三氧化二砷对人乳腺癌雌激素受体表达的影响
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
背景和目的:
     近年来乳腺癌的发病率和死亡率增长速度较快,全世界每年患乳腺癌的女性约130万人,而死于乳腺癌的有50万人。虽然我国的乳腺癌属于低发,但近年来其发病率增长幅度较快,预测20年后将会成为我国发病率最高的恶性肿瘤。
     雌激素受体(estrogen receptor ER)在乳腺癌的发生发展过程中,扮演着重要角色。在体内通过与雌激素的结合,激活含雌激素应答元件基因和含有其他转录因子结合元件基因的表达,进而导致乳腺癌的发生。通常情况下,以ER基因表达情况的不同可将乳腺癌分为ER阴性乳腺癌和ER阳性乳腺癌两种。其中ER阴性乳腺癌约占20%-35%,其恶性程度高、发展迅速、有较高的局部复发率,并易发生肺、肝、骨及脑等重要脏器的转移,预后较差。由于缺乏相应的受体不能进行内分泌和生物治疗,所以这类特殊乳腺癌的治疗仅限于化疗,但疗效欠佳。因此,作为一个分子标记物,ER在乳腺癌的内分泌治疗中进行效果预测、评估激素依赖性等方面具有重要的意义。
     DNA甲基化和染色体重组是当前研究ER阴性乳腺癌中ER基因失活的两大主要学说。甲基化在肿瘤的发生、发展中有着重要作用,但又是一可逆过程。从理论上来说,如果使用药物对甲基化的抑癌基因和癌基因进行人为逆转,即可使因甲基化失活的基因重新获得表达。
     三氧化二砷(As2O3)为中药砒霜的主要成分,近年来发现其对白血病、原发性肝癌等多种肿瘤治疗取得了较好的效果。主要通过诱导细胞凋亡来发挥其细胞毒性作用,如今多项研究证明其具有甲基化抑制作用。
     本实验通过研究不同浓度的As2O3对人ER阴性乳腺癌细胞株MDA-MB-435进行药物诱导,使其基因启动子区CpG岛部分脱甲基而使其重新表达有功能的ERα,从而使得ER阴性乳腺癌细胞恢复对内分泌治疗药物的敏感性。
     材料与方法:
     (1)常规培养雌激素受体(ERα)阴性人乳腺癌细胞株MDA-MB-435,细胞浓度调整为5×105/ml后换含As2O3的培养液,至AS2O3终浓度分别为0.5μmol/L、1.0μmol/L、2.0μmol/L、4.0μmol/L,连续培养72h。以同期培养不加药的MDA-MB-435细胞作为阴性对照,ERα阳性人乳腺癌细胞株MCF-7作为阳性对照。
     (2) RT-PCR方法检测经不同浓度As2O3处理后的MDA-MB-435细胞ERα的mRNA表达情况。
     (3)甲基化特异性PCR (Methylation Specific PCR, MSP)方法检测经不同浓度As2O3处理后的MDA-MB-435细胞ERαDNA的5’-CpG岛甲基化的情况。
     (4) Western blot方法检测经不同浓度As2O3处理后的MDA-MB-435细胞ERα的蛋白表达情况。
     (5)免疫组化方法检测经不同浓度As2O3处理后的MDA-MB-435细胞ERα的蛋白表达情况。
     (6)MTT方法检测经不同浓度As2O3处理后的MDA-MB-435细胞,再经三苯氧胺(TAM)处理,观察各组细胞的生长受抑情况。
     (7)统计学方法:应用统计学处理软件SPSS 16.0,X2检验进行统计学分析,·差异显著性标准设为P<0.05。
     结果:
     (1) RT-PCR法检测阴性对照组无ERαmRNA基因;0.5μmol/L组未扩增出ERαmRNA基因;1.0μmol/L组、2.0μmol/L组、4.0μmol/L组均扩增出ERαmRNA基因。
     (2)甲基化特异性PCR法检测阴性对照组仅甲基化引物扩增出特异PCR条带;0.5μmol/L组、1.0μmol/L组、2.0μmol/L组、4.0μmol/L组甲基化和去甲基化引物均扩增出特异PCR条带。
     (3)经Western blot检测证实阴性对照组未显示蛋白条带;0.5μmol/L组也未显示蛋白条带,但1.0μmol/L组、2.0μmol/L和4.0μmol/L组显示出蛋白条带。
     (4)经免疫组化法检测证实阴性对照组细胞阳性率为7.4%;/L组、/L组、/L组、/L组细胞阳性率分别为7.8%、43.8%、78.2%、41.5%。
     (5)MTT法检测TAM作用于经As203处理的MDA-MB-435细胞后,细胞增殖明显受抑。阴性对照组受抑率为10.7%;0.5μmol/L组、/L组、2.0μmol/L组、/L组细胞受抑率分别是14.9%、30.7%、43.1%、56.6%。
     结论:
     适当浓度的As203通过对部分细胞的去甲基化作用,使ERa阴性人乳腺癌细胞株MDA-MB-435细胞ERa mRNA和蛋白重新表达,并可恢复对内分泌治疗药物(三苯氧胺)的敏感性,从而为ERa阴性乳腺癌患者实现内分泌治疗提供可靠的理论依据。
Background and objective:
     In recent years, the morbidity and mortality of breast cancer are growing rapidly. About 130 million women suffer from breast cancer and 50 million people died of breast cancer each year in the world. Although breast cancer has a low morbidity in our country, the incidence of breast cancer is rising obviously. We predict that it will be the highest incidence of malignant tumor in 20 years.
     Estrogen receptor (ER) is an important role in the development of breast cancer. In vivo, ER combine with estrogen and active the genes which contain estrogen response element and transcription factor response element, then lead to breast cancer. In general, we divide the breast cancer into ER negative breast cancer and ER positive breast cancer. Among about 20~35 percent of breast cancer patients, the estrogen receptor is negative. With the high malignant degree, rapid development, higher local recurrence, and metastases easily occurring in lung, liver, bone, brain and other important viscera, it has poor prognosis. Due to lack of corresponding receptor, the patients cannot apply endocrine and biological treatment. At present, the treatment of this kind of special type of breast cancer limits to chemotherapy, but it is far from perfect. Thus, as an important molecular marker, ER has vital significance to forecast the effect of endocrine therapy and evaluate the dependence of hormone in breast cancer.
     The main theories of ER gene expression silencing are DNA methylation and chromosome restructuring in ER negative breast cancer. Methylation occurs early in the process of cancer, it plays an important role in the occurrence and development of cancer. DNA methylation is a reversible process. By theoretically, if we reverse methylated oncogenes and tumor-suppressor genes with drugs, it will be made the gene re-expressed.
     Arsenic trioxide (AS2O3) is the main ingredient of arsenic's of Traditional Chinese medicine. In recent years, we find that it has better effects on many kinds of tumor cells, such as leukemia, liver cancer and so on. AS2O3 exert it's cytotoxicity by inducing cells apoptosis. Recent research suggests that it can demethylate.
     This study is designed that AS2O3 effect on ERa negative breast cancer cell MDA-MB-435. As2O3 can demethylate gene promoter CpG island partially, and made ERa re-expressed. Consequently, ERa negative breast cancer patients can restore the sensitivity of endocrine drugs.
     Materials and methods:
     (1) ERa negative human breast cancer cell line MDA-MB-435 planted at 37℃and 5% CO2. Adjust cell density of 5×105/ml and treat with 1640 containing AS2O3 0.5μmol/L, 1.0μmol/L,2.0μmol/L,4.0μmol/L respectively. Then plant for 72 hours. MDA-MB-435 without being treated with containing As2O3 was taken as negative control. Human breast cancer cell line MCF-7 (ER positive) was taken as positive control.
     (2) To detect the re-expression of the ERa mRNA by RT-PCR.
     (3) To detect the status of 5'CpG island methylation of ERa gene by Methylation Specific PCR(MSP).
     (4) To detect the re-expression of the ERa protein by Western blot.
     (5) To detect the re-expression of the ERa protein by immunohismchemistry.
     (6) MTT method tested whether Tamoxifen could induce the growth inhibition of MDA-MB-435 cell after treated with As2O3.
     (7) Using SPSS 16.0 statistics processing software andχ2-test for statistical analysis. Significant of difference standard is P<0.05.
     Result:
     (1) It is confirmed by the test of RT-PCR that expression of ERa mRNA were not found in group of negative control, neither in group of 0.5μmol/L. But ERa mRNA were detectable in groups of 1.0μmol/L,2.0μmol/L and 4.0μmol/L.
     (2) By MSP technique, methylation of ERa gene was found in group of negative control, methylation of ERa gene and demethylation of ERa gene were both found in groups of 0.5μmol/L, 1.0μmol/L,2.0μmol/L and 4.0μmol/L.
     (3) It is confirmed by the test of Western blot that expression of ERa protein were not found in group of negative control and in group of 0.5μmol/L. But ERa protein were detectable in groups of 1.0μmol/L,2.0μmol/L and 4.0μmol/L.
     (4) By immunohistochemistry technique, we found that, in group of negative control, the positive rate was 7.4%. In groups of 0.5μmol/L, 1.0μmol/L,2.0μmol/L and 4.0μmol/L, the positive rate is respectively 7.8%,43.8%,78.2% and 41.5%.
     (5) By the test of MTT, we found that tamoxifen can effect on the growth of MDA-MB-435 cells treated with AS2O3. We found that, in group of negative control, the inhibition rate was 10.7%. Inhibition rate was respectively 14.9%,30.7%,43.1% and 56.6% in groups of 0.5μmol/L, 1.0μmol/L,2.0μmol/L and 4.0μmol/L.
     Conclusion:
     A proper concentration of AS2O3 could effect on a part cells through demethylation and induce ERa gene re-expression in human breast cancer cell line MDA-MB-435. And it can restore to sensitivity of endocrine therapy drug(tamoxifen). Consequently, this study provides the reliable theory base for treating ERa negative breast cancer patients with endocrine therapy.
引文
[1]. Jemal A, Siegel R, Xu JQ, et al. Cancer statistics [J]. CA Cancer Clin,2010,60:277-300
    [2]. Yang L, Parkin DM, Ferlay J, et al. Eimates of cancer incidence in China for 2000 and projections for 2005 [J]. Cancer Epidemiol Biomarkers Prev,2005,14 (1):243~250
    [3]. Champagne FA, Curley JP. Maternal regulation of estrogen receptor alpha methylation [J]. Ocurr Opin Pharmacol 2008,8:735~739
    [4].王艳,霍翔,沈洪兵.雌激素受体和孕激素受体与乳腺癌关系的研究进展[J].中国肿瘤,2007,16(12):987-990
    [5].王雅杰,王宁.ER及Her-2对乳腺癌治疗策略的影响[J].中国癌症杂志,2010,20(10):721-726
    [6]. Gonzalgo M L, Hayashida T, Bender C M, et al. The role of DNA methylation in expression of the p19/p16 locus in human bladder cancer cell lines [J]. Cancer Res,1998,58(6): 1245~1252
    [7]. Matsuda Y, Ichida T, Matsuzawa J, et al. p16INK4 is inactivated by extensive CpG methylation in human hepatocellular carcinoma [J]. Gastroenterology,1999,116 (2):394~400
    [8]. Jung Y, Park J, Kim TY, et al. Potential advantages of DNA methyltransferase-1 (DNMT1) targeted inhibition for cancer therapy [J]. Mol Med,2007,85:1137~1148
    [9]. Agrawal A, Murphy RF, Agrawal DK. DNA methylation in breast and colorectal cancers [J]. Modern Pathology,2007,20:711~721
    [10].Han YH, Moon HJ, You BR. et al. The effect of MAPK inhibitors on arsenic trioxide treated Calu-6 lung cells in relation to cell death, ROS and GSH levels [J]. Anticancer Res,2009,29: 3837~3844
    [11].Harvey JM, Clark GM, Osborne CK, et al. Estrogen receptor status by immunohisto-chemistry is superior to the ligand binding assay for predicting response to adjuvant endocrine therapy in breast cance [J]. Clin Oncol,1999,17 (5):1474~1481
    [12].Lawson JS, Field AS, Champion S, et al. Low estrogen receptor alpha expression in normal breast tissue underlies low breast cancer incidence in Japan [J]. Lancet,1999,354 (9192): 1787~1788
    [13].齐小伟.三阴性乳腺癌患者的新辅助化疗疗效及生存分析[J].中华乳腺病杂志,2009,3(1):109-112
    [14]. Yager JD, Davidson NE. Estrogen carcinogenesis in breastcancer [J]. N Engl J Med,2006, 354 (15):270~282
    [15].Roodi NBailey LR, Kao WY, et al. Estrogen receptor gene analysis in estrogen receptor-positive and receptor-negative primary breast cancer [J]. Natl Cancer Inst,1995, 87(6):446-451
    [16].Lwase H, Kobayashi S, Lwata H, et al. Molecular analysis of the estrogen receptor (ER) gene in association with ER negativity in breast cancer [J]. Gan To Kagaku Ryoho,1996, 23(11):61~65
    [17].Lapidus RG, Nass SJ, Butash KA. et al. Mapping of ER gene CpG island methylation-specific polymerase chain reaction [J]. Cancer Res,1998,58 (12):2515~2519
    [18].Baylin SB, Herman JG. DNA hypermethylation in tumorigenesis:epigetics joins genetics [J]. Trends Genet,2000,16 (4):168~174
    [19].Becker K, Kellerq, Hoefler H. The use molecular biology in diagnosis and prognosis of gastric cancer [J]. Surg Oncol,2000,9 (1):5~11
    [20].王欣欣,付丽.DNA去甲基化与肿瘤治疗的研究进展[J].中华病理学杂志,2009,38(5):355-359
    [21].Trasler J M, Bovenzi, Gamete imprinting:setting epigenetic patterns for the next generation [J]. Reprod Fertil Dev,2006,18(12):63~69
    [22].范保星.DNA甲基化检测方法[J].国外医学遗传学分册,2002,25(2):99-101
    [23].苏世振,布立敬,李红智,等.乳腺癌雌激素受体α基因启动子甲基化与其蛋白表达的关系[J].温州医学院学报,2009,39(2):133-136
    [24].Giacinti L, Claudio P, Lopez M, et al. Epigenetic information and estrogen receptor alpha expression in breast cancer [J]. Oncologist,2006,11 (1):1-8
    [25].唐波,彭志红,姜军.5氮2脱氧胞苷与三苯氧胺协同抗雌激素受体α阴性乳腺癌的体外实验研究[J].中华外科杂志,2005,43(23):1545-1549
    [26].Billam M, Sobolewski MD, Davidson NE. Effects of a novel DNA methyltransferase inhibitor zebularine on human breast cancer cells [J]. Breast Cancer Res Treat,2010,120: 581~592
    [27]. Miller Jr WH, Schipper HM, Lee JS, et al. Mechanisms of action of arsenic trioxide [J]. Cancer Res,2002,62, (3):893-903
    [28].Vahter M, Mechanisms of arsenic biotransformation [J]. Toxicology,2002,27(181-182): 211~217
    [29].Gonzalez-FraileMI, Garcia-SanzR, MateosMV, et al. Methylenetetrahy drofolate reductase genotype does not play a role in multiple myeloma pathogenesis [J]. BrJ Haemato,2002,117 (4):890~892
    [30].傅海英,沈建箴,沈松菲等.巢式MSP检测砷剂诱导人多发性骨髓瘤U266细胞系p16 基因去甲基化及转录[J].中国实验血液学杂志,2007,(15):79-85
    [31].JamesS J, Melnyk S, Pogribna M, et al. Elevationin Sadenosylhomocysteine and DNA hypomethylation:potential epigenetic mechanism for homocysteine-related pathology[J]. Nutr,2002,132 (8):2361-2366
    [32].王鸣明,窦红菊,邹丽芳,等.砷剂诱导骨髓瘤细胞周期改变与SOCS-1基因去甲基化作用的研究[J].癌症,2008,27(11):1150-1154
    [33].张丽娜,王留兴,樊青霞,等.三氧化二砷诱导雌激素受体阴性乳腺癌细胞株MDA-MB-435s凋亡和Bc122、Bax表达关系的研究[J].肿瘤基础与临床,2007,20(6):2203-2206
    [34].曲志博,刘连新,陈炜,等.三氧化二砷抑制乳腺癌裸鼠移植瘤的生长及其作用机制的探讨[J].中国普通外科杂志,2007,16(1):265-268
    [35].Klijn JG, Berns PM, Schmitz PI, et al. The clinical significance of epidermal growth factor receptor (EGFR) in human breast cancer:a review on 5232 patients [J]. Endocr Rev,1992, 13(1):3-17
    [36].Nahta R, Yuan L X, Du Y, et al. Lapatinib induces apoptosis in trastuzumab resistant breast cancer cells:effects on insulinlike growth factor signaling [J]. Mol cancer Ther,2007,6 (2): 667~674
    [37].Geyer C E, Forster J, Lindquist D, et al. Laptinib plus capecitabine for HER-2 positive advanced breast cancer [J]. N Eng J Med,2006,355(26):2733~2743
    [38].Cesare G, Paoblo M, Antonio R, et al. The role of bevacizumab in the treatment of nonsmall cell lung cancer:current indications and future developments [J]. The Oncologist, 2007,12(10):1183~1193
    [39].赵胜雷.血管内皮生长因子与乳腺癌靶向治疗的研究进展[J].中国癌症防治杂志,2010,2(2):152-154
    [40].李林蔚.人乳腺癌细胞雌激素受体基因的去甲基化及其蛋白再表达[M].[硕士学位论文].郑州:郑州大学,2005
    [41].王瑞,李林蔚,王瑞林,等.人乳腺癌细胞雌激素受体基因的去甲基化及其表达[J].中华肿瘤杂志,2006,28(12):894-897
    [1]. Reik Wj Dean W, Walter J. Epigenetic reprogramming in mammalian velopment.Science, 2001,93(5532):1089-1093
    [2].丁波泥.天花粉蛋白抑制乳腺癌生长及逆转ERa基因甲基化的研究[D].[博士学位论文].长沙:中南大学,2008
    [3]. Etoh T,Kanai Y,Ushijima S,et al. Increased DNA methyltransferase I (DNMTI) protein ex-pression correlates significantly with poorer tumor differentiation and frequent DNA hyper-methylation of multiple CpG islands in gastric cancers.Am J Pathol,2004,164 (2):689~699
    [4]. Lama G, Angelucci C,Bruzzese N,et al. Sensitivity of human melanoma cells to oestrogens,tamoxifen and quercetin:is there any relationship with type Ⅰ and Ⅱ estrogens binding site expression[J]. Melanoma Res,1998,8(4):313
    [5].柯跃斌,夏菠,等.DNA去甲基化与基因激活过程[J].癌变.畸形.突变,2010,22(2):149-154
    [6], Hsieh CL.Evidence that protein binding specifies sites of DNA demethylation [J]. Mol Cell Biol,1999,19(1):46~56
    [7].李培坤,耿小平.DNA甲基化抑制剂研究进展[J].国际药学研究杂志,2010,37(3):198-203
    [8]. Momparler RL. Pharmacology of 5-aza-2-deoxycytidine (decitabine) [J]. Sem in Hematol, 2005,42(1):9~16
    [9]. Issa JP, Kantarjian HM, Kirkpatrick P. Azacitidine [J]. Nat Rev Drug Discover,2005,4 (4): 275-276
    [10].Kaminskas E, FarrellA, Abraham S, et al. Approval summary:azacitidine for treatment of myelodys plastic syndrome subtypes [J]. Clin Cancer Res,2005,11 (10):3604~3608
    [11].Issa JP, Gharibyan V, Cortes J, et al. Phase Ⅱ study of kwdose decitabine in patients with chronicmyelogenous leukemia resistant to imatinib mesylate [J].Clin Oncol,2005,23 (17):3948~-3956
    [12].Samlowski WE, Leachman SA, WadeM, et al. Evaluation of 72 day continuous intravenous infusion of decitabine:inhibition of promoter specific and global genomic DNA methylation [J]. Clin Oncol,2005,23 (17):3897~3905
    [13].徐昕,戴秋新,徐茂忠,等.地西他滨对淋巴瘤细胞株Raji中P15INK4B基因的去甲基化作用[J].山东大学学报(医学版),2010,48(7):62-68
    [14].Atceq B, Unterberger A, szyf M, et al. 1‰ nacological inhibition of DNA methylation induces proinvasive and prometastatic genes in duo and in vivo[J]. Neoplasia,2008,10(3): 266-278
    [15].Kurkjian C, Kummar S, Murgo AJ, et al. DNA methylation:its role in cancer development and therapy [J]. Curr Probl Cancer,2008,32 (5):187~-235
    [16].Griffiths EA, Gore SD. DNA methyltransferase and histone deacetylase inhibitors in the treatment of myelodysp lastic syndromes [J]. Sem in Hem atol,2008,45 (1):23~30
    [17].Kaminskas E, Farrell AT, Wang YC, et al. FDA drug approval summary:azacitidine (5— azacytidine, Vidaza) for injectable suspension [J]. Oncologist,2005,10 (3):176~182
    [18].Cheng JC, Yoo C B, Weisenberger D J, et al. Preferential response of cancer cells to zebμlarine [J]. Cancer Cel,2004,6 (2):151 ~158
    [19].Hurd P J, Whitmarsh A J, Baldwin G S, et al. Mechanism-based inhibition of 5-cytosine DNA methyltransferases by Hpyrimidinone [J]. Mol Biol,1999,286 (2):389~401
    [20].Marquez V E, Barchij JR, Kelley JA, et al. Zebμlarine:a unique molecule for an epigenetically based strategy in cancer chemotherapy [J]. Nucleo tides nucleic Acids, 2005,24(5-7):305~318
    [21]. Lee G, Wolff E, Miller J H. Mutagenicity of the cytidineanalog zebularine in Escherichia coli [J]. DNA Repair (Amst),2004,3 (2):155-161
    [22].殷鹏,张月飞Zebμlarine抗肿瘤研究进展[J].广东医学,2007,28(4):659-661
    [23].Song Y, Zhang C. Hydralazine inhibits human cervical cancer cell growth in vitro in association with APC demethylation and re-expression [J]. Cancer Chem other Pharmacol, 2009,63(4):605~613
    [24].陈伟.三氧化二砷和肼苯哒嗪体外逆转ERa表达作用效果的比较[M].[硕士学位论文].郑州:郑州大学,2007
    [25]. Mare JM, Wang LJ. Arsenic alters cytosine methylation patterns of the promoter of the tumor suppressor gene P52 in human lung cell:a model for a mechanism of carcinogenesis [J]. Mutat Res,1997,386 (3):263~277
    [26].王冰.三氧化二砷诱导雌激素受体表达的体外研究[M].[硕士学位论文].郑州:郑州大学,2006
    [27].王鸣明,窦红菊,邹丽芳,等.砷剂诱导骨髓瘤细胞周期改变与SOCS-1基因去甲基化作用的研究[J].癌症,2008,27(11):1150-1154
    [28]. Yang L, Luo JM, Wen SP, et al. Effect of As2O3 on demethylation of SHP-1 gene in human lymphoma cell line T2 [J]. Chin Cancer,2009,28 (3):209~213
    [29]. Lee BH, Lin X, et al. Procainamide is a specific inhibitor of DNA methyltransferase [J]. Biol Chem,2005,280 (49):40749-40756
    [30].Fang MZ, Wang Y, et al. Tea Polypheno epigallocateehin gallate inhibits DNA methyltransferase and reactivates methylation silenced genes in cancer cell lines [J]. Cancer Res,2003,63(22):7563~7570
    [31].Gao Z, Xu Z, HungMS, et al. Promoter demethylation of WIF-1 by epigallocatechin gallate in lung cancer cells [J]. Anticancer Res,2009,29 (6):2025~2030
    [32].Gu B, Ding Q, Xia G, et al. EGCG inhibits growth and induces apoptosis in renal cell carcinoma through TFPI-2 over-expression [J]. Oncol Rep,2009,21 (3):635~640
    [33].Ma X, Fang Y, Beklemisheva et al. Phenylhexy isothiocyanate inhibits histone deacetylases and remodels chromatins to induce growth arrest in human leukemia cells [J]. Int Oncol. 2006,28(5):1287~1293
    [34].黄轶群,马旭东,等.异硫氰酸苯己酯Molt-4细胞组蛋白甲基化、乙酰化调控的实验研究[J].中华血液学杂志,2007,28,(9):612-615
    [35].庄志明.异硫氰酸苯已酯(PHI)诱导前列腺癌PC3细胞凋亡的分子机制研究[M].[硕士学位论文].福州:福建医科大学,2009
    [36].范江,陆劲松.药物诱导MDA-MB-435细胞表达ERa及其对内分泌治疗的敏感性[J].中华肿瘤杂志,2006,28(12):886-889
    [37].Siedleeki P,Boy RQMusch T, et al. Discovery of two novel. Small-molecule inhibitors of DNA methylatior [J]. Med Chem,2006,49 (2):678-683
    [38].姜蕊,宋伟.RG108对肺腺癌A549细胞增殖、凋亡及RASSF1A基因表达的影响[J].中国组织化学与细胞化学杂志,2009,18(4):354-357
    [39].黄利鸣,邢学森,黄益玲.P16基因甲基化与Hela细胞凋亡相关性的研究[J].中国肿瘤临床,2005,32(13):721-724
    [40].华芳,单保恩,赵连梅,等.天花粉蛋白抑制人乳腺癌MDA-MB-231细胞生长及逆转syk基因甲基化的研究[J].肿瘤,2009,29,(10):944-949
    [41].柴新娟,栾菊.中药成分CDP对乳腺癌细胞P16,E-CADHERIN去甲基化作用的研究[J].四川大学学报(医学版),2009,40(5):798-802

© 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