应用基因芯片筛选肺癌细胞辐射增敏分子药靶的研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
肺癌是最常见的恶性肿瘤之一,严重威胁着人类的健康。根据组织学分类,肺癌分为小细胞肺癌(SCLC)和非小细胞肺癌(NSCLC)两种,NSCLC又包含多种类型。NSCLC约占支气管肺癌的80%,其5年生存率不足10%。大多数患者在诊断时都已达到中晚期,不能进行手术,只能进行放疗和/或化疗。目前放射治疗多采用直线加速器进行分割照射,每次2Gy,总剂量约为60Gy。虽然可以给予较大的剂量,但是辐射引起的副作用影响患者的预后和生存质量。因此,迫切需要发展新的治疗模式。许多科学家认为将来肺癌的治疗方向将偏重于靶向治疗和靶向治疗与常规治疗相结合的综合治疗模式。目前,传统的细胞毒性药物和分子化合物的研究进展并不理想。如果想提高肺癌的疗效,必须寻找新的靶标和分子化合物。
     同一组织来源的细胞系基因表达具有明显的异质性,以往的研究认为基因表达和放射治疗的疗效以及辐射敏感性密切相关。SCLC和NSCLC是肺癌的两大类,它们的辐射敏感性明显不同。所以,我们选择了两种具有代表性的肺癌细胞系作为研究对象,一种是肺腺癌细胞A549,另一种是小细胞肺癌细胞NCI-H446。我们对两种细胞的辐射敏感性和辐射反应进行了检测,通过细胞克隆形成实验检测了两种细胞的辐射存活曲线;使用流式细胞仪检测了两种细胞受到5Gy照射后不同时间点的细胞周期变化和细胞凋亡的情况。
     人类基因组完成后,DNA芯片已经成了研究转录组的主要方法。鉴于辐
Lung cancer causes many deaths worldwide and is the leading cause of cancer death in China and the most common malignant cancer that threatens human being's health. The long-term survival of lung cancer patients treated with conventional therapies remains poor and has changed little in decades. Lung cancer is classified according to the histology of the cancer cells into SCLC (small cell lung cancer) and NSCLC (non-small cell lung cancer), the latter consisting of several types. NSCLC accounts for nearly 80% of all bronchogenic neoplasms and is characterized by a particularly poor prognosis with almost 90% of patients dying from it within 5 years of diagnosis. Most NSCLC patients are inoperable at diagnosis and submitted to radiotherapy and/or chemotherapy. Radiotherapy is usually delivered by an external beam from a linear accelerator. Standard therapy for unresectable disease consists of approximately 60 Gy, with the dose divided among 30 sessions over a period of six weeks. Although higher doses have been used, the more the toxic effects of radiation in normal organs have been found. Concurrent chemotherapy may increase the efficiency of radiotherapy by sensitizing the tumor to ionizing radiation (IR), but it can also increase the adverse effects (particularly esophagitis). Because of the poor
    prognosis, there is an urgent requirement for research in the development of new therapies, such as gene therapy for lung cancer. Many scientists think future directions for lung cancer treatment are heavily weighted toward targeted therapies - namely, those aimed at molecular abnormalities involved in the pathogenesis of lung cancer- rather than traditional cytotoxic agents and targeted therapies combined with standard therapy. Since little further progress is expected with the use of traditional cytotoxic agents and compounds, such as those that target protein kinase C, epithelial growth factor receptor, vascular endothelial growth factor, cyclooxygenase-2, and farnesyl transferase, new agentsand approaches must be evaluated if we are to advance therapy for lung cancer.Previous studies reported that cell lines of the same pathological origin could be highly heterogeneous in gene expression. Many studies have reported correlations between gene expression and radiotherapeutic response and in vitro radiosensitivity. SCLC and NSCLC represent the two major categories of lung cancer that differ in their sensitivity to IR. We selected two lung cancer cell lines, one representative of adenocarcinomas (A549) and one variant of small cell lung cancer (NCI-H446). Radiosensitivities of both cell lines were measured with a standard clonogenic assay, cell cycle distributions and apoptosis were detected with FCM and radiation survival curves derived from colony formation assays of two cell lines were found.DNA microarrays have become the main technological workhorse for the transcripotome after the completion of the human genome. Given the complexity of IR-induced responses, microarray as a powerful tool for investigation small intracellular changes offers new opportunities to identify a wider range of genes and signaling pathways involved in the IR response. Understanding the mechanism of IR response at the molecular level will help us to provide valuable information for designing effective radiotherapy of various tumor types and improving radiocurability of tumor cells. To better understand the IR-mediated
    gene expression changes that might results in different response to IR, oligonucleotide microarray were designed by our laboratory. To validate the microarray results, RT-PCR was performed using the RNA obtained 6, 24 and 48 h after irradiation.Antisense oligonucleotides have been extensively studied as a research tool in determining gene function and as a novel therapeutic approach to treatment of various human diseases. Microarray results found that MDM2 is interesting, which has been shown to be a critical component of the responses to IR. The design of the ASO complementary to the MDM2 mRNA was completed by a computational neural network model, secondary structure of RNA prediction and molecular mechanisms of action of antisene drug. Five anti-MDM2 ASOs were synthesized in our laboratory and transfected the A549 cells in the presence of Lipofectin and demonstrated specifically knocked-down MDM2 expression, as demonstrated at both mRNA and protein levels. The cell survival and proliferative ability, apoptosis and radiosensitivities of treated cells were detected by MTT assays, FCM and clonogenic assays respectively.Currently, RNAi is a discovered knockdown phenomenon that is induced by double-stranded RNA (dsRNA) in organism and mammalian cells. The mechanism of RNAi is not fully understood, but recent genetic and biochemical studies have revealed some details at the molecular level. The activity of siRNA is high even at low concentrations, without apparent toxicity. In the present study, RNAi strategy was employed to reduce MDM2 in A549 cells. Plasmid pPUR/U6 was the gift of ZY. Shen, University of New Mexicoschool of Medicine. Six RNAi oligos were designed according to the sequence of effective ASOs and principles of siRNA design. Equal amount of sense and antisense template were annealed, and subcloned into the Apal and EcoRI sites of pPUR/U6 vector to
    construct MDM2-siRNA plasmids, Escherichia coli DH5a competent cells were transformed with the resultant plasmids. Plasmid DNA was purified using a commercial DNA purification kit and confirmed by DNA sequence analysis. Two out of three siRNA plasmids were constructed successfully. A549 cells grown in 6-we 11 plates were transfected 3u,g siRNAs by using Lipofectamine? 2000 according to the manufacturer's protocol. pPUR/U6 without insert was used as a control. The cells were selected with puromycin. MDM2 expression was determined by RT-PCR and Western Blotting and the transfected cells response to IR was detected by FCM and MTT assay.The studies found that both lung cancer cell lines that differ in then-sensitivity to IR. The A549 cell line displayed the surviving fraction at 2Gy (SF2) was 0.64 in our colony-forming assays. The NCI-H446 cell line can be considered as relatively sensitive to radiation with a SF2 of 0.51 in our colony-forming assays. By FCM, a significant G2 phase arreast and DNA fragment were found in NCI-H446 cells after IR. Therefore, both cell lines were selected for the analyses. The radiation-related oligonucelotide microarray was fabricated successfully, which has 143 genes, including 127 genes involved in stress response, cell cycle progression, DNA damage and repair, apoptosis and proliferation, together with 11 housekeeping genes and 2 rice genes and 3 luciferase genes as control. Using selected lung cancer cell lines, we identified genes showing altered expression in lung cancer cell lines, as well as several potential ionizing radiation resistance candidate genes. We set a cutoff of > 1.5-fold for "induction" and <0i75 for "repression" in all experiments. According to these criteria, 34 differential expression genes were identified. To verify the expression of the genes identified in microarray experiments, RT-PCR was performed using the RNA at the 6h, 24h and 48h after IR. We tested seven genes ( MDM2, p53, , XRCC5, Bcl-2, PIM2 ,
    NFKBIA and Cyclin Bl) for RT-PCR, and found that the results were inaccordance with those from the microarray data except for NFKBIA gene, eventhough the value for each mRNA level might be different between the twomeasurements. Among the regulated genes, some might be predicted to result ineffects on DNA repair, some on cell cycle arrest, others on apoptosis. Ofnotable interest was MDM2, which has been shown to be a critical component ofthe responses to IR. In our studies, we found that MDM2 was induced in A549 cellsand reduced in NCI-H446 cells after IR. IR can activate p53 to arrest the cell cycle andto stimulate apoptosis. However, p53 is nr the only factor that determinesradiosensitivity. p53 and MDM2 form an autoregulatory feedback loop. The levelof MDM2 expression determines the extent to radiation induces an increase in theactivity of the p53 tumor suppressor. Therefore, the ability of MDM2 to conferradioresistance to tumors is beginning to be assessed. Now, it is important todetermine whether increased levels of MDM2 correlate with poor clinicaloutcome and whether MDM2 can be targeted to increase the efficacy ofradiotherapy. By antisense technology, inhibition of MDM2 using ASO5 hassignificant enhanced radiosensitivity in reducing survival and increasing apoptosisof A549 cells. By RNAi, inhibition of MDM2 using siRNAs has significantlyenhanced the efficacy of IR in reducing survival and increasing cell death ofA549 cells, especially for siRNAl. Although gene silencing by" transfection ofsiRNA is effective in mammalian cells, it reduces gene expression for only a shortperiod. Loss of long-term gene silencing was found despite the cells resistant topuromycin.As described and discussed, Highlights of our study were fabrication of radiation-related oligonucleotide microarray and the detection of a few genes with cell proliferation, such as MDM2, BCL-2, PKCz and PIM2 expression levels
引文
1.鲍云化,高红军。肺癌放射治疗进展。中国肺癌杂志,2000,3(6):419-422。
    2. Koukourakis MI, Kourousis C, Kamilaki M, Koukouraki S, Giatromanolaki A, Kakolyris S, Kotsakis A, Androulakis N, Bahlitzanakis N, Georgoulias V. Weekly docetaxel and concomitant boost radiotherapy for non-small cell lung cancer. A phase Ⅰ/Ⅱ dose escalation trial. Eur J Cancer. 1998, 34(6): 838-844.
    3.闫宇邱,孙伟建,周平坤。放射增敏剂的作用机制及临床研究进展。中华放射医学与防护杂志,2004,24(3):292-294。
    4. Spira A, David S. Ettinger. Multidisciplinary Management of Lung Cancer. N Engl J Med, 2004, 350: 379-392.
    5. The Lung Cancer Study Group. Effects of postoperative mediastinal radiation on completely resected stage Ⅱ and stage Ⅲ epidermoid cancer of the lung. N Engl J Med, 1986, 315: 1377-1381.
    6. PORT Meta-analysis Trialists Group. Postoperative radiotherapy in non-small-cell lung cancer: systematic review and meta-analysis of individual patient data from nine randomised controlled trials. Lancet, 1998, 352: 257-263.
    7.李邦华,周绪堂。肺癌治疗的新靶点及应用前景。中国肿瘤,2003,12(8):460-463。
    8. Michael Baumarm, Mechthild Krause, Daniel Zips, Cordula Petersen, Klaus Dittmann, Wolfgang Dorr and Hans-Peter Rodemann. Molecular targeting in radiotherapy of lung cancer. Lung cancer, 2004, 45: S187-S197.
    9. Rosen EM, Fan S, Goldberg ID, Rockwell S. Biological basis of radiation sensitivity. Part 2: Cellular and molecular determinants of radiosensitivity. Oncology (Huntingt), 2000, 14(5): 741-757.
    10. Collis S J, Swartz MJ, Nelson WG, DeWeese TL. Enhanced Radiation and Chemotherapy-mediated Cell Killing of Human Cancer Cells by Small Inhibitory RNA Silencing of DNA Repair Factors. Cancer Res, 2003; 63(7): 1550-1554.
    11. Kawabe S, Nishikawa T, Munshi A, Roth JA, Chada S, Meyn RE. Adenovirus-mediated mda-7 gene expression radiosensitizes non-small cell lung cancer cells via TP53-independent mechanisms. Molecular therapy, 2002, 6(5): 637-643.
    12. Foder SPA. DNA sequencing: Massively parallel genomics. Science, 1997, 277: 393-395.
    13. Fodor SP, Read JL, Pirrung MC, Stryer L, Lu AT, Solas D. Light-directed, spatially addressable parallel chemical synthesis. Science, 1991, 251(4995): 767-773.
    14. Schena M, Shalon D, Davis RW, Brown PO. Quantitative monitoring of gene expression patterns with a complementary DNA microarray. Science, 1995, 270(5235): 467-470.
    15. Schena M, Heller RA, Theriault TP, Konrad K, Lachenmeier E, Davis RW. Microarrays: biotechnology's discovery platform for functional genomics. Trends Biotechnol. 1998, 16(7): 301-306.
    16.郭万峰,滕光菊,王升启。几种基因芯片技术的比较,中国生物工程杂志,2004,5:15-19.
    17. Brazma A, Hingamp P, Quackenbush J, Sherlock G, Spellman P, Stoeckert C, Aach J, Ansorge W, Ball CA, Causton HC, Gaasterland T, Glenisson R Holstege FC, Kim IF, Markowitz V, Matese JC, Parkinson H, Robinson A, Sarkans U, Schulze-Kremer S, Stewart J, Taylor R, Vilo J, Vingron M. Minimum information about a microarray experiment (MIAME)-toward standards for microarray data. Nat Genet. 2001; 29(4): 365-371.
    18. Stoeckert CJ Jr, Causton HC, Ball CA. Microarray databases: standards and ontologies. Nat Genet. 2002; 32 Suppl: 469-473.
    19. Ball CA, Sherlock G, Parkinson H, Rocca-Sera P, Brooksbank C, Causton HC, Cavalieri D, Gaasterland T, Hingamp P, Holstege F, Ringwald M, Spellman P, Stoeckert CJ Jr, Stewart JE, Taylor R, Brazma A, Quackenbush J; Microarray Gene Expression Data (MGED) Society. Standards for microarray data. Science. 2002; 298(5593): 539.
    20. Andrei V.Gudkov and Elena A.Komarova. The role of p53 in determining sensitivity to radiotherapy. Nature reviews/cancer, 2003, 3:117-128.
    21. Kitahara O, Katagiri T, Tsunoda T, Harima Y, Nakamura Y. Classification of sensitivity or resistance of cervical cancers to ionizing radiation according to expression profiles of 62 genes selected by cDNA microarray analysis. Neoplasia. 2002; 4(4):295-303.
    22. Achary MP, Jaggernauth W, Gross E, Alfieri A, Klinger HP, Vikram B. Cell lines from the same cervical carcinoma but with different radiosensitivities exhibit different cDNA microarray patterns of gene expression. Cytogenet Cell Genet, 2000, 91(1-4): 39-43.
    23. Otomo T, Hishii M, Apai H, Sato K, Sasai K. Microarray analysis of temporal gene responses to ionizing radiation in two glioblastoma cell lines: up-regulation of DNA repair genes. J. Radiat. Res., 2004, 45(1): 53-60.
    24. Amundson SA, Lee RA, Koch-Paiz CA, Bittner ML, Meltzer P, Trent JM, Fornace AJ Jr. Differential responses of stress genes to low dose-rate gamma irradiation. Mol Cancer Res. 2003; 1(6): 445-452.
    25. Kang CM, Park KP, Song JE, Jeoung DI, Cho CK, Kim TH, Bae S, Lee SJ, Lee YS. Possible biomarkers for ionizing radiation exposure in human peripheral blood lymophocytes. Radiation research.2003,159: 312-319.
    26. Hartmann KA, Modlich O, Prisack HB, Gerlach B, Bojar H. Gene expression profiling of advanced head and neck squamous cell carcinomas and two squamous cell carcinoma cell lines under radio/chemotherapy using cDNA arrays. Radiotherapy and Oncology, 2002, 63: 309-320.
    27. Zhao R, Gish K, Murphy M, Yin Y, Notterman D, Hoffman WH, Tom E, Mack DH, Levine AJ. Analysis of p53-regulated gene expression patterns using oligonucleotide arrays. Genes Dev. 2000; 14 (8): 981-993.
    28. Robles AI, Bemmels NA, Foraker AB, Harris CC. APAF-1 is a transcriptional target of p53 in DNA damage-induced apoptosis. Cancer Research, 2001, 61:6660-6664.
    29. Burns TF, El-Deiry WS. Microarray analysis of p53 target gene expression patterns in the spleen and thymus in response to ionizing radiation. Cancer BiolTher. 2003; 2 (4): 431-443.
    30. Anderle P, Duval M, Draghici S, Kuklin A, Littlejohn TG, Medrano JF, Vilanova D, Roberts MA. Gene expression databases and data mining. BioTechniques, 2003, 34:S36-S44.
    31. Ernst Urban, Christian R. Noe. Structural modifications of antisense oligonucleotides. Ⅱ Farmaco, 2003, 58: 243-258.
    32. Marwick C. First "antisense" drug will treat CMV retinitis. JAMA. 1998, 280(10):871.
    33. Perry CM, Balfour JA. Fomivirsen. Drugs. 1999 , 57(3):375-80
    34. Burkhard Jansen and Uwe Zangemeister-Wittke. Antisense therapy for cancer- the time of truth. Lancet Oncol, 2002, 3: 672-683.
    35. Banerjee D. Technology evaluation: G-3139. Curr Opin Mol Ther. 1999 , 1(3):404-408.
    36. Klasa RJ, Gillum AM, Klem RE, Frankel SR. Oblimersen Bcl-2 antisense: facilitating apoptosis in anticancer treatment. Antisense Nucleic Acid Drug Dev. 2002,12(3):193-213.
    37. Koziner B. Potential therapeutic applications of oblimersen in CLL. Oncology (Huntingt). 2004,18(13 Suppl 10):32-38.
    38. Benimetskaya L, Lai JC, Khvorova A, Wu S, Hua E, Miller P, Zhang LM, Stein CA. Relative Bcl-2 independence of drug-induced cytotoxicity and resistance in 518A2 melanoma cells. Clin Cancer Res. 2004,10 (24):8371-8379.
    39. Frantz S. Lessons learnt from Genasense's failure. Nat Rev Drug Discov. 2004, 3(7):542-543.
    40. Wacheck V, Krepler C, Strommer S, Heere-Ress E, Klem R, Pehamberger H, Eichler HG, Jansen B. Antitumor effect of G3139 Bcl-2 antisense oligonucleotide is independent of its immune stimulation by CpG motifs in SCID mice. Antisense Nucleic Acid Drug Dev. 2002, 12(6):359-367.
    41. Lai JC, Benimetskaya L, Santella RM, Wang Q, Miller PS, Stein CA. G3139 (oblimersen) may inhibit prostate cancer cell growth in a partially bis-CpG-dependent non-antisense manner. Mol Cancer Ther. 2003, 2(10):1031-43.
    42. Tortora G, Ciardiello F. Antisense strategies targeting protein kinase C: preclinical and clinical development. Semin Oncol. 2003, 30(4 Suppl 10):26-31.
    43. Villalona-Calero MA, Ritch P, Figueroa JA, Otterson GA, Belt R, Dow E, George S, Leonardo J, McCachren S, Miller GL, Modiano M, Valdivieso M, Geary R, Oliver JW, Holmlund J. A phase Ⅰ/Ⅱ study of LY900003, an antisense inhibitor of protein kinase C-alpha, in combination with cisplatin and gemcitabine in patients with advanced non-small cell lung cancer. Clin Cancer Res. 2004, 10(18 Pt 1): 6086-6093.
    44. Izumi Y, Xu L, di Tomaso E, Fukumura D, Jain RK. Tumour biology: Herceptin acts as an anti-angiogenic cocktail. Nature, 2002, 416 (6878): 279-280.
    45. Slichenmyer WJ, Fry DW. Anticancer therapy targeting the erbB family of receptor tyrosine kinases. Semin Oncol, 2001, 28 (5Suppl 16): 67-79.
    46.陈忠斌,王升启。抗肿瘤反义寡核苷酸药物研究现状和趋势。中国新药杂志,2002,11(9):683-685.
    47.杨英,杜清友,王升启。反义核酸协同化疗药物抑制肝癌细胞的增殖。中华肝脏病杂志,2003,11(12):719-721。
    48. Kasid U, Dritschilo A. RAF antisense oligonucleotide as a tumor radiosensitizer. Oncogene. 2003, 22(37): 5876-5884.
    49. Zhang Z, Wang H, Prasad G, Li M, Yu D, Bonner JA, Agrawal S, Zhang R. Radiosensitization by anti-MDM2 mixed-backbone oligonucleotide in in vitro and in vivo human cancer models. Clin Cancer Res. 2004, 10(4): 1263-1273.
    50. Zhang R, Wang H, Agrawal S. Novel antisense anti-MDM2 mixed-backbone oligonucleotides: proof of principle, in vitro and in vivo activities, and mechanisms. Curr Cancer Drug Targets, 2005, 5(1): 43-49.
    51. Pyo H, Choy H, Amorino GP, Kim JS, Cao Q, Hercules SK, DuBois RN. A selective cyclooxygenase-2 inhibitor, NS-398, enhances the effect of radiation in vitro and in vivo preferentially on the cells that express cyclooxygenase-2. Clin Cancer Res. 2001, 7 (10):2998-3005.
    52. 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(4):611-620.
    53. Fire A, Xu S, Montgomery MK, Kostas SA, Driver SE, Mello CC. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature, 1998, 391(6669):806-811.
    54. Fjose A, Ellingsen S, Wargelius A, Seo HC. RNA interference: mechanisms and applications. Biotechnol Annu Rev, 2001, 7:31-57.
    55. Agami R. RNAi and related mechanisms and their potential use for therapy. Curr Opin Chem Biol, 2002, 6(6): 829-834.
    56. Elbashir SM, Harborth J, Lendeckel W, Yalcin A, Weber K, Tuschl T. Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature, 2001; 411(6836):494-498.
    57. Zamore PD, Tuschl T, Sharp PA, Bartel DP. RNAi: double-stranded RNA directs the ATP-dependent cleavage of mRNA at 21 to 23 nucleotide intervals. Cell, 2000; 101(l):25-33.
    58. Hammond SM, Bernstein E, Beach D, Hannon GJ. An RNA-directed nuclease mediates post-transcriptional gene silencing in Drosophila cells. Nature, 2000, 404(6775):293-296.
    59. Crooke ST. Progress in antisense technology. Annu Rev Med. 2004, 55:61-95.
    60. Holen T, Amarzguioui M, Wiiger MT, Babaie E, Prydz H. Positional effects of short interfering RNAs targeting the human coagulation trigger Tissue Factor. Nucleic Acids Res, 2002; 30(8):1757-1766.
    61. Elbashir SM, Harborth J, Weber K, Tuschl T. Analysis of gene function in somatic mammalian cells using small interfering RNAs. Methods, 2002; 26(2): 199-213.
    62. Brummelkamp TR, Bernards R, Agami R. A system for stable expression of short interfering RNAs in mammalian cells. Science, 2002, 296(5567):550-553.
    63. Jiang M, Milner J. Selective silencing of viral gene expression in HPV-positive human cervical carcinoma cells treated with siRNA, a primer of RNA interference. Oncogene, 2002, 21 (39):6041 -6048.
    64. Siegmund D, Hadwiger P, Pfizenmaier K, Vornlocher HP, Wajant H. Selective inhibition of FLICE-like inhibitory protein expression with small interfering RNA oligonucleotides is sufficient to sensitize tumor cells for TRAIL-induced apoptosis. Mol. Med. 2002, 8(11): 725-732.
    65. Kosciolek BA, Kalantidis K, Tabler M, Rowley PT. Selective inhibition of FLICE-like inhibitory protein expression with small interfering RNA oligonucleotides is sufficient to sensitize tumor cells for TRAIL-induced apoptosis. Mol Cancer Ther, 2003, 2(3):209-216.
    66. Fernet M, Hall J. Genetic biomarkers of therapeutic radiation sensitivity. DNA Repair, 2004, 3: 1237-1243.
    67. Zhang X, Kon T, Wang H, Li F, Huang Q, Rabbani ZN, Kirkpatrick JP, Vujaskovic Z,Dewhirst MW, Li CY. Enhancement of hypoxia-induced tumor cell death in vitro and radiation therapy in vivo by use of small interfering RNA targeted to hypoxia-inducible factor-1 alpha. Cancer Res. 2004, 64(22):8139-8142.
    68. Fakharzadeh, S. S., Trusko, S. P., and George, D. L. Tumorigenic potential associated with enhanced expression of a gene that is amplified in a mouse tumor cell line. EMBO J., 1991,10: 1565-1569.
    69. Picksley, S. M. and Lane, D. P. The p53-mdm2 autoregulatory feedback loop: a paradigm for the regulation of growth control by p53. Bioessays, 1993,2:689-690.
    70. Wu, X., Bayle, J. H., Olson, D., and Levine, A. J. The p53-mdm-2 autoregulatory feedback loop. Genes Dev., 1993, 2: 1126 - 1132.
    71. Patrick Chene. Inhibition of the p53-MDM2 interaction: targeting a protein-protein interface. Molecular cancer reseach, 2004, 2:20-28.
    72. Momand, J., Zambetti, G. P., Olson, D. C, George, D., and Levine, A. J. The mdm-2 oncogene product forms a complex with the p53 protein and inhibits p53-mediated transactivation. Cell, 1992, 2: 1237 - 1245.
    73. Honda, R. and Yasuda, H. Activity of MDM2, a ubiquitin ligase, toward p53 or itself is dependent on the RING finger domain of the ligase. Oncogene, 2000,2: 1473 - 1476.
    74. Fang, S., Jensen, J. P., Ludwig, R. L., Vousden, K. H., and Weissman, A. M. Mdm2 is a RING finger-dependent ubiquitin protein ligase for itself and p53. J. Biol. Chem., 2000, 2: 8945 - 8951.
    75. Roth, J., Dobbelstein, M, Freedman, D. A., Shenk, T., and Levine, A. J. Nucleo-cytoplasmic shuttling of the mdm2 oncoprotein regulates the levels of the p53 protein via a pathway used by the human immunodeficiency virus rev protein. EMBO J., 1998, 2: 554 - 564.
    76. Wang, H., Zeng, X., Oliver, P., Le, L. P., Chen, J., Chen, L., Zhou, W., Agrawal, S., and Zhang, R. MDM2 oncogene as a target for cancer therapy: an antisense approach. Int. J. Oncol., 1999, 2: 653 - 660.
    77. Kondo, S., Barnett, G. H., Hara, H., Morimura, T., and Takeuchi, J. MDM2 protein confers the resistance of a human glioblastoma cell line to cisplatininduced apoptosis. Oncogene, 1995, 10: 2001 -2006.
    78. Meye, A., Wurl, P., Bache, M., Bartel, F., Grunbaum, U., Mansa-ard, J., Schmidt, H., and Taubert, H. Colony formation of soft tissue sarcoma cells is inhibited by lipid-mediated antisense oligodeoxynucleotides targeting the human mdm2 oncogene. Cancer Lett., 2000,149: 181-188,
    79. Chen, L., Agrawal, S., Zhou, W., Zhang, R., and Chen, J. Synergistic activation of p53 by inhibition of MDM2 expression and DNA damage. Proc. Natl. Acad. Sci. USA, 1998, 95: 195-200.
    80. Sato, N., Mizumoto, K., Maehara, N., Kusumoto, M., Nishio, S., Urashima, T., Ogawa, T., and Tanaka, M. Enhancement of drug-induced apoptosis by antisense oligodeoxynucleotides targeted against Mdm2 and p21WAFl/CIP1. Anticancer Res., 2000, 20: 837-842.
    81. Wang, H., Nan, L., Yu, D., Lindsey, J. R., Agrawal, S., and Zhang, R. Antitumor efficacy of a novel antisense anti-MDM2 mixed-backbone oligonucleotide in human colon cancer models: p53-dependent and p53-independent mechanisms. Mol. Med., 2002, 8: 185 - 199.
    82. Tortora, G., Caputo, R., Damiano, V., Bianco, R., Chen, J., Agrawal, S., Bianco, A. R., and Ciardiello, F. A novel MDM2 anti-sense oligonucleotide has anti-tumor activity and potentiates cytotoxic drugs acting by different mechanisms in human colon cancer. Int. J. Cancer, 2000, 88: 804-809.
    83. Prasad, G., Wang, H., Agrawal, S., and Zhang, R. Antisense anti-MDM2 oligonucleotides as a novel approach to the treatment of glioblastoma multiforme. Anticancer Res., 2002, 22: 107-116.
    84. Chen, L., Lu, W., Agrawal, S., Zhou, W., Zhang, R., and Chen, J. Ubiquitous induction of p53 in tumor cells by antisense inhibition of MDM2 expression. Mol. Med., 1999, 5: 21-34.
    85. Li, M, Luo, J., Brooks, C. L., and Gu, W. Acetylation of p53 inhibits its ubiquitination by Mdm2. J. Biol. Chem., 2002, 277: 50607-50611.
    86. Lai, Z., Yang, T., Kin, Y. B., Sielecki, T. M., Diamond, M. A., Strack, P., Rolfe, M., Caligiuri, M., Benfield, P. A., Auger, K. R., and Copeland, R. A. Differentiation of hdm2-mediated p53 ubiquitination and hdm2 autoubiquitination activity by small molecular weight inhibitors. Proc. Natl. Acad. Sci. USA, 2002, 2: 14734-14739.
    87. Xia L, Paik A, Li JJ. p53 activation in chronic radiation-treated breast cancer cells: regulation of MDM2/p14ARF. Cancer Res. 2004, 64(1):221-228.
    88. Midgley, C. A., Desterro, J. M., Saville, M. K., Howard, S., Sparks, A., Hay, R. T., and Lane, D. P. An N-terminal p14ARF peptide blocks mdm2-dependent ubiquitination in vitro and can activate p53 in vivo . Oncogene, 2000, 2: 2312-2323.
    89. Bartel F, Harris LC, Wurl P, Taubert H. MDM2 and its splice variant messenger RNAs: expression in tumors and down-regulation using antisense oligonucleotides. Mol Cancer Res. 2004, 2(l):29-35.
    90. Perry ME, Piette J, Zawadzki JA, Harvey D, Levine AJ. The mdm-2 gene is induced in response to UV light in a p53-dependent manner. Proc. Natl. Acad. Sci. USA, 1993, 90(24): 11623-11627.
    91. MuZ, Hachem P, Agrawal S, Pollack A. Antisense MDM2 sensitizes prostate cancer cells to androgen deprivation, radiation, and the combination. Int J Radiat Oncol Biol Phys, 2004, 58(2):336-343.
    92. Perry ME. Mdm2 in the response to radiation. Mol Cancer Res, 2004;
    93. Pearce AG, Segura TM, Rintala AC, Rintala-Maki ND, Lee H. The generation and characterization of a radiation resistant model system to study radioresistance in human breast cancer cells. Radiat Res, 2001, 156(6):739-750.
    94. Sirzen F, Nilsson A, Zhivotovsky B, Lewensohn R. DNA-dependent protein kinase content and activity in lung carcinoma cell lines: correlation with intrinsic radiosensitivity. Eur J Cancer. 1999, 35(1):111-116.
    95. Guo GZ, Sasai K, Oya N, Shibata T, Shibuya K, Hiraoka M. A significant correlation between clonogenic radiosensitivity and the simultaneous assessment of micronucleus and apoptotic cell frequencies. Int J Radiat Biol. 1999, 75(7):857-864.
    96. Guo GZ, Sasai K, Oya N, Takagi T, Shibuya K, Hiraoka M. Simultaneous evaluation of radiation-induced apoptosis and micronuclei in five cell lines. Int J Radiat Biol. 1998, 73(3):297-302.
    97. Guo G, Yan-Sanders Y, Lyn-Cook BD, Wang T, Tamae D, Ogi J, Khaletskiy A, Li Z, Weydert C, Longmate JA, Huang TT, Spitz DR, Oberley LW, Li JJ. Manganese superoxide dismutase-mediated gene expression in radiation-induced adaptive responses. Mol Cell Biol. 2003, 23(7):2362-2378.
    98. Guo G, Wang T, Gao Q, Tamae D, Wong P, Chen T, Chen WC, Shively JE, Wong JY, Li JJ. Expression of ErbB2 enhances radiation-induced NF-kappaB activation. Oncogene. 2004,23(2):535-545.
    99. Chen XF, Shen BH, Xia LQ, Khaletzkiy A, Chu D, Wang J.Y.C, Li JJ. Activation of nuclear factor NF-kappaB in radioresistance of TP53-inactive human keratinocytes. Cancer res. 2002, 62:1213-1221.
    100. Sheridan MT, Catharine West. Ability to undergo apoptosis does not correlate with the intrinsic radiosensitivity (SF2) of human cervix tumor cell lines. Int J. Radiation Oncology Biol. Phys., 2001, 50(2): 503-509.
    101. Gigante M, Toffoli G, Bertola A, Biscontin G, Dassie A, Zanelli GD, Zanin E, Trovo MG, Muzzio PC. Radiosensitivity in multidrug-resistant and Cisplatin-resistant human carcinoma cell lines. Am J Clin Oncol. 2003, 26(4):E73-79.
    102. Wei K, Kodym R, Jin C. Radioresistant cell strain of human fibrosarcoma cells obtained after long-term exposure to x-rays. Radiat Environ Biophys. 1998, 37(2):133-137.
    103. Teyssier F, Bay JO, Dionet C, Verrelle P. Cell cycle regulation after exposure to ionizing radiation. Bull Cancer. 1999, 86(4):345-357.
    104. Hunakova L, Chorvath M, Duraj J, Bartosova Z, Sevcikova L, Sulikova M, Chovancova J, Sedlak J, Chorvath B, Boljesikova E. Radiation-induced apoptosis and cell cycle alterations in human carcinoma cell lines with different radiosensitivities. Neoplasma. 2000, 47(1):25-31.
    105. Meyn RE, Stephens LC, Ang KK, Hunter NR, Brock WA, Milas L, Peters LJ. Heterogeneity in the development of apoptosis in irradiated murine tumours of different histologies. Int J Radiat Biol 1993; 64(5):583-591.
    106. Dewey W.C., Ling C.C. and Meyn R.E. Radiation-induced apoptosis: Relevance to radiotherapy. Int J Radiat Oncol Biol Phys 1996, 33:781-796.
    107. Peltenburg LT. Radiosensitivity of tumor cells. Oncogenes and apoptosis. Q J Nucl Med 2000; 44(4):355-364.
    108. Schena M, Shalon D, Heller R, Chai A, Brown PO, Davis RW. Parallel human genome analysis: Microarray based expression monitoring of 1000 genes. Proc. Natl. Acad. Sci. USA, 1996, 93(20):10614-10619.
    109. Kruse JJ, te Poele JA, Velds A, Kerkhoven RM, Boersma LJ, Russell NS, Stewart FA. Identification of differentially expressed genes in mouse kidney after irradiation using microarray analysis. Radiat Res. 2004, 161(1): 28-38.
    110. Edwards JS, Battista JR. Using DNA microarray data to understand the ionizing radiation resistance of Deinococcus radiodurans. Trends Biotechnol. 2003, 21(9): 381-382.
    111. Marko NF, Dieffenbach PB, Yan G, Ceryak S, Howell RW, McCaffrey TA, Hu VW. Does metabolic radiolabeling stimulate the stress response? Gene expression profiling reveals differential cellular responses to internal beta vs. External gamma radiation. FASEB J. 2003, 17(11): 1470-1486.
    112. Li W, Huang J, Fan M, Wang S. Mprobe: computer aided probe design for oligonucleotide micrroarray. Appl Bioinformatics, 2002, 1(3): 103- 106.
    113. Wahl GM, Carr AM. The evolution of diverse biological responses to DNA damage: insights from yeast and p53. Nature Cell Biol, 2001, 3, E277-E286.
    114. Lowndes N.F. and Murguia J.R. Sensing and responding to DNA damage. Curr Opin Genet Dev, 2000, 10: 17-25.
    115. Rosen EM, Fan S, Rockwell S, Goldberg ID. The molecular and cellular basis of radiosensitivity: implications for understanding how normal tissues and tumors respond to therapeutic radiation. Cancer Invest, 1999, 17(1): 56-72.
    116. Goping IS, Barry M, Liston P, Sawchuk T, Constantinescu G, Michalak KM, Shostak I, Roberts DL, Hunter AM, Korneluk R, Bleackley RC. Granzyme B-induced apoptosis requires both direct caspase activation and relief of caspase inhibition. Immunity, 2003, 18(3): 355-365.
    117. Sutton VR, Wowk ME, Cancilla M, Trapani JA. Caspase activation by granzyme B is indirect, and caspase autoprocessing requires the release of proapoptotic mitochondrial factors. Immunity, 2003(3), 18: 319-329.
    118. Tan KO, Tan KM, Chan SL, Yee KS, Bevort M, Ang KC, Yu VC. MAP-1, a novel proapoptotic protein containing a BH3-like motif that associates with Bax through its Bcl-2 homology domains. J Biol Chem, 2001, 276(4): 2802-2807.
    119. Wei MC, Zong WX, Cheng EH, Lindsten T, Panoutsakopoulou V, Ross AJ, Roth KA, MacGregor GR, Thompson CB, Korsmeyer SJ. Proapoptotic BAX and BAK: a requisite gateway to mitochondrial dysfunction and death. Science, 2001, 292(5517): 727-730.
    120. Gudkov AV, Komarova EA. The role of p53 in determining sensitivity to radiotherapy. Nat Rev Cancer, 2003,3: 117-128
    121. Perry ME, Mdm2 in the response to radiation, Mol Cancer Res, 2004, 2: 9-19.
    122. Eymin B, Gazzeri S, Brambilla C, Brambilla E. Mdm2 overexpression and pl4 (ARF) inactivation are two mutually exclusive events in primary human lung tumors. Oncogene, 2002, 21 (17) : 2750-2761.
    123. Gorgoulis VG, Zacharatos P, Kotsinas A, Mariatos G, Liloglou T, Vogiatzi T, Foukas P, Rassidakis G, Garinis G, loannides T, Zoumpourlis V, Bramis J, Michail PO, Asimacopoulos PJ, Field JK, Kittas C. Altered expression of the cell cycle regulatory molecules pRb , p53 and MDM2 exert a synergetic effect on tumor growth and chromosomal instability in non-small cell lung carcinomas (NSCLCs) . Mol Med, 2000, 6(3) : 208-237.
    124. Aikawa H, Sato M, Fujimura S, Takahashi H, Endo C, Sakurada A, Chen Y, Kondo T, Tanita T, Matsumura Y, Saito Y, Sagawa M. MDM2 expression is associated with progress of disease and WAFl expression in resected lung cancer. Int J Mol Med, 2000, 5 (6) : 631-633.
    125. Chalk AM and Sonnhammer EL. Computational antisense oligo prediction with a neural network model. Bioinformatics, 2002, 18(12): 1567-1575.
    126. Cooke ST. Molecular mechanisms of action of antisense drugs. Biochim Biophys Acta., 1999,1489(1): 31-44
    127. Scherer LJ, Rossi JJ. Approaches for the sequence-specific knockdown of mRNA. Nat Biotechnol, 2003, 21(12):1457-1465.
    128. Jones SW, Souza PM, Lindsay MA. siRNA for gene silencing: a route to drug target discovery. Current Opinion in Pharmacology 2004, 4:522-527.
    129. Clayton J. RNA interference: the silent treatment. Nature. 2004, 431(7008):599-605.
    130. Ryther RC, Flynt AS, Phillips JA 3rd, Patton JG. siRNA therapeutics: big potential from small RNAs. Gene Ther. 2005, 12(1):5-11.
    131. Soutschek J, Akinc A, Bramlage B, Charisse K, Constien R, Donoghue M, Elbashir S, Geick A, Hadwiger P, Harborth J, John M, Kesavan V, Lavine G, Pandey RK, Racie T, Rajeev KG, Rohl I, Toudjarska I, Wang G, Wuschko S, Bumcrot D, Koteliansky V, Limmer S, Manoharan M, Vomlocher HP. Therapeutic silencing of an endogenous gene by systemic administration of modified siRNAs. Nature. 2004, 432(7014):173-178.
    132. de Fougerolles A, Manoharan M, Meyers R, Vomlocher HP. RNA interference in vivo: toward synthetic small inhibitory RNA-based therapeutics. Methods Enzymol. 2005, 392:278-296.
    133. Gorre ME, Mohammed M, Ellwood K, Hsu N, Paquette R, Rao PN, Sawyers CL. Clinical resistance to STI2571 cancer therapy caused by BCR-ABL gene mutation or amplification. Science, 2001, 293 (5531): 876-880.
    134. Saetrom P, Snove O Jr. A comparison of siRNA effeacy predictors. Biochem Biophys Res Commun., 2004 321 (1) :247-253

© 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