwick E,Daub H, et al. EGFR transactivation by G-protein-coupled receptors requires metalloproteinase cleavage of proHB-EGF. Nature, 1999, 402: 884-888
    [27] Mendelsohn J,Baselga J. Status of epidermal growth factor receptor antagonists in the biology and treatment of cancer.J Clin Oncol, 2003, 21:2787- 2799
    [28] Pai R, Soreghan B, Szabo IL, et al. Prostaglandin E2 transactivates EGF receptor: a novel mechanism for promoting colon cancer growth and gastrointestinal hypertrophy. NatMed, 2002, 8:289-293
    [29] Buchanan FG, Wang D, Bargiacchi F, et al. Prostaglandin E2 regulates cell migration via the intracellular activation of the epidermal growth factor receptor. J Biol Chem, 2003,278:35451-35457
    [30] Ding YB, Shi RH, Tong JD, et a l. PGE2 up-regulates vascular endothelial growth factor expression in MKN28 gastric cancer cells via epidermal growth factor receptor signaling system. Exp Oncol, 2005, 27:108-113
    [31] Shao J, Evers BM, Sheng H. Prostaglandin E2 synergistically enhances receptor tyrosine kinase-dependent signaling system in colon cancer cells.J Biol. Chem,2004,279:14287-14293
    [1] Das D, Arber N, Jankowski JA. Chemoprevention of colorectal cancer. Digestion. 2007,76:51-67
    [2] Wang MT, Honn KV, Nie D. Cyclooxygenases, prostanoids, and tumor pro- gression.Cancer Metastasis Rev. 2007,26:525-534
    [3] Nagata S.Apoptosis mediated by the Fas system. Prog Mol Subcell Biol, 1996, 16:87-103
    [4] O'Connell J, Bennett MW, O'Sullivan GC, et al. Fas ligand expression in primary colon adenocarcinomas: evidence that the Fas counterattack is a prevalent mechanism of immun evasion in human colon cancer. J Pathol,1998,186: 240-246
    [5] Lee SH,Shin MS,Park WS,et al. Immunohistochemical localization of FAP-1,an inhibitor of Fas-mediated apoptosis,in normal and neoplastic human tissues. APMIS,1999,107:1101-1108
    [6] Butler LM,Hewett PJ,Butler WJ,et al. Down-regulation of Fas gene expression in colon cancer is not a result of allelic loss or gene rearrangement. Br J Cancer,1998,77:1454-1459
    [7] von Reyher U, Str?ter J, Kittstein W, et al. Colon carcinoma cell use mechanisms to escape CD95-mediated apoptosis.Cancer Res,1998,58: 526-534
    [8]Yoong KF,Afford SC, Randhawa S,et al. Fas/FasL ligand ineraction in human colorectal hepatic metastases: A mechanism of hepatocyte destruction to facilitate local tumor invasion. Am J Pathol,1999,154: 663-670
    [9] Mollinedo F, Gajate C.Fas/CD95 death receptor and lipid rafts: new targets for apoptosis-directed cancer therapy.Drug Resist Updat.2006,9: 51-73
    [10] Gjertsen BT, Bredholt T, Anensen N, et al.Bcl-2 antisense in the treatment of human malignancies: a delusion in targeted therapy.Curr Pharm Biotechnol. 2007,8: 373-381
    [11] Danial NN.BCL-2 family proteins: critical checkpoints of apoptotic cell death.Clin Cancer Res.2007,13: 7254-7263
    [12]陈卫昌,林茂松,张宝峰等,结肠癌组织中p53和凋亡相关基因bcl-2、bax的表达及其临床意义。胃肠病学,2006,11:195-399
    [1]杨波,庄德成,姜伟青等。人结肠腺癌裸鼠移植瘤转移模型的建立。江苏医药,2006,32:841-843
    [2] Krajewska M, Krajewski S, Epstein JI,et al. Immunohistochemical analysis of bcl-2, bax, bcl-X, and mcl-1 expression in prostate cancers. Am J Pathology,1996,148:1567-1576
    [3] Weidner N. Intratumor microvessel density as a prognostic factor in cancer. Am J Pathol,1995,147:9-19
    [4] Lu PY, Woodle MC.Delivering small interfering RNA for novel therapeutics. Methods Mol Biol.2008,437: 93-107
    [5] Takahashi Y, Nishikawa M, Takakura Y. In vivo siRNA delivery to tumor cells and its application to cancer gene therapy.Yakugaku Zasshi.2007,127: 1525-1531
    [6] Liu B. Exploring cell type-specific internalizing antibodies for targeted delivery of siRNA.Brief Funct Genomic Proteomic. 2007, 2:112-119
    [7] Whalley K. RNA interference: Breakthrough for systemic RNAi. Nat Rev Drug Discov. 2006,5:373
    [8] Wang D, DuBois RN. Cyclooxygenase 2-derived prostaglandin E2 regulates the angiogenic switch. PNAS,2004,101:415-416
    [9] Napoleone F, Robert S. K.Angiogenesis as a therapeutic target. Nature, 2005,438, 967-974
    [10] Ferrara N. Vascular Endothelial Growth Factor as a Target for Anticancer Therapy. Oncologist,2004,9:2-10
    [11] Fang J, Shing Y, Wiederschain D, et al. Matrix metalloproteinase-2 is required for the switch to the angiogenic phenotype in a tumor model. PNAS,2000,97(8):3884-3889
    [1] Hannon GJ. RNA interference.Nature, 2000, 418: 244-251
    [2] Napoli C,Lemieux C,Jorgensen R. Introduction of a chimeric chalcome synthase gene into petunia results in reversible co-suppression of homologous genes in trans. Plant Cell, 1990, 2:279-289
    [3] Guo S, Kemphues KJ. Par-1, a gene required for establishing polarity in C. elegans embryos, encodes a putative Ser/Thr kinase that is asymmetrically distributed. Cell, 1995, 81:611-620
    [4] Fire A, Xu S, Montogomery MK, et al. Potent and specific genetic inforference by double-staranded RNA in Caenorhabditis elegans. Nature, 1998, 391(6669):806-811
    [5] Arenz C, Schepers U. RNA interference: An ancient mechanism or astate of the art therapeutic application. Naturwissenschaften, 2003, 90:345-359
    [6] Hamilton A, Baulcombe DC. A species of small antisense RNA in posttranscriptional gene silencing in plants. Science, 1999, 286: 950-952
    [7] Elabashir SM, Harborth J, Lendeckel W, et al. Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature , 2001, 411:494-498
    [8] Billy E, Brondani V, Zhang H, et al. Specific interference with gene expressioninduced by double-stranded RNA in mouse embryonal teratocarcinoma cells lines. Proc Matl Acad Sci USA, 2001, 98:14428-14433
    [9] Zamore PD, Tuschl T, Sharp PA, et al. RNAi: double-stranded RNA directs the ATP-dependent cleavage of mRNA at 21 to 23 nucleotide intervals. Cell, 2000, 101:25-33
    [10] Bosher JM, Labouess M. RNA interference: genetic wand and genetic watchdog. Nature Cell boil, 2000, 2:E31-E36
    [11] Brummelkamp TR, Bernards R. A system for stable experession of short interfering RNAs in mammalian cells. Science, 2002, 296:550-553
    [12] Brummelkamp TR, Bernards R. New tools for functional mammalian cancer genetics[J]. Nat Rev Cancer, 2003, 3:781-789
    [13] Bernstein E, Caudy AA, Hammond SM, et al. Role for a bidentate ribonuclease in the initation step of RNA interference. Nature, 2001, 409:363-366
    [14] Baulcombe DC, MacFarlane SA. Gene silencing: RNA makes no protein. Curr Biol, 1999, 9: R599-R601
    [15] Elabashir SM, Martinez J, Patkaniowska A, et al. Functional anatomy of siRNAs for mediating efficient RNAi in Drosophila melanogaster embryo lysate. EMBO J, 2001c, 20:6877-6888
    [16] Martinez J, Patkaniowska A, Urlaub H, et al. Single-stranded antisense siRNAs guide target RNA cleavage in RNAi. Cell, 2002, 110:563-574
    [17] Plasterk RH, Ketting RF. The silence of the genes. Curr Opin Genet Dev, 2000, 10:562-567
    [18] Table H, Sarkissian M, Kelly WG, et al. The rde-1 gene, RNA interference, and transposon silencing in C.elegans. Cell, 1999, 99:123-132
    [19] Ketting RF, Haverkamp THA, Luenen HG, et al. Mut-7 of C.elegans, is a homolog of Werner syndrome helicase and Rnase D. Cell, 1999, 99:133-141
    [20] Hong J, Wei N, Chalk A, et al. Focusing on RISC assembly in mammalian cells. Biochem Biophys Res Commun. 2008, 368:703-708
    [21] Dalmay T, Hamilton A, Rudd S, et al. An RNA-dependent RNA polymerase gene in Arabidopsis is required for posttranscriptional gene silencing mediated by atransgene but not by a virus. Cell, 2000, 101:543-553
    [22] Sijen T, Fleenor J, Simmer F, et al. On the role of RNA amplification in dsRNA-triggered gene silencing. Cell, 2001, 107:465-476
    [23] Stein P, Svoboda P, Anger M, et al. RNAi: Mammalian oocytes do it without RNA-dependent RNA polymerase. Rn. a A Publication of the Rn.a Society. 2003, 9:187-192
    [24] Hammond SM. Dicing and slicing: The core machinery of the RNA interference pathway. FEBS Lett. 2005, 579 :5822–5829
    [25] Aufsatz W, Mette MF, van der Winden J, et al. HDA6, a putative histone deacetylase needed to enhance DNA methylation induced by double-stranded RNA. EMBO J, 2002, 21, 6832-6841
    [26] Aravin AA, Hannon GJ, Brennecke J. The Piwi-piRNA pathway provides an adaptive defense in the transposon arms race. Science. 2007, 318:761-764
    [27] Baulcombe D. An RNA microcosom. Science, 2002, 297:2002-2003
    [28] Clemens MJ. PKR-a protein kinase regulated by double-stranded RNA. Int J Biochem Cell Biol. 1997, 29:945-949
    [29] Tuschl T, Zamore PD, Lehmann R, et al. Targeted mRNA degradation by double-stranded RNA in vitro. Genes Dev, 1999, 13:3191-3197
    [30] Reynolds A, Leake D, Boese Q, et al. Rational siRNA design for RNA interference. Nat Biotechnol, 2004, 3:326-330
    [31] Amarzguioui M, Rossi JJ, Kim D. Approaches for chemically synthesized siRNA and vector-mediated RNAi. FEBS Lett, 2005, 579:5974-5881
    [32] Liu C M, Liu D P, Dong W J, et al. Retrovirus vector-mediated stable gene silencing in human cell. Biochemical and Biophysical Research Communications, 2004, 313: 716-720.
    [33] Castanotto D, Li H, Rossi JJ, Functional siRNA expression from transfected PCR products. RNA, 2002, 8:1454-1460
    [34] Castanotto D, Rossi JJ. Construction and transfection of PCR products expressing siRNAs or shRNAs in mammalian cells. Methods Mol Biol. 2004, 252:509-14
    [35] Spencer SC. Electroporation technique of DNA transfection. Applied Biochemistry and Biotechnology, 1993, 42:75-82
    [36] Walters DK, Jelinek DF. The effectiveness of double-stranded short inhibitory RNAs(siRNAs) may depend on the method of transfection. Antisense Nucleic Acid Drug Dev, 2002, 12:411-418
    [37] Wang D, Jing N, Lin Q. Strearylamine liposome as a new efficient reagent for DNA transfection of eukaryotic cells. Biochem Biophy Res Commu, 1996, 226:450-455
    [38] Zhang S, Zhao B, Jiang H, et al. Cationic lipids and polymers mediated vectors for delivery of siRNA. J Control Release. 2007, 123:1-10
    [39] Matsuda T, Cepko CL. Analysis of gene function in the retina. Methods Mol Biol. 2008; 423:259-78
    [40] Fu D, Uauy C, Blechl A, et al. RNA interference for wheat functional gene analysis. Transgenic Res. 2007, 16:689-701
    [41] Farzana L, Brown SJ. Hedgehog signaling pathway function conserved in Tribolium segmentation. Dev Genes Evol. 2008,218:181-192
    [42] Berra E, Pouysségur J. The silencing approach of the hypoxia-signaling pathway. Methods Enzymol. 2007,435:107-121
    [43] Volarevic M, Smolic R, Wu CH, et al.Potential role of RNAi in the treatment of HCV infection. Expert Rev Anti Infect Ther. 2007, 5:823-831
    [44] Ma Y, Chan CY, He ML. RNA interference and antiviral therapy. World J Gastroenterol. 2007, 13:5169-5179
    [45] Watanabe T, Umehara T, Kohara M. Therapeutic application of RNA interference for hepatitis C virus. Adv Drug Deliv Rev. 2007, 59:1263-1276
    [46] Haasnoot J, Westerhout EM, Berkhout B. RNA interference against viruses: strike and counterstrike. Nat Biotechnol. 2007, 25:1435-1443
    [47] Susan L. Uprichard The therapeutic potential of RNA interference. FEBS Letters 2005, 579: 5996–6007
    [48] Micklem DR, Lorens JB. RNAi screening for therapeutic targets in human malignancies. Curr Pharm Biotechnol. 2007, 8:337-43.
    [49] Sumimoto H, Kawakami Y. Lentiviral vector-mediated RNAi and its use forcancer research. Future Oncol. 2007, 3:655-664
    [50] Munkácsy G, Tulassay Z, Gyorffy B. RNA interference and its clinical applications. Orv Hetil. 2007, 148:2235-2240
    [51] Amarsanaa J, Fumihiko K, Hideaki I,et al. Single small-interfering RNA expression vector for silencing multiple transforming growth factor-b pathway components. Nucleic Acids Res, 2005, 33:1-9
    [52] Khoury M, Jorgensen C, Apparailly F. RNAi in arthritis: prospects of a future antisense therapy in inflammation. Curr Opin Mol Ther. 2007, 9:483-489
    [53] Orlacchio A, Bernardi G, Orlacchio A, et al. RNA interference as a tool for Alzheimer's disease therapy. Mini Rev Med Chem. 2007, 7):1166-1176
    [54] Yokota T. Gene therapy of ALS with short interfering RNA. Brain Nerve. 2007, 59:1187-1194
    [55] Kolfschoten IG, Regazzi R. Technology Insight: small, noncoding RNA molecules as tools to study and treat endocrine diseases. Nat Clin Pract Endocrinol Metab. 2007, 3:827-834
    [56] Wang J, Theunissen TW, Orkin SH. Site-directed, virus-free, and inducible RNAi in embryonic stem cells. Proc Natl Acad Sci U S A. 2007, 104:20850-20855
    [57] Ding L, Buchholz F. RNAi in embryonic stem cells. Stem Cell Rev. 2006, 2:11-18
    [58] Lykke-Andersen K. Regulation of gene expression in mouse embryos and its embryonic cells through RNAi. Mol Biotechnol. 2006, 34:271-278
    [59] Heidersbach A, Gaspar-Maia A, McManus MT, et al.RNA interference in embryonic stem cells and the prospects for future therapies.Gene Ther. 2006, 13:478-486
    [60] Elmén J, Lindow M, Schütz S, et al. LNA-mediated microRNA silencing in non-human primates. Nature. 2008, 452: 26-29
    [61] Kleinman ME, Yamada K, Takeda A, et al. Sequence- and target-independent angiogenesis suppression by siRNA via TLR3. Nature. 2008, 452:591-597
    [62] Visintin M, Melchionna T, Cannistraci I, et al. In vivo selection of intrabodies specifically targeting protein-protein interactions: A general platform for an"undruggable" class of disease targets. J Biotechnol. 2008, 134:129-135
    [63] Liu B. Exploring cell type-specific internalizing antibodies for targeted delivery of siRNA. Brief Funct Genomic Proteomic. 2007, 6:112-119
    [64] Whalley K. RNA interference: Breakthrough for systemic RNAi. Nat Rev Drug Discov. 2006, 5:373
    [65] Aagaard L, Rossi JJ. RNAi therapeutics: principles, prospects and challenges. Adv Drug Deliv Rev. 2007, 59:75-86
    [66] Chakraborty C. Potentiality of small interfering RNAs (siRNA) as recent therapeutic targets for gene-silencing. Curr Drug Targets. 2007, 8:469-482