pEgr-1-AIF_(△1-480)基因—放射治疗乳腺癌的体外抑瘤效应研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
恶性肿瘤严重威胁着人类的健康,一直是人们关注的焦点。肿瘤的常规治疗手段包括手术治疗、放射治疗、化学药物治疗及生物治疗等,但是由于多方面原因肿瘤的治疗效果往往不佳。近年来兴起的基因治疗为肿瘤治疗带来了新的希望,尤其是与放射治疗联合应用的肿瘤基因-放射治疗,将放射治疗和基因治疗的各自优点相结合,即能实现电离辐射对肿瘤细胞的直接杀伤目的,又能实现辐射条件下肿瘤杀伤基因的高效表达,继而实现消除肿瘤的作用,产生辐射和基因表达产物的协同抑瘤效果。另外,利用放射治疗具有靶向性和可操控性的特点,可调节杀伤基因表达的时间和位置。
     本研究充分利用早期生长反应基因-1(early growth response gene-1, Egr-1)启动子的辐射诱导表达增强的特性以及凋亡诱导因子(apoptosis inducing factor, AIF)强大的诱导细胞凋亡功能,构建Egr-1介导的截短型AIF真核表达载体pEgr-1-AIF_(△1-480),转染乳腺癌MCF-7细胞,研究其在有和无电离辐射条件下蛋白表达的时程和剂量效应规律,继而将重组质粒与2 Gy X射线联合作用于乳腺癌细胞MCF-7,观察两者联合作用后肿瘤细胞的增殖、侵袭力与凋亡的变化。实验结果表明,重组质粒转染后,细胞中截短型AIF△1-480蛋白的表达具有一定的时程和量效规律,重组质粒对人乳腺癌细胞MCF-7具有明显的抑制增殖、降低侵袭能力及促进凋亡的作用,本研究结果为新的肿瘤基因-放射治疗方案提供了重要的理论基础和实验依据。
At present, the malignant tumor is one of the major diseases, which threats seriously to human health. The primary ways for tumor treatment are surgery, radiotherapy, chemotherapy and biotherapy and so on. And the comprehensive treatment is a big trend. The gene-radiation therapy has been a hot spot in the research field of oncotherapy. Radiotherapy combines with gene therapy to educe synergistic effect. The discovery of radiation-sensitive early growth response-1 (Egr-1) promoter provides a new thinking for the effective combination of radiotherapy and gene therapy, which establishes the theoretical foundation for gene-radiation therapy on tumor. In this study, the plasmid pEgr-1-AIF△1 -480 constructed successfully as the gene therapy introduction system combined with radiotherapy, which could activate the transcription of the Egr-1 promoter, promote apoptosis of tumor cells with the AIF△1-480 gene, and kill tumor cells.
     1. Construction of recombinant plasmids
     1.1 Acquisition of AIF_(△1-480) gene
     The total RNA was extracted from human leukemia Jurkat cells, which was reversely transcribed into cDNA as the template. The specific PCR primers were designed and synthesized according to the sequence of AIF_(△1-480) gene. The AIF_(△1-480) gene was acquired by RT-PCR through agarose gel electrophoresis. The AIF_(△1-480) gene was amplified from the template, and ligated to pMD18T vector. The result of sequencing analysis including enzyme digestion and sequencing process was in coincidence with the anticipated result.
     1.2 Acquisition of Egr-1 promoter
     The pMD19T-Egr-1 was digested into the fragments with EcoRⅠand HindⅢenzymes. Egr-1 fragment was obtained with agarose gel electrophoresis, and identified by cleavage of endonucleases and sequencing analysis. The results of identification confirmed that the sequence of the cloned gene was identical to that published on Genbank.
     1.3 Construction of recombinant plasmids
     The AIF△1-480 gene was ligated to the pcDNA3.1 vector to construct the pcDNA3.1-AIF_(Δ1-480), and the Egr-1 was ligated to the pcDNA3.1-AIF_(Δ1-480) vector to construct the pcDNA3.1-Egr-1-AIF_(Δ1-480) with the technique of genetic engineering. The pcDNA3.1-Egr-1-AIF_(Δ1-480) was identificated by PCR and cleavage of endonucleases. The result of sequencing analysis was in coincidence with the anticipated result. In the following experiments, the gene protein expression rule, the inhibition effects on the tumor growth, apoptosis and invasion in human breast cancer MCF-7 cells were detected after these plasmids combined with radiation.
     2. Experimental grouping and index detection
     There were four groups in the experiment: the control, pcDNA3.1, pcDNA3.1- AIF△1-480 and pcDNA3.1-Egr-AIF_(△1-480). In the time-course experiment, the selected time points were 2, 4, 8, 12, 24 and 48 h, and the irradiation dose was 2 Gy. In the dose-effect experiment, the selected irradiation doses were 0, 0.2, 0.5, 1.0, 2.0 and 5.0 Gy, and the observation was done 24 h after irradiation. Western blot, Transwell, MTT and flow cytometry were used to detect the expressions of protein, cell invasion, cell proliferation, cell cycle progression and apoptosis, respectively.
     3. Radiation-induced expression rule of recombinant plasmids in MCF-7 cells
     3.1 Protein expressions in MCF-7 cells transfected with recombinant plasmids
     The cells were harvested at different time after transfection. The AIF protein expression in MCF-7 cells was detected and analyzed by Western blot. There were not differences in intrinsic AIF protein expressions between all groups. The AIF_(Δ1-480) protein expressions detected in the pcDNA3.1-AIF_(Δ1-480) and pcDNA3.1-Egr-1-AIF_(Δ1-480) groups increased from 4 h after irradiation, and up to the peak value at 48 h. The results indicate that the intrinsic AIF protein expressions could be stable in MCF-7, and were not influenced by transfection with recombinant plasmids and the expressions of Egr-1 and AIF_(Δ1-480). There was time-course law in the AIF_(Δ1-480) protein expressions.
     3.2 Time-course changes of protein expressions in MCF-7 cells transfected with recombinant plasmids after 2.0 Gy X-irradiation
     The cells were harvested at different time after irradiation. The AIF protein expression in MCF-7 cells was detected and analyzed by Western blot. The intrinsic AIF protein expressions increased from 4 h after irradiation in all groups, and up to the peak value at 48 h. The AIF_(Δ1-480) protein expressions detected in the pcDNA3.1-AIF_(Δ1-480) and pcDNA3.1-Egr-1-AIF_(Δ1-480) groups increased from 2 h after irradiation, and up to the peak value at 24 h. And the expression in the pcDNA3.1- Egr-1-AIF_(Δ1-480) group was more than that in the pcDNA3.1-AIF_(Δ1-480) group. The results indicate that ionizing radiation could promote the release of the intrinsic AIF from mitochondria, and the expressions could increase with time extension, and were not influenced by transfection with recombinant plasmids and the expressions of Egr-1 and AIF_(Δ1-480). There was time-course law in the AIF_(Δ1-480) protein expressions. The promoter of Egr-1 could be activated by irradiation, and enhance the AIF_(Δ1-480) protein expressions.
     3.3 Dose-effect changes of protein expressions in MCF-7 cells transfected with recombinant plasmids after X-irradiation with different doses
     The cells were harvested 24 h after X-irradiation with different doses. The protein expression in MCF-7 cells was detected and analyzed by Western blot. The results showed that the intrinsic AIF protein expression increased gradually at same group with the increase of doses. The AIF_(Δ1-480) protein expressions were not detected in the control and pcDNA3.1 groups, but increased with the increase of doses in the pcDNA3.1-AIF_(Δ1-480) and pcDNA3.1-Egr-1-AIF_(Δ1-480) groups. At the same dose, the expression in the pcDNA3.1- Egr-1-AIF_(Δ1-480) group was more than that in the pcDNA3.1-AIF_(Δ1-480) group. The results indicate that there was the dose-effect law in the intrinsic AIF protein expression; ionizing radiation with the increase of doses could promote the more release of the intrinsic AIF. There was dose-effect law in the AIF_(Δ1-480) protein expressions. The promoter of Egr-1 could be activated by irradiation and enhance the AIF_(Δ1-480) protein expressions.
     4 Effects of recombinant plasmids combining with X-irradiation on MCF-7 cells
     4.1 Effects of X-irradiation on proliferation of MCF-7 cells transfected with recombinant plasmids
     The proliferation of MCF-7 cells without irradiation was the fastest in the control and pcDNA3.1 groups. The proliferation was significantly inhibited in the pcDNA3.1-AIF_(Δ1-480) and pcDNA3.1-Egr-1-AIF_(Δ1-480) groups from 12 h after irradiation as compared with that in the control group (P < 0.05). After the cells were irradiated with 2.0 Gy X-ray, the proliferation was significantly slow. In particular, the proliferation was the most slowly and at basically stopping status in the pcDNA3.1-AIF_(Δ1-480) and pcDNA3.1-Egr-1-AIF_(Δ1-480) groups. The results suggest that the AIF_(Δ1-480) protein and radiation, respectively, could inhibit the proliferation of MCF-7 cells, so this could increase with the combination of AIF_(Δ1-480) protein and radiation.
     4.2 Effects of X-irradiation on cell cycle progression of MCF-7 cells transfected with recombinant plasmids
     The percentages of G_0/G_1 phase cells without irradiation increased significantly in the pcDNA3.1-AIF_(Δ1-480) and pcDNA3.1-Egr-1-AIF_(Δ1-480) groups as compared with that in the control group, but those of G_2/M phase cells decreased significantly (P < 0.05). The percentages of G_0/G_1 and G_2/M phase cells irradiated with 2.0 Gy X-ray in all groups increased as compared with those in the control group, particular significantly in the pcDNA3.1-AIF_(Δ1-480) and pcDNA3.1-Egr-1-AIF_(Δ1-480) groups (P < 0.05). The percentages of S phase cells decreased significantly in all groups as compared with those in the control group, specialy in the pcDNA3.1-AIF_(Δ1-480) and pcDNA3.1-Egr-1-AIF_(Δ1-480) groups (P < 0.05). The results indicate that the AIF_(Δ1-480) protein could lead to G1 arrest, and ionizing radiation could lead to G1 and G2 blocking. The G1 and G2 arrests could increase with the combination of AIF_(Δ1-480) protein and ionizing radiation.
     4.3 Effects of X-irradiation on apoptosis of MCF-7 cells transfected with recombinant plasmids
     The early and late apoptotic percentages of the cells without irradiation increased significantly in the pcDNA3.1-AIF_(Δ1-480) and pcDNA3.1-Egr-1-AIF_(Δ1-480) groups (P < 0.05), in particular in later. The early apoptotic percentages of the cells irradiated with 2.0 Gy in all groups increased significantly (P < 0.05). The late apoptotic percentages of the cells irradiated with 2.0 Gy in the control and pcDNA3.1groups increased significantly (P < 0.05), but especial done significantly in the pcDNA3.1-AIF_(Δ1-480) and pcDNA3.1-Egr-1-AIF_(Δ1-480) groups. As compared with those in the pcDNA3.1-AIF_(Δ1-480) and pcDNA3.1-Egr-1-AIF_(Δ1-480) groups without irradiation, the early and late apoptotic percentages of the cells irradiated with 2.0 Gy and transfected with recombinant plasmids were significantly increased (P < 0.05). The results indicate that the AIF_(Δ1-480) protein and ionizing radiation could lead to the early and late apoptosis. The early, late and total apoptosis increased with the combination of AIF_(Δ1-480) protein and radiation.
     4.4 Effects of X-irradiation on invasion of MCF-7 cells transfected with recombinant plasmids
     The number of cells un-irradiated and crossed membrane decreased in the pcDNA3.1-AIF_(Δ1-480) and pcDNA3.1-Egr-1-AIF_(Δ1-480) groups. As compared with that in cells without irradiation, the number of cells irradiated with 2.0 Gy and crossed membrane decreased significantly in the control and pcDNA3.1 groups (P < 0.05), particularly in the pcDNA3.1-AIF_(Δ1-480) and pcDNA3.1-Egr-1-AIF_(Δ1-480) groups (P < 0.01). As compared with that in the control and pcDNA3.1 groups, the number of cells irradiated with 2.0 Gy and crossed membrane decreased significantly in the pcDNA3.1-AIF_(Δ1-480) and pcDNA3.1-Egr-1-AIF_(Δ1-480) groups (P < 0.01). The results indicate that the AIF_(Δ1-480) protein and ionizing radiation could reduce the invasive ability, particularly in the combination of AIF_(Δ1-480) protein and ionizing radiation.
     4.5 Effects of X-irradiation on expression of cytochrome c of MCF-7 cells transfected with recombinant plasmids
     The expression of cytochrome c protein of the cells without irradiation increased in the pcDNA3.1-AIF_(Δ1-480) and pcDNA3.1-Egr-1-AIF_(Δ1-480) groups. As compared with that in the control and pcDNA3.1 groups, the expression of cytochrome c protein of the cells irradiated with 2.0 Gy increased in the pcDNA3.1-AIF_(Δ1-480) and pcDNA3.1-Egr-1-AIF_(Δ1-480) groups, particularly in pcDNA3.1-Egr-1-AIF_(Δ1-480) group. The results indicate that the AIF_(Δ1-480) protein and ionizing radiation could increase Cyt c protein expression, particularly in the combination of AIF_(Δ1-480) protein and radiation.
     Above all, the gene expression plasmid pcDNA3.1-Egr-1-AIF_(Δ1-480) was constructed successfully in present study. The plasmid has the characteristics of irradiation inducibility, and combines with ionizing radiation could induce cell G1 and G2 phase arrests, increase Cyt c protein expression, inhibit the proliferation, promote the apoptosis and reduce the invasive ability in MCF-7 cells. The researches open up a new way to improve the effects of gene-radiation therapy, and provide the theoretical and experimental bases for the clinical application.
引文
[1]殷蔚伯,余子豪,徐国镇,主编.放射肿瘤学[M].第四版.北京:协和医科大学出版社, 2008.
    [2] ASTRO Fact Sheet. Available from: www.astro.org. Accessed March 15, 2008.
    [3]冯作华,药立波,周春燕,主编.医学分子生物学[M].第一版.北京:人民卫生出版社, 2005.
    [4] Mosesso P, Palitti F, Pepe G, et al. Relationship between chromatin structure, DNA damage and repair following X-irradiation of human lymphocytes [J]. Mutat Res, 2010, Epub ahead of print.
    [5] Cariveaua M J, Kalmus G W, Johnke R M, et al. Correlations between radiation-induced double strand breaks and cell cycle checkpoints in X-irradiated NIH/3T3 fibroblasts [J]. Anticancer Res, 2006, 26(5A): 3311-3316.
    [6] Firat E, Heinemann F, Grosu A L, et al. Molecular radiobiology meets clinical radiation oncology [J]. Radiat Biol, 2010, 86(3):252-259.
    [7]刘树铮,主编.医学放射生物学[M].第三版.北京:原子能出版社, 2006.
    [8] Dahm-Daphi J, Sass C, Alberti W. Comparison of biological effects of DNA damage induced by ionizing radiation and hydrogen peroxide in CHO cells [J]. Radiat Biol, 2000, 76(1):67-75.
    [9] Okayasu R, Takakura K, Poole S, et al. Radiosensitization of normal human cells by LY294002: cell killing and the rejoining of DNA and interphase chromosome breaks [J]. Radiat Res, 2003, 44(4):329-333.
    [10] Yokoya A, Fujii K, Ushigome T, et al. Yields of strand breaks and base lesions induced by soft X-rays in plasmid DNA [J]. Radiat Prot Dosim, 2006, 122(1-4):86-88.
    [11] Frankenberg-Schwager M, Gebauer A, Koppe C, et al. Single-strandannealing, conservative homologous recombination, nonhomologous DNA end joining, and the cell cycle-dependent repair of DNA double-strand breaks induced by sparsely or densely ionizing radiation [J]. Radiat Res, 2009, 171(3):265-273.
    [12] Cortes Ledesma F, Khamisy S F, Zuma M C, et al. A human 5'-tyrosyl DNA phosphodiesterase that repairs topoisomerase-mediated DNA damage [J]. Nature, 2009, 461(7264):674-678.
    [13] Valota A, Ballarini F, Friedland W, et al. Modelling study on the protective role of OH radical scavengers and DNA higher-order structures in induction of single- and double-strand break by gamma-radiation [J]. Radiat Biol, 2003, 79(8):643-653.
    [14] Prise K M, Folkard M, Michael B D, et al. Critical energies for SSB and DSB induction in plasmid DNA by low-energy photons: action spectra for strand-break induction in plasmid DNA irradiated in vacuum [J]. Radiat Biol, 2000, 76(7):881-890.
    [15] Nikjoo H, O’Neill P, Wilson W E, et al. Computational approach for determining the spectrum of DNA damage induced by ionizing radiation [J]. Radiat Res, 2001, 156(5 -2):577-583.
    [16] G?tz D, Paytubi S, Munro S, et al. Responses of hyperthermophilic crenarchaea to UV irradiation [J]. Genome Biol, 2007, 8(10):220-222.
    [17] Zhou B B, Elledge S J. The DNA damage response: putting checkpoints in perspective [J]. Nature, 2000, 408(6811):433-439.
    [18] Frankenberg D, Brede H J, Schrewe U J, et al. Induction of DNA double-strand breaks in mammalian cells and yeast [J]. Adv Space Res, 2000, 25(10):2085-2094.
    [19]杨恬主编.细胞生物学[M].第一版.北京:人民卫生出版社, 2005.
    [20] Torreggiani A, Tamba M, Ferreri C. Radical damage involving sulfur- containing enzymes and membrane lipids protein [J]. Pept Lett, 2007,14(7):716-722.
    [21] Mishra K P. Cell membrane oxidative damage induced by gamma-radiation and apoptotic sensitivity [J]. Environ Pathol Toxicol Oncol, 2004, 23(1):61-66.
    [22] Dizdaroglu M, Jaruga P, Birincioglu M, et al. Free radical-induced damage to DNA: mechanisms and measurement [J]. Free Rad Biol Med, 2002, 32(11):1102-1115.
    [23] Watanabe R, Yokoya A, Fujii K, et al. DNA strand breaks by direct energy deposition by Auger and photo-electrons ejected from DNA constituent atoms following K-shell photoabsorption [J]. Radiat Biol, 2004, 80(11-12):823-832.
    [24] Nikjoo H, Bolton C E. Modelling of DNA damage induced by energetic electrons (100 eV to 100 keV) [J]. Radiat Prot Dosim, 2002, 99(1-4):77-80.
    [25] Lacombe S, Le Sech C. Advances in radiation biology: radiosensitization in DNA and living cells. Surf Sci, 2009, 603:1953-1960.
    [26] Prise K M, Schettino G, Folkard M. New insights on cell death from radiation exposure [J]. Lancet Onol, 2005, 6(7):520-528.
    [27] Hall E J, Ed. Cell-survival curves. Radiobiology for the radiobiologist. Sixth Edition. Lippincott Company. Philadephia, 2005.
    [28] Hagemann G, Lipfert C H, Wüppen G. Radiation sensitivity for delayed reproductive death (DRD) following single or split-dose irradiation [J]. Strahlenther Onkol, 2001, 177(10):538-546.
    [29] Li P, Nijhawan D, Wang X. Mitochondrial activation of apoptosis [J]. Cell, 2004, 116(2 Suppl):857-859.
    [30] Schiller M, Blank N, Franz S, et al. Apoptotic bodies derived from apoptotic lymphoblasts contain a distinct pattern of antigens [J]. Autoimmunity, 2007, 40(4):340-341.
    [31] Merritt A, Allen T D, Potten C S, et al. Apoptosis in small intestinal epitheliafrom p53-null mice: evidence for a delayed, p53-independent G2/M- associated cell death after irradiation [J]. Oncogene, 1997, 14(23): 2759-2766.
    [32] Belloni P, Meschini R, Czene S, et al. Studies on radiation-induced apoptosis in G0 human lymphocytes [J]. Radiat Biol, 2005, 81(8):587-599.
    [33] Belloni P, Meschini R, Lewinska D, et al. Apoptosis preferentially eliminates irradiated G0 human lymphocytes bearing dicentric chromosomes [J]. Radiat Res, 2008, 169(2):181-187.
    [34] Mazumder S, Gong B, Chen Q, et al. Proteolytic cleavage of cycline leads to inactivation of associated kinase activity and amplification of apoptosis in hematopoietic cells [J]. Mol Cell Biol, 2002, 22(7):2398-2409.
    [35] Oh S H, Lim S C. A rapid and transient ROS generation by cadmium triggers apoptosis via caspase-dependent pathway in HepG2 cells and this is inhibited through N-acetylcysteine-mediated catalase upregulation [J]. Toxicol Appl Pharmacol, 2006, 12(3):212-223.
    [36] Wang H, Shang L, Hao W. DNA-damaging reagents induce apoptosis through reactive oxygen species-dependent Fas aggregation [J]. Oncogene, 2003, 22(50):8168-8177.
    [37] Yasumoto J, Imai Y, Takahashi A, et al. Analysis of apoptosis-related gene expression after X-ray irradiation in human tongue squamous cell carcinoma cells harboring wild-type or mutated p53 gene [J]. Radiat Res, 2003, 44(1):41-45.
    [38] McAndrew C W, Gastwirt R F, Donoghue D J. The atypical CDK activator Spy1 regulates the intrinsic DNA damage response and is dependent upon p53 to inhibit apoptosis [J]. Cell Cycle, 2009, 8(1):66-75.
    [39] Merritt J A, Roth J A, Logothetis C J. Clinical evaluation of adenoviral- mediated p53 gene transfer: review of INGN 201 studies [J]. Semin Oncol, 2001, 28(5 Suppl 16):105-114.
    [40] Lagarde F, Axelsson G, Dzmber L, et al. Residential radon and lung canceramong never-smokers in Sweden [J]. Epi demiology, 2001, 12(4):396-404.
    [41] Mary N, Mohankumar P, Venkatachalam B, et al. Comparison of UV-induced unscheduled DNA synthesis in lymphocytes exposed to low doses of ionizing radiation in vivo and in vitro [J]. Mutat Res, 2000, 447(2):199-207.
    [42] Pfeiffer P, Goedecke W, Kuhfittig-Kulle S, et al. Pathways of DNA double-strand break repair and their impact on the prevention and formation of chromosomal aberrations [J]. Cytogenet Genome Res, 2004, 104(1-4):7-13.
    [43] Igarashi K, Miura M. Inhibition of a radiation-induced senescence-like phenotype: a possible mechanism for potentially lethal damage repair in vascular endothelial cells [J]. Radiat Res, 2008, 170(4):534-539.
    [44] Fitzpatrick C L, Farese J P, Milner R J, et al. Intrinsic radiosensitivity and repair of sublethal radiation-induced damage in canine osteosarcoma cell lines [J]. Am J Vet Res, 2008, 69(9):1197-1202.
    [45] Ruan K, Song G, Ouyang G. Role of hypoxia in the hallmarks of human cancer [J]. Cell Biochem, 2009, 107(6):1053–1062.
    [46] Kumar P R, Mohankumar M N, Hamza V Z, et al. Dose-rate effect on the induction of HPRT mutants in human G0 lymphocytes exposed in vitro to gamma radiation [J]. Radiat Res, 2006, 165(1):43-50.
    [47] Philip Rubin,主编.临床肿瘤学[M].第一版.北京:人民卫生出版社, 2002.
    [48] Shinomiya N. New concepts in radiation-induced apoptosis:‘premitotic apoptosis’and‘postmitotic apoptosis [J]. Cell Mol Med, 2001, 5(3):240-253.
    [49] Williams J R, Zhang Y, Zhou H, et al. A quantitative overview of radiosensitivity of human tumor cells across histological type and TP53 status [J]. Radiat Biol, 2008, 84(4):253-264.
    [50]沈瑜,糜福顺,主编.肿瘤放射生物学[M].第一版.北京:中国医药科技出版社, 2002.
    [51] Fowler J F. Biological factors influencing optimum fractionation in radiation therapy [J]. Acta Oncol, 2001, 40(6):712-717.
    [52] Guse K, Hemminki A. Cancer gene therapy with oncolytic adenoviruses [J]. BUON, 2009, 14(Suppl 1):7-15.
    [53] Navarro J, Risco R, Toschi M, et al. Gene therapy and intracytoplasmatic sperm injection (ICSI) - a review [J]. Placenta, 2008, 29(Suppl B):193-199.
    [54] http://www.tumor.cn/html/0342/1060.html.
    [55] Wasserfall C H, Herzog R W. Gene therapy approaches to induce tolerance in autoimmunity by reshaping the immune system [J]. Curr Opin Investig Drugs, 2009, 10(11):1143-1150.
    [56]曹雪涛,顾健人,刘德培,等.我国基因治疗的研究前景与战略重点[J].中华医学杂志, 2001, 81(12):705-708.
    [57] Gridley D S, Slater J M. Combining gene therapy and radiation against cancer [J]. Curr Gene Ther, 2004, 4(3):231-248.
    [58] Dubensky T W, Liu M A, Ulmer J B. Delivery systems for gene-based vaccines [J]. Mol Med, 2000, 6 (9): 723-732.
    [59] Pandori M, Hobson D, Sano T. Adenovirus-microbead conjugates possess enhanced infectivity: a new strategy for localized gene delivery [J]. Virology, 2002, 299(2):204-212.
    [60] Russell W C. Update on adenovirus and its vectors [J]. Gen Virol, 2000, 81(11): 2573-2604.
    [61] Weber E, Anderson W F, Kasahara N. Recent advances in retrovirus vector mediated gene therapy: teaching an old vector new tricks [J]. Curr Opin Mol Ther, 2001, 3(5):439-453.
    [62]曹明媚.基因治疗载体的研究进展[J].国外医学·肿瘤分册, 2004, 31(1):22-26.
    [63] Shimotohno K, Temin H M. Formation of infectious progeny virus after insertion of herpes simp lex thymidine kinase gene into DNA of an avianretrovirus [J]. Cell, 1981, 26(Pt1):67-77.
    [64] Sena-Esteves M, Hampl J A, Camp S M, et al. Generation of stable retrovirus packaging cell lines after transduction with herpes simplex virus hybrid amplicon vectors [J]. Gene Med, 2002, 4(3):229-239.
    [65]庞凤.用于肿瘤基因治疗的慢病毒载体研究进展[J].青岛大学医学院学报, 2009, 45(5):493-494, 497.
    [66] Coura R S, Nnardi N B. The state of the art of adeno-associated virus-based vectors in gene therapy [J]. Virol, 2007, 16(4):99.
    [67] Bonsted A, Engesaeter B, Hogset A, et al. Transgene expression is increased by photochemically mediated transduction of polycation-complexed adenoviruses [J]. Gene Ther, 2004, 11(2):152-160.
    [68] Escriou V, Carriere M, Scherman D, et al. NLS bioconjugates for targeting therapeutic genes to the nucleus [J]. Adv Drug Deliv Rev, 2003, 55(2):295-306.
    [69] Kneuer C, Ehrhardt C, Bakowsky H, et al. The influence of physicochemical parameters on the efficacy of non-viral DNA transfection complexes: a comparative study [J]. J Nanosci Nanotechnol, 2006, 6(9-10):2776-2782.
    [70] Yamada T, Iwasaki Y, Tada H, et al. Nanoparticles for the delivery of genes and drugs to human hepatocytes [J]. Nature Biotechnol, 2003, 21(8):885-890.
    [71] Shishido T, Yonezawa D, Iwata K, et al. Construction of arginine-rich peptide displaying bionanocapsules [J]. Bioorg Med Chem Lett, 2009, 19(5):1473-1476.
    [72] Ito A, Matsuoka F, Honda H, et al. Antitumor effects of combined therapy of recombinant heat shock protein 70 and hyperthermia using magnetic nanoparticles in an experimental subcutaneous murine melanoma [J]. Cancer Immunol Immunother, 2004, 53(1):26-32.
    [73] Corsi K, Chellat F, Yahia L, et al. Mesenchymal stem cells, MG63 andHEK293 transfection using chitosan-DNA nanoparticles [J]. Biomaterials, 2003, 24(7):1255-1264.
    [74] Cui Z, Mumper R J. Plasmid DNA-entrapped nanoparticles enginerred from microemulsion erecursors: in vitro and in vivo evaluation [J]. Bioconjug Chem, 2002, 13(6):1319-1327.
    [75] Luo D, Saltzman W M. Enhancement of transfection by physical concentration of DNA at the cell surface [J]. Nat Biotechnol, 2000, 18(8):893-895.
    [76] Juliano R L, Alahari S, Yoo H, et al. Antisense pharmacodynamics: critical issues in the transport and delivery of antisense oligonuclecides [J]. Pharm Res, 1999, 16(4):494-502.
    [77] Anderson W F. Human gene therapy [J]. Nature, 1998, 392 (6679 suppl): 25-30.
    [78]王申五,主编.基因治疗与临床[M].第一版.北京:北京医科大学中国协和医科大学联合出版社, 1996.
    [79] Cavazzana-Calvo M, Hacein-Bey S, de Saint Basile G, et al. Gene therapy of human severe combined immunodeficiency (SCID)-X1 disease [J]. Science, 2000, 288(5466): 669-672.
    [80] Marchisone C, Pfeffer U, Del G F, et al. Progress towards gene therapy for cancer [J]. Exp Clin Cancer Res, 2000, 19(3):261-270.
    [81]来茂德,主编.医学分子生物学[M],第一版.北京:人民卫生出版社, 2001.
    [82] Nemunaitis J M. Potential of Advexin: a p53 gene-replacement therapy in Li-Fraumeni syndrome [J]. Future Oncol, 2008, 4(6):759-768.
    [83] Huang C L, Yokomise H, Miyatake A. Clinical significance of the p53 pathway and associated gene therapy in non-small cell lung cancers [J]. Future Oncol, 2007, 3(1):83-93.
    [84] Xu H J. Retinoblastoma and tumor-suppressor gene therapy [J]. Ophthalmol Clin North Am, 2003, 16(4):621-629.
    [85] Vickers T A, Koo S, Bennett C F, et al. Efficient reduction of target RNAs by small interfering RNA and RNase H-dependent antisense agents [J]. Biol Chem, 2003, 278(9): 7108-7118.
    [86]杨栓平,宋海峰,宋三泰,等.靶向HER-2 mRNA反义寡核苷酸对SK-BR-3乳腺癌细胞Caspase-3蛋白表达的影响[J].中国药理学通报, 2003, 19(5):505-507.
    [87] Niculescu-Duvaz I, Springer C J. Introduction to the background, principles, and state of the art in suicide gene therapy [J]. Mol Biotechnol, 2005, 30(1):71-88.
    [88] Tanaka T, Yamasaki H, Mesnil M. Induction of a bystander effect in Hela cells by using a bigenic vector carrying viral thymidine kinase and connexin32 genes [J]. Mol Carcinog, 2001, 30(3):176-180.
    [89] Kirn D, Niculescu D I, Hallden G, et al. The emerging fields of suicide gene therapy and virotherapy [J]. Trends Mol Med, 2002, 8 (4 Suppl):68-73.
    [90] Hampl M, Tanaka T, Albert P S, et al. Therapeutic effects of viral vector-mediated antiangiogenic gene transfer in malignant ascites. Hum Gene Ther, 2001, 12(14): 1713-1729.
    [91] Mcnally V A, Patterson A V, Williams K J, et al. Antiangiogenic ,bioreductive and gene therapy approaches to the treatment of hypoxic tumours [J]. Curr Pharm Des, 2002, 8 (15):1319-1333.
    [92] Liu K. Breakthroughs in cancer gene therapy [J]. Semin Oncol Nurs, 2003, 19(3):217-226.
    [93] Yoshida J, Mizuno M, Nakahara N, et al. Antitumor effect of an adeno- associated virus vector containing the human interferon-beta gene on experimental intracranial human glioma [J]. Cancer Res, 2002, 93(2):223-228.
    [94] Ahn S J, Jeon Y H, Lee Y J, et al. Enhanced anti-tumor effects of combined MDR1 RNA interference and human sodium/iodide symporter (NIS) radioiodine gene therapy using an adenoviral system in a colon cancer model[J]. Cancer Gene Ther, 2010, 26:Epub ahead of print.
    [95] Yuan H, Li X, Wu J, et al. Reversal of multi-drug resistance by pSUPER- shRNA-mdr1 in vivo and in vitro [J]. Curr Med Chem, 2008, 15(5):470-476.
    [96] Takahashi S, Aiba K, Ito Y, et al. Pilot study of MDR1 gene transfer into hematopoietic stem cells and chemoprotection in metastatic breast cancer patients [J]. Cancer Sci, 2007, 98(10):1609-1616.
    [97] Lumniczky K, Sáfrány G. Cancer gene therapy: combination with radiation therapy and the role of bystander cell killing in the anti-tumor effect [J]. Pathol Oncol Res, 2006, 12(2):118-124.
    [98] Chen J K, Hu L J, Wang D, et al. Cytosine deaminase/5-fluorocytosine exposure induces bystander and radiosensitization effects in hypoxic glioblastoma cells in vitro [J]. Radiat Oncol Biol Phys, 2007, 67(5): 1538-1547.
    [99] Nestler U, Wakimoto H, Siller-Lopez F, et al. The combination of adenoviral HSV TK gene therapy and radiation is effective in athymic mouse glioblastoma xenografts without increasing toxic side effects [J]. Neurooncol, 2004, 67(1-2):177-188.
    [100] Li X J, Wang K M, Xu Y, et al.Ionizing radiation-regulated killing of human hepatoma cells by liposome-mediated CDglyTK gene delivery [J]. Sheng Wu Hua Xue Yu Sheng Wu Wu Li Xue Bao (Shanghai), 2003, 35(1):64-70.
    [101] Ahmed M M. Regulation of radiation-induced apoptosis by early growth response-1 gene in solid tumors [J]. Curr Cancer Drug Targets, 2004, 4(1):43-52.
    [102] Han Z, Wang H, Hallahan D E. Radiation-guided gene therapy of cancer [J]. Technol Cancer Res Treat, 2006, 5(4):437-444.
    [103] Weichselbaum R R, Kufe D. Translation of the radio- and chemo-inducible TNFerade vector to the treatment of human cancers [J].Cancer Gene Ther. 2009,16(8):609-619.
    [104] Gatz S A, Wiesmüller L. p53 in recombination and repair [J]. Cell Death Differ, 2006, 13(6):1003-1016.
    [105] Kato S, Han S Y, Liu W, et al. Understanding the function-structure and function-mutation relationships of p53 tumor suppressor protein by high- resolution missense mutation analysis [J]. Proc Nat1 Acad Sci USA, 2003, 100(14): 8424-8429.
    [106] Talos F, Petrenko O, Mena P, et al. Mitochondrially targeted p53 has tumor suppressor activities in vivo [J]. Cancer Res, 2005, 65(21):9971-9981.
    [107] Swisher S G, Roth J A, Komaki R, et al. Induction of p53-regulated genes and tumor regression in lung cancer patients after intratumoral delivery of adenoviral p53 (INGN 201) and radiation therapy [J]. Clin Cancer Res, 2003, 9(1):93-101.
    [108] Christy B, Nathans D. DNA binding site of the growth factor-inducible protein Zif268 [J]. Proc Natl Acad Sci USA, 1989, 86(22): 8737-8741.
    [109] Datta R, Rubin E, Sukhatme V, et al. Ionizing radiation activates transcription of the EGR-1 gene via CArGelements [J]. Proc Natl Acad Sci, 1992, 89(21):10149-10153.
    [110] Lei N, Heckert L L. Sp1 and Egr-1 regulate transcription of the Dmrt1 gene in Sertoli cells [J]. Biol Reprod, 2002, 66(3):675-684.
    [111] Papanikolaou N A, Sabban E L. Ability of Egr-1 to activate tyrosine hydroxylase transcription in PC12 cells. Cross-talk with AP21 factors [J]. Biol Chem, 2000, 275 (35):26683-26689.
    [112] Weichselbaum R R, Hallahan D E, Beckett M A, et al. Gene therapy by radiation preferentially radiosensitizes tumor cells [J]. Cancer Res, 1994, 54(16):4266-4269.
    [113] Scott S D, Joiner M C, Marples B. Optimizing radiation-responsive gene promoters for radiogenetic cancer therapy [J]. Gene Ther, 2002, 9(20):1396- 1402.
    [114] Scott S D, Marples B, Hendry J H, et al. A radiation-controlled molecular switch for use in gene therapy of cancer [J]. Gene Ther, 2000, 7(13):1121-1125.
    [115]田梅,金光辉,朴春姬,等.辐射诱导表达载体pEgr-hPTEN的构建及其体外抗肿瘤作用的研究[J].中华放射医学与防护杂志, 2003, 23 (6): 423-426.
    [116]朴春姬,田梅,刘林林,等.辐射诱导表达载体pEgr-hTRAIL的构建及其对肿瘤细胞的体外诱导凋亡作用[J].吉林大学学报(医学版), 2005, 31(2): 169-172.
    [117]金光辉,刘树铮,杨建征,等. pEgr-IL18-B7.1联合辐射抗肿瘤作用及其免疫学机制探讨[J].中国病理生理杂志, 2005, 21(10):1883-1887.
    [118]杨巍,刘林林,孙婷,等.小鼠内皮抑素基因克隆、测序及pEgr-IFNγ-mEndostatin重组双基因表达质粒的构建[J].吉林大学学报(医学版), 2004, 30 (1):17-19.
    [119] Susin S A, Zamzami N, Castedo M, et al. Bcl-2 inhibits the mitochondrial release of an apoptogenic protease [J]. Exp Med, 1996, 184:1331-1342.
    [120] Loeffler M, Daugas E, Susin S A, et al. Dominant cell death induction by extramitochondrially targeted apoptosis inducing factor [J]. FASEB, 2001, 15(3):758-767.
    [121] Susin S A, Lorenzo H K, Zamzami N, et al. Molecular characterization of mitochondrial apoptosis-inducing factor [J]. Nature, 1999, 397:441-446.
    [122] Maria J M, Miguel O L, Brigette B, et al. The crystal structure of themouse apoptosis- inducing factorAIF [J]. Nature, 2002, 9(6):442-446.
    [123] Delettre C, Yuste V J, Moubarak R S, et al. AIFsh a novel apoptosis inducing factor (AIF): pro-apoptotic isoform with potential pathological relevance in human cancer [J]. Biol Chem, 2006, 281(10):6413-6427.
    [124] Daugas E, Nochy D, Ravagnan L, et al. Mitochondrio-nuclear redistribution of AIF in apoptosis and necrosis [J]. FASEB, 2000, 14(5):729-739.
    [125] Green D R, Kroemer G. The pathophysiology of mitochondrial cell death [J].Science, 2004, 305(5684):626-629.
    [126] Otera H, Ohsakaya S, Nagaura, et al. Export of mitochondrial AIF in response to proapoptotic stimuli depends on processing at the intermembrane space [J]. EMBO, 2005, 24(7):1375-1386.
    [127] Chiuargi A, Moskowizt M A, Ferrnate M, et al. PARP-l-a perpetrator of apoptotic cell death [J]? Science, 2002, 297:200-201.
    [128] Arnoult D, Parone P, Martinou J C, et al. Mitochondrial release of AIF and EndoG requires caspase activation downstream of Bax/Bak-mediated permeabilization [J]. EMBO, 2003, 22(17):4385-4399.
    [129] Olie R A, Durrieu F, Cornillon S, et al. Apparet caspase independence of programmed cell death in dicytostelium [J]. Curr Biol, 1998, 8(17):955-958.
    [130] Arnoult D, Estaquier J, Tatischeff I, et al. On the evolutionary consevration of the cell death pathway: mitochondrial release of an apoptosis inducing factor during Dieytostelium discoideum cell death [J]. Mol Biol Cell, 2001, 12(10):3016-3030.
    [131] Wang X C, Yang C L, Chai J S, et al. Mechanisms of AIF-mediated apoptotic DNA degradation in Caenorhabditis elegans [J]. Science, 2002, 298(5598): 1587-1592.
    [132] Koonin E V, Aravind L. Origin and evolution of eukaryotic apoptosis:the bacterial connection [J]. Cell Death Differ, 2002, 9(4):394-404.
    [133] Uren R T, Dewson G, Bonzon C, et al. Mitochondrial release of pro-apoptotic proteins: electrostatic interactions can hold cytochrome c but not Smac/ DIABLO to mitochondrial membranes [J]. Biol Chem, 2005, 280(3):2266- 2274.
    [134] Ye H, Cande C, Stephanou, et al. DNA binding as a structural requirement for the apoptogenic action of AIF [J]. Nat Struct Biol, 2002, 9:680-684.
    [135] Vahsen N. Physical interaction of apoptosis inducing factor (AIF) with DNA and RNA [J]. Oncogene, 2000, 25(12):1763-1774.
    [136] Lipton S A, Bossy-Wetzel E. Dueling activities of AIF in cell death vesrus survival: DNA binding and redox activity [J]. Cell, 2002, 111(2):147-150.
    [137] Susin S A, Daugas E, Ravagnan L, et al. Two distinct pathways leading to nuclear apoptosis [J]. Exp Med, 2000, 192(4):571-580.
    [138] Loeffler M, Daugas E, Susin S A, et al. Dominant cell death induction by extramitochondrially targeted apoptosis inducing factor [J]. FASER, 2001, 15(3):758-767.
    [139] Ohiro Y, Garkavtsev L, Kobayashi S, et al. A novel P53-inducible apoptogenic gene,PRG3, encodes a homologue of the apoptosis-inducing factor (AIF) [J]. FEBS Lett, 2002, 524(1-3):163-171.
    [140] Wu M, Xu L G, Li X, et al. AMID,an apoptosis-inducing factor homologous mitochondrion-associated Protein,induces caspase-independent apoptosis [J]. Biol Chem, 2002, 277:25617-25623.
    [141]于翠娟,孟艳玲,桂俊豪,等.重组人凋亡诱导因子基因的构建、表达及对HeLa细胞的促凋亡作用[J].生物化学与生物物理学进展, 2002, 29(6):915-921.
    [142] Ramsay A J, Kent S J, Strugnell R A, et al. Genetic vaccination strategies for enhanced cellular, humeral and mucosal immunity [J]. Immunol Rev, 1999, 171(2):27-44.
    [143]张巍,张勇,韩涛,等.截短型人凋亡诱导因子AIF△1 2400对HeLa细胞的促凋亡作用[J].细胞与分子免疫学杂志, 2007, 23(10):973-975.
    [144] Miramar M D, Costantini P, Ravagnan L, et al. NADH-oxidase activity of mitochondrial apoptosis-inducing factor (AIF) [J]. Biol Chem, 2001, 276(19):16391-16398.
    [145] Joza N, Susin S A, Daugas E, et al. Essential role of the mitochondrial apoptosis-inducing factor in programmed cell death [J]. Nuatre, 2001, 410(6828):549-554.
    [146] Zhang X, Chen J, Graham S H, et al. Intranuclear localization of apoptosisinducing factor r (AIF) and large scale and fragmentation after traumatic brain injury in rats and in neuronal cultures exposed to peroxynitrite [J]. Neuro chem, 2002, 82(1):181-191.
    [147] Zamzami N, Hamel C, Munoz C, et al. Bid acts on the permeability transition pore complex to induce apoptosis [J]. Oncogene, 2000, 19(54):6342-6350.
    [148] Guo Y, Srinivasula S M, Druilhe A, et al. Caspase-2 induces apoptosis by releasing proapoptotic proteins from mitochondria [J]. Biol Chem, 2002, 277(16):13430-13437.
    [149] Susin S A, Zamzami N, Castedo M, et al. The central executioner of apoptosis multiple links between protease activation and mitochondria in Fas/Apo-1/CD95-and ceramide-induced apoptosis [J]. Exp Med, 1997, 186:25-37.
    [150] Ravagan L, Gurbuxani S, Susin S A, et al. Heat shock protein 70 antagonizes apoptosis-inducing factor [J]. Nat Cell Biol, 2001, 3(9):839-843.
    [151] Rashmi R, Santhosh T R, Karunagaran D. Human colon cancer cells differ in their sensitivity to curcumin-induced apoptosis and heat shock Protects them by inhibiting the release of apoptosis-inducing factor and caspases [J]. FEBS Lett, 2003, 538(1-3):19-24.
    [152] Beere H M, Wolf B B, Cain K, et al. Heat-shock protein 70 inhibits apoptosis by preventing recruitment of procaspase-9 to the Apaf-1 [J]. Nat Cell Biol, 2000, 2(8):469-475.
    [153] Rugo H, Shtivelman E, Perez A, et al. PhaseⅠtrial and antitumor effects of BZL 101 for patients with advanced breast cancer [J]. Breast Cancer Res Treat, 2007, 105(1):17-28.
    [154] Wang M, Zhang L, Han X, et al. Atiprimod inhibits the growth of mantle cell lymphoma in vitro and in vivo and induces apoptosis via activating the mitochondrial pathways [J]. Blood, 2007, 109(12):5455-5462.
    [155] Newcomb E W, Tamasdan C, Entzminger Y, et al. Flavopiridol induces mitochondrial- mediated apoptosis in murine glioma GL261 cells via release of cytochrome c and apoptosis inducing factor [J]. Cell Cycle, 2003, 2(3):243-250.
    [156] Lee J H, Park S Y, Shin H K, et al. Poly (ADP-ribose): polymerase inhibition by cilostazol is implicated in the neuroproteetive effect against focal cerebral ischemic infarct in rat [J]. Brain Res, 2007, 1152:182-190.
    [157]龚守良,刘淑春,吕喆,等.低剂量辐射诱导小鼠胸腺细胞周期进程的适应性反应[J].辐射研究与辐射工艺学报, 2004, 22 (3): 176-180.
    [158]刘淑春,赵文举,吕喆,等.低剂量辐射诱导EL-4淋巴瘤细胞凋亡及细胞周期进程适应性反应的剂量率效应[J].吉林大学学报(医学版), 2008, 34(1):24-27.
    [159] Zhang Y, Han T, Zhu Q, et al. The proapoptotic activity of C-terminal domain of apoptosis-inducing factor (AIF) is separated from its N-terminal [J]. Biol Res, 2009, 42(2):249-260.
    [160] Barros M H, Netto L E, Kowaltowski A J, et al. H2O2 generation in Saccharomyces cerevisiae respiratory pet mutants: effect of cytochrome c [J]. Free Radic Biol Med, 2003, 5(2):179-188.
    [161] Singh M H, Brooke S M, Zemlyak I, et al. Evidence for caspase effects on release of cytochrome c and AIF in a model of ischemia in cortical neurons [J]. Neurosci Lett, 2010, 469(2):179-183.

© 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