DNA methylation of mobile genetic elements in human cancers ...

2 downloads 0 Views 262KB Size Report
It is recently considered that the hypomethylation of mobile genetic elements may play a biological role in cancer cells along with the DNA hypermethylation on ...
Genes Genom (2013) 35:265–271 DOI 10.1007/s13258-013-0095-3

REVIEW

DNA methylation of mobile genetic elements in human cancers Kyudong Han • Jungname Lee • Heui-Soo Km Kwangmo Yang • Joo Mi Yi



Received: 13 November 2012 / Accepted: 27 January 2013 / Published online: 14 February 2013 Ó The Genetics Society of Korea 2013

Abstract Mobile genetic elements are responsible for half of the human genome, creating the host genomic instability or variability through several mechanisms. Two types of abnormal DNA methylation in the genome, hypomethylation and hypermethylation, are associated with cancer progression. Genomic hypermethylation has been most often observed on the CpG islands around gene promoter regions in cancer cells. In contrast, hypomethylation has been observed on mobile genetic elements in the cancer cells. It is recently considered that the hypomethylation of mobile genetic elements may play a biological role in cancer cells along with the DNA hypermethylation on CpG islands. Growing evidence has indicated that mobile genetic elements could be associated with the cancer initiation and progression through the hypomethylation. Here we review the recent progress on the relationship between DNA methylation and mobile genetic elements, focusing on the hypomethylation of LINE-1 and HERV elements in various human cancers and suggest that DNA hypomethylation of mobile genetic elements could have potential to be a new cancer therapy target in the future.

K. Han  J. Lee Department of Nanobiomedical Science and WCU Research Center, Dankook University, Cheonan 330-714, South Korea H.-S. Km Department of Biological Science, Pusan National University, Busan 609-735, South Korea K. Yang  J. M. Yi (&) Research Institute, Dongnam Institute of Radiological & Medical Sciences (DIRAMS), Jwadong-gil 40, Jangan-eup, Gijang-gun, Busan, South Korea e-mail: [email protected]

Keywords Mobile genetic element  Hypermethylation  Hypomethylation  LINE elements  HERV elements

Introduction Mobile genetic elements, called junk DNAs or transposable elements, are present in the genome of all known eukaryotes. They account for at least 45 % of mammalian genomes (Gibbs et al. 2007; Gregory et al. 2002; Lander et al. 2001; Mikkelsen et al. 2005; Mills et al. 2011; Scally et al. 2012; Warren et al. 2008). The mobile genetic elements are classified into two major groups, according to their transposition mechanism, the first group is DNA transposon; this element moves into a new genomic region using ‘‘cut and paste’’ mechanism. The second group is retrotransposon; this element transcribes itself and then integrates into a new genomic region via reverse transcription of the RNA intermediates (‘‘copy and paste’’ mechanism). Thus, retrotransposons may well be more ubiquitous than DNA in the mammalian genomes. It has been reported that retrotransposons are capable of retrotransposition in various primate genomes and thus they have a major impact on the architecture and fluidity of their host genomes (Cordaux et al. 2006; Este´cio et al. 2010; Nekrutenko and Li 2001). Retrotransposons are divided into two groups, long terminal repeat (LTR) retrotransposons and non-LTR retrotransposons. Human endogenous retrovirus (HERV) is one of the LTR retrotransposons. In 1981, HERV was first characterized (Martin et al. 1981). It has been suggested that HERV was originated from exogenous retrovirus, inserted into its host genome during primate evolution because its gene structure is similar to that of exogenous retrovirus (50 LTR-gag-pol-env-30 LTR). However, unlike the exogenous virus, most open reading frames of HERV are not

123

266

intact due to mutations such as deletion, insertion codons, and frame shift (Boeke and Stoye 1997; Lander et al. 2001; Stoye 2001). Nonetheless, it has been reported that some of HERVs are actively amplified in the human genome (Mayer and Meese 2005; Yi and Kim 2006; Yi et al 2007). In reality, previous studies about HERV showed that HERV mRNAs are differentially but universally expressed in various cell types and tissues (Yi et al. 2004, 2006, 2007) and HERV expression levels are various among individuals (Andersson et al. 1996). In addition, HERV proteins were found in cancer cells (Yi et al. 2006, 2007) as well as normal cells from placenta and embryo (Muir et al. 2004). In contrast, the LINE-1 elements belong to autonomous nonLTR retrotransposons. They account for approximately 17 % of the human genome (Lander et al. 2001). During the past 6 million years, about 2,000 copies of L1 elements newly inserted into the human genome and some of them are currently active to retrotranspose in the human genome (Beck et al. 2010; Brouha et al. 2003). Since the divergence of human and chimpanzee, L1s have modified the human genome through various mechanisms including de novo insertions, inversions, insertion-mediated deletions, and recombination events (Goodier et al. 2004; Han et al. 2005, 2008). Thus, L1 has acted as an agent causing human genomic variation and instability. Epigenetics is defined as heritable changes in gene expression that are not accompanied by changes in DNA sequence. The heritable changes include DNA methylation and histone modifications, which are now known to regulate a wide range of physiological and pathological processes (Jones and Baylin 2002). Gene silencing at the level of chromatin is necessary for eukaryotic organisms and is specifically important in biological processes, such as differentiation, imprinting, and silencing of large chromosomal domains including the X chromosome, over the life span of female mammals. Like most biological processes, the gene silencing can become dysregulated, resulting in the development of multiple disease states. Recently, there has been much emphasized on the critical role of DNA hypermethylation in human carcinogenesis (Herman and Baylin 2000; Issa 2000). In comparison with the hypermethylation, loss of DNA methylation in cancer has not been much considered in the field of cancer genomics. However, the extensive cancer-associated DNA hypomethylation in the human genome was discovered earlier than cancer-linked DNA hypermethylation (Feinberg and Vogelstein 1983). In cancer cells, DNA hypomethylation often has a higher effect on their genomes than does hypermethylation. As a result, net losses of genomic 5-methylcytosine were observed in many human cancers (Gama-Sosa et al. 1983). Therefore, here, we discuss mobile genetic elements associated with the DNA hypomethylation in cancer genomes and highlight their future clinical potential.

123

Genes Genom (2013) 35:265–271

DNA methylation in cancer DNA methylation is one of the important mechanisms that control gene expression, chromatin structure, genome stability and X-chromosome inactivation (Jones and Baylin 2002). Abnormal patterns of DNA methylation in cancer cells have been recognized for over 20 years. DNA methylation typically occurs at CpG sites, which are CG dinucleotide sequences that occur at a relatively low frequency in the genome (Herman and Baylin 2003). Mammalian DNA contains 5-methylcytosine, the genomic distribution of which is specific for each cell type and is largely established during embryonic development. Mammalian DNA methyltransferases recognize CpG dinucleotides at specific sites within the genome referred to as a ‘‘CpG island’’, a CpG-rich region often encompassing the promoter and transcription start site of the associated gene and possess de novo methylation activity (Bird 1986). Approximately half of all human genes contain CpG islands in the 50 area of their gene promoter (Bird 1986). DNA methylation patterns in normal tissues are in part dependent on the relative levels and activities of DNA methyltransferases and DNA demethylases whose expression is regulated at both the transcriptional and posttranscriptional level (Herman and Baylin 2003). Abnormal DNA methylation can lead to serious imbalance in normal function of cells and can promote pathological conditions. In particular, the cancer genome is known to undergo substantial changes in DNA methylation (Jones and Baylin 2002). Most notable are genome-wide hypomethylation events that preferentially target repetitive DNA elements, and gene-specific hypermethylation of CpG islands. Gene promoter DNA hypermethylation in cancer First classic tumor suppressor gene, the retinoblastoma gene (Rb), raised that DNA hypermethylation of Rb gene could be involved in gene inactivation in retinoblastoma. Sasaki and colleagues have described five retinoblastomas which contains tumor-specific hypermethylation in the promoter region (Sakai et al. 1991). Also, the von Hippel– Lindau gene (VHL) was cloned and shown to be responsible for the inherited syndrome, von Hippel–Lindau disease (Latif et al. 1993). Many groups have reported that VHL gene is associated with DNA promoter hypermethylation in renal cancer and hemangioblastoma (Clifford et al. 1998; Prowse et al. 1997). The most studied of all tumor suppressor genes for promoter hypermethylation is undoubtedly the p16 gene, currently designed CDKN2A, a cyclin-dependent kinase inhibitor which functions in the regulation of the phosphorylation status of the RB protein (Herman 1999). In fact, hypermethylation associated with the loss of

Genes Genom (2013) 35:265–271

Fig. 1 DNA methylation changes during cancer progression. Hypermethylation in CpG island promoter region of tumor suppressor genes induce gene silencing in cancers. In contrast, mobile genetic elements such as LINE-1 and HERV families showed hypomethylation in cancer therefore, lead to global genome instability. Recent evidences suggest that both cases could be contributed for causing human cancers. This schematic is simplified based on the DNA methylation levels in the genome. TSG indicates that tumor suppressor genes

expression of the CDKN2A gene has been found to be one of the most frequent alterations in neoplasia rather than homozygous deletion and some point mutations. First reports describe methylation of the p16 gene in lung, head and neck, gliomas, colorectal, and breast carcinomas (Herman et al. 1995; Merlo et al. 1995; Otterson et al. 1995). Another cyclin-dependent kinase inhibitor lies adjacent to CDKN2A on chromosome 9p21. This gene, p15 or CDKN2B, is structurally and functionally similar to CDKN2A in regulating the phosporylation of Rb. CDNK2B is frequently hypermethylated in many forms of hematological malignancies (Herman et al. 1997). Genes involved in DNA repair have also been founded to be frequently inactivated by promoter region hypermethylation in cancer. For example, methylation of GST-p, MGMT, and MLH1 gene has been found in various types of cancer along with loss of expression of these genes (Esteller et al. 1998; Kane et al. 1997; Qian and Brent 1997) (Fig. 1). Hypomethylation of LINE-1 in cancer Genome-wide DNA hypomethylation and gene-specific hypermethylation of CpG islands are prominent hallmarks of cancer genomes (Ehrlich 2002; Ushijima 2005). Aberrant DNA methylation can cause reactivation of methylationsilenced mobile genetic elements supposed to be methylated (inactive form), which result in increased genomic instability (Ehrlich 2002). Hypermethylation of mobile genetic elements such as LINE1, Alu, and some of HERV families, disperse across the genome in normal cells, but hypomethylation of mobile genetic element is frequently observed in cancers (Table 1). In reality, DNA hypomethylation is more pronounced in tumor progression or malignancy than normal cells (Ehrlich et al. 2006; Qu et al. 1999) although the pattern of the DNA hypomethylation is

267

various, depending on cancer type and individual specimen (Jackson et al. 2004; Widschwendter et al. 2004). DNA hypomethylation of mobile genetic elements contributes to carcinogenesis and chromosomal rearrangements (Dobigny et al. 2004; Guz et al. 2010). Especially, DNA hypomethylation of mobile genetic elements located on centromeric and juxtacentromeric regions is associated with cervical neoplasia (Kim et al. 1994) and hepatocellular carcinoma (Shen et al. 1998). Therefore, it has been suggested that hypomethylations of interspersed repeats (e.g., LINE and SINE) and tandem repeats induce genomic rearrangements responsible for tumor formation or progression (Chen et al. 1998; Gaudet et al. 2003). Several studies have demonstrated that hypomethylation of LINE-1 is associated with cancers (Hoffmann and Schulz 2005; Ostertag and Kazazian 2001). Hypomethylated LINE-1s increase the copy number of their mRNAs and their reactivation can lead to transcriptional regulation of neighboring genes via transcriptional interference (Whitelaw and Martin 2001). Interestingly, being able to activate LINE-1 antisense promoter, hypomethylation of certain L1s is correlated with expression of alternative gene transcripts. In reality, it was reported that hypomethylation of a LINE-1 promoter creates alternative transcripts of the MET oncogene in bladder tumors. The L1-MET promoter was typically methylated in normal cells but most of the bladder carcinoma cell lines showed remarkable hypomethylation on the promoter (Wolff et al. 2010). Thus, the change in LINE-1 methylation level may be a useful marker when monitoring cancer progression. Low expression level of HERVs in various cancers A large and constantly growing number of reports have described detection of RNA transcripts from various HERV families in many different types of human cancers or tumor cell lines (Armbruester et al. 2002; Depil et al. 2002; Patzke et al. 2002; Wang-Johanning et al. 2003a, 2003b; Yi et al. 2004, 2006). Even though the detection of various HERV families transcripts in many cancer types, the biological significance of HERV RNA expression in cancers remains obscure. Specifically, one of class of human retrotransposons that is usually highly methylated and may become hypomethylated in association with carcinogenesis is the human endogenous retroviruses, especially, the HERV-K family. HERV-K family comprises 30–50 full length members per haploid genome, among which several contain intact open reading frames (Lo¨wer et al. 1996). HERV-K elements are the most intact among human endogenous retroviruses and make up 1–2 % of the human genome. Although, HERV-K sequences are relatively poor in CpG dinucleotides, for instance there are fewer than 20 in their LTRs, methylation of these

123

268 Table 1 Hypomethylation of mobile genetic elements in different cancer types

Genes Genom (2013) 35:265–271

Type of cancer Breast cancer

Colorectal cancer

Gastric cancer Leukemia (chronic myelogenous leukemia) Lung cancer (non-small cell lung cancer) Melanoma

Prostate cancer

sequences like that of LINE-1 sequences may be important in controlling their ability to transcribed, to transpose and to participate in recombination. Expansion of our thoughts, we have tried to compare the expression pattern of various HERV families in different human tissues and cancer cell lines (Fig. 2). In human normal tissues, many HERV families have been expressed in human normal tissues (Fig. 2a) compared to various cancer cell lines (Fig. 2b). This gene expression profile of HERV families suggests that expression level of HERV families is losing their basal expression level in disease status such as cancers. As we mentioned earlier, lack of basal expression of genes could be implicated association with regulation by DNA methylation. In our gene expression profile in Fig. 2 could suggest that HERV elements might be regulated by DNA hypo or hypermethylation during cancer progression. However, expression of HERV proteins in tumor tissue provides an argument for some functional significances, some of crucial questions remain mainly how HERV protein expression could contribute to cancer development. Future perspective There is a poor understanding of the pathways that lead to cancer-associated hypomethylation of mobile genetic elements so far. Even though hypomethylation of mobile genetic elements is associated with cancer detection as a biomarker, little is known about biological roles for understanding human cancer. Evidence suggests that this

123

Mobile genetic elements

References

HERV-W Env

Bjerregaard et al. 2006

Alu

Cho et al. 2010

LINE-1

Cho et al. 2010

Alu

Kwon et al. (2010) and Rodriguez et al. (2008)

LINE-1

Chalitchagorn et al. (2004) and Ogino et al. (2008)

Alu

Yoshida et al. 2011

LINE-1 Alu

Yoshida et al. 2011 Roman-Gomez et al. 2008

LINE-1

Roman-Gomez et al. 2008

Alu

Daskalos et al. 2009

LINE-1

Chalitchagorn et al. 2004

HERV-K (HML-2) Env

Buscher et al. 2005

HERV-K(HML-2) Gag

Muster et al. 2003

HERV-K(HML-2) Rec

Buscher et al. 2006

LINE-1

Tellez et al. 2009

Alu

Kim et al. 2011

LINE-1

Chalitchagorn et al. 2004

hypomethylation of DNA in cancer is not a consequence of or a prelude to hypermethylation in cancer. The hypomethylation in repeated DNA sequences may play special roles in carcinogenesis, such as, increasing karyotype instability or affecting expression of genes indirectly. DNA hypermethylation has been known that it is emerging as an important diagnostic and prognostic tool. Like this, DNA hypomethylation analysis may also be useful in detecting cancer and managing the disease (Qu et al. 1999; Santourlidis et al. 1999; Shen et al. 1998). Yang et al. showed the changed in the methylation levels of the LINE-1 repetitive elements could be used as a surrogate marker of genomewide methylation changes (Yang et al. 2004). Also, LINE-1 and HERV-K sequences have been shown to be hypomethylated and transcribed in human teratocarcinoma cells (Bratthauer and Fanning 1992). Recently, the association of global or focal DNA hypomethylation with the early stages of carcinogenesis or with tumor progression provides cancer markers that should be very useful in the clinic (Compare et al. 2011). To discover new genome-wide hypomethylation locus or genes in cancer genome, high-throughput and high-resolution technology for DNA methylation analysis facilitated recently huge development for genome-wide analysis. Over the next 10 years, there will probably be many clinical tests in use that are based on biomarkers using DNA hypomethylation of mobile genetic elements for cancer detection and prognosis. Some type of cancer, DNA hypomethylation is seen as an early indicator of tumorigenesis (Feinberg and Vogelstein 1983). Take all by together, mobile genetic elements such as LINE or HERV

Genes Genom (2013) 35:265–271

269

Fig. 2 Gene expression profile of various HERV families in a human normal tissues and b cancer cell lines. X-axis shows different type of human normal tissues (a) and colon cancer cell lines (b). Y-axis

shows various HER families. Green color indicates HERVs have a basal expression in human normal tissues and cancer cell lines, but red color indicates no expression. (Color figure online)

families for DNA hypomethylation may become a clinically useful addition to hypermethylation analyses of CpG islands in human cancers and future studies should clarify this issue.

Brouha B, Schustak J, Badge RM, Lutz-Prigge S, Farley AH, Moran JV, Kazazian HH Jr (2003) Hot L1s account for the bulk of retrotransposition in the human population. Proc Natl Acad Sci USA 100:5280–5285 Buscher K, Trefzer U, Hofmann M, Sterry W, Kurth R, Denner J (2005) Expression of human endogenous retrovirus K in melanomas and melanoma cell lines. Cancer Res 65:4172–4180 Buscher K, Hahn S, Hofmann M, Trefzer U, Ozel M, Sterry W, Lower J, Lower R, Kurth R, Denner J (2006) Expression of the human endogenous retrovirus-K transmembrane envelope, Rec and Np9 proteins in melanomas and melanoma cell lines. Melanoma Res 16:223–234 Chalitchagorn K, Shuangshoti S, Hourpai N, Kongruttanachok N, Tangkijvanich P, Thong-ngam D, Voravud N, Sriuranpong V, Mutirangura A (2004) Distinctive pattern of LINE-1 methylation level in normal tissues and the association with carcinogenesis. Oncogene 23:8841–8846 Chen RZ, Pettersson U, Beard C, Jackson-Grusby L, Jaenisch R (1998) DNA hypomethylation leads to elevated mutation rates. Nature 395:89–93 Cho YH, Yazici H, Wu HC, Terry MB, Gonzalez K, Qu M, Dalay N, Santella RM (2010) Aberrant promoter hypermethylation and genomic hypomethylation in tumor, adjacent normal tissues and blood from breast cancer patients. Anticancer Res 30:2489–2496 Clifford SC, Prowse AH, Affara NA, Buys CH, Maher ER (1998) Inactivation of the von Hippel–Lindau (VHL) tumour suppressor gene and allelic losses at chromosome arm 3p in primary renal cell carcinoma: evidence for a VHL-independent pathway in clear cell renal tumourigenesis. Genes Chromosom Cancer 22: 200–209 Compare D, Rocco A, Liguori E, D’Armiento FP, Persico G, Masone S, Coppola-Bottazzi E, Suriani R, Romano M, Nardone G (2011) Global DNA hypomethylation is an early event in Helicobacter pylori-related gastric carcinogenesis. J Clin Pathol 64:677–682 Cordaux R, Udit S, Batzer MA, Feschotte C (2006) Birth of a chimeric primate gene by capture of the transposase gene from a mobile element. Proc Natl Acad Sci USA 103:8101–8106 Daskalos A, Nikolaidis G, Xinarianos G, Savvari P, Cassidy A, Zakopoulou R, Kotsinas A, Gorgoulis V, Field JK, Liloglou T

Acknowledgments This work was supported in part by National R&D program (50596-2013) through the Dongnam Institute of Radiological & Medical Sciences (DIRAMS) funded by the Korean Ministry of Education, Science and Technology and by WCU (World Class University) program through the National Research Foundation of Korea funded by the Ministry of Education, Science and Technology (R31-10069).

References Andersson ML, Medstrand P, Yin H, Blomberg J (1996) Differential expression of human endogenous retroviral sequences similar to mouse mammary tumor virus in normal peripheral blood mononuclear cells. AIDS Res Hum Retroviruses 12:833–840 Armbruester V, Sauter M, Krautkraemer E, Meese E, Kleiman A, Best B, Roemer K, Mueller-Lantzsch N (2002) A novel gene from the human endogenous retrovirus K expressed in transformed cells. Clin Cancer Res 8:1800–1807 Beck CR, Collier P, Macfarlane C, Malig M, Kidd JM, Eichler EE, Badge RM, Moran JV (2010) LINE-1 retrotransposition activity in human genomes. Cell 141:1159–1170 Bird AP (1986) CpG-rich islands and the function of DNA methylation. Nature 321:209–213 Bjerregaard B, Holck S, Christensen IJ, Larsson LI (2006) Syncytin is involved in breast cancer-endothelial cell fusions. Cell Mol Life Sci 63:1906–1911 Boeke JD, Stoye JP (1997) Retrotransposons, Endogenous Retroviruses, and the Evolution of Retroelements. In: Coffin JM, Hughes SH, Varmus HE (eds) Retroviruses. Cold Spring Harbor Laboratory Press, Cold Spring Harbor (NY) Bratthauer GL, Fanning TG (1992) Active LINE-1 retrotransposons in human testicular cancer. Oncogene 7:507–510

123

270 (2009) Hypomethylation of retrotransposable elements correlates with genomic instability in non-small cell lung cancer. Int J Cancer 124:81–87 Depil S, Roche C, Dussart P, Prin L (2002) Expression of a human endogenousretrovirus, HERV-K, in the blood cells of leukemia patients. Leukemia 16:254–259 Dobigny G, Ozouf-Costaz C, Waters PD, Bonillo C, Coutanceau JP, Volobouev V (2004) LINE-1 amplification accompanies explosive genome repatterning in rodents. Chromosome Res 12:787– 793 Ehrlich M (2002) DNA methylation in cancer: too much, but also too little. Oncogene 21:5400–5413 Ehrlich M, Woods CB, Yu MC, Dubeau L, Yang F, Campan M, Weisenberger DJ, Long T, Youn B, Fiala ES et al (2006) Quantitative analysis of associations between DNA hypermethylation, hypomethylation, and DNMT RNA levels in ovarian tumors. Oncogene 25:2636–2645 Este´cio MR, Gallegos J, Vallot C, Castoro RJ, Chung W, Maegawa S, Oki Y, Kondo Y, Jelinek J, Shen L et al (2010) Genome architecture marked by retrotransposons modulates predisposition to DNA methylation in cancer. Genome Res 20:1369–1382 Esteller M, Corn PG, Urena JM, Gabrielson E, Baylin SB, Herman JG (1998) Inactivation of glutathione S-transferase P1 gene by promoter hypermethylation in human neoplasia. Cancer Res 58:4515–4518 Feinberg AP, Vogelstein B (1983) Hypomethylation distinguishes genes of some human cancers from their normal counterparts. Nature 301:89–92 Gama-Sosa MA, Slagel VA, Trewyn RW, Oxenhandler R, Kuo KC, Gehrke CW, Ehrlich M (1983) The 5-methylcytosine content of DNA from human tumors. Nucleic Acids Res 11:6883–6894 Gaudet F, Hodgson JG, Eden A, Jackson-Grusby L, Dausman J, Gray JW, Leonhardt H, Jaenisch R (2003) Induction of tumors in mice by genomic hypomethylation. Science 300:489–492 Gibbs RA, Rogers J, Katze MG, Bumgarner R, Weinstock GM, Mardis ER, Remington KA, Strausberg RL, Venter JC, Wilson RK et al (2007) Evolutionary and biomedical insights from the rhesus macaque genome. Science 316:222–234 Goodier JL, Ostertag EM, Engleka KA, Seleme MC, Kazazian HH Jr (2004) A potential role for the nucleolus in L1 retrotransposition. Hum Mol Genet 13:1041–1048 Gregory SG, Sekhon M, Schein J, Zhao S, Osoegawa K, Scott CE, Evans RS, Burridge PW, Cox TV, Fox CA et al (2002) A physical map of the mouse genome. Nature 418:743–750 Guz J, Foksin´ski M, Olin´ski R (2010) Global DNA hipomethylation– the meaning in carcinogenesis. Postepy Biochem 56:16–21 Han K, Sen SK, Wang J, Callinan PA, Lee J, Cordaux R, Liang P, Batzer MA (2005) Genomic rearrangements by LINE-1 insertion-mediated deletion in the human and chimpanzee lineages. Nucleic Acids Res 33:4040–4052 Han K, Lee J, Meyer TJ, Remedios P, Goodwin L, Batzer MA (2008) L1 recombination-associated deletions generate human genomic variation. Proc Natl Acad Sci USA 105:19366–19371 Herman JG (1999) Hypermethylation of tumor suppressor genes in cancer. Semin Cancer Biol 9:359–367 Herman JG, Baylin SB (2000) Promoter-region hypermethylation and gene silencing in human cancer. Curr Top Microbiol Immunol 249:35–54 Herman JG, Baylin SB (2003) Gene silencing in cancer in association with promoter hypermethylation. N Engl J Med 349:2042–2054 Herman JG, Merlo A, Mao L, Lapidus RG, Issa JP, Davidson NE, Sidransky D, Baylin SB (1995) Inactivation of the CDKN2/p16/ MTS1 gene is frequently associated with aberrant DNA methylation in all common human cancers. Cancer Res 55:4525–4530 Herman JG, Civin CI, Issa JP, Collector MI, Sharkis SJ, Baylin SB (1997) Distinct patterns of inactivation of p15INK4B and

123

Genes Genom (2013) 35:265–271 p16INK4A characterize the major types of hematological malignancies. Cancer Res 57:837–841 Hoffmann MJ, Schulz WA (2005) Causes and consequences of DNA hypomethylation in human cancer. Biochem Cell Biol 83:296–321 Issa JP (2000) The epigenetics of colorectal cancer. Ann N Y Acad Sci 910:140–153 Jackson K, Yu MC, Arakawa K, Fiala E, Youn B, Fiegl H, Mu¨llerHolzner E, Widschwendter M, Ehrlich M (2004) DNA hypomethylation is prevalent even in low-grade breast cancers. Cancer Biol Ther 3:1225–1231 Jones PA, Baylin SB (2002) The fundamental role of epigenetic events in cancer. Nat Rev Genet 3:415–428 Kane MF, Loda M, Gaida GM, Lipman J, Mishra R, Goldman H, Jessup JM, Kolodner R (1997) Methylation of the hMLH1 promoter correlates with lack of expression of hMLH1 in sporadic colon tumors and mismatch repair-defective human tumor cell lines. Cancer Res 57:808–811 Kim YI, Giuliano A, Hatch KD, Schneider A, Nour MA, Dallal GE, Selhub J, Mason JB (1994) Global DNA hypomethylation increases progressively in cervical dysplasia and carcinoma. Cancer 74:893–899 Kim SJ, Kelly WK, Fu A, Haines K, Hoffman A, Zheng T, Zhu Y (2011) Genome-wide methylation analysis identifies involvement of TNF-alpha mediated cancer pathways in prostate cancer. Cancer Lett 302:47–53 Kwon HJ, Kim JH, Bae JM, Cho NY, Kim TY, Kang GH (2010) DNA methylation changes in ex-adenoma carcinoma of the large intestine. Virchows Arch 457:433–441 Lander ES, Linton LM, Birren B, Nusbaum C, Zody MC, Baldwin J, Devon K, Dewar K, Doyle M, FitzHugh W et al (2001) International Human Genome Sequencing Consortium. Initial sequencing and analysis of the human genome. Nature 409:860– 921 Latif F, Tory K, Gnarra J, Yao M, Duh FM, Orcutt ML, Stackhouse T, Kuzmin I, Modi W, Geil L et al (1993) Identification of the von Hippel–Lindau disease tumor suppressor gene. Science 260: 1317–1320 Lo¨wer R, Lo¨wer J, Kurth R (1996) The viruses in all of us: characteristics and biological significance of human endogenous retrovirus sequences. Proc Natl Acad Sci USA 93:5177–5184 Martin MA, Bryan T, Rasheed S, Khan AS (1981) Identification and cloning of endogenous retroviral sequences present in human DNA. Proc Natl Acad Sci USA 78:4892–4896 Mayer J, Meese E (2005) Human endogenous retroviruses in the primate lineage and their influence on host genomes. Cytogenet Genome Res 110:448–456 Merlo A, Herman JG, Mao L, Lee DJ, Gabrielson E, Burger PC, Baylin SB, Sidransky D (1995) 50 CpG island methylation is associated with transcriptional silencing of the tumour suppressor p16/CDKN2/MTS1 in human cancers. Nat Med 1:686–692 Mikkelsen TS, Hillier LW, Eichler EE, Zody MC, Zody MC, Jaffe DB, Yang SP, Enard W, Hellmann I, Lindblad-Toh K et al (2005) Initial sequence of the chimpanzee genome and comparison with the human genome. Nature 437:69–87 Mills RE, Walter K, Stewart C, Handsaker RE, Chen K, Alkan C, Abyzov A, Yoon SC, Ye K, Cheetham RK et al (2011) 1000 Genomes Project. Mapping copy number variation by population-scale genome sequencing. Nature 470:59–65 Muir A, Lever A, Moffett A (2004) Expression and functions of human endogenous retroviruses in the placenta: an update. Placenta 25(Suppl A):S16–S25 Muster T, Waltenberger A, Grassauer A, Hirschl S, Caucig P, Romirer I, Fodinger D, Seppele H, Schanab O, Magin-Lachmann C et al (2003) An endogenous retrovirus derived from human melanoma cells. Cancer Res 63:8735–8741

Genes Genom (2013) 35:265–271 Nekrutenko A, Li WH (2001) Transposable elements are found in a large number of human protein-coding genes. Trends Genet 17:619–621 Ogino S, Kawasaki T, Nosho K, Ohnishi M, Suemoto Y, Kirkner GJ, Fuchs CS (2008) LINE-1 hypomethylation is inversely associated with microsatellite instability and CpG island methylator phenotype in colorectal cancer. Int J Cancer 122:2767–2773 Ostertag EM, Kazazian HH Jr (2001) Biology of mammalian L1 retrotransposons. Annu Rev Genet 35:501–538 Otterson GA, Khleif SN, Chen W, Coxon AB, Kaye FJ (1995) CDKN2 gene silencing in lung cancer by DNA hypermethylation and kinetics of p16INK4 protein induction by 5-aza 20 deoxycytidine. Oncogene 11:1211–1216 Patzke S, Lindeskog M, Munthe E, Aasheim HC (2002) Characterization of a novel humanendogenous retrovirus, HERV-H/F, expressed in human leukemia cell lines. Virology 303:164–173 Prowse AH, Webster AR, Richards FM, Richard S, Olschwang S, Resche F, Affara NA, Maher ER (1997) Somatic inactivation of the VHL gene in Von Hippel–Lindau disease tumors. Am J Hum Genet 60:765–771 Qian XC, Brent TP (1997) Methylation hot spots in the 50 flanking region denote silencing of the O6-methylguanine-DNA methyltransferase gene. Cancer Res 57:3672–3677 Qu G, Dubeau L, Narayan A, Yu MC, Ehrlich M (1999) Satellite DNA hypomethylation vs. overall genomic hypomethylation in ovarian epithelial tumors of different malignant potential. Mutat Res 423:91–101 Rodriguez J, Vives L, Jorda M, Morales C, Munoz M, Vendrell E, Peinado MA (2008) Genome-wide tracking of unmethylated DNA Alu repeats in normal and cancer cells. Nucleic Acids Res 36:770–784 Roman-Gomez J, Jimenez-Velasco A, Agirre X, Castillejo JA, Navarro G, San Jose-Eneriz E, Garate L, Cordeu L, Cervantes F, Prosper F et al (2008) Repetitive DNA hypomethylation in the advanced phase of chronic myeloid leukemia. Leuk Res 32:487– 490 Sakai T, Toguchida J, Ohtani N, Yandell DW, Rapaport JM, Dryja TP (1991) Allele-specific hypermethylation of the retinoblastoma tumor-suppressor gene. Am J Hum Genet 48:880–888 Santourlidis S, Florl A, Ackermann R, Wirtz HC, Schulz WA (1999) High frequency of alterations in DNA methylation in adenocarcinoma of the prostate. Prostate 39:166–174 Scally A, Dutheil JY, Hillier LW, Jordan GE, Goodhead I, Herrero J, Hobolth A, Lappalainen T, Mailund T, Marques-Bonet T et al (2012) Insights into hominid evolution from the gorilla genome sequence. Nature 483:169–175 Shen L, Fang J, Qiu D, Zhang T, Yang J, Chen S, Xiao S (1998) Correlation between DNA methylation and pathological changes in human hepatocellular carcinoma. Hepatogastroenterology 45:1753–1759 Stoye JP (2001) Endogenous retroviruses: still active after all these years? Curr Biol 11:R914–R916

271 Tellez CS, Shen L, Estecio MR, Jelinek J, Gershenwald JE, Issa JP (2009) CpG island methylation profiling in human melanoma cell lines. Melanoma Res 19:146–155 Ushijima T (2005) Detection and interpretation of altered methylation patterns in cancer cells. Nat Rev Cancer 5:223–231 Wang-Johanning F, Frost AR, Jian B, Epp L, Lu DW, Johanning GL (2003a) Quantitationof HERV-K env gene expression and splicing in human breast cancer. Oncogene 22:1528–1535 Wang-Johanning F, Frost AR, Jian B, Azerou R, Lu DW, Chen DT, Johanning GL (2003b) Detecting the expression of human endogenous retrovirus E envelope transcripts inhuman prostate adenocarcinoma. Cancer 98:187–197 Warren WC, Hillier LW, Marshall Graves JA, Birney E, Ponting CP, Gru¨tzner F, Belov K, Miller W, Clarke L, Chinwalla AT et al (2008) Genome analysis of the platypus reveals unique signatures of evolution. Nature 453:175–183 Whitelaw E, Martin DI (2001) Retrotransposons as epigenetic mediators of phenotypic variation in mammals. Nat Genet 27: 361–365 Widschwendter M, Jiang G, Woods C, Mu¨ller HM, Fiegl H, Goebel G, Marth C, Mu¨ller-Holzner E, Zeimet AG, Laird PW et al (2004) DNA hypomethylation and ovarian cancer biology. Cancer Res 64:4472–4480 Wolff EM, Byun HM, Han HF, Sharma S, Nichols PW, Siegmund KD, Yang AS, Jones PA, Liang G (2010) Hypomethylation of a LINE-1 promoter activates an alternate transcript of the MET oncogene in bladders with cancer. PLoS Genet 6:e1000917 Yang AS, Este´cio MR, Doshi K, Kondo Y, Tajara EH, Issa JP (2004) A simple method for estimating global DNA methylation using bisulfite PCR of repetitive DNA elements. Nucleic Acids Res 32:e38 Yi JM, Kim HS (2006) Molecular evolution of the HERV-E family in primates. Arch Virol 151:1107–1116 Yi JM, Kim TH, Huh JW, Park KS, Jang SB, Kim HM, Kim HS (2004) Human endogenous retroviral elements belonging to the HERV-S family from human tissues, cancer cells, and primates: expression, structure, phylogeny and evolution. Gene 342:283– 292 Yi JM, Kim HM, Kim HS (2006) Human endogenous retrovirus HERV-H family in human tissues and cancer cells: expression, identification, and phylogeny. Cancer Lett 231:228–239 Yi JM, Schuebel K, Kim HS (2007) Molecular genetic analyses of human endogenous retroviral elements belonging to the HERV-P family in primates, human tissues, and cancer cells. Genomics 89:1–9 Yoshida T, Yamashita S, Takamura-Enya T, Niwa T, Ando T, Enomoto S, Maekita T, Nakazawa K, Tatematsu M, Ichinose M et al (2011) Alu and Satalpha hypomethylation in Helicobacter pylori-infected gastric mucosae. Int J Cancer 128:33–39

123