用户名: 密码: 验证码:
pshuttle-Egr1-shTRAIL-shES双基因—放射治疗的体外抑瘤效应实验研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
放射治疗是目前临床治疗肿瘤的重要手段之一,但是由于肿瘤周边组织的放射损伤及某些肿瘤的辐射抗性问题,使放疗的疗效和应用受到限制。恶性肿瘤基因-放射治疗是近年来肿瘤治疗领域新的研究热点之一,是根据放射治疗和基因治疗的各自特点,将二者联合应用,即将辐射诱导性基因的调控序列与肿瘤杀伤基因相偶联,转染肿瘤细胞,在对肿瘤实施局部放疗的同时诱导肿瘤杀伤基因表达的增强,产生辐射和基因表达产物的协同抑瘤作用。该疗法一方面将放疗与基因治疗有机地结合,发挥协同作用;另一方面,由于辐射具有靶向性和可控性,实现了对杀伤基因表达的时空调控。
     利用Egr-1启动子具有辐射诱导特性,即电离辐射诱导下可启动其下游基因的表达,本实验构建了含Egr-1启动子和TRAIL、endostatin双基因的重组质粒pshuttle-Egr1- shTRAIL-shES,研究在电离辐射诱导下,该重组质粒携带的双基因TRAIL和endostatin mRNA及蛋白表达的时效和量效规律,以及观察其联合X射线照射后,分别对人乳腺癌细胞MCF-7和人血管内皮细胞ECV304增殖、周期和凋亡的影响。结果表明,重组质粒pshuttle-Egr1-shTRAIl-shES中TRAIL和endostatin mRNA和蛋白的表达具有辐射诱导双基因共表达特性,且在联合X射线照射后,对MCF-7和ECV304细胞均具有明显的增殖抑制和促凋亡作用,并可改变细胞周期进程。本研究为提高基因-放射治疗效果开辟了新途径,为双基因-放射治疗的临床应用提供理论和实验依据。
At present, the primary ways for tumor therapy are operation, radiotherapy, chemotherapy and biotherapy, and there are also other effective ways, including endocrine therapy, the traditional Chinese medicine treatment, thermotherapy, radiofrequency ablation therapy and so on. Tumor therapy is aimed to eliminate tumor and (or) prevent the tumor growth, meanwhile, minimize the injury to normal tissues, and improve the recovery rate. Nowadays, with the rapid development of molecular biology and genetic engineering, gene-radiotherapy has been a hot spot in the research of tumor therapy, which combines gene therapy and radiotherapy together to cooperate the effects of each therapeutic mean, so would possess good application prospect for the treatment of regional tumor. The discovery of radiation-sensitive early growth response-1 (Egr-1) promoter provides an opportunity for the effective combination of radiotherapy and gene therapy, and establishes the theoretical foundation for malignant tumor gene-radiotherapy. In this study, in order to combine radiotherapy and gene therapy together to cure cancer, the plasmid pshuttle-Egr1-shTRAIL-shES, which could express double genes (TRAIL and endostatin), would be constructed successfully by taking advantage of multiple-gene introduction system. There are three features in this study, one is that the radiation could kill tumor cells and activate transcription of the Egr-1 promoter to induce the expression of the downstream genes, another one is the shTRAIL gene could promote apoptosis of tumor cells, and the third one is the shES gene could inhibit angiogenesis. In this way, it could lower the radiation dose, and alleviate or avoid damage to normal tissues, and combine radiotherapy, promoting-apoptosis and anti-angiogenesis together, thus achieve the purpose of inhibiting tumor growth and killing tumor cells, which would provide a promising way for tumor treatment.
     1 Construction of recombinant plasmids
     1.1 Acquisition of Egr-1 promoter
     The specific PCR primers were designed and synthesized with the following primers according to the sequence of Egr-1 promoter gene: upstream 5'-ggggtaccgacccggaaacgccatataag- 3', including KpnⅠenzyme digestion sites, downstream 5'-ataagaatgcggccgcccaagttctgcgcgctgg g-3', including NotⅠenzyme digestion sites. The Egr-1 promoter gene was cloned and amplified from T-Egr1 as the template, and identified by cleavage of endonucleases and sequencing process. The results of identification confirmed that the sequence of the cloned gene was identical to that published on Genbank.
     1.2 Acquisition of shTRAIL gene
     The specific PCR primers were designed and synthesized with the following primers according to the sequence of shTRAIL gene: upstream 5'-ctagtctagacaccatgagcactgaaagcatgatc- 3', including XbaⅠenzyme digestion sites; downstream 5'-cgggatccctagttagccaactaaaaa-3', including BamHⅠenzyme digestion sites. The shTRAIL gene was amplified from pACCMV-shTRAIL as the template, and ligated to T vector for sequencing. The result of sequencing analysis was in coincidence with the anticipated result.
     1.3 Acquisition of shES gene
     Firstly, the primers for cloning endostatin gene were designed and synthesized: upstream 5'-ttgatatcatgcacagccaccgcgacttccag-3', including EcoRⅤenzyme digestion sites, downstream 5'-gctctagactacttggaggcagtcatgaa-3', including XbaⅠenzyme digestion sites. The mRNA was extracted from the liver of five-month aborted fetus, and acted as the template of reverse transcription. The endostatin gene was acquired by RT-PCR, and ligated to T vector for sequencing. The result of sequencing analysis was in coincidence with the anticipated result. Then the shES gene was gained by PCR amplification and ligated to T vector for sequencing. The PCR amplification system included the pMD19T-Endostatin plasmid as the template, synthesized IL-2 secretory signal-peptide as the upstream primer, and the downstream primer for cloning endostatin gene as the downstream primer.
     1.4 Construction of recombinant plasmids
     The Egr-1, shTRAIL and shES genes were ligated to pshuttle vector to construct the single-gene recombinant plasmids, pshuttle-Egr1-shTRAIL and pshuttle-Egr1-shES, and the double-gene recombinant plasmids pshuttle-Egr1-shTRAIL-shES by making use of the technique of genetic engineering. In the following experiments, the expression rule of these plasmids in human breast cancer MCF-7 cells and human vascular endothelial ECV304 cells, and their effects on the cellular growth, apoptosis and cell cycle of the two cell lines were detected.
     2 Experimental grouping and index detection
     The experiment was divided into five groups which were the control, pshuttle, pshuttle-Egr1-shTRAIL, pshuttle-Egr1-shES and pshuttle-Egr1-shTRAIL-shES groups. In the time-course experiment, the selected time points were 2, 4, 8, 12, 24 and 48 h, and the irradiation dose was 2.0 Gy. In the dose-effect experiment, the selected irradiation doses were 0, 0.1, 0.5, 1.0, 2.0 and 5.0 Gy. RT-PCR and ELISA were used to detecte the expressions of shTRAIL and shES on both mRNA and protein levels respectively. MTT assay was used to detect the cell proliferation, and flow cytometry to cell cycle and apoptosis.
     3 Radiation-induced expression rule of recombinant plasmids in MCF-7 cells
     3.1 Time-course changes of shTRAIL and shES mRNA and protein expressions in MCF-7 cells or the supernatant transfected with recombinant plasmids after 2.0 Gy X-irradiation
     The cells and supernatant were harvested in different time after 2.0 Gy X-irradiation. The expressions of shTRAIL and shES mRNA in MCF-7 cells and that of shTRAIL and shES protein in the supernatant were detected by RT-PCR and ELISA respectively. The results showed that the expressions of shTRAIL mRNA in the pshuttle-Egr1-shTRAIL and pshuttle-Egr1-shTRAIL-shES groups,and that of shES mRNA in the pshuttle-Egr1-shES and pshuttle-Egr1-shTRAIL-shES groups increased significantly, and manifested the time-effect relationship to some extent. Otherwise, it had no changes in other corresponding groups basically. As compared with those in the 0 h, except for the shTRAIL protein expression in the pshuttle-Egr1-shTRAIL-shES group at 2 and 4 h which increased slightly but without significance, the shTRAIL and shES protein expressions in the supernatant of MCF-7 cells transfected with plasmids increased significantly at other time-points (P < 0.01). The shTRAIL protein expression in the pshuttle-Egr1-shTRAIL and pshuttle-Egr1-shTRAIL-shES groups increased gradually along with the time-prolongation, and the former up to the peak value at 12 h, then decreased, while the later up to the peak value at 48 h. The shES protein expressions in the pshuttle-Egr1-shES and pshuttle-Egr1-shTRAIL-shES groups increased with the time-prolongation, and that at 48 h was the highest.
     3.2 Dose-effect changes of shTRAIL and shES mRNA and protein expressions in MCF-7 cells or the supernatant transfected with recombinant plasmids after X-irradiation with different doses
     The cells and supernatant were harvested at 12 h after X-irradiation with different doses. The expressions of shTRAIL and shES mRNA in MCF-7 cells and that of shTRAIL and shES protein in the supernatant was detected by RT-PCR and ELISA respectively. The results showed that the expressions of shTRAIL mRNA in the pshuttle-Egr1-shTRAIL and pshuttle-Egr1-shTRAIL-shES groups, and that of shES mRNA expressions in the pshuttle-Egr1-shES and pshuttle-Egr1-shTRAIL-shES groups increased significantly after X-irradiation, and even the irradiation with very low dose (0.1 Gy) induced the gene expression, while it had no changes in other corresponding groups basically. As compared with those in the 0 Gy, except for the shTRAIL protein expression in the pshuttle-Egr1-shTRAIL plus 0.5 Gy group with slight increase but no significance, the shTRAIL and shES protein expressions in the supernatant of MCF-7 cells transfected with plasmids increased significantly after X-irradiation with different doses (P < 0.05, P < 0.01), and increased along with the increasing of doses. The shTRAIL protein expression in the pshuttle-Egr1-shTRAIL group was up to the peak value after 2.0 Gy irradiation, while that in the pshuttle-Egr1-shTRAIL-shES group peaked after 0.5 Gy irradiation and sustained at a higher level afterward. The shES protein expression in the pshuttle-Egr1-shES and pshuttle-Egr1-shTRAIL-shES groups sustained at a higher level after 0.1 and 0.5 Gy irradiation respectively.
     4 Radiation-induced expression rule of recombinant plasmids in ECV304 cells
     4.1 Time-course changes of shTRAIL and shES mRNA and protein expressions in ECV304 cells or the supernatant transfected with recombinant plasmids after 2.0 Gy X-irradiation
     The cells and supernatant were harvested in different time after 2.0 Gy X-irradiation. The expressions of shTRAIL and shES mRNA in ECV304 cells and that of shTRAIL and shES protein in the supernatant was detected by RT-PCR and ELISA respectively. The results showed that the expressions of shTRAIL mRNA in the pshuttle-Egr1-shTRAIL and pshuttle-Egr1-shTRAIL-shES groups,and that of shES mRNA in the pshuttle-Egr1-shES and pshuttle-Egr1-shTRAIL-shES groups increased significantly, and manifested the time-effect relationship to some extent. Otherwise, it had no changes in other corresponding groups basically. As compared with those in the 0 h, the shTRAIL and shES protein expressions at other time-points increased significantly (P < 0.01). The shTRAIL protein expression increased gradually in both pshuttle-Egr1-shTRAIL and pshuttle-Egr1-shTRAIL-shES groups, and up to the peak value at 48 and 24 h respectively. The shES protein expression increased gradually and up to the peak value at 48 h in the pshuttle-Egr1-shES group, and up to the peak at 12 h in the pshuttle-Egr1-shTRAIL-shES group.
     4.2 Dose-effect change of shTRAIL and shES mRNA and protein expression in ECV304 cells or the supernatant transfected with recombinant plasmids after X-irradiation with different doses
     The cells and supernatant were harvested 12 h after X-irradiation with different doses. The expressions of shTRAIL and shES mRNA in ECV304 cells and that of shTRAIL and shES protein in the supernatant was detected by RT-PCR and ELISA respectively. The results showed that the expressions of shTRAIL mRNA in the pshuttle-Egr1-shTRAIL and pshuttle-Egr1-shTRAIL-shES groups, and that of shES mRNA expressions in the pshuttle-Egr1-shES and pshuttle-Egr1-shTRAIL-shES groups increased significantly after X-irradiation, and even the irradiation with very low dose (0.1 Gy) induced the gene expression, while it had no changes in other corresponding groups basically. As compared with those in the 0 Gy, the shTRAIL and shES protein expressions increased significantly after X-irradiation with different doses (P < 0.05, P < 0.01). The shTRAIL protein expressions in the pshuttle-Egr1-shTRAIL and pshuttle-Egr1-shTRAIL-shES groups increased along with the increasing of doses, and that in 5.0 Gy was the highest. The shES protein expressions in both the pshuttle-Egr1-shES and pshuttle-Egr1-shTRAIL groups increased along with the increasing of doses, and sustained at a higher level after 0.1 Gy, and up to the peak value after irradiation with 5.0 and 2.0 Gy respectively.
     5 Experimental study on effects of MCF-7 cells by combining recombinant plasmids and X-irradiation
     5.1 Growth inhibitory effects on MCF-7 cells by combining recombinant plasmids and X-irradiation
     5.1.1 Time-effect relationship of the growth inhibitory effects on MCF-7 cells by combining recombinant plasmids and X-irradiation
     The cells were irradiated by 2.0 Gy at 24 h after transfection, then the cellular proliferation was detected by MTT assay at the different time-points after irradiation. The results showed that the growth rates of the MCF-7 cells transfected with recombinant plasmids declined, especially those of the pshuttle-Egr1-shTRAIL-shES group. Under the same time-point, when compared with those in the control group, the growth rates of the pshuttle-Egr1-shTRAIL-shES group declined significantly from the beginning of 4 h, and the differences were significant at other time-points except for 8 h when compared with those in the pshuttle group, and at 8、12、24 and 36 h when compared with those in the pshuttle-Egr1-shES group, and from the beginning of 4 h when compared with those in pshuttle-Egr1-shTRAIL group (P < 0.05 ~ P < 0.001).
     5.1.2 Dose-effect relationship of the growth inhibitory effects on MCF-7 cells by combining recombinant plasmids and X-irradiation
     The cells were irradiated with different doses at 24 h after transfection, then the cellular proliferation was detected by MTT assay at 24 h after irradiation. The results showed that along with the enlargement of the irradiation dose, the inhibition of cellular growth increased gradually. Under the identical irradiation dose, there was significant difference between that of the control and pshuttle groups only after 2.0 Gy X-irradiation (P < 0.05), while that of the pshuttle-Egr1-shES, pshuttle-Egr1-shTRAIL and pshuttle-Egr1-shTRAIL-shES groups was inhibited significantly as compared with that of the control and pshuttle groups respectively (P < 0.05 ~ P < 0.001). Among these, the inhibition of cell growth in the 5.0 Gy + pshuttle-Egr1-shTRAIL-shES group was the strongest.
     5.2 Effects on apoptosis of MCF-7 cells by combining recombinant plasmids and X-irradiation
     The cells were irradiated with 2.0 Gy X-ray at 24 h after transfected with plasmids, and harvested 24 h later. Then the cell apoptotic percentages were detected by FCM. The results showed that as compared with that of the control group, the apoptotic percentages of other groups were increased significantly (P < 0.001), especially that of the pshuttle-Egr1-shTRAIL-shES group which was the highest. Furthermore, the necrotic percentage of the pshuttle-Egr1-shTRAIL-shES group increased significantly when compared with that of all other groups respectively (P < 0.01, P < 0.001).
     5.3 Effects on cell cycle of MCF-7 cells by combining recombinant plasmids and X-irradiation
     The cells were irradiated with 2.0 Gy X-ray at 24 h after transfected with plasmids, and harvested 12 h later. Then the percentage of each phase in cell cycle was detected by FCM. The results showed that as compared with that in the control and pshuttle groups respectively, the percentages of G0/G1 phase decreased, and the percentages of S phase increased significantly in the rest groups (P < 0.001), and the percentages of G2/M phase decreased significantly in the pshuttle-Egr1-shES and pshuttle-Egr1-shTRAIL groups (P < 0.01), but that in the pshuttle-Egr1-shTRAIL-shES groups decreased slightly without significance.
     6 Effects of combining recombinant plasmids and X-irradiation on ECV304 cells
     6.1 Growth inhibitory effects on ECV304 cells by combining recombinant plasmids and X-irradiation
     6.1.1 Time-effect relationship of the growth inhibitory effects on ECV304 cells by combining recombinant plasmids and X-irradiation
     The cells were irradiated by 2.0 Gy X-ray at 24 h after transfection, then the cellular proliferation was detected by MTT assay at the different time-points after irradiation. The results showed that the growth rates in the pshuttle group declined slightly after 2.0 Gy irradiation, but when compared with those of the control group, the differences were no significance. All the growth rates in the pshuttle-Egr1-shES, pshuttle-Egr1-shTRAIL, and pshuttle-Egr1-shTRAIL -shES groups declined, and when compared with those in the control group, the differences began to become significant from the beginning of 12, 12 and 8 h respectively (P < 0.05 ~ P < 0.001). Moreover, as compared with those in the pshuttle-Egr1-shES group respectively, the growth rates in the pshuttle-Egr1-shTRAIL-shES group declined significantly from the beginning of 24 h (P < 0.05 ~ P < 0.001). Moreover, there was significant difference between the growth rates in the pshuttle-Egr1-shTRAIL and pshuttle-Egr1-shTRAIL-shES groups at 36 h (P < 0.05).
     6.1.2 Dose-effect relationship of the growth inhibitory effects on ECV304 cells by combining recombinant plasmids and X-irradiation
     The cells were irradiated with different doses at 24 h after transfection, then the cellular proliferation was detected by MTT assay at 24 h after irradiation. The results showed that along with the enlargement of the irradiation dose, the inhibition of cellular growth increased gradually. As compared with that in the control and pshuttle groups, the cell growth in the rest groups were inhibited significantly from the beginning of 0.5 Gy (P < 0.05, P < 0.01). Among these, the inhibitory effect in the 5.0 Gy + pshuttle-Egr1-shTRAIL-shES group was the strongest, but there was no significant difference when compared with that in the 5.0 Gy + pshuttle-Egr1-shES and 2.0 Gy + pshuttle-Egr1-shTRAIL-shES groups respectively.
     6.2 Effects on apoptosis of ECV304 cells by combining recombinant plasmids and X-irradiation
     The cells were irradiated with 2.0 Gy X-ray at 24 h after transfected with plasmids, and harvested 24 h later. Then the cell apoptotic percentages were detected by FCM. The results showed that as compared with that of the control group, the apoptotic percentages of the pshuttle-Egr1-shES, pshuttle-Egr1-shTRAIL and pshuttle-Egr1-shTRAIL-shES groups were increased significantly (P < 0.05, P < 0.01). As compared with that of the pshuttle group, the apoptotic percentages of the pshuttle-Egr1-shES and pshuttle-Egr1-shTRAIL-shES groups were increased significantly (P < 0.05, P < 0.01). The apoptotic percentage of the pshuttle-Egr1-shTRAIL-shES group was the highest. Moreover, the necrotic percentages of both the pshuttle-Egr1-shTRAIL and pshuttle-Egr1-shTRAIL-shES groups were increased significant when compared with that of the control group, and only that of the later group had significant difference with that of the pshuttle group (P < 0.05).
     6.3 Effects on cell cycle of ECV304 cells by combining recombinant plasmids and X-irradiation
     The cells were irradiated with 2.0 Gy X-ray at 24 h after transfected with plasmids, and harvested 12 h later. Then the percentage of each phase in cell cycle was detected by FCM. The results showed that as compared with that in the control and pshuttle groups, the percentages of G0/G1 phase decreased, and that of G2/M phase increased significantly in the pshuttle-Egr1-shES and pshuttle-Egr1-shTRAIL-shES groups (P < 0.05, P < 0.01). The percentage of G0/G1 phase decreased in the pshuttle-Egr1-shTRAIL group when compared with that in the pshuttle group(P < 0.05), and that of G2/M phase increased in the pshuttle-Egr1-shTRAIL group when compared with that in the control group (P < 0.05).
     Above all, the double-gene coexpression plasmid pshuttle-Egr1-shTRAIL-shES was constructed successfully for the first time in our study, and the plasmid possessed the characteristics of irradiation inducibility and the augumented double-gene coexpression. The effects of growth inhibition and apoptosis promotion on MCF-7 and ECV304 cells by the pshuttle-Egr1-shTRAIL-shES double-gene-radiotherapy were superior to that of both the pure radiotherapy and pshuttle-Egr1-shES or pshuttle-Egr1-shTRAIL single-gene-radiotherapy, and it also could effect the progression of cell cycle. It may be because of the combining action of the promoting apoptosis effect of shTRAIL, the anti-tumor angiogenesis effect of shES, and the directly killing tumor cells by irradiation. The researchs open up a new way to improve the effects of gene-radiotherapy, and provide the theoretical and experimental bases for the clinical application of double-gene-radiotherapy.
引文
[1] http://www.e79.com.cn/xwzx/zlhy/gjhy/20090212/6961.html.
    [2] ANDERSON W F, BLAESE R M, CULVER K, et al. The ADA human gene therapy clinical protocol: points to consider response with clinical protocol [J]. Hum Gene Ther, 1990, 1:331-362.
    [3] CAVAZZANA-CALVO M, HACEIN-BEY S, DE SAINTBASILE G, et al. Gene therapy ofhuman severe combined immunodeficiency (SCID)-X1 disease [J]. Science, 2000, 288(5466):669-672.
    [4] http://www.wiley.co.uk/genmed/clinical/
    [5] 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.
    [6]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.
    [7] PARVEEN Z, KRUPETSKY A, ENGELST?DTER M, et al. Spleen necrosis virus-derived C-type retroviral vectors for gene transfer to quiescent cells [J]. Nat Biotechnol, 2000, 18(6):623-629.
    [8] SANDRIN V, RUSSELL S J, COSSET F L. Targeting retroviral and lentiviral vectors [J]. Curr Top Microbiol Immunol, 2003, 281:137-178.
    [9] 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]. J Gene Med, 2002, 4(3):229-239.
    [10] 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.
    [11] NáSZ I, ADáM E. Recombinant adenovirus vectors for gene therapy and clinical trials [J]. Acta Microbiol Immunol Hung, 2001, 48(3-4):323-348.
    [12]邱江锋,罗春霞,于芙蓉,等.多顺反子腺病毒表达载体PCA_(13)/FasL-IRES-iNOS的构建和鉴定[J].胃肠病学,2003,8(4):204-206.
    [13]王朝霞.重组腺病毒p53基因治疗肺癌的研究进展[J].中国肺癌杂志,2005, 8(2):148-151.
    [14] SCHOGGINS J W, GALL J G, FALCK-PEDERSEN E. Subgroup B and F fiber chimeras eliminate normal adenovirus type 5 vector transduction in vitro and in vivo [J]. J Virol, 2003, 77(2):1039-1048.
    [15] ALEMANY R. Cancer selective adenoviruses [J]. Mol Aspects Med, 2007, 28(1):42-58.
    [16] FUJIWARA T, URATA Y, TANAKA N. Telomerase-specific oncolytic virotherapy for human cancer with the hTERT promoter [J]. Curr Cancer Drug Targets, 2007, 7(2):191-201.
    [17] LOCKLEY M, FERNANDEZ M, WANG Y, et al. Activity of the adenoviral E1A deletion mutant dl922-947 in ovarian cancer: comparison with E1A wild-type viruses, bioluminescence monitoring, and intraperitoneal delivery in icodextrin [J]. Cancer Res, 2006,66(2):989-998.
    [18] COURA RDOS S, NARDI N B.The state of the art of adeno-associated virus-based vectors in gene therapy [J]. Virol J, 2007, 4:99.
    [19] WRIGHT J F, QU G, TANG C, et al. Recombinant adeno-associated virus: formulation challenges and strategies for a gene therapy vector [J]. Curr Opin Drug Discov Devel, 2003, 6(2):174-178.
    [20] ASFOUR B, BABA H A, SCHELD H H, et al. Uniform long-term gene expression using adeno-associated virus (AAV) by ex vivo recirculation in rat-cardiac isografts [J]. Thorac Cardiovasc Surg, 2002, 50(6):347-350.
    [21]李晨.腺相关病毒载体及其在基因治疗中的应用[J].中国康复理论与实践,2006,12(1):51-52.
    [22]邓继先,沈伟.用慢病毒载体制备转基因动物的研究进展[J].中国生物工程杂志,2004,24(9):16-19.
    [23] INDRACCOLO S, HABELER W, TISATO V, et al. Gene transfer in ovarian cancer cells: a comparison between retroviral and lentiviral vectors [J]. Cancer Res, 2002, 62(21):6099-6107.
    [24] WANG S, ZHANG C, ZHANG L, et al. The relative immunogenicity of DNA vaccines delivered by the intramuscular needle injection, electroporation and gene gun methods [J]. Vaccine, 2008, 26(17):2100-2110.
    [25] DIETRICH A, BECHERER L, BRINCKMANN U, et al. Particle-mediated cytokine gene therapy leads to antitumor and antimetastatic effects in mouse carcinoma models [J]. Cancer Biother Radiopharm, 2006, 21(4):333-341.
    [26] MIR L M, BUREAU M F, GEHL J, et al. High-efficiency gene transfer into skeletal muscle mediated by electric pulses [J]. Proc Natl Acad Sci USA, 1999, 96(8):4262-4267.
    [27] WELLS D J. High-efficiency gene transfer into skeletal muscle mediated by electric pulses [J]. Gene Ther, 2004, 11(18):1363-1369.
    [28] TANIYAMA Y, TACHIBANA K, HIRAOKA K, et al. Local delivery of plasmid DNA into rat carotid artery using ultrasound [J]. Circulation, 2002, 105(10):1233-1239.
    [29] F LIU, H CAI, L HUANG, et al. Factors controlling the efficiency of cationic lipid- mediated transfection in vivo via intravenous administration [J]. Gene Ther, 1997, 4(6):517-523.
    [30] KOCH S, POHL P, COBET U, et al. Ultrasound enhancement of liposome-mediated cell transfection is caused by cavitation effects [J]. Ultrasound Med Biol, 2000, 26(5):897-903.
    [31] ESCRIOU V, CARRIèRE M, SCHERMAN D, et al. NLS bioconjugates for targeting therapeutic genes to the nucleus [J]. Adv Drug Deliv Rev, 2003, 55(2):295-306.
    [32] KUNATH K, MERDAN T, HEGENER O, et al. Integrin targeting using RGD-PEI conjugates for in vitro gene transfer [J]. J Gene Med, 2003, 5(7):588-599.
    [33] 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.
    [34] D M KLINMAN, J CONOVER, J M LEIDEN, et al. Safe and effective regulation by gene gun administration of an erytbropoielin-encoding DNA plasmid [J]. Hum Gene Ther, 1999, 10:659-665.
    [35]张晓志,林鸿,杨晓燕,等.重组腺病毒临床级基因治疗制品的质量控制[J].中华医学杂志,2004,84(10):849-852.
    [36] DURRBACH A, ANGEVIN E, PONCET P, et al. Antibody-mediated endocytosis of G250 tumor-associated antigen allows targeted gene transfer to human renal cell carcinoma in vitro [J]. Cancer Gene Ther, 1999, 6(6):564-571.
    [37] DEAS O, ANGEVIN E, CHERBONNIER C, et al. In vivo-targeted gene delivery using antibody-based nonviral vector [J]. Hum Gene Ther, 2002, 13(9):1101-1114.
    [38] XI LI, FU GENG FENG, FAN YAN RONG, et al. Bifidobacterium adolescentis as a delivery system of endostatin for cancer gene therapy: selective inhibitor of angiogenesis and hypoxic tumor growth [J]. Cancer Gene Ther. 2003, 10:105-111
    [39] HAMAJI Y, FUJIMORI M, SASAKI T, et al. Strong enhancement of recombinant cytosine deaminase activity in Bifidobacterium longum for tumor-targeting enzyme/prodrug therapy [J]. Biosci Biotechnol Biochem, 2007, 71(4):874-883.
    [40] HOU X, LIU J E. Construction of Escherichia coli-Bifidobacterium longum shuttle vector and expression of tumor suppressor gene PTEN in B. longum [J]. Wei Sheng Wu Xue Bao, 2006, 46(3):347-352.
    [41] 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.
    [42]胡春生.基因治疗的“3Y”问题与原则[J].遗传,2003,25(5):577-580.
    [43] HUNT K K, VORBURGER S A. TECH.SIGHT. Gene therapy. Hurdles and hopes for cancer treatment [J]. Science, 2002, 297(5580):415-416.
    [44] LI Z, DULLMANN J, SCHIEDLMEIER B, et al. Murine leukemia induced by retroviral gene marking [J]. Science, 2002, 296(5567):497.
    [45] MARSHALL E. Clinical research.gene therapy a suspect in leukemia-like disease [J]. Science, 2002, 298(5591):34-35.
    [46]陶铭.基因治疗及其研究[J].生物学通报,2008,43(4):16-18.
    [47] XU H J. Retinoblastoma and tumor-suppressor gene therapy [J]. Ophthalmol Clin North Am, 2003, 16(4):621-629.
    [48] NEMUNAITIS J M, NEMUNAITIS J. Potential of Advexin: a p53 gene-replacement therapy in Li-Fraumeni syndrome [J]. Future Oncol, 2008, 4(6):759-768.
    [49] 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.
    [50] ASIF-ULLAH M, LéVESQUE M, ROBICHAUD G, et al. Development of ribozyme-based gene-inactivations; the example of the hepatitis delta virus ribozyme [J]. Curr Gene Ther, 2007, 7(3):205-216.
    [51] GU Z P, WANG Y J, LI J G, et al. VEGF165 antisense RNA suppresses oncogenic properties of human esophageal squamous cell carcinoma [J]. World J Gastroenterol, 2002, 8(1):44-48.
    [52] KIM D, ROSSI J. RNAi mechanisms and applications [J]. Biotechniques, 2008, 44(5):613-616.
    [53] PUSHPARAJ P N, AARTHI J J, MANIKANDAN J, et al. siRNA, miRNA, and shRNA: in vivo applications [J]. J Dent Res, 2008, 87(11):992-1003.
    [54] HUESKER M, FOLMER Y, SCHNEIDER M, et al. Reversal of drug resistance ofhepatocellular carcinoma cells by adenoviral delivery of anti-MDR1 ribozymes [J]. Hepatology, 2002, 36(4 Pt 1):874-884.
    [55] NICULESCU-DUVAZ I, SPRINGER CJ. Introduction to the background, principles, and state of the art in suicide gene therapy [J]. Mol Biotechnol, 2005, 30(1):71-88.
    [56] 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.
    [57] 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.
    [58] LI S, YU B, AN P, et al. Combined liposome-mediated cytosine deaminase gene therapy with radiation in killing rectal cancer cells and xenografts in athymic mice [J]. Clin Cancer Res, 2005, 11(9):3574-3578.
    [59] WU D H, LIU L, CHEN L H. Antitumor effects and radiosensitization of cytosine deaminase and thymidine kinase fusion suicide gene on colorectal carcinoma cells [J]. World J Gastroenterol, 2005, 11(20):3051-3055.
    [60] TAKAHASHI M, INANAMI O, KUBOTA N, et al. Enhancement of cell death by TNF alpha-related apoptosis-inducing ligand (TRAIL) in human lung carcinoma A549 cells exposed to x rays under hypoxia [J]. J Radiat Res (Tokyo), 2007, 48(6):461-468.
    [61] ITASAKA S, KOMAKI R, HERBST R S, et al. Endostatin improves radioresponse and blocks tumor revascularization after radiation therapy for A431 xenografts in mice [J]. Int J Radiat Oncol Biol Phys, 2007, 67(3):870-878.
    [62]王贵怀,刘慧风,李储忠,等.肿瘤坏死因子-α基因治疗联合放疗抗大鼠胶质瘤C6细胞作用的实验研究[J].中国神经肿瘤杂志,2006,4(2):118-124.
    [63]吴丛梅,李修义. pEgr-TNFα质粒的构建及基因-放射治疗小鼠黑色素瘤的实验研究[J].中华肿瘤杂志,2004,26(3):143-145.
    [64] WEICHSELBAUM R R, KUFE D W, ADVANI S J, et al. Molecular targeting of gene therapy and radiotherapy [J]. Acta Oncol, 2001, 40(6):735-738.
    [65] PINES A, BIVI N, ROMANELLO M, et al. Cross-regulation between Egr-1 and APE/Ref-1 during early response to oxidative stress in the human osteoblastic HOBIT cell line: evidence for an autoregulatory loop [J]. Free Radic Res, 2005, 39(3):269-228
    [66] CHADDERTON N, COWEN R L, SHEPPARD F C, et al. Dual responsive promoters to target therapeutic gene expression to radiation-resistant hypoxic tumor cells [J]. Int J Radiat Oncol Biol Phys, 2005, 62(1):213-222.
    [67] Yang W, Li X Y. Anti-tumor effect of pEgr-interferon-gamma-endostatin gene-radiotherapy in mice bearing Lewis lung carcinoma and its mechanism [J]. Chin Med J (in English), 2005, 118(4):296-301.
    [68] XIAN J, YANG H, LIN Y, et al. Combination nonviral murine interleukin 2 and interleukin 12 gene therapy and radiotherapy for head and neck squamous cell carcinoma [J]. Arch Otolaryngol Head Neck Surg, 2005, 131(12):1079-1085.
    [69] YANG J, JIN G, LIU X, et al. Therapeutic effect of pEgr-IL18-B7.2 gene radiotherapy in B16 melanoma-bearing mice [J]. Hum Gene Ther, 2007, 18(4):323-332.
    [70] JIN G H, JIN S Z, LIU Y, et al. Therapeutic effect of gene-therapy in combination with local X-irradiation in a mouse malignant melanoma model [J]. Biochem Biophys Res Commun,2005, 330(3):975-981.
    [71] GUO S Y, GU Q L, ZHU Z G, et al. TK gene combined with mIL-2 and mGM-CSF genes in treatment of gastric cancer [J]. World J Gastroenterol, 2003, 9(2):233-237.
    [72] YAO H, HUANG Z H, LI Z, et al. Specific killing effects of combination of double suicide gene and survivin antisense oligonucleotide on breast tumor cells and vein endothelial cells [J]. Zhonghua Wai Ke Za Zhi, 2007, 45(7):476-479.
    [73] YAO H, HUANG Z H, LI Z, et al. Specific killing effects of combination of recombinant adenovirus containing double suicide gene driven by KDR promoter and survivin antisense oligonucleotide on colorectal cancer cells and vascular endothelial cells [J]. Zhonghua Wei Chang Wai Ke Za Zhi, 2008, 11(1):61-66.
    [74] GREENBERGER J S, EPPERLY M W, GRETTON J, et al. Radioprotective gene therapy [J]. Curr Gene Ther, 2003, 3(3):183-195.
    [75] EPPERLY M W, SIKORA C A, DEFILIPPI S J, et al. Pulmonary irradiation-induced expression of VCAM-I and ICAM-I is decreased by manganese superoxide dismutase-plasmid/liposome (MnSOD-PL) gene therapy [J]. Biol Blood Marrow Transplant, 2002, 8(4):175-187.
    [76] EPPERLY M W, WEGNER R, KANAI A J, et al. Effects of MnSOD-Plasmid Liposome Gene Therapy on Antioxidant Levels in Irradiated Murine Oral Cavity Orthotopic Tumors [J]. Radiat Res, 2007, 167(3):289-297.
    [77] WEICHSELBAUM R R, HALLAHAN D E, BECKETT M A, et al. Gene therapy targeted by radiation preferentially radiosensitizes tumor cells [J]. Cancer Res, 1994, 54(16):4266-4269.
    [78] HALLAHAN D E, MAUCERI H J, SEUNG L P, et al. Spatial and temporal control of gene therapy using ionizing radiation [J]. Nat Med, 1995, 1(8):786-791.
    [79] MAUCERI H J, HANNA N N, WAYNE J D, et al. Tumor necrosis factor alpha (TNF-alpha) gene therapy targeted by ionizing radiation selectively damages tumor vasculature [J]. Cancer Res, 1996, 56(19):4311-4314.
    [80] TAKAHASHI T, NAMIKI Y, OHNO T. Induction of the suicide HSV-TK gene by activation of the Egr-1 promoter with radioisotopes [J]. Hum Gene Ther, 1997, 8(7):827-833.
    [81] MANOME Y, KUNIEDA T, WEN P Y, Et al. Transgene expression in malignant glioma using a replication-defective adenoviral vector containing the Egr-1 promoter: activation by ionizing radiation or uptake of radioactive iododeoxyuridine [J]. Hum Gene Ther, 1998, 9(10): 1409-1417.
    [82] 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.
    [83] 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.
    [84] MARPLES B, SCOTT S D, HENDRY J H, et al. Development of synthetic promoters for radiation-mediated gene therapy [J]. Gene Ther, 2000, 7(6):511-517.
    [85] MARPLES B, GRECO O, JOINER M C, et al. Molecular approaches to chemo- radiotherapy [J]. Eur J Cancer, 2002, 38(2):231-239.
    [86] GRECO O, MARPLES B, DACHS G U, et al. Novel chimeric gene promoters responsive to hypoxia and ionizing radiation [J]. Gene Ther, 2002, 9(20):1403-1411.
    [87]魏道严,戴冰冰,陈诗书.放射诱导经Egr-1启动子调控的腺病毒介导CDglyTK基因的肿瘤靶向表达[J].中华医学杂志,2001,81(16):999-1003.
    [88]李秀娟,宋祥福,杨巍,等. pEgr-angiostatin基因辐射诱导表达特性及其抗肿瘤作用[J].吉林大学学报(医学版),2003,29(5):543-546.
    [89]田梅,朴春姬,李修义,等. pEgr-hPTEN体外稳定转染对人脑胶质瘤SHG-44细胞周期及增殖的影响[J].中国实验诊断学,2004,8(1):3-5.
    [90]沈滟,赵大治. TRAIL系统与肿瘤细胞的凋亡[J].河南科技大学学报(医学版),2006,24(4):318-320.
    [91] BAETU T M, KWON H, SHARMA S, et al. Disruption of NF-kappaB signaling reveals a novel role for NF-κB in the regulation of TNF-related apoptosis-inducing ligand expression [J]. J Immunol, 2001, 167(6):3164-3173.
    [92] TRUNEH A, SHARMA S, SILVERMAN C, et al. Temperature sensitive differential affinity of TRAIL for its receptors DR5 is the highest affinity receptor [J]. J Biol Chem, 2000, 275:23319-23325.
    [93] DEGLI-ESPOSTI M A, SMOLAK P J, WALCZAK H, et al. Cloning and characterization of TRAIL-R3, a novel member of the emerging TRAIL receptor family [J]. J Exp Med, 1997, 186(7): 1165-1170.
    [94] SIMONET W S, LACEY D L, DUNSTAN C R, et al. Osteoprotegerin: a novel secreted protein involved in the regulation of bone density [J]. Cell, 1997, 89(2):309-319.
    [95] SHEIKH M S, FORNACE AJ JR. Death and decoy receptors and p53-mediated apoptosis [J]. Leukemia, 2000, 14(8):1509-1513.
    [96] RAVI R, BEDI G C, ENGSTROM L W, et al. Regulation of death receptor expression and TRAIL/Apo2L-induced apoptosis by NF-κB [J]. Nat Cell Biol, 2001, 3(4):409-416.
    [97] BERNARD D, QUATANNENS B, VANDENBUNDER B, et al. Rel/NF-kappaB transcription factors protect against tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL)-induced apoptosis by up-regulating the TRAIL decoy receptor DcR1 [J]. J Biol Chem, 2001, 276(29):27322-27328.
    [98] WU X X , OGAWA O, AND KAKEHI Y. TRAIL and chemothera-peutic drugs in cancer therapy [J]. Vitam Horm, 2004, 67:365-383.
    [99]胡璧,王建军,徐根兴.肿瘤坏死因子诱导凋亡配体(TRAIL)抗肿瘤治疗研究进展[J].药学与临床研究,2008,16(3):491.
    [100] WAJANT H, HAAS E, SCHWENZER R, et al. Inhibition of death receptor-mediated gene induction by a cycloheximide-sensitive factor occurs at the level of or upstream of Fas-associated death domain protein (FADD) [J]. J Biol Chem, 2000, 275(32):24357-24366.
    [101] HU W H, JOHNSON H, SHU H B. Tumor necrosis factor-related apoptosis-inducing ligand receptors signal NF-kappaB and JNK activation and apoptosis through distinct pathways [J]. J Biol Chem, 1999, 274(43):30603-30610.
    [102] G?KE R, G?KE A, G?KE B, et al. Regulation of TRAIL-induced apoptosis by transcription factors [J]. Cell Immunol, 2000, 201(2):77-82.
    [103] LEVERKUS M, NEUMANN M, MENGLING T, et al. Regulation of tumor necrosis factor-related apoptosis-inducing ligand sensitivity in primary and transformed human keratinocytes [J]. Cancer Res, 2000, 60 (3):553-555.
    [104] WANG Q, WANG X, HERNANDEZ A, et al. Regulation of TRAIL expression by thephosphatidylinositol 3-kinase/Akt/GSK-3 pathway in human colon cancer cells [J]. J Biol Chem, 2002, 277(39):36602-36610.
    [105] THAKKAR H, CHEN X, TYAN F, et al. Pro-survival function of Akt/protein kinase B in prostate cancer cells. Relationship with TRAIL resistance [J]. J Biol Chem, 2001, 276(42):38361-38269.
    [106] BORTUL R, TAZZARI P L, CAPPELLINI A, et al. Constitutively active Akt1 protects HL60 leukemia cells from TRAIL-induced apoptosis through a mechanism involving NF-kappaB activation and cFLIP(L) up-regulation [J]. Leukemia, 2003, 17(2):379-389.
    [107] HAMASU T, INANAMI O, ASANUMA T, et al. Enhanced induction of apoptosis by combined treatment of human carcinoma cells with X rays and death receptor agonists [J]. J Radiat Res (Tokyo), 2005, 46(1):103-110.
    [108] NAGANE M, CAVENEE W K, SHIOKAWA Y. Synergistic cytotoxicity through the activation of multiple apoptosis pathways in human glioma cells induced by combined treatment with ionizing radiation and tumor necrosis factor-related apoptosis-inducing ligand [J]. J Neurosurg, 2007, 106(3):407-416.
    [109] GALLIGAN L, LONGLEY D B, MCEWAN M, et al. Chemotherapy and TRAIL-mediated colon cancer cell death: the roles of p53, TRAIL receptors, and c-FLIP [J]. Mol Cancer Ther, 2005, 4(12):2026-2036.
    [110] SRIDHAR S, ALI A A, LIANG Y, et al. Differential expression of members of the tumor necrosis factor alpha-related apoptosis-inducing ligand pathway in prostate cancer cells [J]. Cancer Res, 2001, 61(19):7179-7183.
    [111] BUTTYAN R, MIAN B M. Molecularly targeted therapies for renal cell cancer: TRAIL research advances [J]. J Urol, 2007, 177(5):1606.
    [112] JO M , KIM T H, SEOL D W, et al. Apoptosis induced in normal human hepatocytes by tumor necrosis factor-related apoptosis-inducing ligand [J]. Nat Med, 2000, 6(5):564-567.
    [113] NITSCH R, BECHMANN 1, DEISZ R A, et al. Human brain-cell death induced by tumour-necrosis-factor-related apoptosis-inducing ligand(TRAIL) [J]. Lancet, 2000, 356 (9232): 827-828.
    [114] O'REILLY M S, BOEHM T, SHING Y, et al. Endostatin: an endogenous inhibitor of angiogenesis and tumor growth [J]. Cell, 1997, 88(2):277-285.
    [115] KRUGER E A, DURAY P H, TSOKOS M G, et al. Endostatin inhibits microvessel formation in the ex vivo rat aortic ring angiogenesis assay [J]. Biochem Biophys Res Commun, 2000, 268(1): 183-191.
    [116] GAETZNER S, DECKERS M M, STAHL S, et al. Endostatin’s heparan sulfate-binding site is essential for inhibition of angio-genesis and enhances in situ binding to capillary-like structures in bone explants [J]. Matrix Biol, 2005, 23(8):557-561.
    [117] STAHL S, GAETZNER S, MUELLER T D, et al. Endostatin phenylalanines 31 and 34 define a receptor binding site [J]. Genes Cells, 2005, 10(9):929-939.
    [118] RYCHKOVA N, STAHL S, GAETZNER S, et al. Non-heparan sulfate-binding interactions of endostatin/collagen XVIII in murine development [J]. Dev Dyn, 2005, 232(2):399-407.
    [119] CHANG Z, CHOON A, FRIEDL A. Endostatin binds to blood vessels in situ independent of heparan sulfate and does not compete for fibroblast growth factor-2 binding [J].Am J Pathol, 1999, 155(1):71-76.
    [120] BOEHM T, O'REILLY M S, KEOUGH K, et al. Zinc-binding of endostatin is essential for its antiangiogenic activity [J]. Biochem Biophys Res Commun, 1998, 252(1):190-194.
    [121] YAMAGUCHI N, ANAND-APTE B, LEE M, et al. Endostatin inhibits VEGF-induced endothelial cell migration and tumor growth independently of zinc binding [J]. EMBO J, 1999, 18(16): 4414-4423.
    [122] HOHENESTER E, SASAKI T, MANN K, et al. Variable zinc coordination in endostatin [J]. J Mol Biol, 2000, 297(1):1-6.
    [123] ZHOU H, WANG W, LUO Y. Contributions of disulfide bonds in a nested pattern to the structure, stability, and biological functions of endostatin [J]. J Biol Chem, 2005, 280(12): 11303-11312.
    [124] MIOSGE N, SIMNIOK T, SPRYSCH P, et al. The collagen type XVIII endostatin domain is co-localized with perlecan in basement membranes in vivo [J]. J Histochem Cytochem, 2003, 51(3):285-296.
    [125] GUENTHER U, HERBST H, BAUER M, et al. Collagen type XVIII/endostatin is differentially expressed in primary and metastatic colorectal cancers and ovarian carcinomas [J]. Br J Cancer, 2001, 85(10):1540-1545.
    [126] KIM Y M, HWANG S, KIM Y M, et al. Endostatin blocks vascular endothelial growth factor-mediated signaling via direct interaction with KDR/Flk-1 [J]. J Biol Chem, 2002, 277(31):27872-27879.
    [127] SHI H, HUANG Y, ZHOU H, et al. Nucleolin is a Receptor that Mediates Antiangio-genic and Anti-tumor Activity of Endostatin [J]. Blood, 2007, 110(8):2899-2906.
    [128] HANAI J, DHANABAL M, KARUMANCHI S A, et al. Endostatin causes G1 arrest of endothelial cells through inhibition of cyclin D1 [J]. J Biol Chem, 2002, 277(19):16464-16469.
    [129] HANAI J, GLOY J, KARUMANCHI S A, et al. Endostatin is a potential inhibitor of Wnt signaling [J]. J Cell Biol, 2002, 158(3):529-539
    [130] DIXELIUS J, LARSSON H , SASAKI T, et al. Endostatin induced tyrosine kinase signalling through the Shb adaptor protein regulates endothelial cell apoptosis [J]. Blood, 2000, 95(11):3403-3411.
    [131] MACDONALD N J, SHIVERS W Y, NARUM D L, et al. Endostatin binds tropomysin.A potential modulator of the antitumor activity of endostatin [J]. J Biol Chem, 2001, 276(27):25190-25196.
    [132] KIM Y M, JANG J W, LEE O H, et al. Endostatin inhibits endothelial and tumor cellular invasion by blockingthe activation and catalytic activity of matrix metalloproteinase [J]. Cancer Res, 2000, 60(19):5410-5413.
    [133] LEE S J, JANG J W, KIM Y M, et al. Emtostatin binds to the catalytic domain of matrix metalloproteinase-2 [J]. FEBS Lett, 2002, 519(1-3):147-152.
    [134] WICKSTROM S A, VEIKKOLA T, REHN M, et al. Endostatin-indused modulation of plasminogen activation with concomitant loss of focal adhesions and actin stress fibers in cultured human endothelial cells [J]. Cancer Res, 2001, 61(17):6511-6516.
    [135] REHN M, VEIKKOLA T, KUKK-VALDRE E, et al. Interaction of endostatin with
    integrins implicated in angiogenesis [J]. Proc Natl Acad Sci U S A, 2001, 98(3):1024-1029.
    [136]张立红,葛谊,王旻,等.内皮抑素和血管内皮生长因子在喉鳞癌和声带息肉中的表达及意义[J].北京医学,2003,25(5):813.
    [137] BLEZINGER P, WANG J, GONDO M, et al. Systemic inhibition of tumor growth and tumor metastases by intramuscular administration of the endostatin gene [J]. Nat Biot echnol, 1999, 17(4):343-348.
    [138] CHEN Q R, KUMAR D, STASS S A, et al. Liposomes complexed to plasmids encoding angiostatin and endostatin inhibit breast cancer in nude mice [J]. Cancer Res, 1999, 59(14): 3308-3319.
    [139] FELDMAN A L, RESTIFO N P, ALEXANDER H R, et al. Antiangiogenic gene therapy of cancer utilizing a recombinant adenovirus to elevate systemic endostatin levels in mice [J]. Cancer Res, 2000, 60(6):1503-1506.
    [140] FU G F, LI X, HOU Y Y, et al. Bifidobacterium longum as an oral delivery system of endostatin for gene therapy on solid liver cancer [J]. Cancer Gene Ther, 2005, 12(2):133-140.
    [141] JOKI T, MACHLUF M, ATALA A, et al. Continuous release of endostatin from microencapsulated engineered cells for tumor therapy [J]. Nat Biotechnol, 2001, 19(1):35-39.
    [142] HANNA N N, SEETHARAM S, MAUCERI H J, et al. Antitumor interaction of short course endostatin and ionizing radiation [J]. Cancer J, 2000, 6(5): 287-293.
    [143] BERTOLINI F, FUSETTI L, MANCUSO P, et al. Endostatin, an antiangiogenic drug, induces tumor stabilization after chemotherapy or anti-CD20 therapy in a NOD/SCID mouse model of human high-grade non-Hodgkin lymphoma [J]. Blood, 2000, 96(1): 282-287.
    [144] NAGANE M, PAN G, WEDDLE J J, et al. Increased death receptor 5 Expression by chem. Othe rapeutic agents in human gliom as causes synergistic cytotoxicity with tumor necrosis facor-related apoptosis-inducing ligand in vitro and in vivo [J]. Cancer Res, 2000, 60(4): 847-853.
    [145] DHANABAL M, RAMCHANDRAN R, WATERMAN M J, et al. Endostatin induces endothelial cell apoptosis [J]. J Biol Chem, 1999, 274(17):11721-11726.
    [146] DATTA R, RUBIN E, SUKHATME V, et al. Ionizing radiation activates transcription of the EGR1 gene via CArG elements [J]. Proc Natl Acad Sci USA, 1992, 89(21):10149-10153.
    [147]吴丛梅. pEgr-IFNγ和pEgr-TNFα基因辐射诱导表达特性及其抗肿瘤作用的实验研究[D].博士学位论文.长春:吉林大学,2001.
    [148] NGOI S M, CHIEN A C, LEE C G. Exploiting internal ribosome entry sites in gene therapy vector design [J]. Curr Gene Ther, 2004, 4(1):15-31.
    [149] TROUET D, NILIUS B, VOETS T, et al. Use of a bicistronic GFP- expression vector to characterise ion channels after transfection in mammalian cells [J]. Pflugers Arch., 1997, 434(5):632-638.
    [150] 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.
    [151] SENZER N, MANI S, ROSEMURGY A, et al. TNFerade biologic, an adenovector with a radiation-inducible promoter, carrying the human tumor necrosis factor alpha gene: a phase I study in patients with solid tumors [J]. J Clin Oncol, 2004, 22(4):592-601.
    [152] KUFE D, WEICHSELBAUM R. Radiation therapy: activation for gene transcription and the development of genetic radiotherapy-therapeutic strategies in oncology [J]. Cancer BiolTher, 2003, 2(4):326-329.
    [153] WU X X, JIN X H, ZENG Y, et al. Low concent rations of doxorubicin sensitize human solid cancer cells to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-receptor (R)-mediated apoptosis by inducing TRAIL-R expression [J]. Cancer Sci, 2007, 98:1969-1976.
    [154]李振华,崔为发,刘世超.内皮抑素研究进展及在抗肿瘤中的应用[J].现代诊断与治疗,2008,19(1):34-36.
    [155] XU R, DU P, FAN J J, et al. High-level expression and secretion of recombinant mouse endostatin by Escherichia coli [J]. Protein Expr Purif, 2002, 24(3):453-459.
    [156]汪力慧.血管内皮抑素对lewis肺癌细胞系影响的研究[J].临床肿瘤学杂志,2008,13(2):861.
    [157]王亮,金锡御,潘进洪,等.内皮抑制素抑制内皮细胞增殖的细胞周期调控机制[J].中华实验外科杂志,2003,20(9):128-228.

© 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