Radicales y Dna PDF

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 6

Available online at www.sciencedirect.

com

Cancer Letters 266 (2008) 6–11


www.elsevier.com/locate/canlet

Mini-review

Oxidative stress, DNA methylation and carcinogenesis


Rodrigo Franco a,b,1, Onard Schoneveld b,1, Alexandros G. Georgakilas c,
Mihalis I. Panayiotidis d,*
a
Laboratory of Cell Biology and Signal Transduction, Biomedical Research Unit, FES-Iztacala, UNAM, Mexico, USA
b
Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, National Institutes of Health,
Research Triangle Park, NC 27709, USA
c
Department of Biology Thomas Harriot College of Arts and Sciences, East Carolina University, Greenville, NC 27858, USA
d
School of Public Health, University of Nevada-Reno, MS-274, Reno, NV 89557, USA

Received 12 February 2008; received in revised form 12 February 2008; accepted 14 February 2008

Abstract

Transformation of a normal cell to a malignant one requires phenotypic changes often associated with each of the ini-
tiation, promotion and progression phases of the carcinogenic process. Genes in each of these phases acquire alterations in
their transcriptional activity that are associated either with hypermethylation-induced transcriptional repression (in the
case of tumor suppressor genes) or hypomethylation-induced activation (in the case of oncogenes). Growing evidence sup-
ports a role of ROS-induced generation of oxidative stress in these epigenetic processes and as such we can hypothesize of
potential mode(s) of action by which oxidative stress modulates epigenetic regulation of gene expression. This is of outmost
importance given that various components of the epigenetic pathway and primarily aberrant DNA methylation patterns
are used as potential biomarkers for cancer diagnosis and prognosis.
Ó 2008 Elsevier Ireland Ltd. All rights reserved.

Keywords: Oxidative stress; Free radicals; Reactive oxygen species; Oxidative DNA damage; DNA methylation; Epigenetics; Gene
expression; Cancer

1. Introduction oxygen species (ROS) and/or a reduction in antiox-


idant defenses responsible for their metabolism.
Oxidative stress has long been known to be This generates an imbalance between ROS produc-
involved in the pathophysiology of many human tion and removal in favor of the former.
diseases including, but not restricted, to cancer. Cancer is a multistage process and often involves
The term ‘‘oxidative stress”, refers to a cell’s state ‘‘alterations” or ‘‘changes” in the transcriptional
characterized by excessive production of reactive activity of genes associated with many critical cellu-
lar processes for tumor development including pro-
liferation, senescence, inflammation, metastasis, etc.
*
Corresponding author. Tel.: +1 775 682 7082; fax: +1 775 784 Furthermore, there are known to be both genotoxic
1340.
E-mail address: mpanagiotidis@unr.edu (M.I. Panayiotidis).
and non genotoxic mechanisms contributing to
1
Both authors have contributed equally to the preparation of malignant transformation. Generally, genotoxic
the manuscript. mechanisms involve changes in genomic DNA

0304-3835/$ - see front matter Ó 2008 Elsevier Ireland Ltd. All rights reserved.
doi:10.1016/j.canlet.2008.02.026
R. Franco et al. / Cancer Letters 266 (2008) 6–11 7

sequences that ultimately lead to mutations whereas endogenous ROS formation. More specifically, acti-
non-genotoxic include mechanisms (other than vated macrophages through ‘‘respiratory burst”
directly affecting DNA) capable of modulating gene give rise to various ROS and primarily superoxide
expression. ROS have been implicated at all stages and hydrogen peroxide. For instance, activation of
of the carcinogenic process by involving both types specialized macrophages in liver known as Kupffer
of mechanisms [1]. cells has been implicated in tumor promotion
On the other hand, the term ‘‘epigenetics” refers through ROS release [1].
to altered levels of a gene’s transcriptional activity On the other hand, exogenous sources of ROS
without directly affecting its primary DNA nucleo- generation include those of various xenobiotics
tide sequence. It involves alterations in DNA meth- (with a variable degree of potency), chlorinated
ylation patterns that together with specific histone compounds, various transition metals (participating
modifications (methylations, acetylations, deacety- in Fenton-type chemical reactions) and radiation,
lations, etc.) contribute to a transcriptional inactive all of which have been documented to cause ROS-
chromatin state [2]. In this respect, epigenetic regu- induced damage to cellular macromolecules
lation of gene expression can be viewed as a non (DNA, RNA, lipids and proteins) both in vitro
genotoxic mechanism for promoting tumor and in vivo [1].
formation.
Throughout this review article, we will present 3. Involvement of oxidative stress in carcinogenesis
the evidence for the involvement of oxidative stress
in carcinogenesis, consider the evidence for its role In general, the multistage process of carcinogen-
in inducing DNA methylation changes and assess esis involves the distinct phases of initiation, promo-
the importance of these changes in the multistage tion and progression. The cellular and molecular
process of human carcinogenesis. events underlying each of these phases include
DNA damage, increased proliferation, deficient cell
2. Sources of ROS death and further genetic instability, respectively
[1,4].
There are both endogenous and exogenous Oxidative DNA damage can trigger tumor initia-
sources of ROS generation. Endogenous sources tion. More specifically, studies using ionizing radia-
include those of: (1) mitochondrial oxidative phos- tion have shown multiple hydroxyl radical-induced
phorylation, (2) P450 metabolism, (3) peroxisomes genotoxic by- products in the bases as well as the
and (4) activation of inflammatory cells. It has been deoxyribose backbone of DNA. Among these by-
postulated that during oxidative phosphorylation products, the most characterized one is 8-hydroxy-
1–2% of molecular oxygen is converted to ROS 2-deoxyguanosine (8-OHdG) [5,6]. This DNA lesion
primarily through a series of sequential one-, two- has been shown to be mutagenic by causing G ? T
and three-electron reductions giving rise to superox- transversions in both bacterial and mammalian cells
ide, hydrogen peroxide and hydroxyl radical [7]. However, there are a number of studies indicat-
formation consecutively [3]. In addition, activation ing that oxidative DNA damage could not account
of P450 metabolism has been proposed as a very sig- entirely by itself for tumor development as they
nificant source of ROS formation by mechanisms have shown that elevated levels of 8-OHdG do not
that involve: (1) redox cycling, (2) peroxidase-cata- reflect increased cancer rates [8,9].
lyzed drug oxidations and (3) ‘‘futile cycling” of So, what other mechanisms could account for the
cytochrome P450. In particular, induction of P450 involvement of ROS in carcinogenesis? As it was
2E1 and 2B has been proposed to contribute signif- mentioned earlier on, ROS can affect a number of
icantly to ROS formation primarily through metab- cellular processes critical in tumor development
olism of ethanol and phenobarbital, respectively. such as cell proliferation, senescence, inflammation,
Furthermore, in a number of studies using chemi- metastasis, etc. In terms of cell proliferation, ROS
cals such as peroxisome proliferators, an increased have been shown to modulate cell cycle regulation
production of hydrogen peroxide was observed that through modulation of various cell cycle proteins
contributed to increased levels of oxidative stress including p53 [10] and ATM (ataxia telangiectasia
and their association with cancer induction. Finally, mutated) [11]. ROS have also been shown to modu-
inflammatory cells like neutrophils and macro- late the cell death (senescence) process [12] by acting
phages are perhaps the most significant sources of either as an anti-senescence stimulus [13] or through
8 R. Franco et al. / Cancer Letters 266 (2008) 6–11

the specific induction of AIF (apoptosis-inducing at lysine 16. Among these, the best-characterized
factor) which suppresses apoptosis and conse- histone modification(s) to date is methylation of his-
quently maintains the transformed phenotype of a tone H3 at lysine 9 and how it ‘‘translates” into
cancer cell [14]. On the other hand, inflammation DNA methylation in order to repress transcrip-
has long been known to act as a trigger in cancer tional activation [2,23–25].
development. For example, chronic hepatitis B viral But how is DNA methylation involved in regula-
infection and elevated liver levels of 8-OHdG can tion of transcription? In mammalian cells, the
lead to development of hepatocellular carcinoma enzymes critical for DNA methylation reactions
[15]. The mode of action by which chronic inflam- are DNA methyltransferases 1 (DNMT1), 3a
mation can trigger tumor initiation seems to involve (DNMT3a), and 3b (DNMT3b). These enzymes
NF-jB (nuclear factor jB) activation which, among catalyze the transfer of the methyl donor group
its pleiotrophic effects, can cause inhibition of apop- from S-adenosylmethionine (SAM) to the 50 -carbon
tosis [16]. Finally, metastasis is an integral part of of cytosines within CpG dinucleotide islands
tumor progression during which ROS have been (regions of 0.5–4.0 kb in length) in genomic DNA.
documented to play a major role [17–19]. In fact, More specifically, these enzymes are involved in
various studies have shown that metastatic tumor DNA methylation by means of either maintenance
cells produce higher levels of ROS than primary (DNMT 1) and/or de novo methylation (DNMT
malignant cells which together with increasing levels 3a and DNMT 3b). Thus, they could potentially
of ROS metabolizing enzymes and antioxidant com- contribute not only to increased promoter methyla-
pounds greatly reduce metastasis [20–22]. tion status (through de novo methylation) but also
ensure inheritance of gene silencing (through main-
4. The role of epigenetics in carcinogenesis tenance methylation) both of which could account
for the acquisition of a malignant transformation
In general, increased DNA methylation in the phenotype [26,27]. In general, 5-methylcytosine (by
promoter region of genes causes gene silencing and being a heritable modification) has the potential to
in this respect can contribute to the multistage pro- alter gene expression by means of: (1) causing an
cess of carcinogenesis. In addition, in mammalian increase in mutation rate [28] given that the presence
cells, both DNA methylation and chromatin struc- of 5-methylcytosine in the DNA promotes deamina-
ture are interconnected in specific ways so that genes tion of cytosine to uracil with 5-methylcytosine
will either be transcribed or repressed. Briefly, the deaminating 2–4 times faster than cytosines [29] or
process is initiated by DNA methyltransferases by (2) silencing genes necessary for controlling gene
(DNMTs) bringing together the DNA methylation proliferation [30,31]. Finally, many genes in tumor
machinery to the chromatin through recruitment cells have been shown to contain alterations in their
of histone deacetylases (HDACs) and other chro- DNA methylation patterns. Two such alterations
matin-binding proteins to promoter sites. In this have been proposed to be the common epigenetic
regards, the state of the chromatin’s acetylation sta- mechanisms underlying development of human can-
tus is important in regulating transcriptional activ- cer: (1) global hypomethylation occurring early in
ity in a way that histone acetylation maintains the progression phase of neoplasia and (2) regional
chromatin in a transcriptionally active state whereas hypermethylation of normally unmethylated CpG
histone deacetylation maintains transcriptional islands. Both events can result in the transcriptional
silencing. More specifically, during gene silencing silencing of tumor suppressor gene expression and
DNMTs direct the binding of HDACs and other the transcriptional activation of oncogenes respec-
proteins [methyl-CpG binding proteins (MeCPs); tively. In addition, increased expression of DNMT1
and methyl-CpG binding domain (MBP)] to hyper- has been found in early and late stages of neoplasia
methylated regions of the chromatin where they suggesting its involvement in the generation of
form complexes with other molecules and thus abnormal methylation patterns in cancer cells [32].
blocking access of the transcriptional machinery to Finally, genome-wide hypomethylation has been
the promoter. Specific histone modifications have shown to increase mutation rates and thus leading
been implicated in gene silencing including the fol- to genome instability [33]. It is therefore evident that
lowing: (1) histone H3 methylation at lysines 9 alterations in DNA methylation patterns underlie
and 27 and/or deacetylation at lysine 9, (2) histone aberrant gene expression that is associated with
H4 methylation at lysine 20 and/or deacetylation malignant transformation.
R. Franco et al. / Cancer Letters 266 (2008) 6–11 9

5. Involvement of oxidative stress in DNA chromatin condensation and transcriptional inacti-


methylation vation. Incorporation of 8-OHdG and 5-hydroxym-
ethylcytosine (the oxidation by-product of 5-
Oxidative stress can contribute to tumor develop- methylcytosine) in the MBP recognition sequence
ment not only through genetic but also through epi- resulted in the significant inhibition of the binding
genetic mechanisms. As it was mentioned earlier on, affinity of MBP [46]. These results provide further
generation of the hydroxyl radical can cause a wide evidence about the mechanism underlying ROS-
range of DNA lesions including base modifications, induced DNA damage in interfering with DNA
deletions, strand breakage, chromosomal rearrange- methylation patterns and consequently with tran-
ments, etc. Such DNA lesions have been shown to scriptional activation.
interfere with the ability of DNA to function as a Only recently, there have been a number of stud-
substrate for the DNMTs, resulting in global ies investigating changes in gene expression levels of
hypomethylation [34]. More specifically, X-rays major antioxidant enzymes in an attempt to relate
[35], ultraviolet [36] and c-rays [37] have been shown diminished levels with the pathophysiology underly-
to reduce the methyl-accepting ability of DNA. ing human carcinogenesis. In this respect, it has
Also, the presence of 8-OHdG in CpG dinucleotide been shown that decreased expression of the enzyme
sequences has been shown to strongly inhibit meth- manganese superoxide dismutase (MnSOD; which
ylation of adjacent cytosine residues [38,39], and metabolizes superoxide anion) was associated with
interfere with the ability of restriction nucleases to decreased proliferation in human pancreatic carci-
cleave DNA [40]. More specifically, in a series of ele- noma [47] and breast cancer [48] cells and that its
gant experiments, it was shown that when 8-OHdG inhibition was underlined by DNA hypermethyla-
is substituted for either guanine on the HpaII meth- tion-induced silencing. In addition, in the case of
ylase recognition site (CCGG) such substitution can the metallothionein gene expression (induced by a
inhibit DNA methylation of adjacent cytosines as variety of oxidative stress stimuli), it was also found
well as binding to the methyltransferase. The extent to be silenced through promoter-specific DNA
of this inhibition is dependent on the position of 8- hypermethylation events in both mouse lymphosar-
OHdG [38,39]. In addition, 8-OHdG may not be coma [49] and rat hepatoma [50] cells. Finally, both
recognized by proofreading enzymes and thus may the NAD(P)H: quinone oxidoreductase 1 (NQO1)
persist as a mutation resulting in G ? T transver- and glutathione S-transferase P1 (GSTP1) genes
sions [41]. Another potentially mutagenic lesion in (phase II xenobiotic metabolizing enzymes) have
ROS-induced DNA damage is O6-methylguanine. been shown to be inactivated via promoter hyper-
A number of studies have shown that its presence methylation in hepatocellular carcinoma [51,52],
can inhibit binding of DNA methyltransferases breast, renal [53] and prostate [54] cancers, respec-
and therefore can lead to hypomethylation by tively. These studies clearly demonstrate the associ-
means of inhibiting methylation of adjacent cyto- ation between repressed expression of key
sine molecules [42,43]. Alternatively, O6-methylgua- antioxidant enzymes (in metabolizing ROS genera-
nine can be spontaneously mispaired with thymine tion) and tumor development by means of promoter
and thus also contribute to DNA hypomethylation hypermethylation-induced gene silencing.
[44]. Finally, single-stranded DNA can signal de
novo methylation and, thus, it may be possible that 6. Conclusions and perspectives
formation of single strand breaks by oxidative stress
can contribute to the modifications of DNA methyl- It is evident that ROS-induced oxidative stress is
ation patterns observed in oxidant-transformed cell involved in the multistage process of carcinogenesis
lines [45]. These studies suggest that oxidative DNA by both genetic and epigenetic mechanisms. In par-
damage can affect patterns of DNA methylation ticular, there seems to be a growing interest (by
leading to aberrant gene expression and possibly many investigators) in the involvement of oxidative
contributing to the development of malignancy. stress in the epigenetic regulation of gene expression
As it was mentioned earlier on, DNA methyla- and specifically in controlling DNA methylation.
tion mediates gene expression via binding of MBPs Given the recent findings in detecting circulating
through DNA–protein and protein–protein interac- tumor DNA in patients with cancer [55], it becomes
tions followed by recruitment of histone modifying of paramount importance to reveal the molecular
enzymes. Consequently, these interactions result in mechanisms underlying aberrant DNA methylation
10 R. Franco et al. / Cancer Letters 266 (2008) 6–11

patterns since potentially they could be used as bio- [13] E.C. Vaquero, M. Edderkaoui, S.J. Pandol, I. Gukovsky,
markers in cancer prognosis and diagnosis. A.S. Gukovskaya, Reactive oxygen species produced by
NAD(P)H oxidase inhibit apoptosis in pancreatic cancer
cells, J. Biol. Chem. 279 (2004) 34643–34654.
Acknowledgements [14] A. Urbano, U. Lakshmanan, P.H. Choo, J.C. Kwan, P.Y.
Ng, K. Guo, S. Dhakshinamoorthy, A. Porter, AIF
This work was supported, in part, by the Intra- suppresses chemical stress-induced apoptosis and maintains
mural Research Program of the National Institutes the transformed state of tumor cells, EMBO J. 24 (2005)
2815–2826.
of Health (NIH)/National Institute of Environmen- [15] T.M. Hagen, S. Huang, J. Curnutte, P. Fowler, V. Martinez,
tal Health Sciences (NIEHS) (Onard Schoneveld C.M. Wehr, B.N. Ames, F.V. Chisari, Extensive oxidative
and Rodrigo Franco). DNA damage in hepatocytes of transgenic mice with chronic
Dr. Georgakilas has been supported by funds pro- active hepatitis destined to develop hepatocellular carci-
vided by the Biology Department of East Carolina noma, Proc. Natl. Acad. Sci. USA 91 (1994) 12808–12812.
[16] M. Karin, Nuclear factor-kB in cancer development and
University and a College Research Award (East Car- progression, Nature 441 (2006) 431–436.
olina University). Finally, Dr. Panayiotidis has been [17] P. Storz, Reactive oxygen species in tumor progression,
supported by funds provided by the School of Public Front. Biosci. 10 (2005) 1881–1896.
Health of the University of Nevada at Reno. [18] K. Mori, M. Shibanuma, K. Nose, Invasive potential
induced under long-term oxidative stress in mammary
epithelial cells, Cancer Res. 64 (2004) 7464–7472.
References [19] F. Okada, M. Kobayashi, H. Tanaka, T. Kobayashi, H.
Tazawa, Y. Luchi, K. Onuma, M. Hosokawa, M.C. Dina-
[1] J.E. Klauning, L.M. Kamendulis, The role of oxidative stress uer, N.H. Hunt, The role of nicotinamide adenine dinucle-
in carcinogenesis, Annu. Rev. Pharmacol. Toxicol. 44 (2004) otide phosphate oxidase-derived reactive oxygen species in
239–267. the acquisition of metastatic ability of tumor cells, Am. J.
[2] F. Fuks, DNA methylation and histone modifications: Pathol. 169 (2006) 294–302.
teaming up to silence genes, Curr. Opin. Genet. Dev. 15 [20] M. Nishikawa, K. Hyoudou, Y. Kobayashi, Y. Umeyama,
(2005) 490–495. Y. Takakura, M. Hashiba, Inhibition of metastatic tumor
[3] A. Pappa, R. Franco, O. Schoneveld, A. Galanis, R. growth by targeted delivery of antioxidant enzymes, J.
Sandaltzopoulos, M.I. Panayiotidis, Sulfur-containing com- Control. Release 109 (2005) 101–107.
pounds in protecting against oxidant-mediated lung diseases, [21] I. Heirman, D. Ginneberge, R. Brigellius-Flohe, N. Hend-
Curr. Med. Chem. 14 (2007) 2590–2596. rickx, P. Agostinis, P. Brouckaert, P. Rottiers, J. Grooten,
[4] B. Halliwell, Oxidative stress and cancer: have we moved Blocking tumor cell eicasonoid synthesis by GPx4 impedes
forward?, Biochem J. 401 (2007) 1–11. tumor growth and malignancy, Free Radic. Biol. Med. 40
[5] M.D. Evans, M. Dizdaroglu, M.S. Cooke, Oxidative DNA (2006) 285–294.
damage and disease: induction, repair, and significance, [22] F. Okada, H. Shionoya, M. Kobayashi, T. Kobayashi,
Mutat. Res. 567 (2004) 1–61. H. Tazawa, K. Onuma, Y. Luchi, N. Matsubara, T.
[6] L.J. Marnett, Oxyradicals and DNA damage, Carcinogen- Ijichi, B. Dugas, M. Hosokawa, Prevention of inflam-
esis 21 (2000) 361–370. mation-mediated acquisition of metastatic properties of
[7] M. Moriya, Single-stranded shuttle phagemid for mutagenic benign mouse fibrosarcoma cells by administration of an
studies in mammalian cells: 8-oxoguanine in DNA induces orally available superoxide dismutase, Br. J. Cancer 94
targeted G:C ? T:A transversions in simian kidney cells, (2006) 854–862.
Proc. Natl. Acad. Sci. USA 90 (1993) 1122–1126. [23] H. Santos-Rosa, C. Caldas, Chromatin modifier enzymes,
[8] B. Halliwell, Effect of diet on cancer development: is the histone code and cancer, Eur. J. Cancer 41 (2005) 2381–
oxidative DNA damage a biomarker?, Free Radic Biol. 2402.
Med. 32 (2002) 968–974. [24] M. Esteller, DNA methylation and cancer therapy: new
[9] T. Arai, V.P. Kelly, O. Minowa, T. Noda, S. Nishimura, The developments and expectations, Curr. Opin. Oncol. 17 (2004)
study using wild-type and Ogg1 knockout mice exposed to 55–60.
potassium bromate shows no tumor induction despite an [25] S.B. Baylin, DNA methylation and gene silencing in cancer,
extensive accumulation of 8-hydroxyguanine in kidney Nat. Clin. Pract. Oncol. 2 (2005) S4–S11.
DNA, Toxicology 221 (2006) 179–186. [26] A. Bird, The essentials of DNA methylation, Cell 70 (1992)
[10] K. Bensaad, K.H. Vousden, Savior and slayer: the two faces 5–8.
of p53, Nat. Med. 11 (2005) 1278–1279. [27] M.J. Hitchler, F.E. Domann, An epigenetic perspective on
[11] K. Ito, A. Hirao, F. Arai, K. Takubo, S. Matsuoka, K. the free radical theory of development, Free Radic. Biol.
Miyamoto, M. Ohmura, K. Naka, K. Hosokawa, Y. Ikeda, Med. 43 (2007) 1023–1036.
T. Suda, Reactive oxygen species act through p38 MAPK to [28] W.M. Rideout, G.A. Coetzee, A.F. Olumi, P.A. Jones, 5-
limit the lifespan of hematopoietic stem cells, Nat. Med. 12 Methylcytosine as an endogenous mutagen in the human
(2006) 446–451. LDL receptor and p53 genes, Science 249 (1990) 1288–1290.
[12] J. Chandra, A. Samali, S. Orrenius, Triggering and modu- [29] G.P. Pfeifer, M. Tang, M.F. Denissenko, Mutation hotspots
lation of apoptosis by oxidative stress, Free Radic. Biol. and DNA methylation, Curr. Top. Microbiol. Immunol. 249
Med. 29 (2000) 323–333. (2000) 1–19.
R. Franco et al. / Cancer Letters 266 (2008) 6–11 11

[30] P.A. Jones, DNA methylation errors and cancer, Cancer [44] W. Xiao, L. Samson, In vivo evidence for endogenous DNA
Res. 56 (1996) 2463–2467. alkylation damage as a source of spontaneous mutation in
[31] S.B. Baylin, J.G. Herman, J.R. Graff, P.M. Vertino, J.P. eukaryotic cells, Proc. Natl. Acad. Sci. USA 90 (1993) 2117–
Issa, Alterations in DNA methylation: a fundamental aspect 2121.
of neoplasia, Adv. Cancer Res. 72 (1998) 141–196. [45] J.K. Christman, G. Sheikhnejad, C.J. Marasco, J.R. Sufrin,
[32] W.S. El-Deiry, B.D. Nelkin, P. Celano, R.W. Yen, J.P. 5-Methyl-20 -deoxycytidine in single-stranded DNA can act
Falco, S.R. Hamilton, S.B. Baylin, High expression of the in cis to signal de novo DNA methylation, Proc. Natl. Acad.
DNA methyltransferase gene characterizes human neoplastic Sci. USA 92 (1995) 7347–7351.
cells and progression stages of colon cancer, Proc. Natl. [46] V. Valinluck, H-H. Tsai, D.K. Rogstad, A. Burdzy, A. Bird,
Acad. Sci. USA 88 (1991) 3470–3474. L.C. Sowers, Oxidative damage to methyl-CpG sequences
[33] J.E. Klaunig, L.M. Kamendulis, Y. Xu, Epigenetic mecha- inhibits the binding of the methyl-CpG binding domain
nisms of chemical carcinogenesis, Hum. Exp. Toxicol. 19 (MBD) of methyl-CpG binding protein 2 (MeCP2), Nucleic
(2000) 543–555. Acids Res. 32 (2004) 4100–4108.
[34] J.T. Wachsman, DNA methylation and the association [47] E.M. Hurt, S.B. Thomas, B. Peng, W.L. Farrar, Molecular
between genetic and epigenetic changes: relation to carcino- consequences of SOD2 expression in epigenetically silenced
genesis, Mutat. Res. 375 (1997) 1–8. pancreatic carcinoma cell lines, Br. J. Cancer 97 (2007) 1116–
[35] V.L. Wilson, P.A. Jones, Inhibition of DNA methylation by 1123.
chemical carcinogens in vitro, Cell 32 (1983) 239–246. [48] M.J. Hitchler, K. Wikainapakul, L. Yu, K. Powers, W.
[36] F.F. Becker, P. Holton, M. Ruchirawat, J.-N. Lapeyre, Attatippaholkun, F.E. Domann, Epigenetic regulation of
Perturbation of maintenance and de novo DNA methylation manganese superoxide dismutase expression in human breast
in vitro by UVB (280–340 nm)-induced pyrimidine photodi- cancer cells, Epigenetics 1 (2006) 163–171.
mers, Proc. Natl. Acad. Sci. USA 82 (1985) 6055–6059. [49] S. Majumder, K. Ghosal, Z. Li, Y. Bo, S.T. Jacob, Silencing
[37] J.F. Kalinich, G.N. Catravas, S.L. Snyder, The effect of c of metallothionein-I gene in mouse lymphosarcoma cells by
radiation on DNA methylation, Radiat. Res. 117 (1989) methylation, Oncogene 18 (1999) 6287–6295.
185–197. [50] K. Ghosal, S. Majumder, Z. Li, X. Dong, S.T. Jacob,
[38] S.A. Weitzman, P.W. Turk, D.H. Milkowski, K. Kozlowski, Suppression of metalothionein gene expression in a rat
Free radical adducts induce alterations in DNA cytosine hepatoma because of promoter-specific DNA methylation, J.
methylation, Proc. Natl. Acad. Sci. USA 91 (1994) 1261– Biol. Chem. 275 (2000) 539–547.
1264. [51] M. Tada, O. Yokosuka, K. Fukai, T. Chiba, F. Imazeki, T.
[39] P.W. Turk, A. Laayoun, S.S. Smith, S.A. Weitzman, DNA Tokuhisa, H. Saisho, Hypermethylation of NAD(P)H:
adduct 8-hydroxyl-20 -deoxyguanosine (8-hydroxyguanine) quinine oxidoreductase 1 (NQO1) gene in human hepato-
affects function of human DNA methyltransferase, Carcino- cellular carcinoma, J. Hepatol. 42 (2005) 511–519.
genesis 16 (1995) 1253–1255. [52] S. Zhong, M.W. Tang, W. Yeo, C. Liu, Y.M.D. Lo, P.J.
[40] P.W. Turk, S.A. Weitzman, Free radical DNA adduct 8-OH- Johnson, Silencing of GSTP1 gene by CpG island DNA
deoxyguanosine affects activity of HPA II and MSP I hypermethylation in HBV-associated hepatocellular carcino-
restriction endonucleases, Free Radic. Res. 23 (1995) 255– mas, Clin. Cancer Res. 8 (2002) 1087–1092.
258. [53] M. Esteller, P.G. Corn, J.M. Urena, E. Gabrielso, S.B.
[41] Y. Kuchino, F. Mori, H. Kasai, H. Inoue, S. Iwai, K. Miura, Baylin, J.G. Herman, Inactivation of glutathione S-trans-
E. Ohtsuka, S. Nishimura, Misreading of DNA templates ferase P1 gene by promoter hypermethylation in human
containing 8-hydroxydeoxyguanosine at the modified base neoplasia, Cancer Res. 58 (1998) 4515–4518.
and at adjacent residues, Nature 327 (1987) 77–79. [54] D.S. Millar, K.K. Ow, C.L. Paul, P.J. Russell, P.L. Molloy,
[42] P.A. Hepburn, G.P. Margison, M.J. Tisdale, Enzymatic S.J. Clark, Detailed methylation analysis of the glutathione
methylation of cytosine in DNA is prevented by adjacent O6- S-transferase P1 (GSTP1) gene in prostate cancer, Oncogene
methylguanine residues, J. Biol. Chem. 266 (1991) 7985– 18 (1999) 1313–1324.
7987. [55] R.E. Board, L. Knight, A. Greystoke, F.H. Blackhall, A.
[43] N.-W. Tan, B.F.L. Li, Interaction of oligonucleotides Hughes, C. Dive, M. Ranson, DNA methylation in circu-
containing 6-O-methylguanine with human DNA (cytosine- lating tumor DNA as a biomarker for cancer, Biomark.
5-)-methyltransferase, Biochemistry 29 (1990) 9234–9240. Insights. 2 (2007) 307–319.

You might also like