三氧化二砷对MDA-MB-231细胞系ER-α基因去甲基化的实验研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
ER-α基因定位于染色体6q24~27上,其表达的ER-α蛋白常作为一个重要分子标志物用于评估乳腺癌治疗的激素依赖性和预测内分泌治疗效果;ER-α基因失表达(ER-α阴性)的乳腺癌患者对内分泌治疗不敏感,预后差。
     据文献报道ER-α基因表达沉默与基因重排、突变、缺失等无关,可能与其启动子区CpG岛异常甲基化有关。因此,针对失表达ER-α基因的去甲基化治疗成为雌激素受体阴性乳腺癌治疗的一条新思路。本课题为此进行该方向的实验研究。
     本实验选用国内最早报道原用于诱导治疗的化学物质(As_2O_3)作为去甲基实验药物,开展对ER-α基因高甲基化的乳腺癌细胞系MDA-MB-231细胞去甲基作用机制的实验研究,探讨As_2O_3对乳腺癌细胞MDA-MB-231的去甲基化效果及其同基因表达的关系,旨在研究As_2O_3与乳腺癌细胞DNA甲基化的关系及其去甲基化治疗的可能机制,并为As_2O_3作为去甲基化药物提供实验依据。
     材料与方法:
     (1)选用雌激素受体(ER)阴性人乳腺癌细胞株MDA-MB-231常规培养,将对数生长期的MDA-MB-231细胞调整细胞浓度为5×10~5/mL,换含As_2O_3的L-15培养液,至As_2O_3终浓度分别为0.5μmol/L、2.0μmol/L、4.0μmol/L,连续培养72小时。以不加药同期培养的MDA-MB-231细胞作为阴性对照。以同期培养在RPMI-1640环境中的雌激素受体(ER)阳性人乳腺癌细胞株MCF-7为阳性对照。
     (2)采用REP-PCR方法检测ER-α基因外显子Ⅰ(含过度甲基化的CpG岛)的表达。
     (3)采用Western Blot技术检测经不同浓度As_2O_3处理后的MDA-MB-231细胞ER-α蛋白的表达情况。
     (4)MTT技术检测不同浓度As_2O_3处理后的MDA-MB-231细胞,再经他莫昔芬处理,各组细胞增殖抑制率的差异。
     (5)实验数据经SPSS11.0统计软件处理,采用单因素方差分析,以α=0.05为差异显著性标准。
     结果:
     (1)REP-PCR法检测未经As_2O_3处理的MDA-MB-231细胞在经NotI酶过量消化前后,均可扩出清晰的外显子Ⅰ片段,提示ER-α基因甲基化阳性;一定浓度(2.0μmol/L和4.0μmol/L)的As_2O_3作用72小时后,再次酶切后扩增未见目的条带,提示ER-α基因甲基化阴性;以MCF-7细胞为阳性对照,经NotI酶过量消化后也未扩增出ER-α基因外显子Ⅰ片段。
     (2)经Western Blot检测未经As_2O_3处理的MDA-MB-231细胞未显示ER-α蛋白带,0.5μmol/L As_2O_3处理的MDA-MB-231细胞也未显示ER-α蛋白带,但经2.0μmol/L和4.0μmol/L As_2O_3处理的MDA-MB-231细胞可见ER-α蛋白带。
     (3)MTT法检测他莫昔芬作用于三氧化二砷处理过的MDA-MB-231细胞后,细胞增殖明显受到抑制。浓度2.0μmol/L左右的As_2O_3应当是MDA-MB-231细胞株去甲基化体外实验的较适合剂量,虽然As_2O_3处理后MA-MB-231细胞对TAM的敏感性不及MCF-7细胞,但明显优于未经处理的MA-MB-231细胞,提示TAM对As_2O_3处理过的MDA-MB-231细胞有明显的细胞毒作用。
     结论:
     1.一定浓度的As_2O_3能够使MDA-MB-231细胞系ER-α基因中的CpG岛发生去甲基化。
     2.一定浓度的As_2O_3可诱导MDA-MB-231细胞系ER-α蛋白表达。
     3.As_2O_3诱导MDA-MB-231细胞表达的ER-α是有功能的。
     4.As_2O_3通过去甲基化作用使雌激素受体阴性的乳腺癌细胞表达有功能的雌激素受体,从而为雌激素受体阴性的乳腺癌患者接受内分泌治疗提供理论依据。
Objective: ER-αgene is located on the chromosome 6q24~27.The proteinum expressed by ER-αgene is usually an important molecule marker to evaluate hormonal dependence and predict the effect of endocrine therapy on breast cancer. The patients of breast cancer (ER-αnegative) are not sensitive to endocrine therapy and more malignant poor prognosis.
     Current reasearches report the mechanism of ER-αgene expression silencing is not due to generearrangement, mutation , absence and so on ,but probably associated with the CpG island methylation in the core Promotor of ER-αgene. Thus ,demethylation therapy aimed at ER-αgene becomes a new therapeutic strategy for breast cancer.
     Now we choose arsenic trioxide which used as inductive treatment drug as experiment drug of demethylation. The purpose is to investigate the demethylating effects on ER-αgene in MDA-MB-231 cell and the relations between demethylation and gene expression, to explore the correlation between arsenic trioxide and DNA methylaion and clarify the possible mechanisms of demethylating treatment, also to provide experimental basis for arsenic trioxide as methylation inhibitor.
     Methods and materials:
     (1) Human breast cell line MDA-MB-231 were growing at 37℃and 5% CO_2 MDA-MB-231 were plated at densities of 5×10~5 cells/100-mm dish and were treated with L-15 containing As_2O_3 0.5μmol/L、2.0μmol/L、4.0μmol/L respectively. The media containing the drug was maintained during the 3 days of treatment. MDA-MB-231 without being treated with containing As_2O_3 were taken as negative control . Human breast cell line MCF-7((ER negative) were taken as positive control.
     (2) The MDA-MB-231 cell line can express or not exon I fragment of the estrogen receptor gene (containing the CPG island of hypermethylation) which are determined by REP-PCR technique.
     (3) Western Blot was performed for ER-αprotein after the MDA-MB-231 cell line in which the estrogen receptor gene is silenced due to hypermethylation was treated with different concentration of As_2O_3
     (4) The tamoxifen induced growth inhibition of MDA-MB-231 cell after was treated with different concentration of As_2O_3 by MTT method.
     (5) Experimental data are disposed by SPSS11.0 statistics soft ware,deploying one-way analysis of variance, significance of difference standard (α=0.05).
     Results:
     (1) ER-αgenes of MDA-MB-231 cells of no drugs can clearly amplificate exon I fragment before or behind excessive digestion with restricted enzyme NotI. It hints ER-αgenes containing the hypermethylation part; But ER-αgenes can't amplificate exon I fragment treated with 2.0μmol/L and 4.0umol/L As_2O_3 for three days and excessive digested with restricted enzyme Not I, it hints negative for hypermethylation; Taking MCF-7 cells as positive control, it can't amplificate exon I fragment too behind excessive digestion with restricted enzyme NotI.
     (2) Detectable expression of ER-αprotein was found in the MDA-MB-231 cells treated with 0.5umol/L As_2O_3 determined with Western blot technique But detectable expression of ER-αprotein was found in the MDA-MB-231 cells treated with 1 .0μmol/L、2.0μmol/L and 4.0μmol/L As_2O_3 determined with Western blot technique;
     (3) Tamoxifen could inhibit the growth of the MDA-MB-231 cells treated with As_2O_3 efficiently through MTT methods, arsenic trioxide(2.0μmol/L) should be the better proper dose in vitro experiment of demethylation of the MDA-MB-231 cells,although they are less sensitive to TAM than MCF-7 cells and obviouslay better than the MDA-MB-231 cells without being disposed by arsenic trioxide. so TAM has obviously toxic action of the MDA-MB-231 cells treated by arsenic trioxide.
     Conclusion:
     (1) At definite concentration, As_2O_3 could make the CPG island of ER-αgene shows demethylation in human breast cell line MDA-MB-231 .
     (2) At definite Concentration, As_2O_3 could induce ER-αprotein re-expression in human breast cell line MDA-MB-231 through DNA demethylation.
     (3) The re-expressed genes are functional in human breast cell line MDA-MB-231 through DNA demethylation.
     (4) As_2O_3 could induce ER-αfunctional gene re-expression in human breast cell line MDA-MB-231 through DNA demethylation. Thus provide an evidence in theory for breast cancer patients (ER negative) to take endocrine therapy.
引文
1. Mcinerney EM, Tsai M J, O' Malley BW, et al. Analysis of estrogen receptor transcriptional enhancement by a nuclear receptor coactivator. [J] Proc Natl Acad Sci USA, 1996, 93: 10069-10073.
    2. Bird A P, Taggart M, Frommer M et al. Cell, 1985; 40:91.
    3. Bird A. Trends Genet, 1987;3: 342.
    4. Larsen F, Gundersen G, Lopez R et al. Genomic, 1992:13: 1095.
    5. RazinA, Cedar H DNA methrlationand embryogenesis.EXS, 1993,164:343-57.
    6. Heby O, Persson L, Smeth SS, Polyamines, DNA methylation and cell differentiation. [J]Adv Exp Med Bio,1988,250:291299.
    7. Roger P Sahla ME, Makela S, et al. Decreased expression of estrogen receptor beta protein in proliferative preinvasive mammary tumors. [J]Cancer Res, 2001, 61 (6) 2537-2541.
    8. Yaich L, Dupont WD, Cavener DR, et al, Analysis of the PvuⅡrestriction fragment-length polymorphism and exon structure of the estrogen receptor gene in breast cancer and eripheral blood. [J]Cancer Res, 1992,52(1):77-83.
    9. Kautiainen TL, Jones PA: DNA methyltransferase levels in tumorigenic and nontumorigenic cells in culture. [J]Biol Chem, 1986;261:1594.
    10. Issa JP, Vertino PM, Wu J, Sazawal S, Celano P, Nelkin BD, Hamilton SR,Baylin SB: Increased cytosine DNA-methyltransferase activity duringcolon cancer progression. [J]Natl Cancer Inst, 1993;85:1235.
    11. Laird PW, Jackson-Grusby L, Fazeli A, Dickinson SL, Jung WE, Li E,Weinberg RA,JaenischR:Suppression of intestinal neoplasia by DNA hypomethy-Lation. [J] Cell,1995;81:197
    12. Baylin SB, Herman JG, Graff JR, Vertino PM, Issa JP: Alterations in DNA methylation: A fundamental aspect of neoplasia.[J]Adv Cancer Res,1998;72:141.
    
    13. Laird PW, Jaenisch R: The role of DNA methylation in cancer genetic and epigenetics . [J]Annu Rev Genet, 1996;30:441.
    
    14. Coulondre C, Miller JH, Farabaugh PJ, Gilbert W: Molecular basis of base substitution hotspots in Escherichia coli. [J]Nature,1978;274:775.
    
    15. Gonzalgo ML, Liang G, Spruck CHr, Zingg JM, Rideout WMr, Jones PA:Identification and characterization of differentially methylated regions of genomic DNA by methylation-sensitive arbitrarily primed PCR.[J]CancerRes,1997;57:594.
    
    16. Bender CM, Zingg JM, Jones PA: DNA methylation as a target for drug design.[J]Pharmaceutical Res,1998;15:175.
    
    17. Greenblatt MS, Bennett WP, Hollstein M, Harris CC: Mutations in the p53 tumor suppressor gene: Clues to cancer etiology and molecular pathogenesis. [J] Cancer Res,1994;54:4855.
    
    18. Magewu AN, Jones PA: Ubiquitous and tenacious methylation of the CpG site in codon 248 of the p53 gene may explain its frequent appearance as a mutational hot spot in human cancer. [J]Mol Cell Biol, 1994;14:4225.
    
    19. Gama-Sosa MA, Slagel VA, Trewyn RW, Oxenhandler R, Kuo KC, Gehrke CW,Ehrlich M: The 5-methylcytosine content of DNA from human tumors. [J]Nucleic Acids Res, 1983; 11:6883.
    
    20. Christman JK, Sheikhnejad G, Dizik M, Abileah S, Wainfan E: Reversibility of changes in nucleic acid methylation and gene expression induced in rat liver by severe dietary methyl deficiency. [J]Carcinogenesis,1993;14:551.
    21. Pogdbny IP, Miller BJ, James SJ: Alterations in hepatic p53 gene methylation patterns during tumor progression with folate/methyl deficiency in the rat. [J] Cancer Lett, 1997; 115:31.
    22. Hanada M, Delia D, Aiello A, Stadtmauer E, Reed JC: bcl-2, gene hypomethylation and high-level expression in B-cell chronic lymphocytic leukemia. [J]Blood, 1993;82:1820.
    23. Feinberg AP, Vogelstein B: Hypomethylation distinguishes genes of some human cancers from their normal counterparts. [J]Nature, 1983;301:89.
    24. Ohtani-Fujita N, Fujita T, Aoike A, Osifchin NE, Robbins PD, Sakai T: CpG methylation inactivates the promoter activity of the humanretinoblastoma tumor-suppressor gene. [J]Oncogene, 1993;8:1063
    25. Herman JG, Latif F, Weng Y, Lerman MI, Zbar B, Liu S, Samid D, Duan DS, Gnarra JR, Linehan WM, Baylin SB: Silencing of the VHL tumor-suppressor gene by DNA methylation in renal carcinoma.[J]Proc Natl Acad Sci USA, 1994;91:9700.
    26. Ottariano YL,Issa JP, Parl FF, et al. Methylation of the estrogen receptor gene CpG island marks loss of estrogen receptor expression in human breast cancer. [J]Cancer Res, 1994,54(10):2552-2556.
    27.唐建林,高东宸.用改良LSAB法检测乳腺癌雌激素受体和孕激素受体.[J]中华病理学杂志 1994,23(2):120
    28. Shoker BS, Jarvis C, Sibson DR, et al. Oestrogen receptor expression in the normal and pre-cancerous breast.[J]Pathol, 1999, 188(3):23
    29. Sasano H, Suzuki T, Matsuzaki Y, et al . Messenger ribonucleic acid in situ hybridization analysis of estrogen receptors alpha and beta in human breast carcinoma.[J]Gin Endocxinol Metab, 1999, 84(2):781-785
    30. Knowlden JM, Gee JMN, Robertson JFR, et al. A possible divergent role for the oestrogen receptor alpha and beta subtypes in clinical breast cancer. [J] Int J Cancer, 2000, 89:209-2124.
    31. Speirs V, Malone C, Walton DS,et al. Increased expression of estrogen receptor beta mRNA in tamoxifen-resistant breast cancer patients.[J]Cancer Res, 1999, 59(21):5421.
    32. Speirs V, Parkes AT, Kerin MJ, et al. Coexpression of estrogen receptor alpha and beta: poor prognostic factors in human breast cancer.[J]Cancer Res, 1999, 59(3):525-528.
    33. Leygue E, Dotzlaw H, Watson PH, et al. Expression of the steroid receptor RNA activator in human breast tumors.[J]Cancer Res, 1999, 59(6):1175-1179.
    34. Katsuda SI, Yoshida M, Watanabe T, et al. Estrogen receptor mRNA in uteri of normal estrous cycling and ovariectomized rats by in situ hybridization.[J]Int J Cancer, 2000, 89:92.
    35. Umekita Y, Yoshida A, Sagara Y, Yoshida H. et al. Lipid-Secreting Carcinoma of the Breast: A Case Report and Review of the Literature.[J]Jpn J Canex Res, 1998, 89(1):27-32.
    36. Iwase H, Omoto Y, Iwata H, et al. Clinical Significance of Estrogen Receptor Ain Breast Cancer. [J]Br J Cancer, 1999, 80(12): 1982-1986.
    37. Pasqualine Balmelli-Gallacchi, Fabrice Schoumacher, Jia Wei Liu, et al . A yeast-based bioassay for the determination of functional and non-functional estrogen receptors. [J] Int J Cancer, 1998,79(1):44-48.
    38. Van-Dijk MA, Hart AA, Van't VLJ. Differences in estrogen receptor alpha variant messenger RNAs between normal human breast tissue and primary breast carcinomas.[J]Cancer Res, 2000, 60(3):530-533.
    39.王夫景,李刚.杨雏良.乳腺P53与雌孕激素受体的表达及临床意义.[J]中国普通外科杂志 2002,11(6):273-274.
    40. Vahter M. Mechanisms of arsenic biotransformation. [J]Toxicology. 2002, 27(181-182):211-217.
    41. Gonzalez-FraileMI,Garcia-Sanz R, Mateos MV, et al. Methylenetetrahydrofolate reductase genotype does not play a role in multiple myeloma pathogenesis. [J]Br J Haematol. 2002, 117(4): 890-892.
    42. James SJ, Melnyk S, Pogribna M, et al. Elevation in S-adenosylhomocysteine and DNA hypomethylation: potential epigenetic mechanism for homocysteine-related pathology.[J] Nutr, 2002,132(8 Suppl):2361-2366.
    43. Xie S, Wang Z, OkanoM, et al. Cloning, expression and chromosome locations of the human DNMT3 gene family.[J]Gene, 1999, 236(1):87-95.
    44.黎金庆,李渊,石永进,等.三氧化二砷诱导U266细胞p16基因重新表达.[J]癌症,2004,23(6):626-630.
    45.杨红斌,付京,张盈华,等.乳腺癌组织中孕激素受体与雌激素受体DNA结合功能关系的研究.[J]中国肿瘤临床,1999,26(10):743-746.
    46.齐元富.乳腺癌内分泌治疗的研究进展.[J]国外医学/肿瘤学分册,1998,25(1):32-34.
    47.王冰.三氧化二砷体外诱导雌激素受体表达的初步研究.[J]肿瘤基础与临床,2006,(3):183—184.
    48.杨骅,郁琳琳.三氧化二砷诱导乳癌细胞的凋亡.[J]中国肿瘤生物治疗杂志,1997,(4):245.
    1.张鹏等.中华血液学杂志,1996;17(2):58—60
    2.刘佳等.中国地方病学杂志,1998;17(5):281—283
    3. Wang Z G et al. Blood, 1998;92(5): 1497—1504
    4.李勇等.中国地方病学杂志,1996;15(2):93—96
    5. Labrecque J et al. Blood, 1998; 92 (2): 607—615
    6. Chen G Q et al. Blood, 1996;88(3): 1052—1061
    7. Zhang W et al. Leukemia, 1998; 12(9): 1383—1391
    8. Brown D et al. Proc Nati Acad Sci USA, 1997; 94 (1): 2551—2556
    9. Lallemand B et al. J Exp Med, 1999; 189 (7): 1043—1052
    10. Scott N et al, Chem Res Toxicol, 1996; 6 (1): 102—107
    11. Kitamurs K et al. Int J Hematol, 1997; 65 (1): 179—184
    12. Cantoni O et al. Mlltat Res, 1994; 24 (3): 300—311
    13. Lo J F et al. Biochem J,1992;288(2): 977—982
    14. Dai J et al. Blood, 1999; 93 (1): 268—277
    15. Y L Kwong, D Todd. Delicious posion: Arsenic trioxide for the treatment of leukemia.[J]Blood, 1997,89 (9): 3487.
    16. Mer J. Ancient rcmedy performes now tricks.[J]Science, 1996,273 (2): 578.
    17.黄世林,郭爱霞,向阳,等.复方青黛片为主治疗急性早幼粒细胞白血病的临床研究.[J]中华血液学杂志,1995,16(1):26.
    18. Lu D P, Qin J Y, Chen S S, et al. Effect of treatment of AML -M3 (APL) and their remission maintenance with realgar: a pilot clinical and laboratory study on 38 patients.[J]Blood, 1997,90 (Suppl): 1849.
    19.唐由君.传统抗癌中草药配合中药复方治疗白血病的研究.[J]中国中西医结合杂志,1998,18(12):623.
    20.杨新中.中药复方白血宁1号治疗白血病的疗效观察.[J]临床血液学杂志,1999.12:20.
    21. Soignet SL, RiviR, KonigA, et al. Melarsoprol, an organic arsenical, in the treatment of refractory leukemia: Iaboratory and clinical result.[J]Blood, 1997,90 (Suppl): 2258a.
    22. Akao Y, MizoguchilH, KojimaS, et al. Arsenic-induced apoptosis in B-cell Leukaemic cell Lines in vitro: Activation ofcaspases and down-regulation of Bcl-2 protein.[J]BrJHaematol, 1998,102 (4): 1005-1009.
    23.张日,支雅军,朱子玲,等.三氧化二砷联合木黄酮抑制CML细胞增殖的研究.[J]江苏医药,2000,26(10):763-765.
    24.傅卫军,候健,王东星,等.氧化砷诱导骨髓瘤细胞株(KM3)凋亡的初步研究.[J]中华血液学杂志,1998,19(11):591-592.
    25.伍钢,周云峰,陈同辛,等.三氧化二砷诱导神经母细胞瘤细胞凋亡机理的研究.[J]中华儿科杂志,1998,36(5):265-267.
    26.蔡洪培,邓志华,李石,等.As_2O_3诱导人胃癌细胞凋亡的研究.[J]第二军医大学学报,1999,20(9):636-639.
    27.谭立军,史桂英,石学耕,等.三氧化二砷对人食管癌细胞株凋亡的诱导作用.[J]中国癌症杂志,1999,9(2):85-87.
    28.秦叔达,钱军,何泽明,等.三氧化二砷注射液治疗晚期胆囊癌.[J]临床肿瘤学杂志,2000,5(4):286-287.
    29.朱安龙,刘连新,朴大勋,等.应用三氧化二砷连续区域化疗治疗肝癌.[J]中华肝胆外科杂志,2003,9(4):205-206.
    30.邓友平,林晨,张雪艳,等.三氧化二砷诱导人肺腺癌GLC-82细胞凋亡及分子机制的研究.[J]癌症,1999,18(5):545-549.
    31.邓友平,林晨,梁萧,等.三氧化二砷诱导人宫颈癌Hela细胞凋亡及Bcl-2保护作用的机制研究.[J]中国科学,1999,29(3):33.
    32.杨骅、郁琳琳,王仙平,等.三氧化二砷注射剂诱导乳癌细胞凋亡.[J]中国肿瘤生物治疗杂志,1997,4(3):245.
    33. Koshiuka K, ElstnerE, WilliamsonE, et al. Novel therapeutic aPproach: organic arsenical melarsoprol alone or with all-trans-retinoic acid markedly inhibit growth of human breast and prostatec cancer ceIls in vitro and in vivol.[J]Br J Cancer, 2000,82 (2): 452-458.
    34. Zhang TD, Chen GQ, Wang 2 Q et al. Arsenic trioxide, a therapeutic agent for APL.[J]Oncogene, 2001,20 (49): 7146-7153.
    35. ZhuQ, Zhang J W, Zhu H Q,et al. Synergio effects of arsonic trioxidc and camp during acutc promyelocytic leukemia cell maturation subtends a novel signaling cross-talk.[J]Blood, 2002,99 (3): 1014-1022.
    36. Mackenzie KL, FrancoS, May C. Mass cultured human fibroblasts over expressing hTERT encounter a growth crisis followingan extended period ofproliferation.[J]Exp Cell Res, 2000,259 (2): 336-350.
    37.郝云良,徐从高.三氧化二砷对NB4细胞株端粒酶活性的影响.[J]山东大学学报(医学版),2002,40(6):565-567.
    38. Akao Y, YamadaH, Nakagawa Y. Arsenic-induced apoptosis in malignant cells in vitro.[J]Leuk Lymphoma, 2000,37: 53-63.
    39.刘延芳,江滨,陆道培.硫化砷诱导NB4细胞凋亡及细胞周期阻滞的研究.[J]中华血液学杂志,2000,21:647-648.
    40. Huang SC , Lee T C. Arsenic inhibits mitotic division and perturbs spinale dynarnias in Helas 3 cells.[J]Carcinogenesis, 1998,19: 889-896.
    41. Park WH, Scol J q Kim ES, et al. Arsenic trioxide mediated growth inhibition in MC/CAR myeloma cells via cell cycle arrest in association with induction ofcyclin-dependentkinaseinhibitorp21 andapoptosis. [J]CancerRes,2000,60 (11): 3065-3071.
    42. Seol J G, Park WH, Kim ES, et al. Effect of arsenic trioxide on cell cycle arrest in head and neck cancer cell line PCI-I.[J]Biochemical and BiophysicalResoareh Communications, 1999,265: 400-04.
    43. lshitsukaK, HanadaS, UozumiK, et al. Arsenic trioxide and the growth of human T-ccll leukemia virus type Ⅰ infected T-cell lines. [J] Leuk Lymphoma, 2000,37: 649-655.
    44. COHEN JJ. Overview: mechanisms of apoptosis.[J] Immunol Today, 1993 , 14: 126.
    45. FISHER D E. Apoptosis in cancer therapy:crossing the threshold.[J]Cell,1994,7 8:539.
    46.闻勤生,罗俊卿,焦成松.三氧化二砷诱导肝癌细胞HepG2凋亡作用.[J]第三军医大学学报,2001,23(8):913-914.
    47.娄青林,冷静,彭韬,等.三氧化二砷体外诱导人胃癌SGC-7901细胞凋亡的研究.[J]南京医科大学学报,2001,21(4):299-302.
    48. LOOK A T. Aresnic and a ppotosis in the treatment of acute promyelocytic leukemia. [J] J Nati Cancer Inst, 1998,90 (2): 86.
    49.马丹,葛海燕.三氧化二砷对人结肠癌细胞增殖及凋亡的影响.[J]重庆医学,2003,32(1):55-56.
    50.陈勇,王天铎,陈明,等.三氧化二砷诱导喉癌Hep-2耐药细胞凋亡的研究.[J]山东医科大学基础医学院学报,2002,16(3):139-141.
    51.陈扬超,张月飞,周克元.三氧化二砷诱导鼻咽癌CNE-2Z细胞的凋亡及其作用机制的初步研究.[J]癌症,2001,20(10):1061-1064.
    52.徐洪雨,高媛媛,武俏丽,等.三氧化二砷抑制人肝癌细胞株增殖和诱导凋亡作用.[J]世界华人杂志,2000,8(11):1233-1237.
    53. CLTVAL Z N, MILLIMEN C L, KORSMEYER S J. Bcl-2 heterodimenzes in vivo with a conserved homolog, Bax, that accelerates programmed cell death. [J] Cell,1993,74: 609-617.
    54.王岩,宋云飞,顾冰,等.三氧化二砷诱导膀肤癌BIU-87细胞凋亡作用的研究[J].哈尔滨医科大学学报,2002,36(4):287-289.
    55.涂水平,江石湖,潭继宏,等.三氧化二砷诱导胃癌细胞凋亡及相关基因表达的影响.[J]肿瘤,2001,21(5):327-330.
    56.许勤,汤静燕,伍刚,等.三氧化二砷和高三尖杉诱导神经母细胞瘤细胞凋亡与N-myc表达的关系.[J]癌症,2001,20(3):239-244.
    57. LUTZ W, FULDA S,JEREMIAS I, et al.MycN and IFN gamma cooperate in apoptosis of human neuroblastoma cells.[J]Oncgene, 1998,17 (3): 339-346.
    58. SQUIBB KS , FOLER B A. The toxicity of arsenic and compounds [A]. FOLWER B A, eds Biological and Enviomental effects of Arsenic. 3 rd ed [C] Amsterdam : Elsevier, 1983.236-269.
    59.王展翔,廖军鲜,许勇刚,等.抗氧化剂对三氧化二砷诱导HL-60细胞凋亡作用的影响.[J]白血病,2000,9(3):153-155.
    60.周宇红,黄晓,蔡循,等.三氧化二砷诱导骨髓癌细胞凋亡机制研究.[J]中华肿瘤杂志,2001,23(3):181-183.
    61.高飞,易静,史桂英,等.食管癌、胃癌细胞对氧化砷促凋亡的敏感性与固有活性氧水平有关.[J]中华消化杂志,2002,22(2):70-72.
    62.邢茂,张恩娟三氧化二砷诱导人胃腺癌细胞SCG-7901凋亡机制的研究.[J]中国药房,2001,12(6):333-334.
    63.黄守国,孔北华,马玉燕,等.三氧化二砷诱导人卵巢癌细胞凋亡机制的研究[J].肿瘤防治杂志,2002,9(2):154-156.
    64. JONG WOOK PARK, YUN JUNG CHCI,MIN AH JANG,, et al. Arsenic trioxide induces G2/M growth arrest apoptosis after caspase-3 activation and Blc-2 phosphorylation in pormoncytie U937 cells.[J] Bioc be mical and Biophysical Research Communications,2001,286:726-734.
    65. JAZA N. Essential role of the mitochondrial apoptosis-inducing factor in programmed cell death .[J]Nature, 2001,410: 549.
    66. HOLT S E, GLINSKY V V, IVANOVA A B, et al. Resistense to apoptosis in human cells conferred by telomerase function and tilomere stabilit. [J]J Biol Chem, 1999,274: 7264-7271.
    67.樊华,俞军.三氧化二砷对鼠移植性肝癌端粒酶活性的影响.[J]中国临床医学,2002,9(2):121-123.
    68.廖世兵,金宝翠,刘悦芳,等.三氧化二砷诱导NB4细胞凋亡的端粒酶活性研究.[J]中华血液学杂志,2002,23(1):40-42.
    69.杨仕明,房殿春,罗元辉,等.As2O3对胃癌细胞周期及端粒酶活性的影响.[J]第三军医大学学报,2001,23(9):1030-1033.
    70.钱军,秦叔逵,何泽明,等.三氧化二砷注射液治疗中晚期原发性肝胆癌的临床研究.[J]中华肿瘤杂志,2001,23(6):487.
    71.项颖,黄凤鸣.三氧化二砷注射液治疗中晚期原发性肝癌16例报告.[J]临 床肿瘤学杂志,2003,8(1).
    72.秦叔逵.三氧化二砷治疗原发性肝胆癌的研究进展.[J]肿瘤学杂志,2001,7(2):115.
    73.杨义明,张水兰,宋华勇.亚砷酸联合干扰素治疗中晚期原发性肝癌的临床研究.[J]临床肿瘤学杂志,2002,10(5):355.
    74.朱安龙,刘连新,朴大勋,等.应用三氧化二砷连续区域化疗治疗肝癌.[J]中华肝胆外科杂志,2003,9(4):205.
    75.邵海波,徐克,苏洪英,等.亚砷酸-碘油乳剂介入治疗原发性肝癌的临床观察[J].肿瘤防治杂志,2004,11(6):638.
    76.于志坚,蔡炜,袁洪新,等.诱导肿瘤细胞分化凋亡药物在肝胆肿瘤区域性联合化疗中应用.[J]南通大学学报(医学版),2005,25(4):261.
    77.戚晓军,赵海东,高文斌,等.三氧化二砷瘤内注射联合肝动脉化疗栓塞治疗肝癌.[J]肿瘤学杂志,2003,9(3):147.
    78. Chun YJ, Park IC, Park MJ, et al. Enhancement ofradiationre sponse in human cervical cancer cells in vitro and in vivo by arsenic trioxide (As2O3). [J] FEBs Lett, 2002,519 (13): 195.
    79. Lew YS, KolozsVaryA, Brown SL, et alsynergistic interaction With arsenic trioxide and fractionated radiation in locally ad-vanced murine tumor.[J]cancer Res, 2002,62: 4202.
    80. Monzen H, Griffin RJ, Williams BW, et alstudy ofarsenictriox ideinduced vascular shut down and enhancement with radiationin in solid tumor.[J]Radiat Med, 2004,22 (4): 205.
    81. UsluR, Sanli UA, SezginC, et alArsenic trioxide mediatedcyto-toxicity and apoptosis in prostate and ovarian carcinoma cell lines.[J]Clin Cancer, Res, 2000,6:4957.
    82.黄守国,孔北华,马玉燕,等.二氧化一砷诱导人卵巢癌耐药细胞系3AO/cDDP细胞凋亡及机制的研究.[J]中华医学杂志,2002,12(1):13.
    83. Du YH, Ho PCArsenic compounds induce cytotoxicity and apoptosisin cisplatin-sensitive and-resistant gynccological cancer cell lines.[J]Cancer Chemother Pharmacol,2001,47 (6): 481.
    84. BajG, AmulfoA, Deaflio, et al. Arsenic trioxide and breast cancer: Analysis of the apoptotic, differentiative and immunomodulatory effects.[J]Breast Cancer Res Treat, 2002,73: 6173.
    85. Karasulu HY, KarabulutB, KantarciG, et al. Preparation of arsenic trioxide loaded microemulsion and its enhanced cytotoxicity on MCF-7 breast carcinoma cell line.[J]Drug Deliv, 2004, 11: 345-350.
    86. Chow SK, Chan JY, Fung KP. lnhibitionof cell proliferation and the actlon mechanisms of arsenictrioxide (As_2O_3) onhumanbreast cancer cells.[J]Cell Biochem,2004, 93: 173-187.
    87. LI X, Ding X, Adrian TE. Arsenic trioxide causes redistribution of cell cycle, caspase activation, and GADD expression in human colonic,breast,and pancreatic cancer cells.[J]Cancer Invest, 2004,22: 389-400.
    88.魏玲,王兴武,左文述,等.三氧化二砷抑制人乳腺癌细胞生长及其作用机制的初步研究.[J]中华医学杂志,2005,55:1209-1213.
    89.陈鑫,吴诚义.As_2O_3对人乳腺癌移植瘤模型增殖凋亡的影响.[J]第四军医大学学报,2004,25:698-701.
    90.马银斌,吴诚义,陈鑫.Survinin在人乳腺癌裸鼠移植瘤中的表达及As_2O_3处理后表达变化.[J]第三军医大学学报,2004,26:1423-1425.
    91.陈鑫,吴诚义.砷对人乳腺癌移植瘤血管内皮细胞的影响.[J]中华普通外科杂志,2004,19:252—253.
    92. Vahter M. Mechanisms of arsenic biotransformation. [J]Toxicology. 2002, 27(181-182):211-217.
    93. Gonzalez-Fraile MI, Garcia-Sanz R, Mateos MV, et al. Methylenetetrahydrofolate reductase genotype does not play a role in multiple myeloma pathogenesis.[J]Br J Haematol. 2002, 117(4): 890-892.
    94. James SJ, Melnyk S, Pogribna M, et al. Elevation in S-adenosylhomocysteine and DNA hypomethylation: potential epigenetic mechanism for homocysteine-related pathology. [J]Nutr, 2002,132(8 Suppl):2361-2366.
    95. Xie S, Wang Z, OkanoM, et al. Cloning, expression and chromosome locations of the human DNMT3 gene family.[J]Gene, 1999, 236(1):87-95.
    96.黎金庆,李渊,石永进,等.三氧化二砷诱导U266细胞p16基因重新表达.[J]癌症,2004,23(6):626-630.

© 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