用户名: 密码: 验证码:
PI3K/Akt信号通路对DNA甲基转移酶3B基因表达的调控及其分子机制研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
肿瘤细胞表现广泛而显著的表观遗传学变化,包括DNA甲基化和染色质修饰。DNA甲基化是最广为人知的表观遗传学标志,DNA甲基化主要由DNA甲基转移酶(DNA (cytosine-5)-methyltransferases, DNMTs)(EC 2.1.1.37)所控制。目前,已鉴定的人类具有催化活性的DNMTs有三种:DNMT1, DNMT3A和DNMT3B,其编码基因分别位于染色体19p13.2.、2p23和20q11.2。DNMT1优先选择DNA复制过程中所形成以半甲基化状态存在的DNA双链作为催化底物,而DNMT3A和DNMT3B是从头(de novo) DNA甲基化修饰所必须的,主要为在细胞内建立新DNA甲基化类型所必须。研究发现在肿瘤细胞中存在异常DNA甲基化类型,即广泛的DNA低甲基化与某些基因启动子特异高甲基化。发生在启动子CpG岛高甲基化可影响许多与肿瘤细胞发生发展密切相关基因的表达,在恶性肿瘤以及在癌旁组织向恶性状态转变过程中均发现DNMTs升高。有学者认为具有从头DNA甲基化作用的DNMT3B在肿瘤发生发展中具有更重要作用。在细胞内PI3K/Akt信号通路调控众多信号转导途径,例如细胞生长与存活、细胞间信息传递等。异常PI3K/Akt信号通路参与肿瘤形成与转移。我们实验室已报道PI3K/Akt信号通路可以维持DNMT1蛋白稳定性,从而在肿瘤发生中起促进作用,但PI3K/Akt信号通路对DNMT3B基因表达影响及调控尚未见报道。
     在本文第一部分实验中,我们在6株人肝癌(Human hepatocellular carcinoma, HCC)细胞株中发现DNMT3B mRNA表达水平与Akt1 mRNA或Akt2 mRNA表达水平相关;不同HCC细胞株的DNMT3B3 mRNA水平与DNMT3B4 mRNA水平比值有较大的差异。此外,我们发现人肝癌组织中DNMT3B基因表达明显高于其相对应癌旁组织。
     为了探讨Akt是否参与DNMT3B基因调控,在第二部分实验中,我们首先检测Akt作为上游调节剂及增量调节剂的PI3K对DNMT3B基因影响,研究发现PI3K的小分子特异性抑制剂LY294002在BEL-7404细胞株中下调DNMT3B mRNA和蛋白质水平且呈剂量效应关系和时间效应关系。我们观察到LY294002对DNMT3B mRNA表达的下调降低程度在各个时间点均高于其对DNMT3B蛋白的下调程度,提示LY294002对DNMT3B基因下调可能存在着转录水平调控。此外,我们研究发现LY294002诱导DNMT3B基因表达下调并不伴随DNMT3B亚细胞定位变化。在SMCC7721细胞株及L02细胞株均观察到LY294002降低DNMT3B基因表达。接着我们还应用蛋白质合成抑制剂CHX来检测LY294002对DNMT3B基因表达下调是否与蛋白质从头机器抑制相关,结果发现LY294002不影响DNMT3B蛋白的稳定性。在第二部分实验中,我们还利用本实验室已经建立的持续性活化Akt2的小鼠胚胎成纤维细胞株(mouse embryonic fibroblasts, MEFs)及稳定转染Akt1/2 shRNA的BEL-7404细胞株去研究Akt对DNMT3B基因调控作用,发现Akt2持续性活化导致DNMT3B mRNA及蛋白质水平升高,而在BEL-7404细胞中敲低Aktl/2引起DNMT3B mRNA及蛋白质水平均降低。再者,我们观察到Akt下游信息分子糖原合酶激酶-3β(glycogen synthase kinase-3p, GSK-3β)抑制剂氯化锂可促进DNMT3B基因表达中度增加。以上实验结果表明DNMT3B基因表达受到PI3K/Akt信号通路的调控。
     为了深入阐明PI3K/Akt信号通路调控DNMT3B基因表达的分子机制,.在第三部分实验中,我们研究了LY294002对DNMT3B启动子活性的影响。首先我们依据Yanagisawa报道,构建了四个不同长度DNMT3B启动子片段,包括核心启动子序列及基本启动子序列,并将它们克隆到pGL3-Basic荧光素酶报告基因上游,经测序比对正确后,选择构建成功载体进行下一步试验。我们通过瞬时转染方法及双荧光素酶检测系统研究启动子质粒在BEL-7404细胞中的表达情况,发现核心启动子序列表达荧光素酶活性最高,结果与文献报道中基本相同。接着我们观察了LY294002对这四种DNMT3B启动子活性影响,发现荧火虫荧光素酶及作为内参的海肾荧光素酶活性均比LY294002未处理组低,因此,我们采用LY294002处理pGL3-Basic质粒(具有基本转录活性)前后相对荧光素酶活性比值作为判断标准,若低于此标准被认为启动子活性受到抑制。结果表明,LY294002抑制DNMT3B启动子活性。因此我们认为PI3K/Akt信号通路在转录水平调控了DNMT3B基因表达。在此部分中,我们还通过对DNMT3B的mRNA稳定性研究发现,LY294002可以促进DNMT3B mRNA降解,降低DNMT3B mRNA半衰期,表明LY294002对DNMT3B基因下调也存在转录后水平调控。实验中我们观察到HuR变化亦受LY294002调控,并呈现出时间效应关系,鉴于有研究报道HuR可增加DNMT3B基因稳定性,因此,我们推测HuR可能作为LY294002促进DNMT3B mRNA降解的中介物。此部分的实验结果表明PI3K/Akt信号通路可在转录及转录后水平调控DNMT3B基因表达。
     综上所述,我们首次研究发现在人HCC细胞株BEL-7404中,PI3K/Akt信号通路可以下调DNMT3B基因表达,其下调机制是在转录水平和转录后水平;这一发现对我们认识PI3K/Akt信号通路和DNMT3B基因在肝癌发生机制中的作用具有重要意义。
Cancer cells show widespread and distinguished epigenetic changes when compared with their normal parental tissue, including DNA methylation and chromatin modification. DNA methylation is the best-known epigenetic marker and controlled by DNA methyltransferases (DNMTs). To date, three catalytically active DNMTs have been identified in human, DNMT1, DNMT3A, and DNMT3B, their encoding gene respectively located in 19p13.2.,2p23 and 20q11.2.. DNMT1 prefers hemimethylated DNA substrates and is therefore primarily responsible for maintenance methylation, which must occur during DNA replication. Dnmt3A and Dnmt3B are necessary for de novo methylation and for the establishment of new methylation patterns in mammalian cells. Aberrant DNA methylation in cancer cells includes global DNA hypomethylation and specific promoter hypermethylation of genes. Hypermethylation of the CpG-island promoter can affect genes involved in the development of cancer cells, and the levels of DNMTs are elevated in various malignancies and transition from a precancerous to a malignant state. There are reports about that de novo methyltransferase DNMT3B has more important role in the development of cancers. Within the cells, diverse processes are regulated by phosphatidylinositol 3 kinase (PI3K)/Akt signaling pathway including growth and survival and cell-cell communication. Abnormal PI3K/Akt activity has been linked to the formation of tumors, metastasis. In our previous study we found that phosphatidylinositol 3 kinase (PI3K)/Akt pathway stabilizes DNMT1 protein and advance the carcinogenesis. However, the relationship between PI3K/Akt pathway and DNMT3B gene expression is unknown.
     In first section of our research, we found that Akts (Aktl or Akt2) transcripts are associated with the total DNMT3B transcripts in 6 human hepatocellular carcinoma (HCC) cell lines. We further found that there are various in the ratio of DNMT3B3 mRNA to DNMT3B4 mRNA of 6 HCC cell lines. In addition, we confirmed that DNMT3B protein was strongly higher expression in HCC tissues than each matched adjacent noncancerous tissues.
     To explore the mechanisms of Akt on the regulation of DNMT3B gene expression, in second section of our work, we firstly assessed that PI3K, a upstream molecule of Akt, how to modulate DNMT3B gene expression, and found that LY294002, a small molecule specific inhibitor of PI3K pathway, down-regulated DNMT3B gene expression at the RNA and protein level in hepatocarcinoma cell line BEL-7404 in a dose-and time-dependent manner. We investigated that LY294002 down-regulated DNMT3B mRNA is prior to the down-regulation of DNMT3B protein after LY294002 treatment, which suggested that there was translational regulation. Moreover, we observed that down-regulation of DNMT3B gene expression by LY294002 was not accompanyed with the sub-cellular location of DNMT3B. Furthermore, down-regulation by LY294002 was also found in other two hepatocarcinoma cell lines SMCC7721 and L02. In addition, we performed a CHX chase protein decay assay between LY294002 treatment and without treatment to assesse the relationship between down-regulation of DNMT3B with LY294002 treatment and de novo synthesis of protein machinery. The result showed that LY294002 did not alter the stability of DNMT3B protein. In second section of our work, we found higher DNMT3B mRNA and protein levels in Myr-Akt2-MEFs constructed by our lab and expressed constitute activation of Akt2 than in MSCV-MEFs. Meanwhile, we found that the knockdown of Aktl and Akt2 caused the decrease of DNMT3B mRNA and protein levels in BEL-7404 stably transfected with Aktl/2 shRNA human lentiviral particles (Aktl/2 siRNA). To further confirm Akt regulate the DNMT3B gene expression, we observed Lithium chloride, an inhibitor of GSK3βwhich is a downstreamed molecule of Akt, moderately increased the DNMT3B protein in the BEL-7404 cells. All these findings suggest that PI3K/Akt modulates the DNMT3B gene expression.
     In order to deeply demonstrate the mechanism of regulating DNMT3B gene expression by PI3K/Akt pathway, in the third section of our study, we assessed that how the promoter of DNMT3B was affected by LY294002. Firstly, we constructed four specific DNMT3B promoter fragments which included core promoter region and base promoter sequences according to Yanagisawa's study, and were inserted upstream of the firefly luciferase gene in the pGL3-Basic vector. Next these constructs were sequenced and aligned with GeneBank. Furthermore we assayed luciferase activities of these plasmids with transiently transfection experiments and Dual-Luciferase Reporter Assay, we found the core promoter fragment revealed the highest activity in BEL-7404 cells, the data were similar to that reported in document. Moreover we investigated that both the luciferase activities of firefly and renilla were decreased in BEL-7404 cells transiently transfected with these pGL3 plasmids under with LY294002 treatment, which were consistent with the data of cell cycle assay that represented cycle arrest of the G1 phase in BEL-7404 cell treated with LY294002. Whereas the above investigation, we took the ratio of relative luciferase activity of the pGL3-Basic (occupying base transcription activity) with LY294002 treatment or untreatment to be the criterion, and found the ratio of the DNMT3B promoter constructs were lower than the criterion. These results indicated that PI3K/Akt pathway regulates the expression of DNMT3B gene at transcription control. Furthermore, we observed that LY294002 stimulation induced the increase of DNMT3B degradation using mRNA decay analysis, which suggested that down-regulation of DNMT3B by LY294002 is also post-transcriptional control. Moreover we evaluated that the HuR expression was also modulated by LY294002 in BEL-7404 cells in a time-dependent manner. Whereas DNMT3B mRNA stabilized by HuR was reported, we hypothesized that DNMT3B was down-regulated by LY294002 via HuR decrease. Taken together, these findings in the fourth sections of our study indicated that PI3K/Akt pathway regulates the expression of DNMT3B gene at transcription and post-transcription level.
     In summary, we have, for the first time, demonstrated that PI3K/Akt pathway regulates the expression of DNMT3B gene at transcriptional and post-transcriptional levels, which is particularly important to understand the effects of PI3K/Akt pathway and DNMT3B gene on hepatocarcinogenesis.
引文
1. Esteller, M., Epigenetics in cancer. N Engl J Med,2008.358(11):p.1148-59.
    2. Issa, J.P., CpG-island methylation in aging and cancer. Curr Top Microbiol Immunol,2000.249:p.101-18.
    3. Bird, A., DNA methylation patterns and epigenetic memory. Genes Dev,2002. 16(1):p.6-21.
    4. Jones, P.A. and S.B. Baylin, The epigenomics of cancer. Cell,2007.128(4):p. 683-92.
    5. Fischle, W., Y. Wang, and C.D. Allis, Binary switches and modification cassettes in histone biology and beyond. Nature,2003.425(6957):p.475-9.
    6. Li, E., T.H. Bestor, and R. Jaenisch, Targeted mutation of the DNA methyltransferase gene results in embryonic lethality. Cell,1992.69(6):p. 915-26.
    7. Okano, M., et al., DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell,1999.99(3):p. 247-57.
    8. Esteller, M., CpG island hypermethylation and tumor suppressor genes:a booming present, a brighter future. Oncogene,2002.21(35):p.5427-40.
    9. Csankovszki, G., A. Nagy, and R. Jaenisch, Synergism of Xist RNA, DNA methylation, and histone hypoacetylation in maintaining X chromosome inactivation. J Cell Biol,2001.153(4):p.773-84.
    10. Bestor, T.H., Imprinting errors and developmental asymmetry. Philos Trans R Soc Lond B Biol Sci,2003.358(1436):p.1411-5.
    11. Jones, P.A. and S.B. Baylin, The fundamental role of epigenetic events in cancer. Nat Rev Genet,2002.3(6):p.415-28.
    12. Robertson, K.D., DNA methylation and human disease. Nat Rev Genet,2005. 6(8):p.597-610.
    13. Yoder, J.A., C.P. Walsh, and T.H. Bestor, Cytosine methylation and the ecology of intragenomic parasites. Trends Genet,1997.13(8):p.335-40.
    14. Song, F., et al., Association of tissue-specific differentially methylated regions (TDMs) with differential gene expression. Proc Natl Acad Sci U S A,2005. 102(9):p.3336-41.
    15. Brena, R.M. and J.F. Costello, Genome-epigenome interactions in cancer. Hum Mol Genet,2007.16 Spec No 1:p. R96-105.
    16. Feinberg, A.P. and B. Tycko, The history of cancer epigenetics. Nat Rev Cancer,2004.4(2):p.143-53.
    17. Widschwendter, M., et al., DNA hypomethylation and ovarian cancer biology. Cancer Res,2004.64(13):p.4472-80.
    18. Esteller, M. and J.G. Herman, Cancer as an epigenetic disease:DNA methylation and chromatin alterations in human tumours. J Pathol,2002. 196(1):p.1-7.
    19. Herman, J.G. and S.B. Baylin, Gene silencing in cancer in association with promoter hypermethylation. N Engl J Med,2003.349(21):p.2042-54.
    20. Esteller, M., Cancer epigenomics:DNA methylomes and histone-modification maps. Nat Rev Genet,2007.8(4):p.286-98.
    21. Robertson, K.D., DNA methylation and chromatin-unraveling the tangled web. Oncogene,2002.21(35):p.5361-79.
    22. Ramsahoye, B.H., et al., Non-CpG methylation is prevalent in embryonic stem cells and may be mediated by DNA methyltransferase 3a. Proc Natl Acad Sci U S A,2000.97(10):p.5237-42.
    23. Jeltsch, A., Beyond Watson and Crick:DNA methylation and molecular enzymology of DNA methyltransferases. Chembiochem,2002.3(4):p.274-93.
    24. Yoder, J.A., et al., DNA (cytosine-5)-methyltransferases in mouse cells and tissues. Studies with a mechanism-based probe. J Mol Biol,1997.270(3):p. 385-95.
    25. Tollefsbol, T.O. and C.A. Hutchison,3rd, Control of methylation spreading in synthetic DNA sequences by the murine DNA methyltransferase. J Mol Biol, 1997.269(4):p.494-504.
    26. Xie, S., et al., Cloning, expression and chromosome locations of the human DNMT3 gene family. Gene,1999.236(1):p.87-95.
    27. Chedin, F., M.R. Lieber, and C.L. Hsieh, The DNA methyltransferase-like protein DNMT3L stimulates de novo methylation by Dnmt3a. Proc Natl Acad Sci U S A,2002.99(26):p.16916-21.
    28. Suetake, I., et al., DNMT3L stimulates the DNA methylation activity of Dnmt3a and Dnmt3b through a direct interaction. J Biol Chem,2004.279(26): p.27816-23.
    29. Robertson, K.D., et al., The human DNA methyltransferases (DNMTs) 1,3a and 3b:coordinate mRNA expression in normal tissues and overexpression in tumors. Nucleic Acids Res,1999.27(11):p.2291-8.
    30. Melki, J.R., et al., Increased DNA methyltransferase expression in leukaemia. Leukemia,1998.12(3):p.311-6.
    31. Mizuno, S., et al., Expression of DNA methyltransferases DNMT1,3A, and 3B in normal hematopoiesis and in acute and chronic myelogenous leukemia. Blood,2001.97(5):p.1172-9.
    32. Beaulieu, N., et al., An essential role for DNA methyltransferase DNMT3B in cancer cell survival. J Biol Chem,2002.277(31):p.28176-81.
    33. Eads, C.A., et al., CpG island hypermethylation in human colorectal tumors is not associated with DNA methyltransferase overexpression. Cancer Res,1999. 59(10):p.2302-6.
    34. Girault, I., et al., Expression analysis of DNA methyltransferases 1,3A, and 3B in sporadic breast carcinomas. Clin Cancer Res,2003.9(12):p.4415-22.
    35. Jin, F., et al., Up-regulation of DNA methyltransferase 3B expression in endometrial cancers. Gynecol Oncol,2005.96(2):p.531-8.
    36. Patra, S.K., et al., DNA methyltransferase and demethylase in human prostate cancer. Mol Carcinog,2002.33(3):p.163-71.
    37. Robert, M.F., et al., DNMT1 is required to maintain CpG methylation and aberrant gene silencing in human cancer cells. Nat Genet,2003.33(1):p. 61-5.
    38. Saito, Y., et al., Increased protein expression of DNA methyltransferase (DNMT) 1 is significantly correlated with the malignant potential and poor prognosis of human hepatocellular carcinomas. Int J Cancer,2003.105(4):p. 527-32.
    39. Kanai, Y. and S. Hirohashi, Alterations of DNA methylation associated with abnormalities of DNA methyltransferases in human cancers during transition from a precancerous to a malignant state. Carcinogenesis,2007.28(12):p. 2434-42.
    40. Chen, Z.X., et al., Physical and functional interactions between the human DNMT3L protein and members of the de novo methyltransferase family. J Cell Biochem,2005.95(5):p.902-17.
    41. Soejima, K., W. Fang, and B.J. Rollins, DNA methyltransferase 3b contributes to oncogenic transformation induced by SV40T antigen and activated Ras. Oncogene,2003.22(30):p.4723-33.
    42. Cui, M., et al., Estrogen regulates DNA methyltransferase 3B expression in Ishikawa endometrial adenocarcinoma cells. Mol Biol Rep,2009.36(8):p. 2201-7.
    43. Hervouet, E., et al., Folate supplementation limits the aggressiveness of glioma via the remethylation of DNA repeats element and genes governing apoptosis and proliferation. Clin Cancer Res,2009.15(10):p.3519-29.
    44. Fabbri, M., et al., MicroRNA-29 family reverts aberrant methylation in lung cancer by targeting DNA methyltransferases 3A and 3B. Proc Natl Acad Sci U S A,2007.104(40):p.15805-10.
    45. Garzon, R., et al., MicroRNA-29b induces global DNA hypomethylation and tumor suppressor gene reexpression in acute myeloid leukemia by targeting directly DNMT3A and 3B and indirectly DNMT1. Blood,2009.113(25):p. 6411-8.
    46. Duursma, A.M., et al., miR-148 targets human DNMT3b protein coding region. RNA,2008.14(5):p.872-7.
    47. Vivanco, I. and C.L. Sawyers, The phosphatidylinositol 3-Kinase AKT pathway in human cancer. Nat Rev Cancer,2002.2(7):p.489-501.
    48. Voigt, P., et al., Assigning functional domains within the p101 regulatory subunit of phosphoinositide 3-kinase gamma. J Biol Chem,2005.280(6):p. 5121-7.
    49. Murthy, S.S., et al., Mapping of AKT3, encoding a member of the Akt/protein kinase B family, to human and rodent chromosomes by fluorescence in situ hybridization. Cytogenet Cell Genet,2000.88(1-2):p.38-40.
    50. Masure, S., et al., Molecular cloning, expression and characterization of the human serine/threonine kinase Akt-3. Eur J Biochem,1999.265(1):p.353-60.
    51. Franke, T.F., Intracellular signaling by Akt:bound to be specific. Sci Signal, 2008.1(24):p. pe29.
    52. Nicholson, K.M. and N.G. Anderson, The protein kinase B/Akt signalling pathway in human malignancy. Cell Signal,2002.14(5):p.381-95.
    53. Song, G., G. Ouyang, and S. Bao, The activation of Akt/PKB signaling pathway and cell survival. J Cell Mol Med,2005.9(1):p.59-71.
    54. Manning, B.D. and L.C. Cantley, AKT/PKB signaling:navigating downstream. Cell,2007.129(7):p.1261-74.
    55. Osaki, M., M. Oshimura, and H. Ito, PI3K-Akt pathway:its functions and alterations in human cancer. Apoptosis,2004.9(6):p.667-76.
    56. Qian, C.N., et al., Activation of the PI3K/AKT pathway induces urothelial carcinoma of the renal pelvis:identification in human tumors and confirmation in animal models. Cancer Res,2009.69(21):p.8256-64.
    57. Di Cristofano, A., et al., Pten is essential for embryonic development and tumour suppression. Nat Genet,1998.19(4):p.348-55.
    58. Ramaswamy, S., et al., Regulation of G1 progression by the PTEN tumor suppressor protein is linked to inhibition of the phosphatidylinositol 3-kinase/Akt pathway. Proc Natl Acad Sci U S A,1999.96(5):p.2110-5.
    59. Altomare, D.A. and J.R. Testa, Perturbations of the AKT signaling pathway in human cancer. Oncogene,2005.24(50):p.7455-64.
    60. Sarbassov, D.D., et al., Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science,2005.307(5712):p.1098-101.
    61. Testa, J.R. and A. Bellacosa, AKT plays a central role in tumorigenesis. Proc Natl Acad Sci U S A,2001.98(20):p.10983-5.
    62. Wymann, M.P. and R. Marone, Phosphoinositide 3-kinase in disease:timing, location, and scaffolding. Curr Opin Cell Biol,2005.17(2):p.141-9.
    63. Cha, T.L., et al., Akt-mediated phosphorylation of EZH2 suppresses methylation of lysine 27 in histone H3. Science,2005.310(5746):p.306-10.
    64. Serra, C., et al., Functional interdependence at the chromatin level between the MKK6/p38 and IGF1/PI3K/AKT pathways during muscle differentiation. Mol Cell,2007.28(2):p.200-13.
    65. Foster, K.S., et al., Members of the hSWI/SNF chromatin remodeling complex associate with and are phosphorylated by protein kinase B/Akt. Oncogene, 2006.25(33):p.4605-12.
    66. Sun, L., et al., Phosphatidylinositol 3-kinase/protein kinase B pathway stabilizes DNA methyltransferase I protein and maintains DNA methylation. Cell Signal,2007.19(11):p.2255-63.
    67. Rivenbark, A.G. and W.B. Coleman, The use of epigenetic biomarkers for preclinical detection of hepatocellular carcinoma:potential for noninvasive screening of high-risk populations. Clin Cancer Res,2007.13(8):p.2309-12.
    68. Bosch, F.X., et al., Primary liver cancer:worldwide incidence and trends. Gastroenterology,2004.127(5 Suppl 1):p. S5-S16.
    69. McGlynn, K.A. and W.T. London, Epidemiology and natural history of hepatocellular carcinoma. Best Pract Res Clin Gastroenterol,2005.19(1):p. 3-23.
    70. Tang, Z.Y., et al., A decade's studies on metastasis of hepatocellular carcinoma. J Cancer Res Clin Oncol,2004.130(4):p.187-96.
    71. Thorgeirsson, S.S. and J.W. Grisham, Molecular pathogenesis of human hepatocellular carcinoma. Nat Genet,2002.31(4):p.339-46.
    72. Herath, N.I., B.A. Leggett, and G.A. MacDonald, Review of genetic and epigenetic alterations in hepatocarcinogenesis. J Gastroenterol Hepatol,2006. 21(1 Pt 1):p.15-21.
    73. Dammann, R., G. Yang, and G.P. Pfeifer, Hypermethylation of the cpG island of Ras association domain family 1A (RASSFIA), a putative tumor suppressor gene from the 3p21.3 locus, occurs in a large percentage of human breast cancers. Cancer Res,2001.61(7):p.3105-9.
    74. Xu, G.L., et al., Chromosome instability and immunodeficiency syndrome caused by mutations in a DNA methyltransferase gene. Nature,1999. 402(6758):p.187-91.
    75. Tuck-Muller, C.M., et al., DNA hypomethylation and unusual chromosome instability in cell lines from ICF syndrome patients. Cytogenet Cell Genet, 2000.89(1-2):p.121-8.
    76. Fackenthal, J.D. and L.A. Godley, Aberrant RNA splicing and its functional consequences in cancer cells. Dis Model Mech,2008.1(1):p.37-42.
    77. Ostler, K.R., et al., Cancer cells express aberrant DNMT3B transcripts encoding truncated proteins. Oncogene,2007.26(38):p.5553-63.
    78. Weisenberger, D.J., et al., Role of the DNA methyltransferase variant DNMT3b3 in DNA methylation. Mol Cancer Res,2004.2(1):p.62-72.
    79. Saito, Y., et al., Overexpression of a splice variant of DNA methyltransferase 3b, DNMT3b4, associated with DNA hypomethylation on pericentromeric satellite regions during human hepatocarcinogenesis. Proc Natl Acad Sci U S A,2002.99(15):p.10060-5.
    80. Arsham, A.M., et al., Akt and hypoxia-inducible factor-1 independently enhance tumor growth and angiogenesis. Cancer Res,2004.64(10):p.3500-7.
    81. Alexia, C., et al., Pleiotropic effects of PI-3'kinase/Akt signaling in human hepatoma cell proliferation and drug-induced apoptosis. Ann N Y Acad Sci, 2006.1090:p.1-17.
    82. Chang, H.C., et al., Role of AKT kinase in sphingosine-induced apoptosis in human hepatoma cells. J Cell Physiol,2001.188(2):p.188-93.
    83. de Silanes, I.L., et al., The RNA-binding protein HuR regulates DNA methylation through stabilization of DNMT3b mRNA. Nucleic Acids Res,2009. 37(8):p.2658-71.
    84. Kondo, Y., et al., Alterations of DNA methylation and histone modifications contribute to gene silencing in hepatocellular carcinomas. Hepatol Res,2007. 37(11):p.974-83.
    85. Chang, F., et al., Involvement of PI3K/Akt pathway in cell cycle progression, apoptosis, and neoplastic transformation:a target for cancer chemotherapy. Leukemia,2003.17(3):p.590-603.
    86. Lee, J.W., et al., PIK3CA gene is frequently mutated in breast carcinomas and hepatocellular carcinomas. Oncogene,2005.24(8):p.1477-80.
    87. Karakas, B., K.E. Bachman, and B.H. Park, Mutation of the PIK3CA oncogene in human cancers. Br J Cancer,2006.94(4):p.455-9.
    88. He, X., et al., PIK3IP1, a negative regulator of PI3K, suppresses the development of hepatocellular carcinoma. Cancer Res,2008.68(14):p. 5591-8.
    89. Dong-Dong, L., Z. Xi-Ran, and C. Xiang-Rong, Expression and significance of new tumor suppressor gene PTEN in primary liver cancer. J Cell Mol Med, 2003.7(1):p.67-71.
    90. Hu, T.H., et al., Expression and prognostic role of tumor suppressor gene PTEN/MMAC1/TEP1 in hepatocellular carcinoma. Cancer,2003.97(8):p. 1929-40.
    91. Konishi, H., et al., Molecular Cloning and Characterization of a New Member of the RAC Protein Kinase Family:Association of the Pleckstrin Homology Domain of 3 Types of RAC Protein Kinase with Protein Kinase C Subspecies and [beta][gamma] Subunits of G Proteins. Biochemical and Biophysical Research Communications,1995.216(2):p.526-534.
    92. Nakanishi, K., et al., Critical involvement of the phosphatidylinositol 3-kinase/Akt pathway in anchorage-independent growth and hematogeneous intrahepatic metastasis of liver cancer. Cancer Res,2002.62(10):p.2971-5.
    93. Nakanishi, K., et al., Akt phosphorylation is a risk factor for early disease recurrence and poor prognosis in hepatocellular carcinoma. Cancer,2005. 103(2):p.307-12.
    94. Liu, L.X., et al., Gene expression profiles of hepatoma cell line HLE. World J Gastroenterol,2003.9(4):p.683-7.
    95. Oh, B.K., et al., DNA methyltransferase expression and DNA methylation in human hepatocellular carcinoma and their clinicopathological correlation. Int J Mol Med,2007.20(1):p.65-73.
    96. Rommel, C., M. Camps, and H. Ji, PI3K delta and PI3K gamma:partners in crime in inflammation in rheumatoid arthritis and beyond? Nat Rev Immunol, 2007.7(3):p.191-201.
    97. Hager, M., et al., PTEN expression in renal cell carcinoma and oncocytoma and prognosis. Pathology,2007.39(5):p.482-5.
    98. Aoki, M., et al., The akt kinase:molecular determinants of oncogenicity. Proc Natl Acad Sci U S A,1998.95(25):p.14950-5.
    99. Mende, I., et al., Oncogenic transformation induced by membrane-targeted Akt2 and Akt3. Oncogene,2001.20(32):p.4419-23.
    100. Cheng, J.Q., et al., Transforming activity and mitosis-related expression of the AKT2 oncogene:evidence suggesting a link between cell cycle regulation and oncogenesis. Oncogene,1997.14(23):p.2793-801.
    101. Sun, M., et al., AKT1/PKBalpha kinase is frequently elevated in human cancers and its constitutive activation is required for oncogenic transformation in NIH3T3 cells. Am J Pathol,2001.159(2):p.431-7.
    102. Mirza, A.M., et al., Oncogenic transformation of cells by a conditionally active form of the protein kinase Akt/PKB. Cell Growth Differ,2000.11(6):p. 279-92.
    103. Raucci, A., et al., Osteoblast proliferation or differentiation is regulated by relative strengths of opposing signaling pathways. J Cell Physiol,2008.215(2): p.442-51.
    104. Magun, R., et al., Expression of a constitutively activated form of protein kinase B (c-Akt) in 3T3-L1 preadipose cells causes spontaneous differentiation. Endocrinology,1996.137(8):p.3590-3.
    105. Xu, J. and K. Liao, Protein kinase B/AKT 1 plays a pivotal role in insulin-like growth factor-1 receptor signaling induced 3T3-L1 adipocyte differentiation. J Biol Chem,2004.279(34):p.35914-22.
    106. Kaneko, S., et al., Positive feedback regulation between Akt2 and MyoD during muscle differentiation. Cloning of Akt2 promoter. J Biol Chem,2002. 277(26):p.23230-5.
    107. Vandromme, M., et al., Protein kinase B beta/Akt2 plays a specific role in muscle differentiation. J Biol Chem,2001.276(11):p.8173-9.
    108. Peng, X.D., et al., Dwarfism, impaired skin development, skeletal muscle atrophy, delayed bone development, and impeded adipogenesis in mice lacking Akt1 and Akt2. Genes Dev,2003.17(11):p.1352-65.
    109. Cross, D.A., et al., Inhibition of glycogen synthase kinase-3 by insulin mediated by protein kinase B. Nature,1995.378(6559):p.785-9.
    110. Campbell, P.M. and M. Szyf, Human DNA methyltransferase gene DNMT1 is regulated by the APC pathway. Carcinogenesis,2003.24(1):p.17-24.
    111. Lin, R.K., et al., The tobacco-specific carcinogen NNK induces DNA methyltransferase 1 accumulation and tumor suppressor gene hypermethylation in mice and lung cancer patients. J Clin Invest.120(2):p. 521-32.
    112. Hodge, D.R., et al., Interleukin-6 regulation of the human DNA methyltransferase (HDNMT) gene in human erythroleukemia cells. J Biol Chem,2001.276(43):p.39508-11.
    113. Hodge, D.R., et al., Interleukin 6 supports the maintenance of p53 tumor suppressor gene promoter methylation. Cancer Res,2005.65(11):p.4673-82.
    114. Hodge, D.R., et al., IL-6 enhances the nuclear translocation of DNA cytosine-5-methyltransferase 1 (DNMT1) via phosphorylation of the nuclear localization sequence by the AKT kinase. Cancer Genomics Proteomics,2007. 4(6):p.387-98.
    115. Lu, R., et al., Inhibition of the extracellular signal-regulated kinase/mitogen-activated protein kinase pathway decreases DNA methylation in colon cancer cells. J Biol Chem,2007.282(16):p.12249-59.
    116. Hartmann, W., et al., Phosphatidylinositol 3'-kinase/AKT signaling is activated in medulloblastoma cell proliferation and is associated with reduced expression of PTEN. Clin Cancer Res,2006.12(10):p.3019-27.
    117. Montiel-Duarte, C., et al., Resistance to Imatinib Mesylate-induced apoptosis in acute lymphoblastic leukemia is associated with PTEN down-regulation due to promoter hypermethylation. Leuk Res,2008.32(5):p.709-16.
    118. Leu, Y.W., et al., Double RNA interference of DNMT3b and DNMT1 enhances DNA demethylation and gene reactivation. Cancer Res,2003.63(19):p. 6110-5.
    119. Xing, J., et al., Expression of methylation-related genes is associated with overall survival in patients with non-small cell lung cancer. Br J Cancer,2008. 98(10):p.1716-22.
    120. Phillips, J.M., L.D. Burgoon, and J.I. Goodman, Phenobarbital elicits unique, early changes in the expression of hepatic genes that affect critical pathways in tumor-prone B6C3F1 mice. Toxicol Sci,2009.109(2):p.193-205.
    121. Hajduch, E., G.J. Litherland, and H.S. Hundal, Protein kinase B (PKB/Akt)--a key regulator of glucose transport? FEBS Lett,2001.492(3):p.199-203.
    122. Nemoto, T., et al., Regulation of Akt mRNA and protein levels by glycogen synthase kinase-3beta in adrenal chromaffin cells:effects of LiCl and SB216763. Eur J Pharmacol,2008.586(1-3):p.82-9.
    123. Kunnimalaiyaan, M., M. Ndiaye, and H. Chen, Apoptosis-mediated medullary thyroid cancer growth suppression by the PI3K inhibitor LY294002. Surgery, 2006.140(6):p.1009-14; discussion 1014-5.
    124. Fatrai, S., et al., Akt induces beta-cell proliferation by regulating cyclin Dl, cyclin D2, and p21 levels and cyclin-dependent kinase-4 activity. Diabetes, 2006.55(2):p.318-25.
    125. Xiong, Y., et al., Histone deacetylase inhibitors decrease DNA methyltransferase-3B messenger RNA stability and down-regulate de novo DNA methyltransferase activity in human endometrial cells. Cancer Res,2005. 65(7):p.2684-9.
    126. Yanagisawa, Y., et al., The human DNA methyltransferases DNMT3A and DNMT3B have two types of promoters with different CpG contents. Biochim Biophys Acta,2002.1577(3):p.457-65.
    127. He, L. and G.J. Hannon, MicroRNAs:small RNAs with a big role in gene regulation. Nat Rev Genet,2004.5(7):p.522-31.
    128. Jinawath, A., et al., Transcriptional regulation of the human DNA methyltransferase 3A and 3B genes by Sp3 and Sp1 zinc finger proteins. Biochem J,2005.385(Pt 2):p.557-64.
    129. Park, S.Y, H.M. Shin, and T.H. Han, Synergistic interaction of MEF2D and Spl in activation of the CD14 promoter. Mol Immunol,2002.39(1-2):p. 25-30.
    130. Pore, N., et al., Sp1 is involved in Akt-mediated induction of VEGF expression through an HIF-1-independent mechanism. Mol Biol Cell,2004.15(11):p. 4841-53.
    131. Zhang, X., et al., Sp1 and Sp3 transcription factors synergistically regulate HGF receptor gene expression in kidney. Am J Physiol Renal Physiol,2003. 284(1):p. F82-94.
    132. Qureshi, H.Y., et al., Requirement of phosphatidylinositol 3-kinase/Akt signaling pathway for regulation of tissue inhibitor of metalloproteinases-3 gene expression by TGF-beta in human chondrocytes. Cell Signal,2007.19(8): p.1643-51.
    133. Lin, H.C., T.Y. Song, and M.L. Hu, S-Adenosylhomocysteine promotes the invasion of C6 glioma cells via increased secretion of matrix metalloproteinase-2 in murine microglial BV2 cells. Toxicol Sci,2009.112(2): p.322-30.
    134. Burgering, B.M. and R.H. Medema, Decisions on life and death:FOXO Forkhead transcription factors are in command when PKB/Akt is off duty. J Leukoc Biol,2003.73(6):p.689-701.
    135. Li, Q. and I.M. Verma, NF-kappaB regulation in the immune system. Nat Rev Immunol,2002.2(10):p.725-34.
    136. Du, K. and M. Montminy, CREB is a regulatory target for the protein kinase Akt/PKB. J Biol Chem,1998.273(49):p.32377-9.
    137. Mayo, L.D. and D.B. Donner, A phosphatidylinositol 3-kinase/Akt pathway promotes translocation of Mdm2 from the cytoplasm to the nucleus. Proc Natl Acad Sci U S A,2001.98(20):p.11598-603.
    138. Hervouet, E., F.M. Vallette, and P.F. Cartron, Dnmt3/transcription factor interactions as crucial players in targeted DNA methylation. Epigenetics, 2009.4(7):p.487-99.
    1. Fischle, W., Y. Wang, and C.D. Allis, Binary switches and modification cassettes in histone biology and beyond. Nature,2003.425(6957):p.475-9.
    2. Bird, A., DNA methylation patterns and epigenetic memory. Genes Dev,2002. 16(1):p.6-21.
    3. Jones, P.A. and S.B. Baylin, The epigenomics of cancer. Cell,2007.128(4):p. 683-92.
    4. Yoder, J.A., C.P. Walsh, and T.H. Bestor, Cytosine methylation and the ecology of intragenomic parasites. Trends Genet,1997.13(8):p.335-40.
    5. Song, F., et al., Association of tissue-specific differentially methylated regions (TDMs) with differential gene expression. Proc Natl Acad Sci U S A,2005. 102(9):p.3336-41.
    6. Li, E., T.H. Bestor, and R. Jaenisch, Targeted mutation of the DNA methyltransferase gene results in embryonic lethality. Cell,1992.69(6):p. 915-26.
    7. Okano, M., et al., DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell,1999.99(3):p. 247-57.
    8. Esteller, M., CpG island hypermethylation and tumor suppressor genes:a booming present, a brighter future. Oncogene,2002.21(35):p.5427-40.
    9. Csankovszki, G., A. Nagy, and R. Jaenisch, Synergism of Xist RNA, DNA methylation, and histone hypoacetylation in maintaining X chromosome inactivation. J Cell Biol,2001.153(4):p.773-84.
    10. Bestor, T.H., Imprinting errors and developmental asymmetry. Philos Trans R Soc Lond B Biol Sci,2003.358(1436):p.1411-5.
    11. Jones, P.A. and S.B. Baylin, The fundamental role of epigenetic events in cancer. Nat Rev Genet,2002.3(6):p.415-28.
    12. Ramsahoye, B.H., et al., Non-CpG methylation is prevalent in embryonic stem cells and may be mediated by DNA methyltransferase 3a. Proc Natl Acad Sci U S A,2000.97(10):p.5237-42.
    13. Robertson, K.D., DNA methylation and chromatin-unraveling the tangled web. Oncogene,2002.21(35):p.5361-79.
    14. Jeltsch, A., Beyond Watson and Crick:DNA methylation and molecular enzymology of DNA methyltransferases. Chembiochem,2002.3(4):p.274-93.
    15. Yoder, J.A., et al., DNA (cytosine-5)-methyltransferases in mouse cells and tissues. Studies with a mechanism-based probe. J Mol Biol,1997.270(3):p. 385-95.
    16. Tollefsbol,T.O. and C.A. Hutchison,3rd, Control of methylation spreading in synthetic DNA sequences by the murine DNA methyltransferase. J Mol Biol, 1997.269(4):p.494-504.
    17. Xie, S., et al., Cloning, expression and chromosome locations of the human DNMT3 gene family. Gene,1999.236(1):p.87-95.
    18. Xu, G.L., et al., Chromosome instability and immunodeficiency syndrome caused by mutations in a DNA methyltransferase gene. Nature,1999. 402(6758):p.187-91.
    19. Tuck-Muller, C.M., et al., DNA hypomethylation and unusual chromosome instability in cell lines from ICF syndrome patients. Cytogenet Cell Genet, 2000.89(1-2):p.121-8.
    20. Fabbri, M., et al., MicroRNA-29 family reverts aberrant methylation in lung cancer by targeting DNA methyltransferases 3A and 3B. Proc Natl Acad Sci U S A,2007.104(40):p.15805-10.
    21. Chedin, F., M.R. Lieber, and C.L. Hsieh, The DNA methyltransferase-like protein DNMT3L stimulates de novo methylation by Dnmt3a. Proc Natl Acad Sci U S A,2002.99(26):p.16916-21.
    22. Suetake, I., et al., DNMT3L stimulates the DNA methylation activity of Dnmt3a and Dnmt3b through a direct interaction. J Biol Chem,2004.279(26):p. 27816-23.
    23. Goll, M.G. and T.H. Bestor, Eukaryotic cytosine methyltransferases. Annu Rev Biochem,2005.74:p.481-514.
    24. Gopalakrishnan, S., B.O. Van Emburgh, and K.D. Robertson, DNA methylation in development and human disease. Mutat Res,2008.647(1-2):p.30-8.
    25. Brena, R.M. and J.F. Costello, Genome-epigenome interactions in cancer. Hum Mol Genet,2007.16 Spec No 1:p. R96-105.
    26. Feinberg, A.P. and B. Tycko, The history of cancer epigenetics. Nat Rev Cancer, 2004.4(2):p.143-53.
    27. Widschwendter, M., et al., DNA hypomethylation and ovarian cancer biology. Cancer Res,2004.64(13):p.4472-80.
    28. Esteller, M. and J.G. Herman, Cancer as an epigenetic disease:DNA methylation and chromatin alterations in human tumours. J Pathol,2002. 196(1):p.1-7.
    29. Herman, J.G. and S.B. Baylin, Gene silencing in cancer in association with promoter hypermethylation. N Engl J Med,2003.349(21):p.2042-54.
    30. Esteller, M., Cancer epigenomics:DNA methylomes and histone-modification maps. Nat Rev Genet,2007.8(4):p.286-98.
    31. Rideout, W.M.,3rd, et al.,5-Methylcytosine as an endogenous mutagen in the human LDL receptor and p53 genes. Science,1990.249(4974):p.1288-90.
    32. Magewu, A.N. and P.A. Jones, 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. Mol Cell Biol,1994.14(6):p.4225-32.
    33. Robertson, K.D. and P.A. Jones, DNA methylation:past, present and future directions. Carcinogenesis,2000.21(3):p.461-7.
    34. Yoon, J.H., et al., Methylated CpG dinucleotides are the preferential targets for G-to-T transversion mutations induced by benzo[a]pyrene diol epoxide in mammalian cells:similarities with the p53 mutation spectrum in smoking-associated lung cancers. Cancer Res,2001.61(19):p.7110-7.
    35. Pfeifer, G.P., M. Tang, and M.F. Denissenko, Mutation hotspots and DNA methylation. Curr Top Microbiol Immunol,2000.249:p.1-19.
    36. Gronbaek, K., C. Hother, and P.A. Jones, Epigenetic changes in cancer. APMIS, 2007.115(10):p.1039-59.
    37. Nakayama, M., et al., Hypomethylation status of CpG sites at the promoter region and over expression of the human MDR1 gene in acute myeloid leukemias. Blood,1998.92(11):p.4296-307.
    38. Sakatani, T., et al., Loss of imprinting of Igf2 alters intestinal maturation and tumorigenesis in mice. Science,2005.307(5717):p.1976-8.
    39. Hoffmann, M.J. and W.A. Schulz, Causes and consequences of DNA hypomethylation in human cancer. Biochem Cell Biol,2005.83(3):p.296-321.
    40. Ehrlich, M., DNA hypomethylation, cancer, the immunodeficiency, centromeric region instability, facial anomalies syndrome and chromosomal rearrangements. J Nutr,2002.132(8 Suppl):p.2424S-2429S.
    41. Ehrlich, M., The ICF syndrome, a DNA methyltransferase 3B deficiency and immunodeficiency disease. Clin Immunol,2003.109(1):p.17-28.
    42. Fackenthal, J.D. and L.A. Godley, Aberrant RNA splicing and its functional consequences in cancer cells. Dis Model Mech,2008.1(1):p.37-42.
    43. Ostler, K.R., et al., Cancer cells express aberrant DNMT3B transcripts encoding truncated proteins. Oncogene,2007.26(38):p.5553-63.
    44. Weisenberger, D.J., et al., Role of the DNA methyltransferase variant DNMT3b3 in DNA methylation. Mol Cancer Res,2004.2(1):p.62-72.
    45. Saito, Y., et al., Overexpression of a splice variant of DNA methyltransferase 3b, DNMT3b4, associated with DNA hypomethylation on pericentromeric satellite regions during human hepatocarcinogenesis. Proc Natl Acad Sci U S A,2002.99(15):p.10060-5.
    46. Robertson, K.D., et al., The human DNA methyltransferases (DNMTs) 1,3a and 3b:coordinate mRNA expression in normal tissues and overexpression in tumors. Nucleic Acids Res,1999.27(11):p.2291-8.
    47. Kanai, Y. and S. Hirohashi, Alterations of DNA methylation associated with abnormalities of DNA methyltransferases in human cancers during transition from a precancerous to a malignant state. Carcinogenesis,2007.28(12):p. 2434-42.
    48. Arai, E., et al., Regional DNA hypermethylation and DNA methyltransferase (DNMT) 1 protein overexpression in both renal tumors and corresponding nontumorous renal tissues. Int J Cancer,2006.119(2):p.288-96.
    49. Peng, D.F., et al., DNA methylation of multiple tumor-related genes in association with overexpression of DNA methyltransferase 1 (DNMT1) during multistage carcinogenesis of the pancreas. Carcinogenesis,2006.27(6):p. 1160-8.
    50. Melki, J.R., et al., Increased DNA methyltransferase expression in leukaemia. Leukemia,1998.12(3):p.311-6.
    51. Mizuno, S., et al., Expression of DNA methyltransferases DNMT1,3A, and 3B in normal hematopoiesis and in acute and chronic myelogenous leukemia. Blood,2001.97(5):p.1172-9.
    52. Beaulieu, N., et al., An essential role for DNA methyltransferase DNMT3B in cancer cell survival. J Biol Chem,2002.277(31):p.28176-81.
    53. Eads, C.A., et al., CpG island hypermethylation in human colorectal tumors is not associated with DNA methyltransferase overexpression. Cancer Res,1999. 59(10):p.2302-6.
    54. Girault, I., et al., Expression analysis of DNA methyltransferases 1,3A, and 3B in sporadic breast carcinomas. Clin Cancer Res,2003.9(12):p.4415-22.
    55. Jin, F., et al., Up-regulation of DNA methyltransferase 3B expression in endometrial cancers. Gynecol Oncol,2005.96(2):p.531-8.
    56. Patra, S.K., et al., DNA methyltransferase and demethylase in human prostate cancer. Mol Carcinog,2002.33(3):p.163-71.
    57. Robert, M.F., et al., DNMT1 is required to maintain CpG methylation and aberrant gene silencing in human cancer cells. Nat Genet,2003.33(1):p. 61-5.
    58. Saito, Y., et al., Increased protein expression of DNA methyltransferase (DNMT) 1 is significantly correlated with the malignant potential and poor prognosis of human hepatocellular carcinomas. Int J Cancer,2003.105(4):p.527-32.
    59. Watt, F. and P.L. Molloy, Cytosine methylation prevents binding to DNA of a HeLa cell transcription factor required for optimal expression of the adenovirus major late promoter. Genes Dev,1988.2(9):p.1136-43.
    60. Klose, R.J. and A.P. Bird, Genomic DNA methylation:the mark and its mediators. Trends Biochem Sci,2006.31(2):p.89-97.
    61. Sarraf, S.A. and I. Stancheva, Methyl-CpG binding protein MBD1 couples histone H3 methylation at lysine 9 by SETDB1 to DNA replication and chromatin assembly. Mol Cell,2004.15(4):p.595-605.
    62. Esteller, M., Epigenetics in cancer. N Engl J Med,2008.358(11):p.1148-59.
    63. He, L. and G.J. Hannon, MicroRNAs:small RNAs with a big role in gene regulation. Nat Rev Genet,2004.5(7):p.522-31.
    64. Calin, G.A. and C.M. Croce, MicroRNA signatures in human cancers. Nat Rev Cancer,2006.6(11):p.857-66.
    65. Chen, C.Z., MicroRNAs as oncogenes and tumor suppressors. N Engl J Med, 2005.353(17):p.1768-71.
    66. Lu, J., et al., MicroRNA expression profiles classify human cancers. Nature, 2005.435(7043):p.834-8.
    67. Lujambio, A. and M. Esteller, CpG island hypermethylation of tumor suppressor microRNAs in human cancer. Cell Cycle,2007.6(12):p.1455-9.
    68. Lujambio, A., et al., Genetic unmasking of an epigenetically silenced microRNA in human cancer cells. Cancer Res,2007.67(4):p.1424-9.
    69. Saito, Y. and P.A. Jones, Epigenetic activation of tumor suppressor microRNAs in human cancer cells. Cell Cycle,2006.5(19):p.2220-2.
    70. Saito, Y., et al., Specific activation of microRNA-127 with downregulation of the proto-oncogene BCL6 by chromatin-modifying drugs in human cancer cells. Cancer Cell,2006.9(6):p.435-43.
    71. Johnson, S.M., et al., RAS is regulated by the let-7 microRNA family. Cell, 2005.120(5):p.635-47.
    72. Cimmino, A., et al., miR-15 and miR-16 induce apoptosis by targeting BCL2. Proc Natl Acad Sci U S A,2005.102(39):p.13944-9.
    73. Issa, J.P., Aging, DNA methylation and cancer. Crit Rev Oncol Hematol,1999. 32(1):p.31-43.
    74. Wilson, V.L. and P.A. Jones, DNA methylation decreases in aging but not in immortal cells. Science,1983.220(4601):p.1055-7.
    75. Bollati, V., et al., Decline in genomic DNA methylation through aging in a cohort of elderly subjects. Mech Ageing Dev,2009.130(4):p.234-9.
    76. Casillas, M.A., Jr., et al., Transcriptional control of the DNA methyltransferases is altered in aging and neoplastically-transformed human fibroblasts. Mol Cell Biochem,2003.252(1-2):p.33-43.
    77. Issa, J.P., Age-related epigenetic changes and the immune system. Clin Immunol,2003.109(1):p.103-8.
    78. Nguyen, C., et al., Susceptibility of nonpromoter CpG islands to de novo methylation in normal and neoplastic cells. J Natl Cancer Inst,2001.93(19):p. 1465-72.
    79. Issa, J.P., et al., Methylation of the oestrogen receptor CpG island links ageing and neoplasia in human colon. Nat Genet,1994.7(4):p.536-40.
    80. Issa, J.P., et al., Accelerated age-related CpG island methylation in ulcerative colitis. Cancer Res,2001.61(9):p.3573-7.
    81. Calvanese, V., et al., The role of epigenetics in aging and age-related diseases. Ageing Res Rev,2009.8(4):p.268-76.
    82. Choi, S.W., et al., Folate supplementation increases genomic DNA methylation in the liver of elder rats. Br J Nutr,2005.93(1):p.31-5.
    83. Pufulete, M., et al., Effect of folic acid supplementation on genomic DNA methylation in patients with colorectal adenoma. Gut,2005.54(5):p.648-53.
    84. Hervouet, E., et al., Folate supplementation limits the aggressiveness of glioma via the remethylation of DNA repeats element and genes governing apoptosis and proliferation. Clin Cancer Res,2009.15(10):p.3519-29.
    85. Kaminskas, E., et al., Approval summary:azacitidine for treatment of myelodysplastic syndrome subtypes. Clin Cancer Res,2005.11(10):p.3604-8.
    86. Marquez, V.E., et al., Zebularine:a unique molecule for an epigenetically based strategy in cancer chemotherapy. Ann N Y Acad Sci,2005.1058:p. 246-54.
    87. Lu, Q., et al., Epigenetics, disease, and therapeutic interventions. Ageing Res Rev,2006.5(4):p.449-67.
    88. Winquist, E., et al., Phase Ⅱ trial of DNA methyltransferase 1 inhibition with the antisense oligonucleotide MG98 in patients with metastatic renal carcinoma:a National Cancer Institute of Canada Clinical Trials Group investigational new drug study. Invest New Drugs,2006.24(2):p.159-67.
    89. Brueckner, B., et al., Epigenetic reactivation of tumor suppressor genes by a novel small-molecule inhibitor of human DNA methyltransferases. Cancer Res, 2005.65(14):p.6305-11.
    90. Leu, Y.W., et al., Double RNA interference of DNMT3b and DNMT1 enhances DNA demethylation and gene reactivation. Cancer Res,2003.63(19):p. 6110-5.
    91. Suzuki, M., et al., RNA interference-mediated knockdown of DNA methyltransferase 1 leads to promoter demethylation and gene re-expression in human lung and breast cancer cells. Cancer Res,2004.64(9):p.3137-43.

© 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