慢病毒介导Med19基因沉默对胃癌细胞生物学行为的影响
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
[目的]:探讨Med19基因在胃癌组织中的表达,以及应用RNA干扰技术抑制Med19基因表达对胃癌细胞生物学行为的影响。
     [方法]:检测胃癌组织中Med19的表达并分析其与胃癌临床病理的关系,利用基因重组技术构建靶向Med19的shRNA慢病毒载体,感染胃癌SGC7901和MGC803细胞,检测Med19 mRNA及蛋白抑制效率,并观察其对胃癌细胞增殖、周期和克隆形成能力的影响。
     [结果]:胃癌组织中的Med19的表达显著高于癌旁组织,与肿瘤的大小、细胞分化程度及TNM分期相关。重组shRNA慢病毒载体能有效感染胃癌细胞,下调Med19基因的表达,并抑制肿瘤细胞增殖,使细胞周期阻滞于G1期,克隆形成能力显著减弱,但对肿瘤的侵袭转移没有明显的影响。
     [结论]:Medl9在肿瘤组织中特异性高表达可能是胃癌形成的一个原因,慢病毒介导的RNAi能有效沉默胃癌细胞Med19基因及蛋白表达,抑制体内胃癌细胞增殖并影响细胞周期,有望成为肿瘤基因治疗的新分子靶标。
Background
     Gastric cancer is the most common malignant tumor of the gastrointestinal tract, according to statistics, in the world, the mortality rate ranks second malignant tumors, about 60%of the world's new cases of gastric cancer in China, Japan and other Southeast Asian Countries. Surgical treatment of gastric cancer is still being considered the most direct and most effective means, but after 5-year survival rate is still low. According to 2009 census, although the treatment of gastric cancer has been significant progress, but the cancer patient's overall survival rate after 5 years is still less than 25%.
     Tumor invasion and metastasis of tumor cells and tumor tissue micro-environment interaction. Now that the line of gastric cancer recurrence and metastasis of postoperative gastric cancer patients is the most fundamental reason of death, but on the process of gastric cancer metastasis and the molecular mechanisms of signal pathway is still not very clear, the process may involve multiple molecular mechanisms and Signal transduction pathways, such as MAPK/ERK, NF-κB and PI3K/AKT and so on.
     Large number of experiments have shown that the incidence of gastric cancer process is the interaction between environmental and genetic factors in the outcome, is a cancer gene activation and inactivation of tumor suppressor gene-based multi-step, multi-stage process, and these genes the role of expression products, that is, changes in the body and disrupt cell growth, differentiation and metabolism and other important processes, which lead to cell cancer. Therefore, if you can find a way to block oncogene activation and/ or tumor suppressor gene inactivation siRNA, then there may inhibit gastric cancer. More traditional, such as gene knockout, antisense and ribozymes and other methods is often impossible to completely inhibit or reverse rumor cell growth, comparatively speaking, RNAi technology is different, because the technology can also suppress a number of different genes, and the inhibitory effect of these genes also interfere with each other. In addition, RNAi can be precisely identified to a nucleotide, for the wild-type form of cancer gene mutation, such as ras, p53, etc., are able to produce accurate and effective seal, while the wild-type gene is not affected. A large number of experimental results indicate that, RNAi technology can inhibit the cancer-specific gene mutations in cancer-related genes or gene over-expression and inhibit tumor development, and rapid identification of new tumor target goal, so this technology in cancer Gene therapy has a bright application prospect.
     Objective
     Of Med19 gene in gastric cancer, cancer tissue and normal tissue, as well as application of RNA interference inhibition of Medl9 expression in gastric cancer cell biological behavior.
     Methods
     1. collection of our department 60 cases of human gastric cancer tissue specimens, including 36 male,24 female, mean age 58.2 years, all the specimens were pathologically confirmed gastric adenocarcinoma, all specimens were not received preoperative radiotherapy and chemotherapy. Histological examination of 20 cases of normal gastric mucosa from endoscopic biopsy specimens. Pathological types:adenocarcinoma in 13 cases, 15 cases of differentiated adenocarcinoma, poorly differentiated adenocar-cinoma in 26 cases. TNM stage:Ⅰ8 cases,ⅡPhaseⅢof the 13 cases 19 cases 18 cases ofⅣ. The group of 60 patients were clinically followed up for 48-62 months, the survival time 8-62 months, the median survival time of 37 months.
     Immunohistochemistry and real-time PCR to detect Med19 gene in gastric cancer, gastric cancer and adjacent normal gastric tissues.
     2. re si-Med19 lentiviral vector and packaging
     The recombinant lentiviral SGC7901 gastric cancer cells, quantitative PCR analysis of RNAi on SGC7901 the Med19 mRNA Abundance.
     3. application to build a good Med19 shRNA lentiviral gene and MGC803 SGC7901 cells, the experiment is divided into blank control, negative control and RNAi three groups of three duplicate wells, selected by preliminary experiments, the cells infected with reasonable parameters. Infection in more than 80%efficiency, to ensure normal cell morphology, cell outlines clear, relatively clean background. Successful expansion of infected cells cultured for subsequent functional experiments. Med19 in gene silencing by MTT method five days of continuous cell growth data, and the cell growth curve. The experimental group using PI staining of gastric cancer cells and the cell cycle by flow cytometry.
     Results
     1. Med19 positive expression in gastric cancer was 71.7%(43/60), significantly higher than in adjacent normal gastric tissue and gastric tissue (P <0.01). Gastric tissue and adjacent normal gastric tissues were negative and more visible expression of Medl9, a small number of weakly positive, no significant difference between the two sex (P> 0.05). Med19 expression in gastric cancer cell differentiation, tumor size, TNM staging (P<0.05), but in a different age, gender, lymph node metastasis, invasive depth of gastric cancer samples showed no significant expression (P> 0.05).
     2. successfully constructed the MED19-shRNA lentiviral vectors, lentiviral transfected SGC7901, the mRNA can inhibit gene expression of Medl9 up to 81.8%, while in Western experiments also confirmed the Med19 protein was significantly inhibited, indicating that the recombinant lentiviral shRNA Med19 significantly reduced the expression of genes, the RNAi effect is effective.
     3. application to build a good Med19 shRNA lentiviral gene and MGC803 SGC7901 cells, gene silencing Med19 by MTT method five days of continuous cell growth data, and the cell growth curve and found that inhibition of Med19 gene can significantly inhibit cell Proliferation, RNAi gene silencing group that Med19 group, compared with the negative control group, cell proliferation significantly decreased from the second day since. Flow cytometry showed that cells were cell cycle arrest in Gl cell cycle phase, that is, DNA synthesis phase, a large number of cells can not properly copied into the division process.
     Conclusion
     1. Med19 gene in gastric cancer tissues showed high expression, and its expression was significantly higher than adjacent tissues and normal tissues. Med19 expression in gastric cancer and gastric cancer tumor size, differen-tiation, TNM stage, but not with age, sex, depth of invasion, lymph node metastasis has nothing to do. Med19 expression in larger tumors, poor differentiation, TNM stage were significantly higher than the tumor is small, well differentiated and those with low TNM stage.
     2. recombinant lentiviral shRNA significantly reduced Med19 gene expression.
     3. shRNA slow virus infection MGC803 cells SGC7901 and Med19 gene silencing, can significantly inhibit cell proliferation, cell cycle cells were arrested in G1 phase, which concluded that Med19 gene in gastric cancer is a tumor promoting gene. We found that, Med19 gene silencing ability of colony formation of gastric cancer is weak, a single clone of cells decreased signifi-cantly. Med19 suppression gene can significantly reduce the formation of SGC7901 and MGC803 cell clones, suggesting that gene expression can be reduced Med19 abnormal proliferation of cells to be controlled.
     4. Med19 highly specific expression in gastric tumor formation may be a cause of lentivirus-mediated RNAi can effectively inhibit gastric cancer cell Med19 gene and protein expression, inhibit cell proliferation in vivo and influence the cell cycle, is expected to be tumor-targeting New molecular pathway of gene therapy.
引文
[1]Kim Y J. A multiprotein mediator of transcriptional activation and its interaction with the C-terminal repeat domain of RNA polymerase Ⅱ. Cell, 1994,77 (4):599-608
    [2]Bjorklund S, Gustafsson CM. The yeast Mediator complex and its regulation [J]. Trends Biochem Sci,2005; 30:240.
    [3]Chadick, J.Z. and F.J. Asturias, Structure of eukaryotic Mediator complexes. Trends Biochem Sci,2005.30(5):264-71.
    [4]Boyer T G, Mammalian Srb/Mediator complex is targeted by adenovirus E1 A protein. Nature,1999, IJJ (6733):276-279
    [5]Boube, M., et al., Evidence for a mediator of RNA polymerase Ⅱ transcriptional regulation conserved from yeast to man. Cell,2002.110(2): 143-51.
    [6]Rosenblum-Vos, L.S., et al., The ROX3 gene encodes an essential nuclear protein involved in CYC7 gene expression in Saccharomyces cerevisiae. Mol Cell Biol,1991.11(11):5639-47.
    [7]Gustafsson, CM., et al., Identification of Rox3 as a component of mediator and RNA polymerase Ⅱ holoenzyme. J Biol Chem,1997.272(1): 48-50.
    [8]Beve, J., et al., The structural and functional role of Med5 in the yeast Mediator tail module. J Biol Chem,2005.280(50):p.41366-72.
    [9]Singh, H., et al., A functional module of yeast mediator that governs the dynamic range of heat-shock gene expression. Genetics,2006.172(4): 2169-84.
    [10]Baidoobonso, S.M., B.W. Guidi, and L.C. Myers, Med19(Rox3) regulates Intermodule interactions in the Saccharomyces cerevisiae mediator complex. J Biol Chem,2007.282(8):p.5551-9.
    [11]Ding, N., et al., MED 19 and MED26 are synergistic functional targets of the RE1 silencing transcription factor in epigenetic silencing of neuronal gene expression. J Biol Chem,2009.284(5):p.2648-56.
    [12]Sato S, Tomomori-Sato C, Parmely TJ, Florens L, Zybailov B, Swanson SK et al. A set of consensus mammalian mediator subunits identified by multidimensional protein identification technology. Mol Cell 2004; 14: 685-691.
    [13]Baidoobonso SM, Guidi BW, Myers LC. Med19(Rox3) regulates Intermodule interactions in the Saccharomyces cerevisiae mediator complex. J Biol Chem 2007; 282:5551-5559.
    [14]Sato S, Tomomori-Sato C, Banks CA, Parmely TJ, Sorokina I, Brower CS et al., A mammalian homolog of Drosophila melanogaster transcriptional coactivator intersex is a subunit of the mammalian Mediator complex. J Biol Chem 2003; 278:49671-49674
    [15]唐恩洁,杨健.基因组免疫系统[J].国外医学免疫学分册,2005,28(1):31-37.
    [16]PERSENGIEV S P, ZHU X, GREEN M R. Nonspecific, concentration-dependent stimulation and repression of mammalian gene expression by small interfering RNAs (siRNAs) [J]. RNA,2004,10 (1): 12-18.
    [17]谭余良,殷勤伟.RNAi在癌症治疗中的应用[J].药学学报,2005,40(3):193-198.
    [18]钱海利.RNA干扰技术的应用[J].国外医学肿瘤学分册,2005,32(4):261-264.
    [19]HAHN W C, WEINBERG R A. Modelling the molecular circuitry of cancer [J]. Nat Rev Cancer,2002,2 (5):331-341.
    [20]Jemal A, Siegel R, Ward E, Hao Y, Xu J, Thun MJ. Cancer statistics. CA Cancer J Clin,2009,59:225-249.
    [21]Brummelkamp TR, Nijman SM, Dirac AM, Bernards R. Loss of the cylindromatosis tumour suppressor inhibits apoptosis by activating NF-kappaB. Nature 2003; 424:797-801.
    [22]Framp A. Diffuse gastric cancer. Gastroenterol Nurs 2006; 29: 232-236; quiz 236-238.
    [23]Jinawath N, Furukawa Y, Nakamura Y. Identification of NOL8, a nucleolar protein containing an RNA recognition motif (RRM), which was overexpressed in diffuse-type gastric cancer. Cancer Sci 2004; 95:430-435.
    [24]刘娜,毕锋,潘阳林,薛妍,韩者艺,刘长江,樊代明.RhoC在胃癌细胞中的表达及其小干扰RNA表达载体的构建与鉴定.细胞与分子免疫学杂志2004;20:148-151.
    [25]Ito R, Nakayama H, Yoshida K, Matsumura S,Oda N, Yasui W. Expression of Cbl linking withthe epidermal growth factor receptor system is associated with tumor progression and poor prognosis of human gastric carcinoma. Virchows Arch 2004; 444:324-331.
    [26]Tanida S, Joh T, Itoh K, Kataoka H, Sasaki M, Ohara H, Nakazawa T, Nomura T, Kinugasa Y, Ohmoto H, Ishiguro H, Yoshino K, Higashiyama S, Itoh M. The mechanism of cleavage of EGFR ligands induced by inflammatory cytokines in gastric cancer cells. Gastroenterology 2004; 127:559-569.
    [27]Yan C, Zhu ZG, Yu YY, Ji J, Zhang Y, Ji YB, Yan M, Chen J, Liu BY, Yin HR, Lin YZ. Expression of vascular endothelial growth factor C and chemokine receptor CCR7 in gastric carcinoma and their values in predicting lymph node metastasis. World J Gastroenterol 2004; 10:783-790.
    [28]徐文华,葛银林,徐宏伟,王秀丽,耿芳宋.VEGF基因表达抑制对胃腺癌细胞SGC-7901增殖的影响.世界华人消化杂志2006;14:655-659.
    [29]Xue Y, Bi F, Zhang X, Pan Y, Liu N, Zheng Y, Fan D. Inhibition of endothelial cell proliferation by targeting Racl GTPase with small interference RNA in tumor cells. Biochem Biophys Res Commun 2004; 320:1309-1315.
    [30]Meng F, Ding J, Liu N, Zhang J, Shao X, Shen H, Xue Y, Xie H, Fan D. Inhibition of gastric cancer angiogenesis by vector-based RNA interference for Raf-1. Cancer Biol Ther 2005; 4:113-117.
    [31]Katoh M, Katoh M. Pharmacogenomics on gastric cancer. Cancer Biol Ther 2004; 3:566-567.
    [32]Thomas H, Coley HM. Overcoming multidrug resistance in cancer: an update on the clinical strategy of inhibiting p-glycoprotein. Cancer Control 2003; 10:159-165.
    [33]Rittierodt M, Harada K. Repetitive doxorubicin treatment of glioblastoma enhances the PGP expression-a special role for endothelial cells. Exp Toxicol Pathol 2003; 55:39-44.
    [34]Stege A, Priebsch A, Nieth C, Lage H. Stable and complete overcoming of MDR1/P-glycoproteinmediated multidrug resistance in human gastric carcinoma cells by RNA interference. Cancer Gene Ther 2004; 11: 699-706.
    [35]Hao Z, Li X, Qiao T, Du R, Hong L, Fan D. CIAPIN1 confers multidrug resistance by upregulating the expression of MDR-1 and MRP-1 in gastric cancer cells. Cancer Biol Ther 2006; 5:261-266.
    [36]Shen H, Pan Y, Han Z, Hong L, Liu N, Han S, Yao L, Xie H, Zhaxi C, Shi Y, Fan D. Reversal of multidrug resistance of gastric cancer cells by downregulation of TSG101 with TSG101siRNA. Cancer Biol Ther 2004; 3: 561-565.
    [37]Ning H, Li T, Zhao L, Li T, Li J, Liu J, Liu Z, Fan D. TRF2 promotes multidrug resistance in gastric cancer cells. Cancer Biol Ther 2006; 5:950-956.
    [38]Shankar P, Manjunath N, Lieberman J. The prospect of silencing disease using RNA interference. JAMA 2005; 293:1367-1373.
    [1]Latchman DS. Transcription factors:an overview [J]. Int J Biochem Cell Biol 1997,29(12):1305-1312.
    [2]Cramer P. RNA polymerase Ⅱ structure from core to functional complexes [J]. Curr Opin Genet Dev,2004,14(2):218-226.
    [3]Bjorklund S, Gustafsson CM. The yeast Mediator complex and its regulation [J]. Trends Biochem Sci 2005,30(5):240-244.
    [4]Kelleher RJ, Flanagan PM, Kornberg RD. A novel Mediator between activator proteins and the RNA polymerase Ⅱ transcription apparatus [J]. Cell 1990,61(7):1209-1215.
    [5]Flanagan PM, Kelleher RJ, Sayre MH, et al. A mediator required for activation of RNA polymerase Ⅱ transcription in vitro [J]. Nature 1991, 350(6317):436-438.
    [6]Kim YJ, Bjorklund S, Li Y, et al. A multiprotein mediator of transcriptional activation and its interaction with the C-terminal repeat domain of RNA polymerase Ⅱ [J]. Cell 1994,77 (4):599-608.
    [7]Bourbon HM, Aguilera A, Ansari A, et al. A unified nomenclature for protein subunits of mediator complexes linking transcriptional regulators to RNA polymerase Ⅱ [J]. Mol Cell,2004,14 (5):553-557.
    [8]Boube M, Joulia L, Cribbs DL, et al. Evidence for a mediator of RNA polymerase Ⅱ transcriptional regulation conserved from yeast to man [J]. Cell 2002,110(2):143-151.
    [9]Myers LC, Gustafsson CM, Bushnell DA, et al. The Med proteins of yeast and their function through the RNA polymerase Ⅱ carboxy-terminal domain [J]. Genes Dev 1998,12(1):45-54.
    [10]Guglielmi B, Berkum NL, Klapholz B, et al. A high resolution protein interaction map of the yeast Mediator complex [J]. Nucleic Acids Res, 2004,32(18):5379-5391.
    [11]Koleske AJ, Young RA. The RNA polymerase Ⅱ holoenzyme and its implications for gene regulation [J]. Trends Biochem Sci,1995,20(6):113-116.
    [12]Naar AM, Beaurang PA, Zhou S, et al. Composite co-activator ARC mediates chromatin-directed transcriptional activation [J]. Nature 1999, 398(6730):828-832.
    [13]Rachez C, Lemon BD, Suldan Z, et al. Ligand-dependent transcription activation by nuclear ccurate receptors requires the DRIP complex [J]. Nature 1999,398(6730):824-828.
    [14]Malik S, Gu W, Wu W, et al. The USA derived transcriptional coactivator PC2 is a submodule of TRAP/SMCC and acts synergistically with other PC's [J]. Mol Cell 2000,5(4):753-760.
    [15]Ryu S, Tjian R. Purification of transcription cofactor complex CRSP [J]. ProcNatl Acad Sci USA 1999,96(13):7137-7142.
    [16]Jiang Y, Veschambre P, Erdjument-Bromage H, et al. Mammalian mediator of transcriptional regulation and its possible role as an end-point of signal transduction pathways [J]. Proc Natl Acad Sci USA 1998,95(15): 8538-8543.
    [17]Brower CS, Sato S, Tomomori-Sato C, et al. Mammalian mediator subunit MED8 is an Elongin BC interacting protein that can assemble with Cul2 and Rbx1 to reconstitute a ubiquitin ligase. Proc Natl Acad Sci USA 2002, 99(16):10353-10358.
    [18]Dotson MR, Yuan CX, Roeder RG, et al. Structural organization of yeast and mammalian mediator complexes [J]. Proc Natl Acad Sci USA 2000, 97(26):14307-14310.
    [19]Taatjes DJ, Naa AM, Andel F, et al. Structure, function, and activator-induced conformations of the CRSP coactivator [J]. Science,2002, 295(5557):1058-1062.
    [20]Ito M, Yuan, CX, Okano HJ, et al. Involvement of the TRAP220 Component of the TRAP/SMCC Coactivator Complex in Embryonic Development and Thyroid Hormone Action [J]. Mol Cell 2000,5(4):683-693.
    [21]Baek HJ, Malik S, Qin J, et al. Requirement of TRAP/Mediator for Both Activator-Independent and Activator-Dependent Transcription in Conjunction with TFIID-Associated TAFIIs [J]. Mol Cell Biol 2002,22(8): 2842-2852.
    [22]Sato S, Tomomori-Sato C, Banks CA, et al. Identification of mammalian Mediator subunits with similarities to yeast Mediator subunits Srb5, Srb6, Med11, and Rox3 [J]. J Biol Chem 2003,278(17):15123-5127.
    [23]Bosveld F, van Hoek S, Sibon OCM. Establishment of cell fate during early Drosophila embryogenesis requires transcriptional mediator subunit dMED31 [J]. Dev Biol 2008,313(2),802-813.]
    [24]Risley MD, Clowes C, Yu M, et al. The Mediator complex protein Med31 is required for embryonic growth and cell proliferation during mammalian development [J]. Dev Biol,2010 Mar 27. [Epub ahead of print].
    [25]Casamassimi A, Napoli C. Mediator complexes and eukaryotic transcription regulation:An overview [J]. Biochimie 2007,89(12):1439-1446.
    [26]Asturias FJ, Jiang YW, Myers LC, et al. Conserved structures of mediator and RNA polymerase Ⅱ holoenzyme [J]. Science,1999,283 (5404): 985-987.
    [27]Akoulitchev S, Chuikov S, Reinberg D, et al. TFIIH is negatively regulated by cdk8- containing complexes [J]. Nature,2000,407 (6800): 102-106.
    [28]Park JM, Kim HS, Han SJ, et al. In vivo requirement of activator-specific binding targets of mediator [J]. Mol Cell Biol,2000,12 (23): 8709-8719.
    [29]Yudkovsky N, Ranish J A, Hahn S. A transcription reinitiation intermediate that is stabilized by activator [J]. Nature,2000,324 (6809): 225-229.
    [30]Bjorklund S, Gustafsson CM. The yeast Mediator complex and its regulation [J]. Trends Biochem Sci,2005,52 (5):240-244.
    [31]Gu W, Malik S, Ito M, et al. A novel human SRB/MED-containing cofactor complex, SMCC, involved in transcription regulation [J]. Mol Cell, 1999,5(1):97-108.
    [32]Yoon CK, Qiu H, Govind H, et al. Simultaneous recruitment of coactivators by Gcn4p stimulates multiple Steps of Transcription in vivo [J]. Mol Cell Biol,2005,16 (13):5626-5638.
    [33]Malik S, Roeder RG. Dynamic regulation of pol II transcription by the mammalian Mediator complex [J]. Trends Biochem Sci,2005,52 (5): 256-263.
    [34]Rosenblum-Vos LS, Rhodes L, Evangelista CC, et al. The ROX3 gene encodes an essential nuclear protein involved in CYC7 gene expression in Saccharomyces cerevisiae [J]. Mol Cell Biol,1991,11(11):5639-5647.
    [35]Gustafsson CM, Myers LC, Li Y, et al. Identification of Rox3 as a component ofmediator and RNA polymerase Ⅱ holoenzyme [J]. Biol Chem, 1997,272(1):48-50.
    [36]Brown TA, Evangelista C, Trumpower BL. Regulation of nuclear genes encoding mitochondrial proteins in Saccharomyces cerevisiae [J]. J Bacteriol,1995,177(23):6836-6843.
    [37]Qiu H, HU C, Yoon S, et al. An array of coactivators is required for optimal recruitment of TATA binding protein and RNA polymerase Ⅱ by promoter-bound Gcn4p [J]. Mol Cell Biol,2004,24 (10):4104-4117.
    [38]Singh H, Kremer SB, Duttweiler C, et al. A functional module of yeast mediator that governs the dynamic range of heat-shock gene expression [J]. Genetics,2006,172 (4):2169-2184.
    [39]Song W, Treich I, Qian N, et al. SSN genes that affect transcriptional repression in Saccharomyces cerevisiae encode SIN4, ROX3, and SRB proteins associated with RNA polymerase Ⅱ [J]. Mol Cell Biol,1996,16(1):115-120.
    [40]Tabtiang RK, Herskowitz Ⅰ. Nuclear proteins Nut1p and Nut2p cooperate to negatively regulate a Swi4p-dependent lacZ reporter gene in Saccharomyces cerevisiae [J]. Mol Cellular Biol,1998,18(8):4707-4718.
    [41]Becerra M, Lombardia Ferreira LJ, Hauser NC, et al. The yeast transcriptome in aerobic and hypoxic conditions:effects of hap1, rox1, rox3 and srb10 deletions [J]. Mol Microbiol,2002,43(2):545-555.
    [42]van de Peppel J, Kettelarij N, van Bakel H, et al. Mediator expression profiling epistasis reveals a signal transduction pathway with antagonistic submodules and highly specific downstream targets [J]. Mol Cell 2005,19 (4): 511-522.
    [43]Baidoobonso SM, Guidi BW, Myers LC. Med19 (Rox3) regulates Intermodule interactions in the Saccharomyces cerevisiae mediator complex [J]. J Biol Chem,2007,282(2):5551-5559.
    [44]Lee YC, Park JM, Min S, et al. An activator binding module of yeast RNA polymerase Ⅱ holoenzyme [J]. Mol Cell Biol,1999,19 (4):2967-2976.
    [45]Linder T, Zhu X, Baraznenok V, et al. The classical srb4-138 mutant allele causes dissociation of yeast Mediator [J]. Bioch Biophys Res Commun, 2006,349 (3):948-953.
    [46]Ding N. MED 19 and MED26 are synergistic functional targets of the RE1 silencing transcription factor in epigenetic silencing of neuronal gene expression [J]. J Biol Chem,2009.284(5):2648-2656.
    [47]Sato S, Tomomori-Sato C, Parmely TJ, et al. A set of consensus mammalian mediator subunits identified by multidimensional protein identification technology [J]. Mol Cell,2004,14 (5):685-691.
    [48]梁志欣.肺癌转移相关蛋白LCMR1的原核表达、多克隆抗体制 备及其应用的初步研究[C].中国人民解放军军医进修学院(硕士毕业论文),2004.
    [49]张郁.LCMR1反义寡核苷酸对高转移肺大细胞癌细胞系295D细胞生物学行为的影响及LCMR1基因功能的初步研究[C].中国人民解放军军医进修学院(硕士毕业论文),2004.
    [50]李金东,金成彦,王荣有等.siRNA-Med19对人肺癌A549细胞Med19转录、表达的影响[J].中国老年学杂志,2009,29(9):1072-1074.
    [1]Kornberg Roger D. Mediator and the mechanism of transcriptional activation. Trends in Biochemical Sciences.2005,30(5):235-239
    [2]Dotson M R, Yuan C X, Roeder R G, et al. Structural organization of yeast and mammalian mediator comp lexes [J]. Proc NatiAcad Sci USA, 2000;97:14307-14310.
    [3]Bjorklund Stefan, Gustafsson Claes M.The yeast Mediator complex and its regulation. Trends in Biochemical Sciences.2005,30(5):240-244
    [4]Conaway, R. C., Sato, S., Tomomori-Sato, C., Yao, T., and Conaway, J. W. The mammalian Mediator complex and its role in transcrip tional regulation [J].Trends Biochem Sci,2005; 30:250-251
    [5]Samuelsen C O, Baraznenok V, Khorosjutina O, et al. TRAP230 /ARC240 and TaRAP240/ARC250 Mediator subunits are functionally conserved through evolution [J].Pro Nati Acad Sci USA,2003; 100:6422-6427
    [6]Brown T A, Evangelista C, Trumpower B L. Regulation of nuclear genes encoding mitochondrial proteins in saccharomyces cerevisiae. JOURNAL OF BACTERIOLOGY.1995,177(23):6836-6843
    [7]Qiu, H., Hu, C., Yoon, S., Natarajan, K., Swanson, M. J., and Hinnebusch, A. G An array of coactivators is required for optimal recruitment of TATA binding p rotein and RNA polymerase Ⅱ by promoter-bound Gcn4p [J] 1Mol Cell Biol,2004; (24):4104-4117.
    [8]Harpreet Singh, Alexander M. Erkine, Selena B. Kremer, Harry M. Duttweiler, Donnie A. Davis, Jabed Iqbal, Rachel R. Gross and David S. Gross A functional module of yeast mediator that governs the dynamic range of heat shock gene expression [J]. Genetics,2006; (172):2169-2184.
    [9]Song W, Treich I,Qian N, et al. SSN genes that affect transcrip tional repression in Saccharomyces cerevisiae encode SIN4, ROX3, and SRB proteins associated with RNA polymerase Ⅱ [J]. Mol Cell Biol,1996; 16(1):115-120
    [10]Tabtiang R K, Herskowitz Ⅰ. Nuclear proteins Nutlp and Nut2p cooperate to negatively regulate a Swi4p-dependent lacZ reporter gene in Saccharomyces cerevisiae [J]. Mol Cellular biol,1998; 18:4707-4718.
    [11]van de Peppel J, Kettelaarij N, van Bakel H, et al. Mediator expression profiling epistasis reveals a signal transduction pathway with antagonistic submodules and highly specific downstream targets. MOLECULAR CELL.2005,19(4)511-522
    [12]Shamara M. Baidoobonso, Benjamin W. Guidi, and Lawrence C. Myers. Med19(Rox3) Regulates Intermodule Interactions in the Saccharomyces cerevisiae Mediator Complex. JBC.2007,282(8):5551-5559
    [13]Ding, N, Tomomori-Sato C, Sato, S, et al, MED19 and MED26 Are Synergistic Functional Targets of the RE1 Silencing Transcription Factor in Epigenetic Silencing of Neuronal Gene Expression, JBC,2009,284(5): 2648-2656
    [1]Leirdal M, Sioud M. Gene silencing in mammalian cells by preformed small RNA duplexes.Biochem Biophy Res Commun,2002,295(3):744-748
    [2]Schoenberger J, Grimm D, Kossmehl P, et al. Effects of PTK787/ ZK222584 a tyrosine kinase inhibitor on the growth of a poorly differentiated thyroid carcinoma:An animal study. Endocrinology,2004,145(3):1031-1038.
    [3]Gordon MS, Margolin K, Talpaz M, et al. Phase I safety and pharmacokinetic study of recombinant human anti-vascular endothelial growth factor in patients with advanced cancer. J Clin Oncol,2001,19(3):843-850.
    [4]Arenz C, Schepers U. RNA interference:from an ancient mechanism to a state of the art therapeutic application? Naturwissenschaften,2003,90(8): 345-359
    [5]Kwon M, Scholey JM. Spindle mechanics and dynamics during mitosis in Drosophila. Trends Cell Biol,2004,14(4):194-205.
    [6]Schuck S, Manninen A, Honsho M, et al. Generation of single and double knockdowns in polarized epithelial cells by retrovirus-mediated RNA interference.ProcNatl Acad Sci USA,2004,101(14):4912-4917.
    [7]Xia H, Mao Q, Eliason SL, et al. RNAi suppresses polyglutamine-induced neurodegeneration in a model of spinocerebellar ataxia. Nat Med, 2004,10(8):816-820.
    [8]Rutz S, Scheffold A. Towards in vivo application of RNA interference-new toys, old problems. Arthritis Res Ther,2004,6(2):78-85.
    [9]Elbashir SM, Harborth J, Lendeckel W, et al.Duplexes of 21-nucleotide RNAs medate RNA interference in cultured mammalian cells. Nature,2001,411(6936):494-498.
    [10]Myers JW, Jones JT, Meyer T, et al. Recombinant Dicer efficiently converts large dsRNAs into siRNAs suitable for gene silencing. Nat biotechnol,2003,21(3):324-328.
    [11]Matsukura S, Jones PA, Takai D. Establishment of conditional vectors for hairpin siRNA knockdowns. Nucleic Acids Res,2003,31(15):e77.
    [12]Paddison PJ, Caudy AA, Bernstein E, et al.Short hairpin RNAs (shRNAs) induce sequence-specific silencing in mammalian cells. Genes Dev. 2002,16(8):948-958.
    [13]Elbashir SM, Lendeckel W, Tuschl T. RNA interference is mediated by 21-and 22-nucleotide RNAs. Genes Dev,2001,15(2):188-200.
    [14]Sui G, Soohoo C, Affar el B, et al. A DNA vector based RNAi technology to suppress gene expression in mammalian cells. Proc Natl Acad Sci USA,2002,99(8):5515-5520.
    [15]Brummelkamp TR, Bernards R, Agami R. A system for stable expression of short interfering RNAs in mammalian cells. Science,2002, 296(5567):550-553.
    [16]Sioud M, Leirdal M. Potential design rules and enzymatic synthesis of siRNAs. Methods Mol Biol.2004,252:457-469.
    [17]Berezhna SY, Supekova L, Supek F, et al.siRNA in human cells selectively localizes to target RNA sites. Proc Natl Acad Sci USA.2006, 103(20):7682-7687.
    [18]Robinson DA, Dillon CP, Kwiatkowski AV, et al. A lentivirus-based system to functionally silence genes in primary mammalian cells, stem cells and transgenic mice by RNA interference. Nat Genet,2003,33(3):401-406.
    [19]Cascallo M, Alemany R. Adenovirus-mediated gene transfer to tumor cells.Methods Mol Biol,2004,246:121-138.
    [1]Pancoska P, Moravek Z, Moll UM. Efficient RNA interference depends on global context of the target sequence:quantitative analysis of silencing efficiency using Eulerian graph representation of siRNA. Nucleic Acids Res, 2004,32(4):1469-1479.
    [2]Shankar P, Manjunath N, Lieberman J.The prospect of silencing disease using RNA interference. JAM A,2005,293(11):1367-1373.
    [3]Pulukuri SM, Gondi CS, Lakka SS, et al. RNA interference-directed knockdown urokinase plasminogen activator and urokinase plasminogen activator receptor inhibits prostate cancer cell invasion, survival, and tumorigenicity in vivo. J Bial Chem,2005,280(43):36529-36540.
    [4]Elbashir SM, Harborth J, Lendeckel W, et al.Duplexes of 21-nucleotide RNAs medate RNA interference in cultured mammalian cells. Nature,2001, 411(6936):494-498.
    [5]Pham JW, Pellino JL, Lee YS, et al. A Dicer-2-dependent 80s complex cleaves targeted mRNAs during RNAi in Drosophila. Cell,2004,117(1): 83-94.
    [6]Schwarz DS, Hutvagnerq, Du T, et al. Asymmetry in the assembly of the RNAi enzyme complex. Cell,2003,115(2):199-208.
    [7]Meister G, Landthaler M, Patkaniowska A, et al. Human Argonaute2 mediates RNA cleavage targeted by miRNAs and siRNAs. Mol Cell,2004, 15(2):185-197.
    [8]Cheng CP, Pogany J, Nagy PD. Mechanism of DI RNA formation in tombusviruses:dissecting the requirement for primer extension by the tombusvirus RNA dependent RNA polymerase in vitro. Virology,2002, 304(2):460-473.
    [9]Dykxhoorn DM, Lieberman J. The silent revolution:RNA interference as basic biology, research tool, and therapeutic.Annu Rev Med,2005,56: 401-423.
    [10]Lewis DL, Hagstrom JE, Loomis AG, et al.Efficient delivery of siRNA for inhibition of gene expression in postnatal mice. Nat Genet,2002, 32(1):107-108.
    [11]McCaffrey AP, Meuse L, Pham TT, et al.RNA interference in adult mice. Nature,2002,418(6893):38-39.
    [12]Castanotto D, Li H, Rossi JJ. Functional siRNA expression from transfected PCR prouducts.RNA,2002,8(11):1454-60.

© 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