抑癌基因p16、Bcl11b及Ikaros与辐射诱发胸腺淋巴瘤的相关研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
辐射致癌是电离辐射重要的生物效应之一,目前,随着核能在国民经济和军事领域中的广泛应用,辐射致癌的危险性受到更广泛关注。近年来,学者们对辐射致癌及其机制进行了深入研究,但辐射致癌的机理至今尚未彻底阐明。本研究利用辐射致癌的经典动物模型-辐射诱发小鼠胸腺淋巴瘤模型,探讨抑癌基因p16、Bcl11b及Ikaros基因在辐射致癌中的作用。本研究的创新点在于将分子生物学、遗传学及表观遗传学相结合探讨辐射致癌机制。
     本研究采用基因芯片技术筛查基因表达谱改变;采用实时定量PCR及RT-PCR方法检测基因转录水平变化;采用Western Blot方法检测蛋白表达变化;采用PCR-RFLP方法检测基因SNPs;采用MSP及BSP方法检测基因DNA甲基化改变。结果证实,电离辐射可诱发胸腺淋巴瘤,其发生率BALB/c小鼠为58.51%,C57BL/6J小鼠为43.01%,两个品系小鼠辐射致癌敏感性存在差异。
     基因芯片检测表明,BALB/c小鼠胸腺淋巴瘤细胞中基因表达谱发生改变,共有3063个差异表达基因,其中上调基因1591个,下调基因1472个,涉及KEGG数据库145个通路,BioCarta数据库76个通路及GenMAPP数据库218个通路。研究表明,胸腺淋巴瘤细胞中P16及Bcl11b蛋白表达均明显降低。时效研究表明,胸腺细胞中Bcl11b蛋白表达于照射后1~3个月明显降低。胸腺细胞中Ikaros mRNA水平于照射后3个月显著降低(P < 0.01)。遗传学研究表明,p16基因rs3695947位点、Ikaros基因rs28185870及rs28185923位点基因多态性的差别可能与电离辐射诱发胸腺淋巴瘤发生率的种系差异相关。表观遗传学研究证实,胸腺淋巴瘤细胞中p16基因启动子区CpG岛所检测的23个CpG位点中,第10及第11个CpG位点的DNA甲基化发生率均显著增高(P < 0.05,P < 0.01)。
     以上结果提示,抑癌基因表达下调及DNA高度甲基化改变,可能在电离辐射诱发胸腺淋巴瘤中起重要作用。本研究所获结果将为进一步阐明辐射致癌机制提供新的生物学依据。
Ph.D. Student Liu Yongzhe Supervisor Prof. Ju Guizhi Radiation carcinogenesis is an important biological effect of ionizing radiation. Although many studies have been conducted, the mechanism of radiation carcinogenesis is still remaining unclear. In the present study we established a mouse thymic lymphoma model induced byγ-ray; employed gene chip to detect the difference of gene expression; used real-time PCR and RT-PCR to detect the level of gene transcription; adopted Western Blot to check the changes of protein expression; took PCR-RFLP to detect SNPs on target gene; and used MSP and BSP to detect the changes of DNA methylation. All these findings may explain the relationships among tumor suppressor genes p16, Bcl11b, Ikaros, and thymic lymphoma induced by ionizing radiation.
     1 The establishment of mouse thymic lymphoma model induced by ionizing radiation
     1.1 Mouse thymic lymphoma model induced by ionizing radiation We usedγ-ray to systemically irradiate mice BALB/c and C57BL/6J. The single dose was 1.75Gy per week continuing for 4 weeks. After 6 months irradiation, mice were executed. The thymus was taken out and undertaken gross specimen observation and pathology detection. At the meanwhile, we used the normal thymus of BALB/c and C57BL/6J as controls. According to the gross specimen observation results the mice with thymic lymphoma had an enlarged thymus with gray color, tough texture, adhesion significantly with the surrounding tissue, and some of mice were accompanied by pleural effusion by comparison with normal mice. Pathology results showed that the thymic organizational structure of the mice with thymic lymphoma was replaced by the diffuse proliferation of lymphoma cells. After calculation, the thymic lymphoma incidences induced by ionizing radiation were 58.51% (55/94) for BALB/c mice and 43.01% (40/93) for C57BL/6J mice. We also established 1-month model and 3-month model exposed to ionizing radiation for later use.
     2 Changes of gene expression profile in thymic lymphoma cells induced by ionizing radiation
     2.1 Differential expression genes in thymic lymphoma cells induced by ionizing radiation
     We mixed BALB/c mouse thymic lymphoma and normal thymus respectively and sent to Shanghai Boxing Company for detection. The BeadChip Test of MouseWG-6 v2.0 genome-wide expression showed that a total of 3063 genes expressed differently between thymic lymphoma and normal thymus, including 1591 up-regulated genes, 1472 down-regulated genes.
     2.2 Pathways of differential expression genes in thymic lymphoma cells induced by ionizing radiation
     Differential expression genes in thymic lymphoma and normal thymus cells involved 145 pathways in the KEGG database, 76 pathways in the BioCarta database, and 218 pathways in the GenMAPP database according to the bio-molecular function annotation system on-line analysis of the BOAO MAS.
     3 Gene transcription changes of p16, Bcl11b and Ikaros in thymic lymphoma cells induced by ionizing radiation
     3.1 Changes of p16 mRNA in thymic lymphoma cells The real-time PCR was used to detect the changes of p16 mRNA in thymic lymphoma cells in BALB/c mice. There was no significant difference on p16 mRNA level between thymic lymphoma and normal thymus cells.
     3.2 Changes of Bcl11b mRNA in thymic lymphoma cells The real-time PCR was used to detect the changes of Bcl11b mRNA in thymic lymphoma cells in BALB/c mice. There was no significant difference on Bcl11b mRNA level between thymic lymphoma and normal thymus cells.
     3.3 Changes of Ikaros mRNA in thymic lymphoma cells
     The real-time PCR was used to detect the changes of Ikaros mRNA in thymic lymphoma cells in BALB/c mice. There was no significant difference on Ikaros mRNA level between thymic lymphoma and normal thymus cells.
     4 Protein expression changes of p16 and Bcl11b in thymic lymphoma cells induced by ionizing radiation
     4.1 Changes of p16 protein in thymic lymphoma cell Western Blot was used to detect the changes of p16 protein expression in thymic lymphoma cells in BALB/c mice. We found that the p16 protein expression decreased in thymic lymphoma compared to normal thymus cell.
     4.2 Changes of Bcl11b protein in thymic lymphoma cell Western Blot was used to detect the changes of Bcl11b protein expression in thymic lymphoma calls in BALB/c mice. We found that the Bcl11b protein expression decreased in thymic lymphoma compared to normal thymus cell.
     5 Changes of gene transcription of p16, Bcl11b and Ikaros in mouse thymocytes before tumor formation
     5.1 Changes of p16 mRNA in thymocytes
     We used RT-PCR to detect the changes of p16 mRNA in thymus cells in BALB/c mice before tumor formation. The results indicated that there were no significant differences on p16 mRNA level in thymocytes among 1-month, 3-months irradiation and normal groups.
     5.2 Changes of Bcl11b mRNA in thymocytes
     We used RT-PCR to detect the changes of Bcl11b mRNA in thymus cells in BALB/c mice before tumor formation. The results indicated that there were no significant differences on Bcl11b mRNA level in thymocytes among 1-month, 3-months irradiation and normal groups.
     5.3 Changes of Ikaros mRNA in thymocytes
     We used RT-PCR to detect the changes of Ikaros mRNA in thymus cells in BALB/c mice before tumor formation. The results indicated that there were no significant differences on Ikaros mRNA level in thymocytes between 1-month irradiation and normal groups. The Ikaros mRNA level in 3-month irradiation group was significantly reduced compared to the normal thymocytes (P<0.01).
     6 Changes of protein expression of p16, Bcl11b and Ikaros in mouse thymocytes before tumor formation
     6.1 Changes of p16 protein expression in thymocytes Western Blot was employed to detect the changes of p16 protein expression in thymus cells in BALB/c mice before tumor formation. The results indicated that there were no significant differences on p16 protein expression in thymocytes among 1-month, 3-months irradiation and normal groups.
     6.2 Changes of Bcl11b protein expression in thymocytes Western Blot was employed to detect the changes of Bcl11b protein expression in thymus cells in BALB/c mice before tumor formation. The results indicated that there were significant differences on Bcl11b protein expression in thymocytes among 1-month, 3-months irradiation and normal groups. The protein expression level of Bcl11b both in 1-month and 3-months irradiation groups were significantly reduced compared to the normal thymocytes.
     7 The differences of germ-line in single nucleotide polymorphisms (SNPs)
     7.1 The differences of p16 in SNPs between BALB/c and C57BL/6J mice The PCR-RFLP method was used to detect rs3695947 polymorphism on p16. The results showed that the genotype of rs3695947 is G/G in BALB/c mice and A/A in C57BL/6J mice.
     7.2 The differences of Bcl11b in SNPs between BALB/c and C57BL/6J mice The PCR-RFLP method was used to detect rs33254541 polymorphism on Bcl11b. The results showed that the genotype of rs33254541 is T/T both in BALB/c and C57BL/6J mice.
     7.3 The differences of Ikaros in SNPs between BALB/c and C57BL/6J mice The PCR-RFLP method was used to detect rs28185870 and rs28185923 polymorphism on Ikaros. The results showed that the genotypes of rs28185870 and rs28185923 are C/C, T/T in BALB/c mice respectively, and T/T, C/C in C57BL/6J mice respectively.
     8 DNA methylation changes of p16, Bcl11b and Ikaros genes in thymic lymphoma cells
     8.1 DNA methylation changes of CpG island in p16 promoter region in thymic lymphoma
     MSP was used to detect the DNA methylation changes of CpG island in p16 promoter region in thymic lymphoma. The results showed that there were no DNA methylation changes of CpG island in p16 promoter region either in thymic lymphoma or normal thymic cells.
     We used BSP methods to detect the DNA methylation changes of CpG island in p16 promoter region for thymic lymphoma. The results showed that the DNA methylations were significantly increased at the tenth and the eleventh among 23 CpG sites detected in p16 promoter region in thymic lymphoma (P<0.05, P<0.01respectively).
     8.2 DNA methylation changes of CpG island in Bcl11b promoter region in thymic lymphoma BSP was used to detect the DNA methylation changes of CpG island in Bcl11b promoter region in thymic lymphoma. The results showed that there were no DNA methylation changes of CpG island in Bcl11b promoter region either in thymic lymphoma or normal thymic cells among 20 CpG sites.
     8.3 DNA methylation changes of CpG island in Ikaros promoter region in thymic lymphoma
     BSP was used to detect the DNA methylation changes of CpG island in Ikaros promoter region in thymic lymphoma. The results showed that there were one DNA methylation changes of CpG island in Ikaros promoter region both in thymic lymphoma and normal thymic cells among 50 CpG sites detected.
     The present study will provide new biological evidence to elucidate the mechanisms for radiation carcinogenesis.
引文
[1]刘树铮.医学放射生物学[M].北京:原子能出版社, 2006.
    [2]周平坤,霍艳英,吴德昌.辐射致癌效应与机制[J].辐射防护通讯, 2007, 27(1): 7-12.
    [3]李雨,韩玲,蔡建明.辐射致癌研究进展[J].国外医学肿瘤学分册, 2000, 27(4): 219-220.
    [4] KAPLAN HS, BROWN MB. A quantitative dose-response study of lymphoid-tumor development in irradiated C57 black mice[J]. J Natl Cancer Inst, 1952, 13(1): 185-208.
    [5] KAPLAN HS, BROWN MB, PAULL J. Influence of bone borrow injections on involution and neoplasia of mouse thymus after systemic irradiation[J]. J Natl Cancer Inst, 1953, 14(2): 303-316.
    [6] KAPLAN HS. On the natural history of the murine leukemia[J]. Cancer Res, 1967, 17(8): 1325-1340.
    [7] AIZAWA S, TANAKA K, MORI M, et al. Direct detection of p53-/- thymocyte appearing at an early stage of radiation-induced thymic lymphomagenesis in p53+/- heterozygous B10 mice[J]. Int J Radiat Biol, 2007, 83(4): 259-267.
    [8] SANTOS J, VAQUERO C, REYES J, et al. Lack of correlation between DNA copy number and mRNA expression levels of c-myc in gamma-radiation- induced mouse thymic lymphomas by using quantitative real-time PCR[J]. Clin Transl Oncol, 2006, 8(5): 349-353.
    [9] MORALES JC, FRANCO S, MURPHY MM, et al. 53BP1 and p53 synergize to suppress genomic instability and lymphomagenesis[J]. Proc Natl Acad Sci U S A, 2006, 103(9): 3310-3315.
    [10] VILLA-MORALES M, SANTOS J, FERNáNDEZ-PIQUERAS J. Functional Fas (Cd95/Apo-1) promoter polymorphisms in inbred mouse strains exhibiting different susceptibility to gammaradiation-induced thymic lymphoma[J]. Oncogene, 2006, 25(14): 2022-2029.
    [11] LABI V, ERLACHER M, KIESSLING S, et al. Loss of the BH3-only protein Bmf impairs B cell homeostasis and accelerates gamma irradiation-induced thymic lymphoma development[J]. J Exp Med, 2008, 205(3): 641-655.
    [12] TAMURA Y, MARUYAMA M, MISHIMA Y, et al. Predisposition to mouse thymic lymphomas in response to ionizing radiation depends on variant alleles encoding metalresponsive transcription factor-1 (Mtf-1)[J]. Oncogene, 2005, 24(3): 399-406.
    [13] ISHII-OHBA H, KOBAYASHI S, NISHIMURA M, et al. Existence of a threshold-like dose for gamma-ray induction of thymic lymphomas and no susceptibility to radiation-induced solid tumors in SCID mice[J]. Mutat Res, 2007, 619(1-2): 124-133.
    [14] DANGE P, SARMA H, PANDEY BN, et al. Radiation-induced incidence of thymic lymphoma in mice and its prevention by antioxidants[J]. J Environ Pathol Toxicol Oncol, 2007, 26(4): 273-279.
    [15] UTSUYAMA M, HIROKAWA K. Radiation-induced-thymic lymphoma occurs in young, but not in old mice[J]. Exp Mol Pathol, 2003, 74(3): 319-325.
    [16]李秀娟,赫荣华,李修义,等.低剂量辐射适应性反应对辐射诱发小鼠胸腺淋巴瘤的影响[J].中华放射医学与防护杂志, 2000, 20(3): 153-156.
    [17] INA Y, TANOOKA H, YAMADA T, et al. Suppression of thymic lymphoma induction by life-long low-doserate irradiation accompanied by immune activation in C57BL/6 mice[J]. Radiat Res, 2005, 63(2): 153-158.
    [18] HOSOI Y. Antitumor effects by low dose total body irradiation[J]. Yakugaku Zasshi, 2006, 126(10): 841-848.
    [19] BONIVER J, HUMBLET C, RONGY AM, et al. Cellular events in radiationinduced lymphomagenesis[J]. Int J Radiat Biol, 1990, 57(4): 693-698.
    [20] SADO T, KAMISAKU H, KUBO E. Bone marrow-thymus interactions during thymic lymphomagenesis induced by fractionated radiation exposurein B10 mice: analysis using bone marrow transplantation between Thy 1 congenic mice[J]. J Radiat Res (Tokyo), 1991, 32(2): 168-180.
    [21] KOMINAMI R, NIWA O. Radiation carcinogenesis in mouse thymic lymphomas[J]. Cancer Sci, 2006, 97(7): 575-581.
    [22] DE YZAGUIRRE MM, HERNáNDEZ JS, NAVARRO PF, et al. Epigenetic silencing of E-and N-cadherins in the stroma of mouse thymic lymphomas[J]. Carcinogenesis, 2006, 27(5):1081-1089.
    [23] SANTOS J, GONZáLEZ-SáNCHEZ L, MATABUENA-DEYZAGUIRRE M, et al. A role for stroma-derived annexin A1 as mediator in the control of genetic susceptibility to T-cell lymphoblastic malignancies through prostaglandin E2 secretion[J]. Cancer Res, 2009, 69(6): 2577-2587.
    [24] KANG HM, JANG JJ, LANGFORD C, et al. DNA copy number alterations and expression of relevant genes in mouse thymic lymphomas induced by gamma-irradiation and N-methyl-N-nitrosourea[J]. Cancer Genet Cytogenet, 2006, 166(1): 27-35.
    [25] TAKABATAKE T, KAKINUMA S, HIROUCHI T, et al. Analysis of changes in DNA copy number in radiation-induced thymic lymphomas of susceptible C57BL/6, resistant C3H and hybrid F1 Mice[J]. Radiat Res, 2008, 169(4): 426-436.
    [26] YOSHIDA MA, NAKATA A, AKIYAMA M, et al. Distinct structural abnormalities of chromosomes 11 and 12 associated with loss of heterozygosity in X-ray-induced mouse thymic lymphomas[J]. Cancer Genet Cytogenet, 2007, 179(1): 1-10.
    [27] KOTURBASH I, POGRIBNY I, KOVALCHUK O. Stable loss of global DNA methylation in the radiation-target tissue-a possible mechanism contributing to radiation carcinogenesis[J]? Biochem Biophys Res Commun, 2005, 337(2): 526-533.
    [28] SUZUKI G, SHIMADA Y, HAYASHI T, et al. An association between oxidative stress and radiation-induced lymphomagenesis[J]. Radiat Res,2004, 161(6): 642-647.
    [29]曾益新.肿瘤学[M].北京:人民卫生出版社, 1999.
    [30] OHI H, MISHIMA Y, KAMIMURA K, et al. Multi-step lymphomagenesis deduced from DNA changes in thymic lymphomas and atrophic thymuses at various times after gamma-irradiation[J]. Oncogene, 2007, 26(36): 5280-5289.
    [31] TANAKA K, WATANABE K, MORI M, et al. Cytogenetic and cellular events during radiation-induced thymic lymphomagenesis in the p53 heterozygous (+/-) B10 mouse[J]. Int J Radiat Biol, 2002, 78(3): 165-172.
    [32] KUBOTA T, YOSHIKAI Y, TAMURA Y, et al. Comparison of properties of spontaneous and radiationinduced mouse thymic lymphomas: Role of Trp53 and radiation[J]. Radiat Res, 2005, 163(2): 159-164.
    [33] DANO L, GUILLY MN, M ULERISM, et al. CGH analysis of radon-induced rat lung tumors indicates similarities with lung cancers[J]. Genes Chromosomes Cancer, 2000, 29(1): 1-8.
    [34]菅复春,张子宏,肖乃淼,等.基因芯片技术的应用[J].河南畜牧兽医, 2006, 27(8): 11-12.
    [35]刘红岩,从玉文,善亚君,等.电离辐射诱导BALB/c小鼠骨髓细胞基因表达谱的研究[J].辐射研究与辐射工艺学报, 2003, 21(4): 272-275.
    [36]李智恒李雨. C57小鼠受照后胸腺细胞基因表达谱的变化[J].国际放射医学核医学杂志, 2007, 31(4): 240-244.
    [37]龙贤辉,徐勤枝,贺性鹏,等. 2 Gy和10 Gyγ射线照射正常人淋巴母细胞基因表达转录谱的比较[J].中华放射医学与防护杂志, 2006, 26(2): 110-113.
    [38]李建国,闻建华,段志凯,等.不同剂量γ射线照射人肝细胞差异基因表达谱的研究[J].辐射防护, 2009, 29(3): 148-152.
    [39]刘青杰,陈德清.辐射诱导的基因表达改变及其在生物剂量学中的应用前景[J].中华放射医学与防护杂志, 2008, 28(6): 665-668.
    [40]韩玲,李雨,陈杞.应用DNA芯片技术结合核技术对辐射致癌分子机制的研究[J].国外医学·放射医学核医学分册, 2001, 25(2): 78-81.
    [41]王心如.毒理学基础[M].北京:人民卫生出版社, 2008.
    [42] COLLINS FS, PATRINOS A, JORDAN E, et al. New goals for the U.S. human genome project: 1998-2003[J]. Science, 1998, 282(5389): 682-689.
    [43]仪军玲,李彩霞,胡兰.单核苷酸多态性及其检测方法[J].证据科学, 2008, 16(6): 757-763.
    [44]赵静,周韧.单核苷酸多态性及其数据库的应用[J].国际病理科学与临床杂志, 2006, 26(2): 152-155.
    [45] RISCH NJ. Searching for genetic determinants in the new millennium[J]. Nature, 2000, 405(6788): 847-856.
    [46] TAYLOR JG, CHOI EH, FOSTER CB, et al. Using genetic variation to study human disease[J]. Trends Mol Med, 2001, 7(11): 507-512.
    [47]刘越,吕社民.单核苷酸多态性影响基因功能的机制[J].生命的化学, 2008, 28(2): 214-216.
    [48]杨永强,王巍杰,徐长波.单核苷酸多态性研究进展[J].化学与生物工程, 2009, 26(8): 19-21.
    [49] GABRIEL SB, SCHAFFNER SF, NGUYEN H, et al. The structure of haplotype blocks in the human genome[J]. Science, 2002, 296(5576): 2225-2229.
    [50] HAN W, YIP S P,WANG J, et al. Using denaturing HPLC for SNP discovery and establishing the linkage disequilibrium pattern for the all-trans-retinol dehydrogenase (RDH8) gene[J]. J Hum Genet, 2004, 49(1): 16-23.
    [51] HAN W, LOU DH, WANG J, et al. Application of DHPLC in human genomic SNP screening and genotyping for all-trans-retinol dehydrogenase[J]. Zhejiang Da Xue Xue Bao Yi Xue Ban, 2005, 34(3): 248-254.
    [52] GUZOWSKI D, CHANDRASEKARAN A, GAWEL C, et al. Analysis of single nucleotide polymorphisms in the promoter region of interleukin-10 by denaturing high-performance liquid chromatography[J]. J Biomol Tech, 2005, 16(2): 154-166.
    [53] LIU D, ZHANG Y, DU Y, et al. Identification and characterization of single nucleotide polymorphisms in 6 growth-correlated genes in porcine by denaturing high performance liquid chromatography[J]. DNA Seq, 2007, 18(3): 220-227.
    [54] DING C, JIN S. High-throughput methods for SNP genotyping[J]. Methods Mol Biol, 2009, 578: 245-254.
    [55] BUCASAS KL, PANDYA GA, PRADHAN S, et al. Assessing the utility of whole-genome amplified serum DNA for array-based high throughput genotyping[J]. BMC Genet, 2009, 18(10): 85.
    [56] ANANTHARAMAN R, CHEW FT. Validation of pooled genotyping on the Affymetrix 500 k and SNP6.0 genotyping platforms using the polynomial- based probe-specific correction[J]. BMC Genet, 2009, 14(10): 82.
    [57]翟仙敦,刘之江,侯一平.焦磷酸测序技术及其法医学应用[J].中国司法鉴定, 2009, 4: 39-42, 63.
    [58] ROYO JL, GALáN JJ. Pyrosequencing for SNP genotyping[J]. Methods Mol Biol, 2009, 578:123-133.
    [59] ANJUM GM, DU W, KLEIN R, et al. Pyrosequencing-based strategy for a successful SNP detection in two hypervariable regions: HV-I/HV-II of the human mitochondrial displacement loop[J]. Electrophoresis, 2010, 31(2): 309-314.
    [60] HIRUNSATIT R, KONGRUTTANACHOK N, SHOTELERSUK K, et al. Polymeric immunoglobulin tecep tor polymorphisms and risk of nasopharyngeal cancer[J]. BMC Genet, 2003, 4(1): 3.
    [61] PAN C, LAN X, CHEN H, et al. A TaqI PCR-RFLP detecting a novel SNP in exon 2 of the bovine POU1F1 gene[J]. Biochem Genet, 2008, 46(7-8): 424-432.
    [62]周涛,李庆.单核苷酸多态性遗传标记(SNP)的检测及其应用[J].中国优生与遗传杂志, 2010, 18(2): 3-4.
    [63] WENZEL T, FR?HLICH T, STRASSBURGER K, et al. Novelpropargylamine -linked nucleosides for high throughput SNP genotyping by MALDI-TOF MS[J]. Nucleosides Nucleotides Nucleic Acids, 2001, 20(4-7): 883-887.
    [64]张莉,王绿化.单核苷酸多态性与放射性损伤[J].癌症进展杂志, 2008, 6(2): 141-146, 162.
    [65] WILLEMS P, CLAES K, BAEYENS A, et al. Polymorphisms in nonhomologous end-joining genes associated with breast cancer risk and chromosomal radiosensitivity[J]. Genes Chromosomes Cancer, 2008, 47(2): 137-148.
    [66] WILDING CS, CURWEN GB, TAWN EJ, et al. Influence of polymorphisms at loci encoding DNA repair proteins on cancer susceptibility and G2 chromosomal radiosensitivity[J]. Environ Mol Mutagen, 2007, 48(1): 48-57.
    [67] YUAN X, LIAO Z, LIU Z, et al. Single nucleotide polymorphism at rs1982073: T869C of the TGFbeta 1 gene is associated with the risk of radiation pneumonitis in patients with non-small-cell lung cancer treated with definitive radiotherapy[J]. J Clin Oncol, 2009, 27(20): 3370-3378.
    [68] IWATA M, IWAKAWA M, NODA S, et al. Correlation between single nucleotide polymorphisms and jejunal crypt cell apoptosis after whole body irradiation[J]. Int J Radiat Biol, 2007, 83(3): 181-186.
    [69] OKAMOTO K, IWASAKI N, NISHIMURA C, et al. Identification of KCNJ15 as a susceptibility gene in Asian patients with type 2 diabetes mellitus[J]. Am J Hum Genet, 2010, 86(1): 54-64.
    [70] GRANT SF, LI M, BRADFIELD JP, et al. Association analysis of the FTO gene with obesity in children of Caucasian and African ancestry reveals a common tagging SNP[J]. PLoS One, 2008, 3(3): e1746.
    [71] NAGANUMA T, NAKAYAMA T, SATO N, et al. Haplotype-based case-control study on human apurinic/apyrimidinic endonuclease 1/redox effector factor-1 gene and essential hypertension[J]. Am J Hypertens, 2010,23(2): 186-191.
    [72] POTKIN SG, GUFFANTI G, LAKATOS A, et al. Hippocampal atrophy as a quantitative trait in a genome-wide association study identifying novel susceptibility genes for Alzheimer's disease[J]. PLoS One, 2009, 4(8): e6501.
    [73] OHI K, HASHIMOTO R, YASUDA Y, et al. The chitinase 3-like 1 gene and schizophrenia: Evidence from a multi-center case-control study and meta-analysis[J]. Schizophr Res, 2010, 116(2-3): 126-132.
    [74] CHIN LJ, RATNER E, LENG S, et al. A SNP in a let-7 microRNA complementary site in the KRAS 3' untranslated region increases non-small cell lung cancer risk[J]. Cancer Res, 2008, 68(20): 8535-8540.
    [75] KATOH M. Dysregulation of stem cell signaling network due to germline mutation, SNP, Helicobacter pylori infection, epigenetic change and genetic alteration in gastric cancer[J]. Cancer Biol Ther, 2007, 6(6): 832-839.
    [76] WADDINGTON CH. Preliminary notes on the development of the wings in normal and mutant strains of drosophila[J]. Proc Natl Acad Sci U S A, 1939, 25(7): 299-307.
    [77] ESTELLER M. Epigenetics in cancer[J]. N Engl J Med, 2008, 358(11): 1148-1159.
    [78] HERMANN A, GOWHER H, JELTSCH A. Biochemistry and biology of mammalian DNA methyltransferases[J]. Cell Mol Life Sci, 2004, 61(19-20): 2571-2587.
    [79] SULEWSKA A, NIKLINSKA W, KOZLOWSKI M, et al. DNA methylation in states of cell physiology and pathology[J]. Folia Histochem Cytobiol, 2007, 45(3): 149-158.
    [80] PARRY TE. Mutagenic mechanisms in leukemia and cancer: A new concept cytosine lack could be as mutagenic as cytosine deamination[J]. Leuk Res, 2006, 30(9): 1079-1083.
    [81] GAUDET F, HODGSON JG, EDEN A, et al. Induction of tumors in mice bygenomic hypomethylation[J]. Science, 2003, 300(5618): 489-492.
    [82] SIDIROPOULOS M, PAMPALAKIS G, SOTIROPULOU G, et al. Downregulation of human kallikrein 10 (KLK10/NES1) by CpG island hypermethylation in breast, ovarian and prostate cancers[J]. Tumour Biol, 2005, 26(6): 324-336.
    [83]周翠兰,殷宇芳,张佳,等. DNA甲基化的生物学意义及其检测方法[J].南华大学学报(医学版), 2005, 33(2): 148-153.
    [84] SULEWSKA A, NIKLINSKA W, KOZLOWSKI M, et al. Detection of DNA methylation in eucaryotic cells[J]. Folia Histochem Cytobiol, 2007, 45(4): 315-324.
    [85] HERMAN JG, GRAFF JR, MYOHANEN S, et al. Methylation-specific PCR: A novel PCR assay for methylation status of CpG islands[J]. Proc Natl Acad Sci USA, 1996, 93(18): 9821-9826.
    [86] LI LC, DAHIYA R. MethPrimer: Designing primers for methylation PCRs[J]. Bioinformatics, 2002, 18(11): 1427-1431.
    [87] LI LC. Designing PCR primer for DNA methylation mapping[J]. Methods Mol Biol, 2007, 402: 371-384.
    [88] ZHANG Y, ROHDE C, TIERLING S. DNA methylation analysis by bisulfite conversion, cloning, and sequencing of individual clones[J]. Methods Mol Biol, 2009, 507: 177-187.
    [89] KALINICH JF, CATRAVAS GN, SNYDER SL. The effect of gamma radiation on DNA methylation[J]. Radiat Res, 1989, 117(2): 185-197.
    [90] TAWA R, KIMURA Y, KOMURA J, et al. Effects of X-ray irradiation on genomic DNA methylation levels in mouse tissues[J]. J Radiat Res (Tokyo), 1998, 39(4): 271-278.
    [91] POGRIBNY I, RAICHE J, SLOVACK M, et al. Dose-dependence, sex-and tissue-specificity, and persistence of radiation-induced genomic DNA methylation changes[J]. Biochem Biophys Res Commun, 2004, 320(4): 1253-1261.
    [92] KOVALCHUK O, BURKE P, BESPLUG J, et al. Methylation changes in muscle and liver tissues of male and female mice exposed to acute and chronic low-dose X-ray-irradiation[J]. Mutat Res, 2004, 548(1-2): 75-84.
    [93] RAICHE J, RODRIGUEZ-JUAREZ R, POGRIBNY I, et al. Sex- and tissue-specific expression of maintenance and de novo DNA methyltransferases upon low dose X-irradiation in mice[J]. Biochem Biophys Res Commun, 2004, 325(1): 39-47.
    [94] POGRIBNY I, KOTURBASH I, TRYNDYAK V, et al. Fractionated low-dose radiation exposure leads to accumulation of DNA damage and profound alterations in DNA and histone methylation in the murine thymus[J]. Mol Cancer Res, 2005, 3(10): 553-561.
    [95] SWAFFORD DS, MIDDLETON SK, PALMISANO WA, et al. Frequent aberrant methylation of p16INK4a in primary rat lung tumors[J]. Mol Cell Biol, 1997, 17(3): 1366-1374.
    [96] MORGAN WF. Non-targeted and delayed effects of exposure to ionizing radiation: II. Radiation-induced genomic instability and bystander effects in vivo, clastogenic factors and transgenerational effects[J]. Radiat Res, 2003, 159(5): 581-596.
    [97] HAMASAKI K, IMAI K, HAYASHI T. Radiation sensitivity and genomic instability in the hematopoietic system: Frequencies of micronucleated reticulocytes in whole-body X-irradiated BALB/c and C57BL/6 mice[J]. Cancer Sci, 2007, 98(12): 1840-1844.
    [98] KOTURBASH I, BAKER M, LOREE J, et al. Epigenetic dysregulation underlies radiation-induced transgenerational genome instability in vivo[J]. Int J Radiat Oncol Biol Phys, 2006, 66(2): 327-330.
    [99] SHARPLESS NE, BARDEESY N, LEE KH, et al. Loss of p16Ink4a with retention of p19Arf predisposes mice to tumorigenesis[J]. Nature, 2001, 413(6851): 86-91.
    [100] VIALLARD JF, LACOMBE F, BELLOC F, et al. Molecular mechanismscontrolling the cell cycle: Fundamental aspects and implications for oncology[J]. Cancer Radiother, 2001, 5(2): 109-129.
    [101]鞠桂芝,王晓梅,付士波,等.电离辐射对p16基因转录及蛋白表达的影响[J].中华放射医学与防护杂志, 2003, 23(1): 4-6.
    [102] BIANCHI T, RUFER N, MACDONALD HR, et al. The tumor suppressor p16Ink4a regulates T lymphocyte survival[J]. Oncogene, 2006, 25(29): 4110-4115.
    [103] WAKABAYASHI Y, WATANABE H, INOUE J, et al. Bcl11b is required for differentiation and survival of alphabeta T lymphocytes[J]. Nat Immunol, 2003, 4(6): 533-539.
    [104] ALBU DI, FENG D, BHATTACHARYA D, et al. BCL11B is required for positive selection and survival of double-positive thymocytes[J]. J Exp Med, 2007, 204(12): 3003-3015.
    [105] YOSHIKAI Y, SATO T, MORITA S, et al. Effect of Bcl11b genotypes and gamma-radiation on the development of mouse thymic lymphomas[J]. Biochem Biophys Res Commun, 2008, 373(2): 282-285.
    [106] KAMIMURA K, OHI H, KUBOTA T, et al. Haploinsufficiency of Bcl11b for suppression of lymphomagenesis and thymocyte development[J]. Biochem Biophys Res Commun, 2007, 355(2): 538-542.
    [107] WAKABAYASHI Y, INOUE J, TAKAHASHI Y, et al. Homozygous deletions and point mutations of the Rit1/Bcl11b gene in gamma-ray induced mouse thymic lymphomas[J]. Biochem Biophys Res Commun, 2003, 301(2): 598-603.
    [108] SAKATA J, INOUE J, OHI H, et al. Involvement of V(D)J recombinase in the generation of intragenic deletions in the Rit1/Bcl11b tumor suppressor gene in gamma-rayinduced thymic lymphomas and in normal thymus of the mouse[J]. Carcinogenesis, 2004, 25(6): 1069-1075.
    [109] THOMAS RM, CHEN C, CHUNDER N, et al. Ikaros silences T-bet expression and interferon-gamma production during T helper 2differentiation[J]. J Biol Chem, 2010, 285(4): 2545-2553.
    [110] QUIRION MR, GREGORY GD, UMETSU SE, et al. Cutting edge: Ikaros is a regulator of Th2 cell differentiation[J]. J Immunol, 2009, 182(2): 741-745.
    [111] UMETSU SE, WINANDY S. Ikaros is a regulator of Il10 expression in CD4+ T cells[J]. J Immunol, 2009, 183(9): 5518-5525.
    [112] KATHREIN KL, LORENZ R, INNES AM, et al. Ikaros induces quiescence and T-cell differentiation in a leukemia cell line[J]. Mol Cell Biol, 2005, 25(5): 1645-1654.
    [113] OKANO H, SAITO Y, MIYAZAWA T, et al. Homozygous deletions and point mutations of the Ikaros gene in gamma-ray-induced mouse thymic lymphomas[J]. Oncogene, 1999, 18(48): 6677-6683.
    [114] SHIMADA Y, NISHIMURA M, KAKINUMA S, et al. Radiation-associated loss of heterozygosity at the Znfn1a1 (Ikaros) locus on chromosome 11 in murine thymic lymphomas[J]. Radiat Res, 2000, 154(3): 293-300.

© 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