Electronic journal of oncology
Volume 98 • N◦ 7 • juillet 2011 John Libbey Eurotext
©
Epigenetic changes in osteosarcoma Article received on September 12, 2010, accepted on December 14, 2010 Reprints: J. Cui
Juncheng Cui1 , Wanchun Wang1 , Zhihong Li1 , Zhaogui Zhang1 , Bei Wu2 , Li Zeng3 1
Central South University, The Second Xiangya Hospital, Department of Orthopedics, Changsha, Hunan, China 2 Central South University, Xiangya Hospital, Department of Orthopedics, Changsha, Hunan, China 3 The First Affiliate Hospital of Nanhua University, Department of Medical Cosmetology, Hengyang, Hunan, China
To cite this article: Cui J, Wang W, Li Z, Zhang Z, Wu B, Zeng L. Epigenetic changes in osteosarcoma. Bull Cancer 2011 ; 98 : E62-E68. doi : 10.1684/bdc.2011.1400.
Abstract. Osteosarcoma is one of the most prevalent primary bone tumors. The pathogenesis and molecular development of this tumor remains elusive. The prognosis is unfavorable due to lack of effective treatment methods. Recent advances in the epigenetics have brought a profound impact on the understanding of molecular mechanisms that lead to osteosarcoma. In this review, we summarized the current literature on
O
steosarcoma is one of the most prevalent primary bone tumors. The age distribution of this tumor is bimodal, with peaks in adolescence and above the age of 50 years [1]. The pathogenesis and molecular mechanisms of this tumor remain elusive. Although several effective treatment options are available, local recurrence (4-8%) and postoperative pulmonary metastasis of osteosarcoma (about 30%) are frequent, the prognosis of osteosarcoma is unfavorable [2]. Recent progresses in the epigenetics research have shown that epigenetic changes appear to contribute to the malignant transformation and progression of osteosarcoma, providing a better understanding of the molecular mechanisms of this tumor.
E62
Key words: osteosarcoma, epigenetics, DNA methylation, histone modifications
DNA methylation DNA methylation refers to the addition of a methyl group to the 5 -carbon of cytosine in CpG sequences, catalyzed by DNA methyltransferases (DNMTs) [4, 5]. A study of the human genome revealed that the distribution of CpG dinucleotides is not random. Some of CpG dinucleotides cluster together to form CpG-rich DNA regions called CpG islands, where methylcytosine residues are often found in CpG islands are 0.5 to 2 kb long and found in the 5 region of approximately 60% of genes [6]. Most CpG islands in actively expressed genes are unmethylated, with the exception of certain imprinted genes and genes on the inactive X chromosomes of females [7, 8]. However, during cancer development, DNA methylation in cancer cells exhibits inverse profiles compared with normal cells [9, 10]. DNA methylation aberrations can occur as either hypermethylation or hypomethylation. Both forms can lead to chromosomal instability and transcriptional gene silencing, both forms have been implicated in a variety of human malignancies, including osteosarcoma.
DNA hypermethylation DNA hypermethylation is a well-investigated epigenetic abnormality seen in several malignancies. Aberrant methylation of gene promoters has been Bull Cancer vol. 98 • N◦ 7 • juillet 2011
doi : 10.1684/bdc.2011.1400
Epigenetics refers to the research of heritable changes in gene expression without the change in gene sequence [3]. It is one of the most rapidly expanding fields in cancer related research. DNA methylation and histone modifications are two important, well-studied epigenetic mechanisms in osteosarcoma. These two processes can act either independently or dependently affecting the gene expression and in turn the tumorigenesis. Here, we provide an overview of the current understanding of epigenetic changes associated with osteosarcoma, and the potential diagnostic and therapeutic applications in osteosarcoma.
epigenetic changes that are thought to contribute to the carcinogenesis of osteosarcoma, and discussed the potential diagnostic and therapeutic applications as well as future areas of research.
Epigenetic changes in osteosarcoma
hypothesized as an early event in the neoplastic progression [11]. DNA hypermethylation contributes to gene silencing by preventing the binding of activating transcription factors and by attracting repressor complexes that induce the formation of inactive chromatin structures. Numerous tumor suppressor genes have been identified as being regulated by DNA hypermethylation in different types of cancer, such as breast tumor and malignant melanoma [12, 13]. With regard to osteosarcoma, the exact function of most of these aberrantly silenced genes is still unknown; however, a few studies have indicated that some aberrantly silenced genes are closely associated with osteosarcoma progression. Besides, these genes involved in a number of cellular pathways such as cell cycle regulation, apoptosis, signal transduction, tumor cell proliferation and differentiation and growth arrest. For many of these genes, CpG island DNA hypermethylation seems to play a central role in osteosarcoma progression and metastasis, as summarized in table 1.
by demethylases including 5-methylcytosine glycosylase and MBD2b [39], it can causes breakdown of such a defense mechanism, and lead to structural and functional alterations of the genome. There are two types of hypomethylation: global or localized hypomethylation. Global hypomethylation refers to overall decrease in 5-methylcytosine content in the genome. Localized or gene specific hypomethylation denotes to a decrease in cytosine methylation relative to the “normal” methylation level. It can affect several specific regions of the genome, such as the promoter regions of oncogenes or normally highly methylated sequences such as repetitive sequences and oncogenes [40]. Both global hypomethylation and localized hypomethylation have been implicated in human cancer. Genes, which have been identified to be aberrantly hypomethylated in osteosarcoma are summarized in table 2.
Histone modification DNA hypomethylation DNA hypomethylation is another type of methylation change related to epigenetic aberration in many malignancies including osteosarcomas. DNA methylation of normal genomes is a defensive mechanism by which repetitive DNA is transcriptionally silenced to prevent it from propagating [38]. Hypomethylation is catalyzed
The basic structural unit of chromatin is the nucleosome, a complex structure composed of an octamer of four core histones, H2A, H2B, H3 and H4. Histones have been critically implicated in regulation of chromatin structure and gene expression. They comprise a globular domain and a more flexible and charged NH2 terminal, which is called as histone
Table 1. Hypermethylated genes in osteosarcomas. Genes
Common names
Genes ID
Functions
CDK4
Cyclin-dependent kinase 4
1019
Amplification and cell cycle apoptosis [14]
ChM-I
Chondromodulin-I
162840
Growth arrest
[15,16]
EGFR
Epidermal growth factor receptor
1956
Amplifications
[17–19]
GADD45A Growth arrest and DNA-damage-inducible 45 alpha 13197
Apoptosis
[20–22]
IGF2
Insulin-like growth factor 2
3481
Signal transduction
[23,24]
INK4A
Cyclin-dependent kinase inhibitor 2A
1029
Cell cycle regulation
[25–28]
P53
p53 tumor suppressor homologue
100384887 Cell cycle regulation and apoptosis
[29,30]
RASSF1A
RAS association domain family protein 1A
11186
Signal transduction
[31,32]
RECQL4
RecQ protein-like 4
9401
Mutations
[33,34]
RUNX2
Runt-related transcription factor 2
367218
Signal transduction
[35,36]
WIF-1
WNT inhibitory factor 1
11197
Proliferation and differentiation
[37]
Bull Cancer vol. 98 • N◦ 7 • juillet 2011
References
E63
J. Cui, et al. Table 2. Hypomethylated genes in osteosarcomas. Genes
Common names
Genes ID
Functions
References
FOS
FBJ osteosarcoma oncogene
314322
Amplifications
[41,42]
H19
H19, imprinted maternally expressed transcript
283120
Growth arrest
[43]
MYC
V-myc myelocytomatosis viral oncogene homologue
4609
Amplifications
[44–46]
PHLDA2
Pleckstrin homology-like domain, family A, member 2
22113
Apoptosis
[43]
“tail”. These tails are subject to a variety of covalent modifications including acetylation, methylation, phosphorylation, ubiquitylation and sumoylation [47, 48]. These modifications are involved in regulation of gene expression. The most widely studied modifications are histone acetylation and deacetylations. They are regulated by histone acetyltransferases (HAT) and histone deacetylases (HDAC), respectively. It has been recognized in recent years that the HDACs play a fundamental role in keeping the balance between the acetylated and deacetylated state of chromatin [49]. Consequently, considerable effort has been devoted to developing HDAC inhibitors (HDACi), which have been found to induce numerous anti-proliferative effects including apoptosis, cytostasis and differentiation [50]. In osteosarcomas, several genes of biological significance may be potentially regulated by histone modification. One such gene is p21WAF1, which arrests cell growth by inhibiting cyclin/Cdk complexes and can act as tumor suppressor gene by negatively regulating the cell cycle [51]. Decreased expression of the p21WAF1 has been detected in osteosarcoma and is associated with the malignancy [52]. Maeda et al. had shown that p21WAF1 was upregulated by HDACi including TSA and sodium butyrate in MG63 cells. Moreover, the cell cycle analysis revealed that the p21WAF1 arrested MG63 cells in the G2/M phase [53]. Cellular FLICE-inhibitory protein (cellular FLIP) is another gene regulated by histone modification in osteosarcomas. It is an inhibitor of Fas-mediated activation of caspase-8. HDACi including FR901228 were shown to induce downregulation of cellular FLIP through inhibiting generation of FLIP mRNA, The downregulation of cellular FLIP renders Fas-resistant osteosarcoma cell lines become more sensitive to Fasmediated apoptosis [54].
E64
Interactions between DNA methylation and histone modifications Both DNA methylation and histone acetylation has been suggested to modulate gene expression [55-57]. It has been shown that methylated DNA recruits HDAC through methyl-DNA binding proteins (MBPs). This interaction then recruits HDAC activity to methylated promoters which results in gene silencing [58]. Furthermore, DNMTs can directly recruit HDAC activity to silence gene expression [59, 60]. Cheng et al. have demonstrated that monomethyl histone H3 lysine 27 is expressed in staurosporine-induced apoptotic osteosarcoma cell lines in vitro [61]. Chondromodulin-I (ChM-I) is a vascular endothelial cell growth inhibitor purified from bovine epiphyseal cartilage [62], the expression of this gene is regulated by the binding of Sp3 to the core promoter region, which is inhibited by the methylation of CpG in the target genome in the osteogenic lineage, osteosarcoma cells [15]. The histone tails associated with the hypermethylated promoter region of the ChM-I gene were deacetylated by HDAC2 in three ChM-I- negative osteosarcoma cell lines. Treatment with an HDACi induces the binding of Sp3 in one cell line, which became ChM-I-positive. This process was associated with acetylation instead of the dimethylation of histone H3 at lysine 9 (H3-K9) and the demethylation of the core promoter region. The demethylation was transient. It is gradually replaced by methylation after a rapid recovery of histone deacetylaion [63]. These studies provide evidence that the plasticity of differentiation being regulated by the cell- specific plasticity of epigenetic regulation and provide a rationale for a combined treatment with DNA methylation and HDACi in reversing the epigenetic silencing of key tumor suppressor genes. Bull Cancer vol. 98 • N◦ 7 • juillet 2011
Epigenetic changes in osteosarcoma
Epigenetic changes as biomarkers Early detection is the most efficient way to decrease mortality of osteosarcoma. Specific epigenetic changes have been found occurring early in multistep carcinogenesis of osteosarcomas. These changes have been considered as potential and promising biomarkers for early detection of this tumor. To be clinically applicable, the biomarkers must be readily detectable in clinical specimens such as cancer tissues, serum and body fluids. MTA is a component of histone deacetylation (NuRD) complex, which is associated with ATP-dependent chromatin remodeling and HDAC activity [64]. Park et al. demonstrated in their study that the ezrin and MTA positivity can be additional prognostic markers for osteosarcoma of the jaw [65]. p16INK4a is the G1 cell-cycle checkpoint protein that is responsible for maintaining the balance of cellular proliferation. It is a subject of frequent involvement in osteosarcomas. Importantly, hypermethylation of the p16INK4A promoter is significantly associated with malignancy and metastasis [66]. Maitra et al. examined a series of 38 pediatric osteosarcomas with 16% loss of p16INK4a expression, while absence of p16INK4a expression significantly correlated with decreased survival. These findings suggested that immuno-histochemical analysis of p16INK4a expression in osteosarcomas may be a useful biomarker of prognosis [67]. Furthermore, studies are needed so that new biomarkers may be able not only to prognosticate osteosarcoma patients but also to serve as therapeutic targets and thereby improve survival rates in the routine clinical practice.
Therapeutic targets Unlike genetic events that occur at tumor suppressor genes, epigenetic modifications are heritable and potentially reversible. Therefore, epigenetic modifications can be used as potential therapeutic targets. At present, the most widely investigated drugs can be classified in two types: (1) DNMT inhibitors, such as 5-azacytidine (azacitidine) and 5-aza-2 -deoxycytidine (5-aza-dC), (2) HDACi.
deoxynucleotide triphosphates. Eventually, deoxynucleotide triphosphates can incorporate into RNA (5AC) and/or DNA (5AC and DAC) in place of cytosine [68]. Nucleoside analogue inhibitors can also induce hypomethylation by depletion of cellular DNMTs [69]. Azacitidine is the first DNA hypomethylating agent approved by FDA for the treatment of myelodysplastic syndromes. Marcucci et al. [70] demonstrated a single dose of azacitidine at 75 mg/m2 that is well tolerated in adult population. Myelosuppression is a known side effect of this drug, which may limit their clinical utility. Zebularine is the first orally bioavailable drug in this group, it is chemically stable and significantly less cytotoxic [71]. Other categories drugs are being studied, such as non-nucleoside analogue DNA methylation inhibitors, including epigallocatechin-3-gallate (EGCG), procaine, procainamide and hydralazine, DNMT1 antisense oligonucleotides (MG98).
HDAC inhibitors A number of compounds exhibit HDAC inhibitory activity, which are commonly used including the valproic acid (VPA), the trichostatin A (TSA), the benzamides SNDX-275 (formerly MS-275) and MGCD0103 and the cyclic peptide FK-228. A recent research by Wittenburg et al. demonstrated that pretreatment of OS cell lines with VPA followed by doxorubicin (DOX) resulted in significant growth inhibition and potentiation of apoptosis, associated with a dose-dependent increase in nuclear DOX accumulation [72]. Suberoyl anilide hydroxamic acid (SAHA), which is structurally similar to TSA, is the first approved HDACi for clinical use in cancer patients by the FDA [73]. Kisseberth et al. provided evidence that SAHA induced apoptosis in cells from osteosarcoma and histiocytic sarcoma cell lines in vitro [74]. Blattmann et al. demonstrated that SAHA induced an inhibition of cell proliferation and clonogenic survival and led to a significant radiosensitization in OS cell lines [75]. Nevertheless, as a class there are of limited clinical application due to side effects, such as myelosuppression, fatigue, somnolence and gastrointestinal symptoms [76].
Combination therapy DNMT inhibitors Azacitidine and 5-aza-dC are nucleoside analogue inhibitors known to inhibit DNA methylation. The mechanism of action is that they can be converted to the Bull Cancer vol. 98 • N◦ 7 • juillet 2011
The single epigenetic drug activity currently is limited and associated with significant side effects. Thus, it prompted investigation of the pharmacodynamic interactions between DNMTs and HDACi. The
E65
J. Cui, et al.
combinatorial use of these two agents in the clinic is based on their synergistic effects in reestablishing the expression of tumor suppressor genes. Some hypermethylated genes cannot be transcriptionally reactivated with HDACi (TSA) alone in tumor cells, but sequential treatment with 5-azacytosine nucleosides and subsequently with an HDACi (TSA), results in more potent re-expression of methylated tumor suppressor genes than either agent alone [77]. These results indicate that the combined strategies of anticancer therapy may make these drugs more versatile and result in a synergistic effect on various tumor cells. Additionally, the combination therapy may lead to reduction in side effects and the dose of each individual agent.
Summary and future directions Currently, epigenetic changes in tumors are being extensively studied and genome wide analysis of mutated genes in breast and colorectal cancers has been well-annotated [78]. The advance in the human cancer epigenetic field has encouraged us to undertake osteosarcoma epigenome projects to map its epigenetic modifications. The outcome of these projects will enable us to understand the definitely progressive changes in epigenomic patterns during the development of osteosarcoma. And it is essential for us to master the development of epigenetic markers and therapeutics, which will be very important for the application of these advances to diagnosis, prognosis and therapy of osteosarcoma patients. Furthermore, although the current development of epigenetic diagnosis and therapeutics has been demonstrated to be a promising prospect, the more detailed comprehension of the molecular mechanisms underlying the correlation between reversion of epigenetic changes, gene re-expression, therapeutic response and epigenetic drugs is essential. Conflict of interest : none.
References 1. Marcove RC, Heelan RT, Huvos AG, Healey J, Lindeque BG. Osteoid osteoma. Diagnosis, localization, and treatment. Clin Orthop Relat Res 1991 ; 267 : 197-201. 2. Grimer RJ, Taminiau AM, Cannon SR. Surgical outcomes in osteosarcoma. J Bone Joint Surg Br 2002 ; 84 : 395-400. 3. Wolffe AP. Epigenetics: regulation through repression. Science 1999 ; 286 : 481-6.
E66
4. Okano M, Bell DW, Haber DA, Li E. DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell 1999 ; 99 : 247-57. 5. Herman JG, Baylin SB. DNA hypermethylation in tumorigenesis epigenetics joins genetics. Trends Genet 2006 ; 16 : 168-74. 6. Gardiner-Garden M, Frommer M. CpG islands in vertebrate genomes. J Mol Biol 1987 ; 196 : 261-82. 7. Riggs AD, Pfeifer GP. X-chromosome inactivation and cell memory. Trends Genet 1992 ; 8 : 169-74. 8. Razin A, Cedar H. DNA methylation and genomic imprinting. Cell 1994 ; 77 : 473-6. 9. Baylin SB, Ohm JE. Epigenetic gene silencing in cancer – a mechanism for early oncogenic pathway addiction? Nat Rev Cancer 2006 ; 6 : 107-16. 10. Esteller M. Cancer epigenomics: DNA methylomes and histonemodification maps. Nat Rev Genet 2007 ; 8 : 286-98. 11. Warnecke PM, Bestor TH. Cytosine methylation and human cancer. Curr Opin Oncol 2000 ; 12 : 68-73. 12. Yang X, Yan L, Davidson NE. DNA methylation in breast cancer. Endocr Relat Cancer 2001 ; 8 : 115-27. 13. Muthusamy V, Duraisamy S, Bradbury CM, et al. Epigenetic silencing of novel tumor suppressors in malignant melanoma. Cancer Res 2006 ; 66 : 11187-93. 14. Wei G, Lonardo F, Ueda T, et al. CDK4 gene amplification in osteosarcoma: reciprocal relationship with INK4A gene alterations and mapping of 12q13 amplicons. Int J Cancer 1999 ; 80 : 199-204. 15. Aoyama T, Okamoto T, Nagayama S, et al. Methylation in the core-promoter region of the chondromodulin-I gene determines the cell-specific expression by regulating the binding of transcriptional activator Sp3. J Biol Chem 2004 ; 279 : 28789-97. 16. Villagra A, Gutierrez J, Paredes R, et al. Reduced CpG methylation is associated with transcriptional activation of the bone-specific rat osteocalcin gene in osteoblasts. J Cell Biochem 2002 ; 85 : 112-22. 17. Akatsuka T, Wada T, Kokai Y, et al. ErbB2 expression is correlated with increased survival of patients with osteosarcoma. Cancer 2002 ; 94 : 1397-404. 18. Montero AJ, Diaz-Montero CM, Mao L, et al. Epigenetic inactivation of EGFR by CpG island hypermethylation in cancer. Cancer Biol Ther 2006 ; 5 : 1494-501. 19. Hayashi Y, Ueki K, Waha A, Wiestler OD, Louis DN, von Deimling A. Association of EGFR gene amplification and CDKN2 (p16/MTS1) gene deletion in glioblastoma multiforme. Brain Pathol 1997 ; 7 : 871-5. 20. Al-Romaih K, Somers GR, Bayani J, et al. Modulation by decitabine of gene expression and growth of osteosarcoma U2OS cells in vitro and in xenografts: identification of apoptotic genes as targets for demethylation. Cancer Cell Int 2007 ; 7 : 14. 21. Takai D, Jones PA. The CpG island searcher: a new WWW resource. In Silico Biol 2003 ; 3 : 235-40. 22. Al-Romaih K, Sadikovic B, Yoshimoto M, Wang Y, Zielenska M, Squire JA. Decitabine-induced demethylation of 5 CpG island in GADD45A leads to apoptosis in osteosarcoma cells. Neoplasia 2008 ; 10 : 471-80. 23. Li Y, Meng G, Huang L, Guo QN. Hypomethylation of the P3 promoter is associated with up-regulation of IGF2 expression in human osteosarcoma. Hum Pathol 2009 ; 40 : 1441-7. 24. Savage SA, Woodson K, Walk E, et al. Analysis of genes critical for growth regulation identifies insulin-like growth factor 2 receptor variations with possible functional significance as risk factors for osteosarcoma. Cancer Epidemiol Biomarkers Prev 2007 ; 16 : 166774. 25. Esteller M, Tortola S, Toyota M, et al. Hypermethylationassociated inactivation of p14(ARF) is independent of p16(INK4a) methylation and p53 mutational status. Cancer Res 2000 ; 60 : 129-33.
Bull Cancer vol. 98 • N◦ 7 • juillet 2011
Epigenetic changes in osteosarcoma 26. Sarbia M, Geddert H, Klump B, Kiel S, Iskender E, Gabbert HE. Hypermethylation of tumor suppressor genes (p16INK4A, p14ARF and APC) in adenocarcinomas of the upper gastrointestinal tract. Int J Cancer 2004 ; 111 : 224-8.
47. Schübeler D, MacAlpine DM, Scalzo D, et al. The histone modification pattern of active genes revealed through genomewide chromatin analysis of a higher eukaryote. Genes Dev 2004 ; 18 : 1263-71.
27. Badal V, Menendez S, Coomber D, Lane DP. Regulation of the p14ARF promoter by DNA methylation. Cell Cycle 2008 ; 7 : 112-9.
48. Shilatifard A. Chromatin modifications by methylation and ubiquitination: implications in the regulation of gene expression. Annu Rev Biochem 2006 ; 75 : 243-69.
28. Miller CW, Aslo A, Campbell MJ, Kawamata N, Lampkin BC, Koeffler HP. Alterations of the p15, p16, and p18 genes in osteosarcoma. Cancer Genet Cytogenet 1996 ; 86 : 136-42.
49. Thiagalingam S, Cheng KH, Lee HJ, Mineva N, Thiagalingam A, Ponte JF. Histone deacetylases: unique players in shaping the epigenetic histone code. Ann N Y Acad Sci 2003 ; 983 : 84-100.
29. Hu N, Huang J, Emmert-Buck MR, et al. Frequent inactivation of the TP53 gene in esophageal squamous cell carcinoma from a high-risk population in China. Clin Cancer Res 2001 ; 7 : 883-91.
50. Kelly WK, Marks PA. Drug insight: histone deacetylase inhibitors – development of the new targeted anticancer agent suberoylanilide hydroxamic acid. Nat Clin Pract Oncol 2005 ; 2 : 150-7.
30. Pogribny IP, James SJ. Reduction of p53 gene expression in human primary hepatocellular carcinoma is associated with promoter region methylation without coding region mutation. Cancer Lett 2002 ; 176 : 169-74.
51. Sowa Y, Orita T, Minamikawa S, et al. Histone deacetylase inhibitor activates the WAF1/Cip1 gene promoter through the Sp1 sites. Biochem Biophys Res Commun 1997 ; 241 : 142-50.
31. Lim S, Yang MH, Park JH, et al. Inactivation of the RASSF1A in osteosarcoma. Oncol Rep 2003 ; 10 : 897-901. 32. Harada K, Toyooka S, Maitra A, et al. Aberrant promoter methylation and silencing of the RASSF1A gene in pediatric tumors and cell lines. Oncogene 2002 ; 21 : 4345-9. 33. Nishijo K, Nakayama T, Aoyama T, et al. Mutation analysis of the RECQL4 gene in sporadic osteosarcomas. Int J Cancer 2004 ; 111 : 367-72. 34. Wang LL, Gannavarapu A, Kozinetz CA, et al. Association between osteosarcoma and deleterious mutations in the RECQL4 gene in Rothmund-Thomson syndrome. J Natl Cancer Inst 2003 ; 95 : 669-74. 35. Young DW, Hassan MQ, Pratap J, et al. Mitotic occupancy and lineage-specific transcriptional control of rRNA genes by Runx2. Nature 2007 ; 445 : 442-6. 36. Young DW, Hassan MQ, Yang XQ, et al. Mitotic retention of gene expression patterns by the cell fate-determining transcription factor Runx2. Proc Natl Acad Sci U S A 2007 ; 104 : 3189-94. 37. Rubin EM, Guo Y, Tu K, Xie J, Zi X, Hoang BH. Wnt inhibitory factor 1 decreases tumorigenesis and metastasis in osteosarcoma. Mol Cancer Ther 2010 ; 9 : 731-41. 38. Urnov FD. Methylation and the genome: the power of a small amendment. J Nutr 2002 ; 132 : 2450S-6S. 39. Bhattacharya SK, Ramchandani S, Cervoni N, Szyf M. A mammalian protein with specific demethylase activity for mCpG DNA. Nature 1999 ; 397 : 579-83. 40. Dunn BK. Hypomethylation: one side of a larger picture. Ann N Y Acad Sci 2003 ; 983 : 28-42. 41. Papaggeli PC, Kortsaris AC, Matsouka PT. Aberrant methylation of c-myc and c-fos protooncogenes and p53 tumor suppressor gene in myelodysplastic syndromes and acute non-lymphocytic leukemia. J BUON 2003 ; 8 : 341-50. 42. Wu JX, Carpenter PM, Gresens C, et al. The proto-oncogene c-fos is over-expressed in the majority of human osteosarcomas. Oncogene 1990 ; 5 : 989-1000. 43. Li Y, Meng G, Guo QN. Changes in genomic imprinting and gene expression associated with transformation in a model of human osteosarcoma. Exp Mol Pathol 2008 ; 84 : 234-9. 44. Al-Kuraya K, Novotny H, Bavi P, et al. HER2, TOP2A, CCND1, EGFR and C-MYC oncogene amplification in colorectal cancer. J Clin Pathol 2007 ; 60 : 768-72. 45. Bhattacharya N, Sabbir MG, Roy A, Dam A, Roychoudhury S, Panda CK. Approximately 580 Kb surrounding the MYC gene is amplified in head and neck squamous cell carcinoma of Indian patients. Pathol Res Pract 2005 ; 201 : 691-7. 46. Ladanyi M, Park CK, Lewis R, Jhanwar SC, Healey JH, Huvos AG. Sporadic amplification of the MYC gene in human osteosarcomas. Diagn Mol Pathol 1993 ; 2 : 163-7.
Bull Cancer vol. 98 • N◦ 7 • juillet 2011
52. Liao WM, Zhang CL, Li FB, Zeng BF, Zeng YX. p21WAF1/CIP1 gene DNA sequencing and its expression in human osteosarcoma. Chin Med J (Engl) 2004 ; 117 : 936-40. 53. Maeda T, Nagaoka Y, Kawai Y, et al. Inhibitory effects of cancer cell proliferation by novel histone deacetylase inhibitors involve p21/WAF1 induction and G2/M arrest. Biol Pharm Bull 2005 ; 28 : 849-53. 54. Watanabe K, Okamoto K, Yonehara S. Sensitization of osteosarcoma cells to death receptor-mediated apoptosis by HDAC inhibitors through downregulation of cellular FLIP. Cell Death Differ 2005 ; 12 : 10-8. 55. Bird A. DNA methylation patterns and epigenetic memory. Genes Dev 2002 ; 16 : 6-21. 56. Jones PA, Baylin SB. The fundamental role of epigenetic events in cancer. Nat Rev Genet 2002 ; 3 : 415-28. 57. Fahrner JA, Eguchi S, Herman JG, Baylin SB. Dependence of histone modifications and gene expression on DNA hypermethylation in cancer. Cancer Res 2002 ; 62 : 7213-8. 58. Antequera F, Bird A. CpG islands as genomic footprints of promoters that are associated with replication origins. Curr Biol 1999 ; 9 : R661-667. 59. Fuks F, Burgers WA, Godin N, Kasai M, Kouzarides T. Dnmt3a binds deacetylases and is recruited by a sequence-specific repressor to silence transcription. EMBO J 2001 ; 20 : 2536-44. 60. Robertson KD, Ait-Si-Ali S, Yokochi T, Wade PA, Jones PL, Wolffe AP. DNMT1 forms a complex with Rb, E2F1 and HDAC1 and represses transcription from E2F-responsive promoters. Nat Genet 2000 ; 25 : 338-42. 61. Cheng MF, Lee CH, Hsia KT, Huang GS, Lee HS. Methylation of histone H3 lysine 27 associated with apoptosis in osteosarcoma cells induced by staurosporine. Histol Histopathol 2009 ; 24 : 1105-11. 62. Hiraki Y, Tanaka H, Inoue H, Kondo J, Kamizono A, Suzuki F. Molecular cloning of a new class of cartilage-specific matrix, chondromodulin-I, which stimulates growth of cultured chondrocytes. Biochem Biophys Res Commun 1991 ; 175 : 971-7. 63. Aoyama T, Okamoto T, Kohno Y, et al. Cell-specific epigenetic regulation of ChM-I gene expression: crosstalk between DNA methylation and histone acetylation. Biochem Biophys Res Commun 2008 ; 365 : 124-30. 64. Toh Y, Pencil SD, Nicolson GL. A novel candidate metastasisassociated gene, mta1, differentially expressed in highly metastatic mammary adenocarcinoma cell lines. cDNA cloning, expression, and protein analyses. J Biol Chem 1994 ; 269 : 22958-63. 65. Park HR, Cabrini RL, Araujo ES, Paparella ML, Brandizzi D, Park YK. Expression of ezrin and metastatic tumor antigen in osteosarcomas of the jaw. Tumori 2009 ; 95 : 81-6. 66. Kiss NB, Geli J, Lundberg F, et al. Methylation of the p16INK4A promoter is associated with malignant behavior in abdominal extraadrenal paragangliomas but not pheochromocytomas. Endocr Relat Cancer 2008 ; 15 : 609-21.
E67
J. Cui, et al. 67. Maitra A, Roberts H, Weinberg AG, Geradts J. Loss of p16(INK4a) expression correlates with decreased survival in pediatric osteosarcomas. Int J Cancer 2001 ; 95 : 34-8.
73. Marks PA, Breslow R. Dimethyl sulfoxide to vorinostat: development of this histone deacetylase inhibitor as an anticancer drug. Nat Biotechnol 2007 ; 25 : 84-90.
68. Christman JK. 5-azacytidine and 5-aza-2 -deoxycytidine as inhibitors of DNA methylation: mechanistic studies and their implications for cancer therapy. Oncogene 2002 ; 21 : 5483-95.
74. Kisseberth WC, Murahari S, London CA, Kulp SK, Chen CS. Evaluation of the effects of histone deacetylase inhibitors on cells from canine cancer cell lines. Am J Vet Res 2008 ; 69 : 938-45.
69. Zhou L, Cheng X, Connolly BA, Dickman MJ, Hurd PJ, Hornby DP. Zebularine: a novel DNA methylation inhibitor that forms a covalent complex with DNA methyltransferases. J Mol Biol 2002 ; 321 : 591-9.
75. Blattmann C, Oertel S, Ehemann V, et al. Enhancement of radiation response in osteosarcoma and rhabomyosarcoma cell lines by histone deacetylase inhibition. Int J Radiat Oncol Biol Phys 2010 ; 78 : 237-45.
70. Marcucci G, Silverman L, Eller M, Lintz L, Beach CL. Bioavailabi -lity of azacitidine subcutaneous versus intravenous in patients with the myelodysplastic syndromes. J Clin Pharmacol 2005 ; 45 : 597-602.
76. Kelly WK, O’Connor OA, Marks PA. Histone deacetylase inhibitors: from target to clinical trials. Expert Opin Investig Drugs 2002 ; 11 : 1695-713.
71. Cheng JC, Weisenberger DJ, Gonzales FA, et al. Continuous zebularine treatment effectively sustains demethylation in human bladder cancer cells. Mol Cell Biol 2004 ; 24 : 1270-8.
77. Cameron EE, Bachman KE, Myohanen S, Herman JG, Baylin SB. Synergy of demethylation and histone deacetylase inhibition in the re-expression of genes silenced in cancer. Nat Genet 1999 ; 21 : 103-7.
72. Wittenburg LA, Bisson L, Rose BJ, Korch C, Thamm DH. The histone deacetylase inhibitor valproic acid sensitizes human and canine osteosarcoma to doxorubicin. Cancer Chemother Pharmacol 2010 ; 67 : 83-92.
E68
78. Wood LD, Parsons DW, Jones S, et al. The genomic landscapes of human breast and colorectal cancers. Science 2007 ; 318 : 1108-13.
Bull Cancer vol. 98 • N◦ 7 • juillet 2011