放射线抵抗宫颈癌细胞株DNA损伤修复基因表达谱的研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
宫颈癌是我国女性发病率较高的恶性肿瘤之一,每年新增宫颈癌病例130000余例、死亡50000余例,其发病率与死亡率分别占我国妇科肿瘤的第一位和第二位,对中晚期或术后复发的宫颈癌患者,放射治疗是主要的治疗方法。1998年,我院全国首家应用体外照射联合中子后装治疗宫颈癌,肿瘤局部控制率和5年存活率分别为85.2%和88.7%,取得了良好的疗效,但在治疗过程中,我们观察到部分宫颈癌患者存在放射抵抗的问题,而且在临床实践中也发现治疗过程中有耐放射性改变,因此,研究影响宫颈癌细胞放射敏感性的因素具有十分重要的意义。
     DNA损伤修复是影响放射敏感性的重要因素,因此深入研究DNA修复基因的表达谱,有助于阐明宫颈癌抵抗放疗的分子机制,为寻找提高宫颈癌放疗敏感性的分子靶点提供实验基础。但DNA损伤修复机制十分复杂多样,小数量的基因不能阐述清楚,需要参与DNA损伤修复过程基因组的大量信息,这样才能真正了解其分子机制。而基因芯片技术是一个强有力的获得癌细胞中成千上万的基因表达的综合性信息的工具,通过该技术可为研究DNA损伤修复基因致宫颈癌放疗抵抗的分子机制提供有效方法。因此本研究首先检测了DNA损伤修复基因APE1在宫颈癌中的表达情况,分析APE1与宫颈癌的临床病理因素及锎-252中子放疗的预后的关系,初步探讨了DNA损伤修复基因APE1与宫颈癌放射抵抗的相关性。然后通过诱导建立宫颈癌耐放射细胞株,采用基因芯片技术对宫颈癌放DNA损伤修复信号通路基因表达谱进行研究,筛选出GADD45α等差异表达基因,构建GADD45α表达载体并初步证明其增加宫颈癌放疗敏感性的作用。
     研究目的
     1.探讨DNA损伤修复基因APE1与宫颈癌放射抵抗的相关性;
     2.探讨DNA损伤修复相关基因致宫颈癌放射抵抗的机制;
     3.初步探讨GADD45α表达载体增强宫颈癌放疗敏感性的作用。
     研究内容和方法
     1. DNA损伤修复基因APE1在宫颈癌中的表达及其与锎-252中子放疗预后的关系:采用采用免疫组化方法APE1在宫颈癌中的表达情况,分析APE1与宫颈癌的临床病理因素及锎-252中子放疗的预后的关系;
     2.诱导并建立放射线照射耐受株:应用直线加速器和锎-252中子后装治疗机对宫颈癌Hela细胞株进行剂量个体化的反复放射线照射,剂量呈梯度增加,最后使其耐放射性具有一定的稳定遗传能力,建立耐中子射线细胞株HelaNR和耐X射线细胞株HelaXR,并通过克隆形成分析、超微结构观察、细胞倍增时间、细胞周期分布和凋亡这些指标检测其耐放射特性。
     3.耐放射宫颈癌细胞株DNA损伤修复相关差异表达基因的筛选:采用DNA损伤信号通路基因芯片(OHS-029)检测Hela、HelaNR和HelaXR细胞的基因表达谱,分析其差异表达基因,并采用western blot和Real-time PCR技术验证基因芯片结果的可靠性。
     4. GADD45α表达载体增强宫颈癌放疗敏感性的初步实验研究:构建特异性pcDNA3.1-GADD45α真核表达载体,Real-time PCR检测转染后宫颈癌细胞GADD45αmRNA表达情况,流式细胞仪检测转染后细胞凋亡情况。
     研究结果
     1. DNA损伤修复基因APE1在宫颈癌中的表达及其与锎-252中子放疗预后的关系:宫颈癌组织APE1表达水平明显高于正常宫颈组织和CIN病例(P<0.01)。APE1在正常宫颈组织和CIN病例均呈胞核表达,宫颈癌组织中APE1呈胞核表达(59)、单纯胞浆表达(8)或核浆共同表达(22)。APE1表达强度与FIGO分期、病理分级和淋巴结转移情况有关(P<0.05),与年龄和病理分型无关。APE1亚细胞定位情况与FIGO分期、病理分级有关(P<0.01),与淋巴结转移情况无关。生存分析显示在APE1核表达组(中位生存时间70.9月)和APE1低表达组(中位生存时间75.8月)的生存时间明显长于APE1浆表达组(中位生存时间57.8月)和APE1高表达组(中位生存时间56.5月)(P=0.025,0.001)。
     2.诱导并建立放射线照射耐受株:(1)克隆形成分析结果发现耐放射株与亲本株相比,SF2值、D0(平均致死剂量)、Dq(准域剂量)值均明显增高,结果说明诱导的耐放射细胞株的放射敏感性降低了,其耐放射性具有一定的稳定遗传能力。(2)通过电子显微镜观察细胞,可发现耐放射细胞的形态与结构均发生了变化,细胞表面可见伪足样突起;细胞浆内可见大量空泡,核蛋白体、线粒体、染色体、粗面内质网、核仁均出现了形态变化,细胞骨架排列紊乱。(3)Hela, HelaNR和HelaXR的细胞倍增时间分别是(28.62±2.77) h、(33.12±3.67) h和(36.94±3.16) h,放射耐受株的细胞倍增时间明显长于亲本株(P <0.05)。(4)我们用流式细胞仪检测细胞周期分布的结果显示,相对于亲本株Hela,中子耐受细胞株HelaNR和X射线耐受细胞株HelaXR的各周期百分比变化并不明显;但在接受4Gy射线照射后,亲本株G2期分布明显升高,G1期分布减少,而放射耐受细胞株G2升高不明显;在接受16Gy射线照射后,放射耐受细胞株G2期也明显升高,但亲本株G2期升高更加明显。这也说明放射耐受细胞株较亲本株的放射敏感性更低。(5)凋亡检测结果显示,随着照射剂量的增加,细胞的凋亡率也升高,但在相同剂量射线照射后,放射耐受株的细胞凋亡率明显低于亲本株细胞,两者间有显著性差异(P <0.05),表明在相同放射剂量的照射下,放射耐受株对放射线更加抗拒,进一步证明了诱导的耐放射株具有放射抵抗性。
     3.耐放射宫颈癌细胞株DNA损伤修复相关差异表达基因的筛选:与亲本株Hela细胞相比,耐放射株HelaNR和HelaXR细胞基因表达改变的总趋势是一致的,表明中子射线和X射线照射诱导Hela细胞发生放射抵抗,引起相似的DNA损伤修复基因表达的改变。筛选亲本株与耐放射株差异在两倍以上的基因:HelaNR细胞有24个差异表达基因,其中19个上调,有5个下调;HelaXR细胞有41个差异表达基因,其中38个上调,有3个下调。通过生物信息和参考文献挖掘得到GADD45α和BTG2等可能与宫颈癌放射耐受密切相关的基因,并采用western blot和Real-time PCR检测了Hela、HelaNR和HelaXR细胞中GADD45α和BTG2两个基因的蛋白和mRNA表达情况,结果显示:在HelaNR和HelaXR细胞中,GADD45α表达下调,BTG2表达上调,与基因芯片的结果是一致的。
     4. GADD45α表达载体增强宫颈癌放疗敏感性的初步实验研究:通过测序和在PUBMED上做BLASTn比对,结果表明,重组质粒中插入的序列与已知GADD45α基因序列同源性达到99%,我们成功构建pcDNA3.1-GADD45α真核表达载体,Real-time PCR检测结果显示转染后细胞GADD45αmRNA含量明显增加(P<0.05),说明pcDNA3.1-GADD45α真核表达载体能增加GADD45α基因在这3个细胞株中的表达。进一步用流式细胞仪检测0、4和16Gy X射线照射后Hela细胞的凋亡情况,结果显示随着照射剂量的增加,细胞的凋亡率也升高,但在相同剂量X射线照射后,对照组的细胞凋亡率与脂质体组无显著差异(P>0.05),转染组的细胞凋亡率明显高于对照组和脂质体组,它们之间有显著性差异(P <0.05),表明转染pcDNA3.1-GADD45α质粒,增加Hela细胞GADD45αmRNA表达,可增加X射线照射后细胞的调亡率。
     结论
     1. APE1表达强度和亚细胞定位情况与宫颈癌的发生、发展和锎-252中子放疗的预后有关,APE1表达强度与宫颈癌的侵袭和转移有关,提示APE1的DNA损伤修复功能可能是导致宫颈癌放疗抵抗的重要因素;
     2.我们采用梯度增加剂量照射宫颈癌Hela细胞进行诱导的方法,建立了耐中子射线细胞株HelaNR和耐X射线宫颈癌细胞株HelaXR,并通过检测其放射生物学特性,证明了诱导的耐放射株具有放射抵抗性;
     3.通过基因芯片技术检测宫颈癌Hela细胞株和诱导的耐放射亚株HelaNR及HelaXR的基因表达谱,筛选出与宫颈癌放射抵抗相关的DNA损伤修复信号通路差异表达基因,为宫颈癌放疗耐受的干预及基因放射治疗提供可能的靶点。
     4.构建pcDNA3.1-GADD45α真核表达载体,转染入宫颈癌细胞,可显著增加其GADD45α基因的表达,并且能显著增加X射线诱导的细胞凋亡,初步说明了提高宫颈癌细胞GADD45α基因表达可增加其对放射线的敏感性。
Cervical cancer is one of the most common malignancies in China, with a high incidence and mortality and more than 130,000 new cases reported and 50,000 women dying of the disease per year. Radiotherapy is particularly effective for patients with cervical cancers at an advanced stage or that cannot be cured surgically. In 1998, we first treated cervical cancer with 252Cf rays combined with X-rays in China. It obtained satisfactory clinical effect, tumor local control rate was 85.2% and 5-year survival rate was 88.7% respectively. But we observed the existence of radioresistance phenomenon in some primarily cervical cancer patients and secondary radioresistance in course of treatment. Therefore, it was very important that we investigated influencing factors of radiosensitivity in cervical cancer.
     The activity of the DNA damage repair pathway is one of the most important factors leading to radioresistance in tumors, including cervical cancer. In many human tumor lines, radiosensitivity correlated with DNA damage induction and repair, so the research of DNA damage repair gene expression profiles is helpful to elucidate the mechanism of radioresistance in cervical cancer and supports to look for the targets of improving radiosensitivity. But the mechanism of DNA damage repair so complex that it couldn’t be explained by a little of genes and need much information of DNA repair genes. DNA microarray technology is a powerful technique to detect the biological response of thousands of genes and can be used to study the mechanism that DNA damage repair gene leads to radioresistance in cervical cancer. Hence, we first investigate the expression feature of APE1, a DNA damage repair gene, and its correlation with clinicopathology and prognostic significance after 252Cf radiotherapy in cervical cancer. Then, long-term 252Cf neutron ray and X-ray irradiation of Hela cells was used to generate two radioresistant cell sub-lines, HelaNR and HelaXR, which provided a model system for studying the radioresistance mechanisms of cervical cancer cells. Whereafter, microarray analysis was used to identify the gene expression patterns of two radioresistant sub-lines derived from Hela cells and the differentially expressed genes were selected, for example GADD45αand BTG2. Finally, we constructed the eukaryotic expression vector of GADD45αgene and investigated its role in radiosensitivity to radiotherapy in cervical cancer.
     Objective
     1. To explore the relationship between APE1 and radioresistance in cervical cancer.
     2. To investigate the mechanism that DNA damage repair gene leads to radioresistance in cervical cancer.
     3. To investigate primarily the function that eukaryotic expression vector of GADD45αgene enhance radiosensitivity in cervical cancer.
     Materials and Methods
     1. The expression feature of APE1, a DNA damage repair gene, and its correlation with clinicopathology and prognostic significance after 252Cf radiotherapy in cervical cancer: The expression of APE1 was detected by immunohistochemistry technique in 89 cases of cervical cancer (treated by 252Cf), 15 cases cervical intraepithelial neoplasia(CIN) and 10 cases of normal cervical tissue, and its association with clinicopathological data as well as prognosis was analyzed.
     2. The generation of two radioresistant cell sub-lines: Hela cells were treated with fractionated 252Cf neutron and X-rays, with a cumulative dose of 75 Gy each, over 8 months, yielding the sub-lines HelaNR and HelaXR. Radioresistant characteristics were detected by clone formation assay, ultrastructural observations, cell doubling time, cell cycle distribution, and apoptosis assay.
     3. The screening of differentially expressed genes relate to DNA damage repair genes in cervical cancer: Gene expression patterns of the radioresistant sub-lines were studied through microarray analysis and verified by western blotting and real-time PCR.
     4. The investigation of eukaryotic expression vector of GADD45αgene enhancing radiosensitivity in cervical cancer: we constructed the eukaryotic expression vector of GADD45αgene. Then, The expression of GADD45αmRNA was detected after eukaryotic expression vector of GADD45αgene was transfected into cervical cancer cell strains by real-time PCR and the apoptosis was detected after eukaryotic expression vector of GADD45αgene was transfected into Hela cells by FCM.
     Results
     1. The expression feature of APE1, a DNA damage repair gene, and its correlation with clinicopathology and prognostic significance after 252Cf radiotherapy in cervical cancer: The expression of APE1 in cervical cancer is higher significantly than that in normal cervical tissue and CIN(P<0.01). In normal cervical tissue and CIN, the APE1 express was located in the nucleus. In cervical cancer, the APE1 express was located in the nucleus (59), cytoplasm (8) or nucleus and cytoplasm (22), the location of APE1 was related with FIGO stage and pathological grade (P<0.01), and not related with lymph node metastasis. The level of APE1 express related with FIGO stage, pathological grade and lymph node metastasis (P<0.05), and not related with age and pathological type. The Kaplan-Meier survival analysis showed that the survival time of the group of APE1 nucleus expression (median survival time is 70.9 months) and the group of APE1 low expression (median survival time is 75.8 months) is longer significantly than that of the group of APE1 cytoplasm expression (median survival time is 57.8 months) and the group of APE1 high expression (median survival time is 56.5 months) (P=0.025, 0.001).
     2. The generation of two radioresistant cell sub-lines: (1) Under equal doses of 252Cf neutron ray and X-ray irradiation, the D0, Dq, and SF2 values of the HelaNR and HelaXR cells were higher than those of the Hela cells, indicating that the sub-lines were more radioresistant than the parent cells. (2) In Hela cells, microvilli were present on the surface and the cytoplasm contained abundant mitochondria and ribosome. In the radioresistant sub-lines, swelling of mitochondria, vacuolization, dilatation of the endoplasmic reticulum, and myelin figures were observed. Thus, in the HelaNR and HelaXR sub-lines, the ultrastructural changes induced by long-term irradiation persisted even > 2 months after the last radiation treatment. (3) The cell doubling times of the HelaNR and HelaXR cells (33.12±3.67 h, and 36.94±3.16 h, respectively) were longer than those of the parent Hela cells (28.62±2.77 h). (4) In Hela cells, exposure to a radiation dose of 4 Gy significantly increased the proportion of cells in G2 and decreased the proportion of cells in G1. The proportion of G2-arrested Hela cells was even greater following 16-Gy irradiation. In the HelaNR and HelaXR sub-lines, however, the proportion of cells in G2 did not increase after irradiation with 4 Gy. At 16 Gy, cells of both sub-lines became arrested in G2 but the proportion was much lower than in Hela cells. Thus, cells of the radioresistant sub-lines probably arrested in the G1 phase of the cell cycle. (5) At 0 Gy, the apoptosis rate was 0.93–2.71% for all three cell lines. At 4 and 16 Gy, the apoptosis rate of Hela cells was much higher than that of the radioresisitant sub-lines (4Gy: 9.32 vs 3.84, 7.94 vs 5.43; 16Gy: 22.47 vs 7.28, 20.03 vs 11.1, p < 0.05). Thus, the sub-lines HelaXR and HelaNR were more radioresistant than the parental Hela cell line.
     3. The screening of differentially expressed genes relate to DNA damage repair genes in cervical cancer: Using stringent criteria for array analysis (≥2-fold change in expression), we identified 113 genes related to DNA damage signaling pathways that were differentially expressed in the parental Hela cells compared to radioresistant HelaXR and HelaNR cells. Of the 24 genes significantly altered by at least 2-fold in HelaNR cells, 19 were up-regulated and 5 down-regulated. Likewise, of the 41 genes significantly altered by at least 2-fold in HelaXR cells, 38 were up-regulated and 3 down-regulated. For the two radioresistant sub-lines, the similar overall trend in gene-expression changes indicated that long-term exposure to 252Cf neutron and X-rays had resulted in a similar induction of genes involved in DNA damage signaling pathways. The number of genes that underwent homologous recombination and nonhomologous end-joining, processes that result in altered gene expression, was higher in HelaXR cells (8 and 3, respectively) than in HelaNR cells (2 and 1, respectively). Furthermore, the number of cell cycle arrest and mismatch repair genes expressed was higher in HelaXR cells (7 and 3, respectively) than in HelaNR cells (3 and 1, respectively) whereas the number of base excision repair genes expressed in the two sub-lines was almost the same (4 in HelaXR cells and 5 in HelaNR cells). Thus, genes encoding double-strand break (DSB) repair, mismatch repair, and cell cycle arrest functions were more highly expressed in cells resistant to X-rays than in cells resistant to 252Cf neutron rays. Two genes, BTG2 and GADD45α, were chosen for further analysis and validation of the microarray data by Western blotting and real-time PCR. As shown by Western blotting, BTG2 protein expression was up-regulated in the resistant sub-lines, especially in HelaNR cells, while the expression of GADD45αprotein was down-regulated in both resistant sub-lines. PCR analysis showed that BTG2 and GADD45αmRNA expression paralleled that of the respective proteins.
     4. The investigation of eukaryotic expression vector of GADD45αgene enhancing radiosensitivity in cervical cancer: Through gene sequencing and BLASTn in PUBMED, we determined that eukaryotic expression vector of GADD45αgene was constructed successfully. The expression of GADD45αmRNA increased significantly after eukaryotic expression vector of GADD45αgene was transfected into cervical cancer cell strains (P<0.05). Apoptosis rate for almost 0.81–1.91% was found at 0Gy dose point in three groups,which was increased with higher radiation doses. A significant difference was observed between the transfected group and control group, lipofectin group (p < 0.05). There was no significant difference between control group and lipofectin group (p> 0.05).
     Conclusion
     1. The dystopic express of APE1 might play a pivotal role in carcinogenesis and progression of cervical cancer, and the express of APE1 might estimate the prognosis after 252Cf radiotherapy. It showed that the DNA damage repair function of APE1 might correlate with radioresistance in cervical cancer.
     2. Hela cells were irradiated with fractionated 252Cf neutron and X-rays, yielding two radioresistant cell sub-lines HelaNR and HelaXR. The radioresistant sub-lines were more radioresisitant to parental Hela cells by detecting their radioresistant characteristics.
     3. Microarray analysis was used to identify the gene expression patterns of two radioresistant sub-lines derived from Hela cells and the differentially expressed genes relate to DNA damage repair genes in cervical cancer were selected. It supported to look for the targets of improving radiosensitivity in cervical cancer.
     4. The eukaryotic expression vector of GADD45αgene was constructed. The expression of GADD45αmRNA and radiation-induced apoptosis increased significantly after eukaryotic expression vector of GADD45αgene was transfected into cervical cancer cell strains. It showed that eukaryotic expression vector of GADD45αgene might enhance radiosensitivity in cervical cancer.
引文
1.孙建衡.做好妇科肿瘤的放射治疗[J].中国实用妇科与产科杂志,2006;22(8):564-5.
    2.单锦露,雷新,王东,王阁,李力,赵可伟.锎252中子腔内后装加盆腔外照射治疗子官颈癌临床分析[J].中华妇产科杂志,2005;40(4):223-6.
    3. Schena M, Shalon D, Heller R, et al. Parallel human genome analysis: microarray-based expression monitoring of 1000 genes. Proc Natl Acad Sci U S A, 1996, 93(20):10614-9.
    4. Shiu SH, Borevitz JO. The next generation of microarray research: applications in evolutionary and ecological genomics. Heredity, 2006: 1-9.
    5. Fishel ML, Kelley MR. The DNA base excision repair protein Ape1/Ref-1 as a therapeutic and chemopreventive target. Mol Aspects Med,2007;28(3-4):375-395.
    6. Raffoul JJ, Banerjee S, Singh-Gupta V, et al. Down-regulation of apurinic/apyrimidinic endonuclease 1/redox factor-1 expression by soy isoflavones enhances prostate cancer radiotherapy in vitro and in vivo. Cancer Res,2007;67(5):2141-2149.
    7.王东,卿毅,仲召阳等. APE1 RNA干扰提高骨肉瘤中子放疗敏感性的实验研究.中华放射医学与防护杂志, 2007;27(4): 322-324.
    8.卿毅,王东,仲召阳等. pSilence APE1提高骨肉瘤放疗敏感性的动物实验研究.中国肿瘤临床, 2007;34(4): 230-233.
    9. Bhosle SM, Huilgol NG, Mishra KP. Enhancement of radiation-induced oxidative stress and cytotoxicity in tumor cells by ellagic acid [J]. Clin Chim Acta. 2005, 359(1-2): 89-100.
    10. Turesson I, Carlsson J, Brahme A, et al. Biological response to radiation therapy. Acta Oncol. 2003, 42(2): 92-106.
    11.沈瑜,糜福顺等.肿瘤放射生物学[M].中国医药科技出版社, 2001: 22~77.
    12. Ward JF. Mechanisms of DNA repair and their potential modification for radiotherapy. Int J Radiat Oncol Biol Phys. 1986, 12(7): 1027-32.
    13. Elshaikh M, Ljungman M, Ten Haken R, et al. Advances in radiation oncology. Annu Rev Med. 2006, 57: 19-31.
    14. Connell PP, Kron SJ, Weichselbaum RR. Relevance and irrelevance of DNA damage response to radiotherapy. DNA Repair (Amst). 2004, 3(8-9): 1245-51.
    15. Verheij M, Bartelink H. Radiation-induced apoptosis. Cell Tissue Res. 2000, 301(1):133-42.
    16. Moeller BJ, Dewhirst MW. Raising the bar: how HIF-1 helps determine tumor radiosensitivity. Cell Cycle. 2004, 3(9): 1107-10.
    17. Ma BB, Bristow RG, Kim J, et al. Combined-modality treatment of solid tumors using radiotherapy and molecular targeted agents. J Clin Oncol. 2003, 21(14): 2760-76.
    18. Olive PL. The role of DNA single-strand and double-strand breaks in cell killing by ionizing radiation. Radiat Res. 1998, 150: 942-45.
    19.周平坤,项晓琼,刘学英等. AT细胞DNA双链断裂重接修复效率及其忠实性.生物化学杂志. 1995, 11: 412.
    20. Shi CM, Su YP, Cheng TM. Recent advances in the pathological basis and experimental management of impaired wound healing due to total-body irradiation. Med Sci Monit. 2006, 12(1): RA1-4.
    21. Rosenberg A, Knox S. Radiation sensitization with redox modulators: a promising approach. Int J Radiat Oncol Biol Phys. 2006, 64(2): 343-54.
    22.罗刚,郑成位,单锦露等. 252锎中子腔内后装治疗子宫颈癌的临床观察.中华妇产科杂志, 2001;36(12 ): 742-744.
    23.单锦露,雷新,王东等. 252Cf中子腔内后装加盆腔外照射治疗子宫颈癌临床分析.中华妇产科杂志, 2005;40(4): 223-226.
    24. Martin RC, Knauer JB, Balo PA. Production, distribution and applications of californium-252 neutron sources. Appl Radiat Isot,2000;53(4-5): 785-792.
    25. Bobola MS, Finn LS, Ellenbogen RG, et al. Apurinic/apyrimidinic endonuclease activity is associated with response to radiation and chemotherapy in medulloblastoma and primitive neuroectodermal tumors. Clin Cancer Res, 2005;11(20): 7405-7414.
    26. Sak SC, Harnden P, Johnston CF, et al. APE1 and XRCC1 protein expression levels predict cancer-specific survival following radical radiotherapy in bladder cancer. Clin Cancer Res,2005;11(17):6205-11.
    27. Xu Y, Moore DH, Broshears J, et al. The apurinic/apyrimidinic endonuclease (APE/ref-1) DNA repair enzyme is elevated in premalignant and malignant cervicalcancer. Anticancer Res, 1997;17(5B):3713-3719.
    28. Herring CJ, West CM, Wilks DP, et al. Levels of the DNA repair enzyme human apurinic/apyrimidinic endonuclease (APE1, APEX, Ref-1) are associated with the intrinsic radiosensitivityof cervical cancers. Br J Cancer, 1998;78(9):1128-1133.
    29. Biaglow JE. The effects of ionizing radiation on mammalian cells. J Chem Educ, 1981(58): 144-156.
    30. Painter RB, Ginoza HS. Some characteristics of the resistance transfer factor (RTF) episome as determined by inactivation with tritium, P32, and gamma radiation. Biophys J. 1966; 6(2): 153-62.
    31. Lee JU, Hosotani R, Wada M, et al. Role of Bcl-2 family proteins (Bax, Bcl-2 and Bcl-X) on cellular susceptibility to radiation in pancreatic cancer cells. Eur J Can. 1999; 35(9): 1374-80.
    32. Asanuma K, Moriai R, Yajima T, et al. Survivin as a radioresistance factor in pancreatic cancer. Jpn J Cancer Res. 2000; 91(11): 1204-9.
    33. Hall EJ. Cell survival curves. Radiobiology for the radiologist (3th edn). J B Lippincott, Company, Philadelphia, 1982: 29-44.
    34.汪俊,李育华,糜富顺.细胞存活曲线数学模型的三种拟合方法的比较和改进.中华放射医学和防护杂志,1988, 8 (2), 117-119.
    35. Choi N, Baumann M, Flentjie M. Predietive factors in radiotherapy for non-small cell lung cancer: present status. Lung Cancer; 2001, 31(l): 43-56.
    36. Hall EJ. Radiosensitivity and cell age in the mitotic cucle. Radiobiology for the radiologist (3th edn). J B Lippincott, Company, Philadelphia, 1982: 91-106.
    37. Verheij M, Bartelink H. Radiation-induced apoptosis. Cell Tissue Res 2000; 301:133-42.
    38. Mayn RE,StePhens LC,Ang KK. Heterogeneity in the development of apoptosis in irradiated murine tumors of different histologies. Int J Radiat Biol. 1993, 63: 583.
    39. Schena M, Shalon D, Heller R, et al. Parallel human genome analysis: microarray-based expression monitoring of 1000 genes. Proc Natl Acad Sci U S A, 1996, 93(20):10614-9.
    40. Maduro JH, de Vries EG, Meersma GJ, et al. Targeting pro-apoptotic trail receptors sensitizes Hela cervical cancer cells to irradiation-induced apoptosis. Int J RadiatOncol Biol Phys. 2008; 72(2):543-52.
    41. Liu SS, Chan KY, Leung RC, et al. Enhancement of the radiosensitivity of cervical cancer cells by overexpressing p73alpha. Mol Cancer Ther. 2006; 5(5):1209-15.
    42. Abdulkarim B, Sabri S, Deutsch E, et al. Antiviral agent Cidofovir restores p53 function and enhances the radiosensitivity in HPV-associated cancers. Oncogene. 2002; 21(15):2334-46.
    43. Song H, Xin XY, Xiao F, et al. Influence of survivin gene repression by RNA interference on the radiosensitivity and chemosensitivity to cisplatin of cervical cancer cell Hela. Zhonghua Fu Chan Ke Za Zhi. 2006; 41(8):554-8.
    44. Haediman G. Microarray platforms-comparisons and contrasts. Pharmacogenomics. 2004; 5(5): 487-502.
    45. Dalma-Weiszhausz DD, Chicurel ME, Gingeras TR. Microarrays and genetic epidemiology: a multipurpose tool for a multifaceted field. Genet Epidemiol. 2002; 23(1): 4-20.
    46. Kitahara O, Katagiri T, Tsunoda T, et al. 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.
    47. Wong YF, Selvanayagam ZE, Wei N, et al. Expression genomics of cervical cancer: molecular classification and prediction of radiotherapy response by DNA microarray. Clin Cancer Res, 2003, 9(15):5486-92.
    48. Harima Y, Togashi A, Horikoshi K, et al. Prediction of outcome of advanced cervical cancer to thermoradiotherapy according to expression profiles of 35 genes selected by cDNA microarray analysis. Int J Radiat Oncol Biol Phys, 2004, 60(1):237-48.
    49. Haber JE. Partners and pathwaysrepairing a double-strand break. Trends Genet, 2000, 16(6):259-64.
    50. Hwang A, Muschel RJ. Radiation and the G2 phase of the cell cycle. Radiat Res. 1998; 150(Suuppl 5): S52-59.
    51. Jung M, Kern FG, Jorgensen TJ, et al. Fibroblast growth factor-4 enhanced G2 arrest and cell survival following ionizing radiation. Cancer Res. 1994; 54(19):94-7.
    52. Cheong N, Wang Y, Iliakis G. Radioresistance induced in rat embryo cells by transfection with the oncogenes H-ras plus v-myc is cell cycle dependent and maximalduring S and G2. Int J Radiat Biol. 1993; 63(5): 623-9.
    53. Consiglio Nazionale delle Ricerche, Istituto di Neurobiologia,et al.The Gene PC3TIS21/BTG2, Prototype Member of the PC3/BTG/TOB Family:Regulator in Control of Cell Growth, Differentiation,and DNA Repair? J Cellular Phy. 2001; 187: 155-165.
    54. Guardavaccaro D, Corrente G, Covone F, et al. Arrest of G(1)-S progression by the p53-inducible gene PC3 is Rb dependent and relies on the inhibition of cyclin D1 transcription. Mol Cell Biol 2000; 20:1797-815.
    55. Rouault JP, Falette N, Guéhenneux F, et al. Identification of BTG2, an antiproliferative p53-dependent component of the DNA damage cellular response pathway. Nat Genet 1996; 14:482-6.
    56. el-Ghissassi F, Valsesia-Wittmann S, Falette N, et al. BTG2(TIS21/PC3) induces neuronal differentiation and prevents apoptosis of terminally differentiated PC12 cells. Oncogene 2002; 21:6772-8.
    57. Olive PL, Banath JP, Durand RE. Development of apoptosis and polyploidy in human lymphoblast cells as a function of position in the cell cycle at the time of irradiation. Radiat Res. 1996; 146(6): 595-602.
    58. Russell J, Wheldon TE, Stanton P. A radioresistant variant derived from a human neuroblastoma cell line is less prone to radiation-induced apoptosis. Cancer Res. 1995; 55(21): 4915-21.
    59. Hendry JH, West CM. Apoptosis and mitotic cell death: their relative contributions to normal-tissue and tumour radiation respose. Int J Radiat Biol. 1997; 71(6): 709-19.
    60. Bustin SA. Quantification of mRNA using real-time reverse transcription PCR (RT-PCR): trends and problems. J Mol Endocrinol. 2002; 29: 23-39.
    61. Bubner B, Baldwin IT. Use of real-time PCR for determining copy number and zygosity in transgenic plants. Plant Cell Rep. 2004; 23(5): 263-71.
    62. Ponchel F, Toomes C, Bransfield K, et al. Real-time PCR based on SYBR-GreenⅠfluorescence: an alternative to the TaqMan assay for a relative quantification of gene rearrangements, gene amplifications and micro gene deletions. BMC Biotechnol. 2003; 3(1): 18.
    63. Zhan Q. Gadd45a, a p53- and BRCA1-regulated stress protein, in cellular response to DNA damage. Mutat Res. 2005; 569(1-2):133-43.
    64. Jin S, Fan F, Fan W, Zhao H, Tong T, Blanck P, Alomo I, Rajasekaran B, Zhan Q. Transcription factors Oct-1 and NF-YA regulate the p53-independent induction of the GADD45 following DNA damage[J]. Oncogene. 2001 May 10; 20(21):2683-90.
    65. Fan W, Jin S, Tong T, Zhao H, Fan F, Antinore MJ, Rajasekaran B, Wu M, Zhan Q. BRCA1 regulates GADD45 through its interactions with the OCT-1 and CAAT motifs [J]. J Biol Chem. 2002 Mar 8;277(10):8061-7.
    66. Weichselbaum R, Hallahan DE, Beckett MA, et al. Gene therapy targeted by radiation preferentially radiosensitizes tumor cells [J]. Cancer Res. 1994, 54 (16): 4266–4269.
    67. Cuddy K, et al. RT-PCR with affinity-captured mRNA. Nucleic Acids Res. 1993; 21(19): 2281-2287.
    68. Morgon RA, Anderson WF. Human gene therapy. Ann Rev Biochem. 1993; 62: 191-217.
    69. Mulligan RC. The badic science of gene therapy. Science. 1993; 260: 926-932.
    70. Lyerly HK, Dimaio JM. Gene delivery systems in surgery. Arch Surg.1993; 28: 1197-1206.
    71. Gauchez AS, Du Moulinet D'Hardemare A, Lunardi J, et al. Potential use of radiolabeled antisense oligonucleotides in oncology [J]. Anticancer Res. 1999, 19(6B): 4989-97.
    72. Wang XW, Zhan Q, Coursen JD, et al. GADD45 induction of a G2/M cell cycle checkpoint. Proc Natl Acad Sci U S A. 1999; 96(7): 3706-11.
    73. Gao H, Jin S, Song Y, Fu M, Wang M, Liu Z, Wu M, Zhan Q. B23 regulates GADD45a nuclear translocation and contributes to GADD45a-induced cell cycle G2-M arrest[J]. J Biol Chem. 2005 Mar 25; 280(12):10988-96.
    74. Jin S, Antinore MJ, Lung FD, Dong X, Zhao H, Fan F, Colchagie AB, Blanck P, Roller PP, Fornace AJ Jr, Zhan Q. The GADD45 inhibition of Cdc2 kinase correlates with GADD45-mediated growth suppression [J]. J Biol Chem. 2000 Jun 2; 275(22):16602-8.
    75. Jin S, Tong T, Fan W, Fan F, Antinore MJ, Zhu X, Mazzacurati L, Li X, Petrik KL, Rajasekaran B, Wu M, Zhan Q. GADD45-induced cell cycle G2-M arrest associates with altered subcellular distribution of cyclin B1 and is independent of p38 kinase activity. Oncogene[J]. 2002 Dec 12; 21(57):8696-704.
    76. Hildesheim J, Bulavin DV, Anver MR, Alvord WG, Hollander MC, Vardanian L, Fornace AJ Jr. Gadd45a protects against UV irradiation-induced skin tumors, and promotes apoptosis and stress signaling via MAPK and p53[J]. Cancer Res. 2002 Dec 15; 62(24):7305-15.
    77. Zou C, Guan Y, Zou C, Wang J, Wang LE, Liebert M, Grossman HB, Wei Q. N-(4-hydroxyphenyl)retinamide (4-HPR) modulates GADD45 expression in radiosensitive bladder cancer cell lines[J]. Cancer Lett. 2002 Jun 28; 180(2):131-7.
    78. Yin F, Bruemmer D, Blaschke F, Hsueh WA, Law RE, Herle AJ. Signaling pathways involved in induction of GADD45 gene expression and apoptosis by troglitazone in human MCF-7 breast carcinoma cells[J]. Oncogene. 2004 Jun 3; 23(26):4614-23.
    79. Jiang T, Soprano DR, Soprano KJ. GADD45A is a mediator of CD437 induced apoptosis in ovarian carcinoma cells[J]. J Cell Physiol. 2007 Sep; 212(3):771-9.
    80. Zhang D, Song L, Li J, Wu K, Huang C. Coordination of JNK1 and JNK2 is critical for GADD45alpha induction and its mediated cell apoptosis in arsenite responses[J]. J Biol Chem. 2006 Nov 10; 281(45):34113-23.
    81. Al-Romaih K, Sadikovic B, Yoshimoto M. Decitabine-induced demethylation of 5' CpG island in GADD45A leads to apoptosis in osteosarcoma cells. Neoplasia. 2008 May; 10(5):471-80.
    82. Yang C, Yang S, Wood KB. Multidrug resistant osteosarcoma cell lines exhibit deficiency of GADD45alpha expression. Apoptosis. 2009 Jan; 14(1):124-33.
    83. Li Y, Qian H, Li X. Adenoviral-mediated gene transfer of Gadd45a results in suppression by inducing apoptosis and cell cycle arrest in pancreatic cancer cell. J Gene Med. 2009 Jan; 11(1):3-13.
    84. Takekawa M, Saito H. A family of stress-inducible GADD45-like proteins mediate activation of the stress-responsive MTK1/MEKK4 MAPKKK[J]. Cell. 1998 Nov 13; 95(4):521-30.
    85. Tong T, Ji J, Jin S, Li X, Fan W, Song Y, Wang M, Liu Z, Wu M, Zhan Q. Gadd45a expression induces Bim dissociation from the cytoskeleton and translocation to mitochondria[J]. Mol Cell Biol. 2005 Jun; 25(11):4488-500.
    1. Morgenbesser SD, Williams BO, Jacks T, et al. P53-depdendent apoptosis produced by Rb-deficiency in the developing mouse lens. Nature 1994; 871: 72–74.
    2. Lane D. P53, guardian of the genome. Nature 1992; 858: 15–16.
    3. Oleinick NL, Chiu S, Friedman LR, et al. DNA–protein crosslinks: new insights into their formation and repair in irradiated mammalian cells. In: Simic MG, Grossman L, Upton AC, eds. Mechanism of DNA Damage and Repair,Plenum, New York, 1986:181–192.
    4. Radford IR. Effect of radiomodifying agents on the ratios of X-ray-induced lesions in cellular
    1. DNA: use in lethal lesions determination. Int J Radiat Biol 1986; 49: 621–637.
    5. Fornace AJ, Little JB. DNA crosslinking induced by X-rays and chemical agents. Biochim Biophys Acta 1977; 477: 343–355.
    6. Cutler RG. Crosslinkage hypothesis of aging: DNA adducts in chromatin as a primary aging process. In: Smith KD, ed. Aging, Carcinogenesis, and Radiation Biology. Plenum, New York, 1976; 443–492.
    7. Cerutti PA. Base damage induced by ionizing radiation. In: Wang SY, ed. Photochemistry and Photobiology of Nucleic Acids. Wang. Academic, New York, 1976; vol II: 375–401.
    8. Cerutti PA. Effects of ionizing radiation on mammalian cells. Naturwissenschaften 1974; 61: 51–59.
    9. Patil MS, Locher SE, Hariharan PV. Radiation induced thymine base damage and its excision repair in active and inactive chromatin of Hela cells. Int J Radiat Biol 1985; 48:691–700.
    10. Brent TP. Purification and characterization of human endonucleases specific for damaged DNA. Analysis of lesions induced by UV or X-radiation. Biochim Biophys Acta 1976; 454: 172–183.
    11. Grosovsky AJ, De Boer JG, Drobetsky EA, et al. DNA sequence analysis of ionizing radiation induced mutation in mammalian cells. Proc. 8(th) Int Congr Radiat Res 1987; abstract 217.
    12. Ahnstr?m, G Edvarsson KA. Radiation-induced single-strand breaks in DNA determined by rate of alkaline strand separation and hydroxylapatite chromatography: an alternative to velocity sedimentation. Int J Radiat Biol 1974; 26: 493–497.
    13. Coquerelle T, Bopp A, Kessler B, et al. Strand breaks and 5'-end groups in DNA of irradiated thymocytes. Int J Radiat Biol 1973; 24: 397–404.
    14. Kohn KW, Erickson, LC, Ewing RAG, et al. Fractionation of DNA from mammalian cells by alkaline elution. Biochemistry 1976; 15: 4629–4637.
    15. Roots R, Yang, TC, Craise I, et al. Impaired repair capacity of DNA breaks induced in mammalian cellular DNA by accelerated heavy ions. Radiat Res 1979; 78: 38–49.
    16. Sakai K, Okada S. Radiation-induced DNA damage and cellular lethality in cultured mammalian cells. Radiat Res 1984; 98 : 479–490.
    17. Woods WG, Lopez, M, Kalvonjian M. Normal repair of gamma-radiation induced single- and double-strand DNA breaks in retinoblastoma fibroblasts. Biochim Biophys Acta 1982; 698: 40–48.
    18. Lennartz M, Coquerelle T, Bopp A, et al. Oxygen effect on strand breaks and specific endgroups in DNA of irradiated thymocytes. Int J Radiat Biol 1975; 27: 577–587.
    19. Palcic B, Skarsgard LD. The effects of oxygen on DNA single-strand breaks produced by ionizing radiation in mammalian cells. Int J Radiat Biol 1972; 21: 417–433.
    20. Koch CJ, Painter RB. The effects of extreme hypoxia on the repair DNA single-strand breaks in mammalian cells. Radiat Res 1975; 64: 256–269.
    21. Van der Schans GP, Centen HB, Lohman PHM. The induction of gamma endonucleasesusceptible sites by gamma rays in CHO cells and their cellular repair are not affect by the presence of thiol compounds during irradiation. Mutat Res 1979; 59: 119–122.
    22. Chiu S, Oleinick NL, Friedman LR, et al. Hypersensitivity of DNA in transcriptionally active chromatin to ionizing radiation. Biochim Biophys Acta 1982; 699: 15–21.
    23. Hariharan PV, Eleczko S, Smith BP, et al. Normal rejoining of DNA strand breaks in ataxia telangiectasia fibroblast lines after low X-ray exposure. Radiat Res 1981; 86: 589–597.
    24. Paterson MC, Smith BP, Lohman PHM, et al. Defective excision repair of gamma-ray damaged DNA in human (ataxia telangiectasia) fibroblasts. Nature 1976; 260: 444–446.
    25. Edgren M, Revesz L, Larsson A. Induction and repair of single-strand DNA breaks after X-irradiation of human fibroblasts deficient in glutathione. Int J Radiat Biol 1981; 40: 355–363.
    26. Revesz L, Edgren M. Glutathione dependent yield and repair of single-strand breaks in irradiated cells. Br J Cancer Res 1984; 49(Suppl VI): 55–60.
    27. Bl?cher D. DNA double-strand breaks in Ehrlich ascites tumour cells at low doses of X-rays. Determination of induced breaks by centrifugation at reduced speed. Int J Radiat Biol 1982; 42: 317–328.
    28. Corry PM, Cole A. Radiation-induced double-strand scissions of the DNA of mammalian metaphase cells. Radiat Res 1968; 36: 528–543.
    29. Lehman AR, Stevens S. The production and repair of double-strand breaks in cells from normal humans and from patients with Ataxia telangiectasia. Biochim Biophys Acta 1977; 474: 49–60.
    30. Elkind MM. DNA repair and cell repair, are they related? Int J Radiat Oncol Biol Phys 1979; 5: 1089–1094.
    31. Fankenberg-Schwager M, Frankenberg D, et al. Repair of DNA double-strand breaks in irradiated yeast cells under nongrowth conditions. Radiat Res 1980; 82: 498–510.
    32. Van der Schans GP, Centen HB, Lohman PHM. DNA lesions induced by ionizing radiation. In: Natarajan AT, Obe G, Altmann H, eds. Progress in Mutation Research. Elsevier, Amsterdam, 1982; vol 4, 285–299.
    33. Van der Schans GP, Paterson MC, Cross WG. DNA strand breaks and rejoining in cultured human fibroblasts exposed to fast neutron or gamma rays. Int J Radiat Biol 1983; 44: 75–85.
    34. Coquerelle T, Weibezahn KF. Rejoining of DNA double-strand breaks in humanfibroblasts and its impairment in one ataxia telangiectasia and two Fanconi strains. J Supramol Struct Cell Biochem 1981; 17: 369–376.
    35. Resnick MA. The repair of double strand breaks in DNA: a model involving recombination. J Theor Biol 1976; 59: 97–106.
    36. Weibezahn KF, Coquerelle T. Radiation-induced DNA double-strand breaks are rejoined by ligation and recombination process. Nucleic Acids Res 1981; 9: 3139–3150.
    37. Radford IR. The level of induced DNA double-strand breakage correlates with cell killing after X-irradiation. Int J Radiat Biol 1990; 48: 45–54.
    38. Schwartz JL, Rotmensch J, Giovanazzi SM, et al. Faster repair of DNA double-strand breaks in radioresistant human tumour cells. Intl J Radiat Oncol Biol Phys 1988; 15: 907–912.
    39. Kelland LR, Edwards SM, Steel G. Induction and rejoining of DNA double-strand breaks in human cervix carcinoma cell lines of differing radiosensitivity. Radiat Res 1988; 116: 526–538.
    40. Green A, Pager A, Stout PM, et al. Relationships between DNA damage and the survival of radiosensitive mutant Chinese hamster cell lines exposed to gamma-radiation. Part I: intrinsic radiosensitivity. Intl J Radiat Biol 1992; 61: 465–472.
    41. Alaoui-Jamali MA, Batist G, Lehnert S. Radiation-induced damage to DNA in drug and radiation-resistant sublines of a human breast cancer cell line. Radiat Res 1992; 129: 37–42.
    42. Chang EH, Pirollo KF, Zou ZQ, et al. Oncogenes in radioresistant, noncancerous skin fibroblasts from a cancer-prone family. Science 1987; 234: 1036–1039.
    43. Kasid UN, Pfeifer A, Weichselbaum RR, et al. The ras oncogene is associated with a radiationresistant human laryngeal cancer. Science 1987; 234: 1039–1041.
    44. FitzGerald TJ, Daugherty C, Kase K, et al. Activated human N-ras oncogene enhances X-irradiation repair of mammalian cells in vitro less effectively at low dose rate. Am J Cln Oncol 1985; 8: 517–522.
    45. Sklar MD. The ras oncogenes increase the intrinsic resistance of NIH 3T3 cells to ionizing radiation. Science 1988; 239: 645–647.
    46. Ling CC, Endlich B. Radioresistance induced by oncogenic transformation. Radiat Res 1989; 120: 267–279.
    47. McKenna WG, Weiss MC, Bakanauskas VJ, et al. The role of the H-ras oncogene in radiation resistance and metastasis. Int J Radiat Oncol Biol Phys 1990; 18: 849–859.
    48. Iliakis G, Metzger L, Muschel RJ, et al. Induction and repair of DNA double strand breaks in radiation-resistant cells obtained by transformation of primary rate embryo cells with oncogenes H-ras and v-myc. Cancer Res 1990; 50: 6575–6579.
    49. Gordon DJ, Milner AW, Beaney RP, et al. The increase in radioresistance of V79 cells cultured as spheroids is correlated to changes in nuclear morphology. Radiat Res 1990; 121: 174–179.
    50. Kapiszewska M, Wright WD, Lange CS, et al. DNA supercoiling changes in nucleoids from irradiated L5178Y-S and -R cells. Radiat Res 1989; 119: 569–575.
    51. Burzio LO, Riquelme PT, Koide SS. ADP ribosylation of rate liver nucleosomal core histones. J Biol Chem 1979; 254: 3029–3037.
    52. Riquelme PT, Burzio LO, Koide SS. Poly(ADP-ribose) synthetase activity in rat testis mitochondria. J Biol Chem 1979; 254: 3018–3028.
    53. Endres M, Wang ZQ, Namura S, et al. Ischemic brain injury is mediated by the activation of poly(ADP-ribose) polymerase. J Cereb Blood Flow Metab 1997; 17 : 1143.
    54. Eliasson MJ, Sampei K, Mandir AS, et al. Poly(ADP-ribose) polymerase gene disruption renders mice resistant to cerebral ischemia. Nature Med 1997; 3: 1089.
    55. Szabo C, Dawson VL. Role of poly(ADP-ribose) synthetase in inflammation and ischaemiareperfusion. Trends Pharmacol Sci 1998; 19: 287.
    56. Paterson MC, Smith BP, Lohman PH, et al. Defective excision repair of gamma-ray-damaged DNA in human (ataxia telangiectasia) fibroblasts. Nature 1976; 260: 444–447.
    57. Lavin MF, Davidson M. Repair of strand breaks in superhelical DNA of ataxia telangiectasia lymphoblastoid cells. Cell J Sci 1981; 48: 383–391.
    58. Cole J, Arlett CF, Green MH, et al. Comparative human cellular radiosensitivity: II. The survival following gamma-irradiation of unstimulated (G0) T-lymphocytes, T-lymphocyte lines, lymphoblastoid cell lines and fibroblasts from normal donors, from ataxia–telangiectasia patients and from ataxia–telangiectasia heterozygotes. Int J Radiat Biol 1988; 54: 929–943.
    59. Cornforth MN, Bedford JS. On the nature of a defect in cells from individuals with ataxiatelangiectasia. Science 1985; 227: 1589–1591.
    60. Marecki JC, McCord JM. The inhibition of poly (ADP-ribose) polymerase enhances growth rates of ataxia telangiectasia cells. Arch Biochem Biophys 2002; 402: 227–234.
    61. Yu SW, Wang H, Poitras MF, et al. Mediation of poly(ADP-ribose) polymerase-1-dependent cell death by apoptosis-inducing factor. Science 2002; 297(5579): 259–263.
    62. Tsujimoto Y, Cossman E, Jaffe E, et al. Involvement of the bcl-2 gene in human follicular lymphoma. Science 1985; 228: 1440–1443.
    63. McDonnell T, Troncosco P, Brisbay S, et al. Expression of the protooncogene bcl-2 in the prostate and its association with emergence of androgen-independent prostate cancer. Cancer Res 1992; 52: 6940–6944.
    64. Bhargava V, Kell D, Van de Rijn M, et al. Bcl-2 immunoreactivity in breast carcinoma correlates with hormone receptor positivity. Am J Pathol 1994; 145:535– 540.
    65. Reed J, Meister L, Tanaka S, et al. Differential expression of bcl-2 protooncogene in neuroblastoma and other human tumor cell lines of neural origin. Cancer Res 1991; 51: 6529–6538.
    66. Hockenberry D, Oltvai Z, Yin XM, et al. Bcl-2 functions in an antioxident pathway to prevent apoptosis. Cell 1993; 75: 241–251.
    67. Sandstrom P, Mannic M, Buttke T. Inhibition of activation-induced death in T cell hybridomas by thiol antioxidants: oxidative stress as a mediator of apoptosis. J Leukocyte Biol 1994; 55: 221–226.
    68. Kane D, Saralin T, Auton S, et al. Bcl-2 inhibition of neural cell death: decreased generation of reactive oxygen species. Science 1993; 262: 1274–1276.
    69. Story M, Vochringer D, Malone C, et al. Radiation-induced apoptosis in sensitive and resistant cells isolated from a mouse lymphoma. Int J Radiat Biol 1994; 66: 659–669.
    70. Overgaard J, Horsman MR. Modification of hypoxia-induced radioresistance in tumors by the use of oxygen and sensitizers. Semin Radiat Oncol 1996; 6: 10–21.
    71. Saunders M, Dische S. Clinical results of hypoxic cell radiosensitization from hyperbaric oxygen to accelerated radiotherapy, carbogen and nicotinamide. Br J Cancer 1996; 27(Suppl): S271–S278.
    72. Overgaard J. Sensitization of hypoxic tumor cells-clinical experience. Int J Radiat Biol 1989; 56: 801–811.
    73. Werner-Wasik M, Schmid CH, Bornstein L, et al. Prognostic factors for local and distant recurrence in stage I and II cervical carcinoma. Int J Radiat Oncol Biol Phys 1995; 32: 1309–1317.
    74. Fein DA, Le WR, Henlon AL, et al. Pretreatment hemoglobin level influences local control and survival of T1–T2 squamous cell carcinomas of the glottic larynx. J Clin Oncol 1995; 13: 2077–2083.
    75. Dubray B, Mosseri V, Brunin F, et al. Anemia is associated with lower local–regional control and survival after radiation therapy for head and neck cancer: a prospective study. Radiology 1996; 201: 553–558.
    76. Dische S, Warburton MF, Saunders MI. Radiation myelitis and survival in the radiotherapy of lung cancer. Int J Radiat Oncol Biol Phys 1988; 15: 75–81.
    77. Sasai K, Ono K, Hiraoka M, et al. The effect of arterial oxygen content on the results of radiation therapy for epidermoid bronchogenic carcinoma. Int J Radiat Oncol Biol Phys 1989; 16: 1477–1481.
    78. Cole CJ, Pollack A, Zagars GK, et al. Local control of muscle-invasive bladder cancer: preoperative radiotherapy and cystectomy versus cystectomy alone. Int J Radiat Oncol Biol Phys 1955; 32: 331–340.
    79. Greven KM, Solin LJ, Hanks GE. Prognostic factors in patients with bladder carcinoma treated with definitive irradiation. Cancer 1990; 65: 908–912.
    80. Dunphy EP, Peterson IA, Cox RS, et al. The influence of initial hemoglobin and blood pressure levels on results of radiation therapy for carcinoma of the prostate. Int J Radiat Oncol Biol Phys 1989; 16: 1173–1178.
    81. Siemann DW, Horsman MR, Chaplin DJ. The radiation response of KHT sarcomas following nicotinamide treatment and carbogen breathing. Radiother Oncol 1994; 31: 117–122.
    82. Fenton BM, Lord EM, Paoni SF. Enhancement of tumor perfusion and oxygenation by carbogen and nicotinamide during single- and multifraction irradiation. Radiat Res 2000; 153: 75–83.
    83. Kaanders JH, Pop LA, Marres HA, et al. Accelerated radiotherapy with carbogen andnicotinamide (ARCON) for laryngeal cancer. Radiother Oncol 1998; 48: 115–122.
    84. Hoskin PJ, Saunders MI, Dische S. Hypoxic radiosensitizers in radical radiotherapy for patients with bladder carcinoma: hyperbaric oxygen, misonidazole, and accelerated radiotherapy, carbogen, and nicotinamide. Cancer 1999; 86: 1322–1328.
    85. Overgaard J, Hansen HS, Overgaard M, et al. A randomized double-blind phase III study of nimorazole as a hypoxic radiosensitizer of primary radiotherapy in supraglottic larynx and pharynx carcinoma. Results of the Danish Head and Neck Cancer Study (DAHANCA) Protocol 5–85. Radiother Oncol 1998; 46: 135–146.
    86. Von Pawel J, von Rosemeling R, Gatzemeier U, et al. Tirapazamine plus cisplatin versus cisplatin in advanced non-small-cell lung cancer: a report of the international CATAPULT I study group. Cisplatin and tirapazamine in subjects with advanced previously untreated nonsmall-cell lung tumors. J Clin Oncol 2000; 18: 1351–1359.
    87. Rischin D, Peters L, Hicks R, et al. Phase I trial of concurrent tirapazamine, cisplatin, and radiotherapy in patients with advanced head and neck cancer. J Clin Oncol 2001; 19: 535–542.
    88. Craighead PS, Pearcey R, Stuart G. A phase I/II evaluation of tirapazamine administered intravenously concurrent with cisplatin and radiotherapy in women with locally advanced cervical cancer. Int J Radiat Oncol Biol Phys 2000; 48: 791–795.
    89. Kovacs MS, Hocking DJ, Evans JW, et al. Cisplatin anti-tumor potentiation by tirapazamine results from a hypoxia-dependent cellular sensitization to cisplatin. Br J Cancer 1999; 80: 1245–1251.
    90. Goldberg Z, Evans J, Birrell G, et al. An investigation of the molecular basis for the synergistic interaction of tirapazamine and cisplatin. Int J Radiat Oncol Phys 2001; 49: 175–182.

© 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