You are on page 1of 7

Mutation Research 711 (2011) 167–173

Contents lists available at ScienceDirect


Mutation Research/Fundamental and Molecular
Mechanisms of Mutagenesis
journal homepage: www.elsevier.com/locate/molmut
Community address: www.elsevier.com/locate/mutres

Review

Reactive Oxygen Species (ROS)––Induced genetic and epigenetic alterations in


human carcinogenesis
Dominique Ziech a , Rodrigo Franco b , Aglaia Pappa c,∗ , Mihalis I. Panayiotidis d,e,∗∗
a
Department of Student Success Services, University of Nevada, Reno, NV 89557, USA
b
Redox Biology Center & School of Veterinary Medicine & Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA
c
Department of Molecular Biology & Genetics, Democritus University of Thrace, Dragana Campus, Alexandroupolis 68100, Greece
d
School of Community Health Sciences, University of Nevada, Reno, NV 89557, USA
e
Department of Pathology, Medical School, University of Ioannina, University Campus, Ioannina 45110, Greece

a r t i c l e i n f o a b s t r a c t

Article history: Cancer is a multistage and complex process characterized by molecular alterations that underlie all three
Received 19 November 2010 phases of its development: (i) initiation, (ii) promotion and (iii) progression. Some of these molecular
Received in revised form 26 February 2011 events include alterations in gene expression that are regulated by both genetic and epigenetic mecha-
Accepted 28 February 2011
nisms. On the other hand, “oxidative stress” implies a cellular state where ROS production exceeds the
Available online 16 March 2011
cell’s ability to metabolize them resulting in excessive accumulation of ROS that overwhelms cellular
defenses. Such state has been shown to regulate both genetic and epigenetic cascades underlying altered
Keywords:
gene expression in human disease including cancer. Throughout this manuscript, we review the current
Reactive Oxygen Species (ROS)
Oxidative stress
state of knowledge on the role of ROS-induced oxidative stress in altering the genetic and epigenetic
Epigenetics involvement during human carcinogenesis.
DNA methylation © 2011 Elsevier B.V. All rights reserved.
Histone modifications
DNA damage
DNA repair
DNA methyltransferases
HATs
HDACs
Tumor suppressor genes
Oncogenes
Human carcinogenesis

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 168
2. Oxidative stress-induced genetic alterations in human carcinogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 168
3. ROS-induced oxidative stress based epigenetic alterations in human carcinogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 170
4. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 172
Conflict of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 172
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 172
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 172

∗ Corresponding author at: Department of Molecular Biology & Genetics, Democritus University of Thrace, Dragana Campus, Alexandroupolis 68100, Greece.
Tel.: +30 25510 30625.
∗∗ Corresponding author at: Department of Pathology, Medical School, University of Ioannina, University Campus, Ioannina 45110 Greece. Tel.: +30 25510 35689.
E-mail addresses: apappa@mbg.duth.gr (A. Pappa), mpanagiotidis@unr.edu (M.I. Panayiotidis).

0027-5107/$ – see front matter © 2011 Elsevier B.V. All rights reserved.
doi:10.1016/j.mrfmmm.2011.02.015
168 D. Ziech et al. / Mutation Research 711 (2011) 167–173

1. Introduction genome. Major oxidative DNA damage products include those of


8-oxo-7,8-dihydroadenine (8-oxoAde), 8-oxo-7,8-dihydroguanine
The term “free radicals” refer to any molecular species with one (8-oxoGua), 8-oxo-7,8-dihydro-2 -deoxyguanosine (8-oxodG), and
or more unpaired electron(s), a characteristic that provides them 5,6-dihydroxy-5,6-dihydrothymine as well as the ring-open
with an “unstable” chemical nature and thus confers them with the lesions of 4,6-diamino-5-formamido-pyrimidine and 2,6-diamino-
property of being very reactive. It is indeed this property that when 4-hydroxy-5-formamido-pyrimidine [4]. Elevated levels of such
they react with DNA, RNA, proteins, and lipids, leads to cellular DNA lesions have been recorded in many tumor types and are
toxicity. Free radicals include both reactive oxygen (ROS; such as, strongly implicated in all stages of carcinogenesis [5]. Of these
superoxide (O2 − ), hydroxyl (OH− ), hydroperoxyl (HOO− ), peroxyl oxidative lesions, 8-oxoGua has received most attention due to
(ROO− ) and alkoxyl (RO− ) radicals [4,7]) and nitrogen (RNS) species. its potential to serve as a reliable biomarker of oxidative stress
In particular, ROS production can be of endogenous (mitochondrial and thus as a promising biomarker of ROS-induced carcinogenesis
and P450 metabolism, inflammatory cell activation, etc.) as well [2,4]. It is hypothesized to have strong mutagenic and carcinogenic
as of exogenous (radiation, ozone, etc.) origin. In fact, the major- potential, and has been steadily observed in tissues containing low
ity of environmental, occupational and industrial pollutants are concentrations of antioxidant enzymes and high concentrations of
chemicals able to generate free radical species (primarily through ROS [4]. While 8-oxoGua adducts occur at approximately one in
their metabolic activation) that contribute to the pathophysiology 105 guanidine residues, in normal human cells, its rate of forma-
of human disease including cancer. Finally, when ROS generation tion increases by 35–50% in individuals using tobacco smoke – a
exceeds the cell’s ability to metabolize and detoxify them, a state well known carcinogenic source of ROS [2]. In addition, almost 50%
of “oxidative stress” emerges [45–47]. higher rates of 8-oxoGua levels have been observed in lung, breast
In general, cancer development is a multi-stage process char- or prostate cancer patients when compared to otherwise healthy
acterized by the cumulative action of a number of altered cellular individuals [4]. Furthermore, recent investigations have showed
processes including those of replication, angiogenesis, apoptosis, higher endogenous levels of 8-oxoGua in tumor tissues when com-
metastasis etc. However, although some of these altered cellu- pared to controls, thus suggesting oxidative DNA damage as a
lar processes have been extensively studied, there is still much contributing factor in cancer development [12]. In addition, high
to learn in regards to the molecular mechanism(s) involved. This levels of 8-oxoGua and possibly other DNA lesions are suggested as
is of great importance in enhancing our understanding about the reliable risk factors associated with the transformation of benign to
three main stages of human carcinogenesis, namely initiation, malignant tumors [2]. Furthermore, 8-oxoGua lesions are known
promotion and progression, and how they contribute to the com- to induce aberrant modifications in adjacent DNA a hypothesized
plexity of human cancer biology [45]. To this end, for example, mechanism that significantly contributes to the genetic instability
ROS-mediated DNA damage plays a role in the initiation of car- and metastatic potential of tumor cells [4]. For example, forma-
cinogenesis as well as in malignant transformation [8,42] and tion of 8-oxoGua lesions have been shown to induce a cascade
thus, it may represent a major contributor in the pathogenesis of of adjacent DNA base mutations, such as GC → TA transversions
human carcinogenesis [7]. Finally, because human carcinogene- in the RAS and the TP53 oncogene and tumor suppressor genes
sis is rather complex and multi-stage in origin, there are several respectively in both lung and liver cancer cells [4]. Under normal
different molecular mechanisms that contribute to its pathogen- conditions, DNA repair mechanisms including the DNA glycosy-
esis that include, but are not limited to, those that regulate lases of 8-oxoguanine glycosylase (OGG1) and MutY glycosylase
gene expression and involve both genetic and epigenetic alter- homologue (MUTYH) (in the base excision repair pathway) excise
ations. It would certainly be of major interest to identify such 8-oxodG lesions when paired with cytosine and adenine respec-
mechanism(s) in an attempt to elucidate their potential role as tively [39]. However, in the presence of cumulative oxidative stress,
biomarkers for better cancer diagnostics and therapeutic strategies repair mechanisms often are ineffective. Mutations known to accu-
[30,48]. mulate in response to failed repair mechanisms are those found in
the Ras oncogene and the p53 tumor suppressor gene. Mutations
in the Ras oncogene (a membrane-bound G protein whose main
2. Oxidative stress-induced genetic alterations in human function is to regulate cell growth and oppose apoptotic effects)
carcinogenesis have been well documented in about 30% of lung, skin, liver, blad-
der and colon cancers [2]. On the other hand, mutations in the p53
The carcinogenicity of oxidative stress is primarily attributed to tumor suppressor gene have been established as common genetic
the genotoxicity of ROS in diverse cellular processes [7]. For exam- alterations whereby they lead to gene’s inactivation in more than
ple, hydroxyl radicals can react with pyrimidines and/or purines half of all human cancers. For instance, a recent study has observed
as well as chromatin proteins, resulting in base modifications and a direct relationship between oxidative stress, DNA damage and
genomic instability respectively all of which can cause alterations in elevated frequency of p53 mutations in human dysplastic colon
gene expression [42]. In addition, data have implicated ROS accu- and in colorectal carcinoma [34]. In general, p53 is known to reg-
mulation as being a common phenomenon in many cancer cells. ulate cell cycle entry and also act as a redox-active transcription
Such accumulation can cause direct DNA damage by increasing a factor whose function is linked in regulating ROS generation as
cell’s mutation rate and/or promoting and maintaining the onco- well as mediating ROS-induced apoptotic cell death. Several cyto-
genic phenotype by acting as a secondary messenger in intracellular sine residues in the central domain of the protein are critical for
signaling cascades [2,3]. p53 binding and it is in these regions where mutations most com-
ROS-mediated DNA damage in addition to ineffective DNA monly occur [41]. ROS-mediated posttranslational modifications
repair mechanisms are well established lesions common to human have been shown to inhibit p53, pro-apoptotic enzymes’ (i.e. cas-
cancers such as in the case of breast cancer cell lines and human pases) function and also activate telomerases, metalloproteinases,
breast tumor tissue samples [37]. DNA alterations like strand and DNA methyltransferases [34]. During normal cell proliferation,
breaks, base modifications and DNA–protein cross linkages, are all p53 triggers mechanisms that eliminate oxidative DNA damage
strongly implicated in the initiation stage of carcinogenesis [1]. in comparison to the presence of oxidative damage where p53
For example, hydroxyl radicals are known to attack DNA caus- triggers apoptotic cell death. However, during the carcinogenic
ing single and/or double strand breaks as well as pyrimidine process an imbalance between cell proliferation and cell death
and purine lesions both of which can affect the integrity of the exists that shifts towards uncontrolled cell proliferation [2]. Finally,
D. Ziech et al. / Mutation Research 711 (2011) 167–173 169

p53-depleted cells exhibit significant disruption of cellular ROS As discussed earlier, a significant effect of cumulative oxida-
homeostasis characterized by reduced mitochondrial and cellu- tive DNA damage is the impairment of normal cellular repair
lar superoxide and increased cellular hydrogen peroxide levels mechanisms. In fact, one of the main etiological hypotheses link-
[40]. ing genomic instability, mutagenesis and tumorigenesis is that
Exogenous sources of oxidative stress like tobacco smoking, of deficient cellular repair mechanisms due to extensive oxida-
alcohol consumption, and ionizing radiation are all involved in cel- tive DNA damage and cellular injury. Complex DNA damage, such
lular toxicity induced by oxidative stress [49,50]. Ionizing radiation, as DSBs and OCDLs are considered the hallmarks of irradiation-
for example, alone, can induce genomic instability in cells, which is induced oxidative damage [51,52]. OCDLs are hypothesized to
hypothesized to enhance the rate by which mutations and other challenge the normal cellular repair machinery via cumulative
genetic alterations occur. In fact, recent work has revealed that cytotoxic and mutagenic effects. Additionally, endogenous forma-
after irradiation of prostate and cervical cancers, the risk for both tion of OCDLs are known to accumulate in X-irradiated mouse
rectal and bladder tumors increase respectively [35]. Exogenous skin tissues and human breast cancer cells, leading to induction
exposure to ionizing radiation is also known to induce cumulative of genetic instability and mutagenesis [9]. In general, the base
oxidative stress, over time, and result in DNA damage, cell death, excision pathway repairs DSBs and OCDLs efficiently. In addition,
altered proliferation states and ultimately carcinogenesis. More the DNA-dependent protein kinase catalytic subunit (DNA-PKcs)
specifically, ionizing radiation-induced oxidative stress has been is known to play an important role during the processing of DSBs.
shown to induce PI3K, MAPK, JNK and p38 signaling pathways as Only recently DNA-PKcs interactions with traditional BER proteins,
well as inhibit protein tyrosine phosphatase activity, both of which like XRCC1, APE1 or Pol␤, have generated much interest in the
play an important role in controlling cell proliferation and induc- ability and involvement of DNK-PKcs to process oxidative DNA
ing metastasis [35]. In addition, a recent study has suggested that lesions [43]. Nevertheless, it is well established that when left
oxidative stress-induced cellular damage (via occupational expo- unrepaired, these complex DNA lesions play critical roles in the
sure to low doses of ionizing radiation) results in the generation initiation, promotion and progression of carcinogenesis. In fact, to
of a variant of the PTSG2 gene that may serve as a strong indi- date, such complex DNA lesions are thought to be able to serve as:
cator of increased breast cancer risk [36]. Furthermore, it is well (i) biomarkers in human acute lymphoblastic leukemia cells [44],
established that ionizing radiation induces a wide spectrum of (ii) human tumor tissues from patients with liver, ovary, kidney,
DNA damage lesions, including single strand breaks (SSBs), dam- breast and colon cancer [53] as well as (iii) elevated breast cancer
aged bases, abasic sites and high levels of complex DNA damage risk [43].
such as double strand breaks (DSBs) and oxidatively induced non- Surmounting evidence has implicated the involvement of mito-
DSB clustered DNA lesions (OCDLs) lesions. DSBs are known to be chondrial oxidative DNA damage in the carcinogenesis process.
highly genotoxic DNA lesions because potentially they can lead to In fact, ROS-mediated mutations in mitochondrial DNA (mtDNA)
chromosomal breakage and can arise as the result of exposure to have recently emerged as an important contributor to human
exogenous and endogenous sources of free radical production [43]. carcinogenesis [42]. Mutations in mitochondrial genes encoding
These lesions are generally repaired by the base excision repair complexes I, III, IV and V, as well as within the hypervariable
pathway however, data have implicated that unsuccessful lesions’ region of mtDNA, have been identified in various human can-
repair contributes to significantly increased rates of mutations cers. In general, mtDNA is more susceptible to oxidative damage
and genomic instability [44]. In a separate study, alcohol-induced than nuclear DNA because (i) mitochondrial DNA is not protected
oxidative stress has been shown to generate excess hepatic DNA by histones, (ii) mitochondrial DNA repair capacity is limited,
damage leading to hepatocellular carcinoma [34]. Alcohol is known and (iii) under physiological conditions, the mitochondria con-
to facilitate ROS production and the formation of an in vivo oxida- verts roughly 3–5% of oxygen consumed into superoxide anion and
tive microenvironment and thus enabling suitable conditions for subsequently hydrogen peroxide. In addition, mtDNA is located
the development of cancer. While the underlying mechanism of the in close proximity to the respiratory chain and thus is conse-
relationship between oxidative stress, alcohol and cancer has yet to quently readily exposed to ROS-induced oxidative damage. As a
be elucidated, it is hypothesized that excess ROS production results result, mtDNA has more than two orders of magnitude higher
from the oxidation of ethanol whereby a variety of by-products frequency of oxidative damage than that of nuclear DNA and signif-
cause antioxidant depletion and oxidative macromolecular dam- icantly correlates with the development of cancer [41]. While the
age [35]. In addition, exposure to tobacco smoke has also been underlying mechanism(s) by which mitochondrial DNA alterations
strongly linked to oxidative stress-induced DNA damage and car- participate in the carcinogenic process has yet to be revealed, data
cinogenesis [6]. Cigarette smoking is known to contain increased have suggested that aberrant mtDNA fragment insertion within
levels of free radicals leading to the induction of DNA lesions and regions of nuclear DNA can contribute as a potential mecha-
various oxidized bases (i.e. 8-oxoGua). A vast number of data have nism for oncogenic activation [2]. On another note, exciting new
indicated the central role of tobacco smoke in the development research has suggested that under conditions of excessive DNA
of a number of different human cancers like lung, oral, pharyn- damage, p53 directly activates the mitochondrial death signaling
geal, laryngeal, esophageal, bladder, stomach, pancreatic, kidney, pathways in cancer cells [41]. This process is controlled by the
uterine, cervical and myeloid leukemic ones [38]. Additionally, it (i) MDM family of p53-binding regulatory proteins, (ii) associa-
has been known that free radicals produced during auto-oxidation tion between p53 and mitochondrial proteins of the BCL-2 family
of polyphenols in saliva of tobacco users are crucial to the ini- and (iii) VDAC (known to influence permeability of the mitochon-
tiation and promotion of oral, pharynx, larynx and esophageal drial outer membrane). Taken together, these findings imply a
cancers. More specifically, cigarette smoke has been observed to: novel role for p53 (that is, its involvement in maintaining nor-
(i) cause the oxidation of glutathione (GSH; a common antioxidant mal mtDNA copy number and cellular ROS homeostasis) and thus
known to protect against ROS damage), (ii) decrease blood antiox- proving how loss of p53 function may promote the carcinogenic
idant levels and (iii) increase superoxide radical release. Taken process [40]. Moreover, VDAC has been recently identified as a
together, the ability of genotoxic carcinogens to induce muta- critical target of ROS buildup (susceptible to superoxide injury)
tions coupled with the failure of repairing this damage results in leading to the subsequent stimulation of apoptosis. These find-
“gain-of-function” activity of oncogenes (i.e. K-ras) and/or “loss- ings support the hypothesis that ROS-mediated cellular damage
of-function” activity of tumor suppressor genes (i.e. p53 and p16) induces a state of redox imbalance and pro-carcinogenic activity
[35]. [42].
170 D. Ziech et al. / Mutation Research 711 (2011) 167–173

3. ROS-induced oxidative stress based epigenetic stress-induced carcinogenesis [10]. In fact, many tumor suppres-
alterations in human carcinogenesis sor genes are inactivated via ROS-mediated aberrant methylation
of CpG island-rich promoter regions [16]. For example, studies
In addition to causing genetic changes, ROS may lead to epige- have showed that when exposed to oxidative stress, tumor sup-
netic alterations that affect the genome and play a key role in the pressor genes p15INK4B and p16INK4A accrued aberrant methylation
development of human carcinogenesis [3]. More specifically, ROS patterns and ultimately their expression was silenced [10]. E-
production is associated with alterations in DNA methylation pat- cadherin, another tumor suppressor gene, has also been shown to
terns [17,54]. In particular, hydroxyl radical-induced DNA lesions undergo ROS-mediated hypermethylation leading to its silenced
(such as 8-hydroxyl-2-deoxyguanosine; 8-hydroxyguanine; 8- expression. Such promoter hypermethylation is hypothesized to
OHdG [56–59], O6 -methylguanine [60,61] and single stranded DNA occur via the recruitment of methyl-CpG binding proteins that
[62]) have all been shown to contribute to decreased DNA methy- then recruit histone deacetylases, histone methyltransferases, het-
lation by means of interfering with the ability of DNA to function erochromatin proteins etc. [16]. These epigenetic events alter the
as a substrate for the DNA methyltransferases (DNMTs) and thus chromatin structure in ways that block the access of transcription
resulting in global hypomethylation [13]. In addition, ROS-induced factor complexes to DNA and thus repressing transcription [16]. For
oxidative stress can contribute to gene silencing by mechanisms example, down-regulation of E-cadherin has been correlated with
that involve aberrant hypermethylation of tumor suppressor gene epithelial-to-mesenchymal transitions, metastasis, and poor prog-
promoter regions and thus lead towards progression to a malig- nosis in hepatocellular carcinoma [15]. Other tumor suppressor
nant phenotype. For example, a recent study has shown that genes, such as CDKN2A, RB, VHL and BRAC1, have also been identified
exposure of hepatocellular carcinoma cells to hydrogen peroxide in cancer cells as being inactivated via oxidative-induced aber-
(H2 O2 ) induced hypermethylation of the E-cadherin gene promoter rant CpG island promoter methylation [21,55]. In addition, recent
by increasing the expression of Snail (a transcription factor that studies have demonstrated that both MHL1 (a mismatch repair
down-regulates the expression of E-cadherin) which then induced gene) and MGMT (responsible for the removal of O6 -methylguanine
methylation of the gene’s promoter by recruiting histone deacety- adducts) gene promoter regions are hypermethylation-induced
lase 1 and DNA methyltransferase 1 as well [15]. Thus, we may silenced thus leading to increased mutation rates in human colorec-
conclude that whether by means of DNA hypo- and/or hyperme- tal cancers [63–65]. Genome-wide screening of tumor DNA has led
thylation ROS-induced epigenetic alterations have been implicated to the identification of several tumor-suppressor genes displaying
with malignant transformation and the progression of numer- oxidative stress-induced alterations in methylation patterns. Such
ous tumors [3]. More than 20 years ago, the impact of epigenetic gene products include silencing of SOCS1 in hepatocellular carci-
contribution to disease was first realized when various studies noma, SLC5A8 in colon cancer and BMP3B in non-small-cell lung
distinguished human cancer from normal tissue based upon DNA cancer [21]. In a recent study, 68% of patients with non-small-cell
methylation levels [20]. Since then, a wide range of studies have lung cancer, showed aberrant promoter methylation in at least one
determined major epigenetic alterations in cancer cells, such as of the following genes: p16, RASSF1A, FHIT, H-cadherin and RARˇ
silencing of tumor suppressor genes, activation of oncogenes and [24]. Moreover, in patients with endometrial cancer aberrant DNA
defects in DNA repair [28]. DNA methylation, in part, is perhaps the hypermethylation was observed in three or more genes from a
most extensively studied epigenetic modification, and is hypothe- panel of five analyzed (CDH13, HSPA2, MHL1, RASSF1A, SOCS2) [24].
sized to play an important role in the regulation of gene expression Finally, in patients with cervical neoplasia, detection of a panel of
and chromatin architecture, in association with histone modifi- three genes (DAPK1, RARˇ, TWIST1) associated with improved diag-
cation(s) and other chromatin-associated proteins [17]. In fact, nostics characterized by 95% specificity and sensitivity depending
aberrant DNA methylation is the most common molecular lesion primarily on tumor stage: 74% for invasive cancer and 52% for cervi-
of cancer cells [25]. In general, during DNA replication, methy- cal intraepithelial neoplasia and carcinoma in situ, respectively [24].
lation status is transmitted to corresponding daughter DNA by Overall, aberrant methylation of RB, p16, VHL, hMLH1, E-cadherin
maintenance of DNA methyltransferases, which recognize hemi- and BRAC1 have all been implicated in various cancers, including
methylated CpG sites at the replication fork [23]. Overall, CpG stomach, colon, liver, breast, uterine, renal and those of hema-
islands are kept unmethylated, however, when methylated, are tological origin [23]. However, it is also important to note that
known to repress the expression of the gene due to changes in chro- while methylation of promoter CpG islands leads to gene silencing,
matin structure and the resulting decreased affinity of transcription aberrant methylation of non-promoter CpG islands is also fre-
factors [23]. Methylated cytosines account for approximately 1% quently observed in cancer cells. For example, non-promoter CpG
of total nucleotides and about 75% of all CpG dinucleotides in islands overlapping exon 2 of the CDKN2A gene, are generally kept
the human genome. About 50–60% of gene promoters lie within unmethylated in normal cells while in cancer cells these regions
CpG islands, and it is estimated that the human genome contains are susceptible to ROS-mediated aberrant methylation. Because
approximately 29,000 of such CpG sequences [17,28]. these non-promoter CpG regions are known to be highly suscepti-
DNA methylation changes in cancer cells are characterized by (i) ble to ROS-mediated aberrant methylation, much interest has been
regional CpG island hypermethylation and (ii) generalized genomic invested in their use as diagnostic tools for tumor development
hypomethylation [19]. Such hypomethylation is thought to induce [21].
both genomic instability and activation of protooncogenes. Alterna- On the other hand, genome-wide hypomethylation has also
tively, promoter CpG island hypermethylation is thought to induce been detected in cancer cells. In fact, virtually all tumors exhibit
the silencing of tumor suppressor genes [13,24]. Such altered pat- a global decrease in genomic DNA methylation compared to nor-
terns of methylation lead to aberrant gene expression, in part, mal cells. So far, no relationship between local hypermethylation
by affecting the ability of methylated DNA-binding proteins to and genomic hypomethylation has been found, thus suggesting
interact with nearby transcription factors [14]. For example, a that these processes are independent [24]. However, only recently,
recent study has shown that various oxidized products (dimethyl evidence has linked genomic hypomethylation with oncogene
and methionine sulfoxide) may accumulate in the cytosol dur- overexpression in conjunction with chromosomal instability [25].
ing the initial stages of carcinogenesis and react with nearby In turn, DNA hypomethylation-induced chromosomal instability
nucleotides, leading to aberrant methylation-induced gene silenc- has also been linked to increased mutation rates and abnormal
ing [29]. Exciting new research has suggested tumor suppressor gene expression [26]. Over the last two decades, research has fur-
genes as major target genes of aberrant methylation in oxidative ther solidified hypomethylation to be strongly linked with global
D. Ziech et al. / Mutation Research 711 (2011) 167–173 171

genomic instability and tumor formation [21] and also causing the fication and other chromatin-associated proteins [17]. Histone
activation of proto-oncogenes in many, if not almost all, human modifications have been the subject of intense investigations for
tumors [17,23]. The majority of the large decreases in the methyl- many years and are currently defined as epigenetic modifiers
cytosine content of the genome are mainly due to hypomethylation [26]. The importance of histone modifications lies in the fact
of repetitive sequences, as hypomethylation of single-copy genes that they are essential during development and their deregulation
accounts for <5% of human DNA. Repetitive sequences (defined can lead to human malignancies. Thus, histones have emerged as
as CpG-rich non-coding sequences of the genome) are known to key carriers of epigenetic information, constituting a fundamen-
be involved in oxidative-stress induced aberrant hypomethylation tal and critical regulatory system that extends beyond the genetic
and include (i) satellites (SAT2 and SAT3) and (ii) interspersed system [28]. In general, histones have amino-terminal tails that
repeat sequences (LINEs, SINEs and LTRs) [22]. The CpG-rich SAT2 are targets of several posttranslational modifications, including:
and SAT3 satellites are normally densely methylated and are methylation, acetylation, phosphorylation, sumolyation and ubiq-
known to contribute to the establishment of heterochromatin. They uitination, among others [18,26]. Alterations in histone acetylation
are located as tandem repeats in the pericentromeric and juxta- patterns are present in many tumors [27]. In fact, a recent study
centromeric heterochromatin of several chromosomes but with has reported that cigarette smoke exposure to normal bronchial
chromosomes 1, 9 and 16 containing large arrays of these repeats epithelial or pulmonary carcinoma cells triggers aberrant histone
at the base of their long arms [22]. Diminished methylation at H2AX phosphorylation on serine 139 and/or phosphorylation on
these sites has been described in several human cancers and was serine 198 [38]. Other studies have also observed the phosphory-
also hypothesized to include mechanisms involving ROS-induced lation of histone H3T11 in prostate cancer as well as the global loss
oxidative stress [22,24]. Alternatively, hypomethylation of LINE of monoacetylation and trimethylation of histone H4, a common
sequences has been reported in many human germ cell cancers hallmark in many human tumor cells [18]. Typically, acetylated his-
and appears to generally parallel that of overall hypomethylation tones are associated with relaxed and transcriptionally competent
[22]. Furthermore, hypomethylation of repetitive sequences leads chromatin regions whereas hypoacetylated histones, on the other
to chromosomal rearrangements and, indirectly, to changes in gene hand, are associated with transcriptionally silent regions and are
expression patterns like in the case of hepatocellular carcinoma characterized by condensed chromatin structure [26]. The acetyla-
where such hypomethylation was highly correlated with disease tion of specific lysine residues by histone acetyltransferases (HATs),
recurrence [24]. While hypomethylation of repetitive sequences for example, is associated with transcriptionally active regions
seems to account for the greater part in cancer cells, decreased whereas their hypoacetylation is associated with a transcriptional
methylation of single-copy genes has attracted most of the atten- repressed chromatin. There are, however, instances whereby mod-
tion. It is believed that single-copy genes known to be silenced in ifications on the histone tails are shown to be both activators and
normal tissues become re-activated when aberrantly hypomethy- inhibitors of transcription. The methylation of lysine residues, for
lated. More specifically, reports have suggested the possibility of example, has been shown to lead to either transcriptional activation
major oncogenes like MYC and RAS, to be activated by hypomethy- or repression, depending on the site of methylation [18]. Overall,
lation in cancer cells [22]. HATs are enzymes that alter chromatin-based processes and thus
In addition, oxidative stress-induced damage has been shown attributing to mutations in oncogenes, tumor suppressor genes
not only to interfere with the ability of DNA to function as and/or DNA repair genes resulting in genomic instability, oncogenic
a substrate for DNA methyltransferases (DNMTs), but also to transformation and the development of cancer [28]. Modifications
reduce the methyl-accepting ability of DNA [13,17]. In mam- of histone proteins are strongly linked to DNMTs as they recruit
malian cells, DNMTs consist of DNMT1, DNMT2, DNMT3a, DNMT3b histone deacetylases (HDACs) leading to histone deacteylation and
and DNMT3L. DNMTs 2 and 3L lack enzymatic function due to subsequent transcriptional repression. These synergistic effects
the presence of an animo-terminal regulatory domain in DNMT2 between DNMTs and HDACs have been shown to induce differenti-
and a catalytic domain in DNMT3L. On the other hand, DNMT1 ation, apoptosis and cell growth arrest in several cancer cell lines, as
is regarded as a maintenance methyltransferase and recognizes well as induce the expression of specific cancer-related genes [18].
hemi-methylated DNA. In addition, DNMTs 3a and 3b are classified For example, re-expression of the hypermethylated genes MLH1,
as de novo methyltransferases since they bind to both hemi- TIMP3, CSKN2B and CDKN2A has been enhanced when exposed to
methylated and unmethylated CpG sites and add methyl groups to inhibitors of both DNMTs and HDACs. In particular, HDAC1 has been
previously unmethylated cytosines. Finally, there are reports that over-expressed in a number of tumors, and is likely a contributor to
DNMT3L also participates in de novo methylation [20]. Together, tumorigenic gene silencing [27]. Finally, recent studies have shown
these enzymes catalyze the transfer of the methyl donor group from that both HATs and HDACs are involved in the process of an effi-
S-adenosylmethionine (SAM) to the 5 -carbon of cytosines within cient DNA repair and that inhibition of enzymatic activity and/or
CpG islands in genomic DNA [13]. These enzymes are known to inhibition of the removal of histone acetylation respectively can
contribute to the increase of promoter methylation status (via de compromise the DNA repair machinery thus leading to mutation
novo methylation) as well as ensuring inheritance of gene silenc- fixation and genomic instability [66–70].
ing (via maintenance methylation), both of which could account for Because epigenetic alterations are potentially reversible there
the acquisition of a malignant transformation phenotype [13,31]. is great potential for the development of effective strate-
Increased expression of DNMT1 has been found in early and late gies in preventing against cancer formation [32]. Moreover,
stages of neoplasia suggesting its involvement in the generation because methylation status can be measured accurately, molec-
of abnormal methylation patterns in cancer cells [13]. In addi- ular biomarkers of diagnosis, prognosis, prediction of treatment
tion, it has been reported that patients with mutations in DNMT3b and those related to risk assessment could be developed alongside
have numerous chromosomal aberrations [25]. Data, from vari- “state-of-the-art” molecular techniques [11,33]. DNA methylation
ous research groups, have also revealed an over-expression of all markers hold great promise as disease detectors, not only for
DNMTs in several cancer types suggesting that methyltransferase the early detection of undiagnosed malignancies but also for the
cooperativity (especially between DNMT1 and DNMT3b) maintains monitoring of therapeutic efficacy [33]. DNA methylation markers
DNA methylation and gene silencing in human cancer cells [30]. have several advantages when compared to gene expression pro-
As it was mentioned earlier on, DNA methylation is hypothe- filing and proteomic approaches: (i) stability of the DNA molecule
sized to play an important role in the regulation of gene expression across extraction and handling procedures, (ii) measurement of
and chromatin architecture, in association with histone modi- DNA methylation can be compared with absolute reference points
172 D. Ziech et al. / Mutation Research 711 (2011) 167–173

(i.e. a baseline of unmethylated or completely methylated DNA), [4] B. Tudek, A. Winczura, J. Janik, A. Siomek, M. Foksinski, R. Olinski, Involvement
(iii) abnormal methylation patterns in cancer cells differ qualita- of oxidatively damaged DNA and repair in cancer development and aging, Am.
J. Transl. Res. 2 (2010) 254–284.
tively from normal cells, which allow for the development of assays [5] D. Trachootham, J. Alexander, P. Huang, Targeting cancer cells by ROS-mediated
with high sensitivity and specificity, (iv) DNA methylation patterns mechanism: a radical therapeutic approach? Nat. Rev. 8 (2009) 579–591.
are fairly stable over time, and lastly, (v) once a gene is hypermethy- [6] B.P. Patel, U.M. Rawal, R. Rawal, A. Shukla, P.S. Patel, Tobacco, antioxidant
enzymes, oxidative stress, and genetic susceptibility in oral cancer, Am. J. Clin.
lated in a developing tumor, the methylation signal tends to persist Oncol. 31 (2008) 454–459.
as the tumor grows [32]. [7] K.W. Lee, H.J. Lee, Biphasic effects of dietary antioxidants on oxidative stress-
In summary, epigenetic therapies hold great promise for the mediated carcinogenesis, Mech. Ageing Dev. 127 (2006) 424–431.
[8] N.I. Herath, B. Leggett, G. Macdonald, Review of genetic and epigenetic alter-
treatment of a wide variety of tumors. Strategies combining his- ations in hepatocarcinogenesis, J. Gastroenterol. Hepatol. 21 (2006) 15–21.
tone deactylase and methyltransferase inhibitors appear to be [9] E. Gollapalle, R. Wang, R. Adetolu, D. Tsao, D. Francisco, G. Sigounas, A. Georgak-
ideal in achieving maximal re-expression of silenced tumor sup- ilas, Detection of oxidative clustered DNA-lesions in X-irradiated mouse skin
tissues and human MCF-7 breast cancer cells, Radiat. Res. 167 (2007) 207–216.
pressor genes. Such inhibitors when combined with conventional
[10] A. Toyokuni, Molecular mechanisms of oxidative stress-induced carcinogene-
chemotherapies may represent a powerful strategy for the man- sis: from epidemiology to oxygenomics, IUMBM Life 60 (2008) 441–447.
agement of both early- and late-stage cancers [27]. [11] A. Risch, C. Plass, Lung cancer epigenetics and genetics, Int. J. Cancer. 123 (2008)
1–7.
[12] H.I. Chen, S.H. Liou, S.F. Ho, K.Y. Wu, C.W. Sun, M.F. Chen, L.C.
Cheng, T.S. Shih, C.H. Loh, Oxidative DNA damage estimated by plasma
4. Conclusions
8-hydroxyxeoxyguanosine (8-OHdG): influence of 4,4 -methylenebis (2-
chloroaniline) exposure and smoking, J. Occup. Health. 49 (2007) 389–398.
Cancer is a rather complex, multifactorial and multistage dis- [13] R. Franco, O. Schoneveld, A. Georgakilas, M. Panayiotidis, Oxidative stress, DNA
methylation and carcinogenesis, Cancer Lett. 266 (2008) 6–11.
ease with a number of molecular alterations involved in each stage
[14] G. Egger, G. Liang, A. Aparicio, P.A. Jones, Epigenetics in human disease and
(namely initiation, promotion and progression) of its development. prospects for epigenetic therapy, Nature 429 (2004) 457–463.
Oxidative stress (a state of persistent generation of ROS over- [15] S.O. Lim, J.M. Gu, M.S. Kim, H.S. Kim, Y.N. Park, C.K. Park, J.W. Cho, Y.M. Park,
whelming cellular defenses) has long been known to contribute to G. Jung, Epigenetic changes induced by reactive oxygen species in hepatocellu-
lar carcinoma: methylation of the E-cadherin promoter, Gastroenterology 135
human carcinogenesis. Such contribution has been shown to reg- (2008) 2128–2140.
ulate altered carcinogenic gene expression at the genetic as well [16] T. Ushijima, E.O. Takada, Aberrant methylations in cancer cells: where do they
the epigenetic level by very distinct mechanisms. ROS-induced come from? Cancer Sci. 96 (2005) 206–211.
[17] K.V. Donkena, C.Y. Young, D.J. Tindall, Oxidative stress and DNA methylation in
genetic alterations involve modifications in DNA bases, strand prostate cancer, Obstet. Gynecol. Int. 2010 (2010) 302051.
breaks, DNA–protein cross linkages etc. all of which increase muta- [18] A.T. Hauser, M. Jung, Targeting epigenetic mechanisms: potential of natural
tional rates in genes involved in major cellular pathways like products in cancer chemoprevention, Planta Med. 74 (13) (2008) 1593–1601.
[19] S.Y. Park, E.J. Yoo, N.Y. Cho, N. Kim, G.H. Kang, Comparison of CpG island hyper-
DNA repair etc. thereby making them ineffective. On the other methylation and repetitive DNA hypomethylation in premalignant stages of
hand, ROS-induced epigenetic mechanisms involve changes in DNA gastric cancer, stratified for Helicobacter pylori infection, J. Pathol. 201 (2009)
methylation profiles and/or histone modifications both of which 410–416.
[20] H. Kirk, W.T. Cefalu, D. Ribnicky, Z. Liu, K. Eilertsen, Botanicals as epigenetic
lead to silencing of tumor suppressor genes and/or oncogenic acti-
modulators for mechanisms contributing to development of metabolic syn-
vation and thus contributing to malignant transformation. Finally, drome, Metab. Clin. Exp. 57 (2008) S16–S23.
given that epigenetic alterations could potentially be reversible, [21] T. Ushijima, Detection and interpretation of altered methylation patterns in
cancer cells, Nat. Rev. 5 (2005) 223–231.
there is great potential for the development of effective strategies
[22] M. Hoffman, W. Schulz, Causes and consequences of DNA hypomethylation in
in preventing against human carcinogenesis. human cancer, Biochem. Cell Bio. 83 (2005) 296–321.
[23] K. Miyamoto, T. Ushijima, Diagnostic and therapeutic applications to epigenet-
ics, Jpn. J. Clin. Oncol. 35 (2005) 293–301.
Conflict of interest [24] J. Paluszczak, W.B. Dubowska, Epigenetic diagnostics of cancer – the application
of DNA methylation markers, J. Appl. Genet. 47 (2006) 365–375.
[25] M. Esteller, Aberrant DNA methylation as a cancer-inducing mechanism, Annu.
None. Rev. Pharmacol. Toxicol. 45 (2005) 629–656.
[26] R.M. Brena, T.H.M. Huang, Quantitative assessment of DNA methylation: poten-
tial applications for disease diagnosis, classification, and prognosis in clinical
Acknowledgments settings, J. Mol. Med. 84 (2006) 365–377.
[27] C. Balch, J. Montgomery, H.I. Paik, S. Kim, T.H.M. Huang, K.P. Nephew, New anti-
cancer strategies: epigenetic therapies and biomarkers, Front Biosci. 10 (2005)
This work was supported, in part, by the National Institutes 1897–1931.
of Health (Grant# P20RR17675), Centers of Biomedical Research [28] Z. Herceg, Epigenetics and cancer: towards an evaluation of the impact of envi-
ronmental and dietary factors, Mutagenesis 22 (2007) 91–103.
Excellence (COBRE) and Layman Award form the University of
[29] K. Kawai, Y.S. Li, M.F. Song, H. Kasai, DNA methylation by dimethyl sulfoxide
Nebraska-Lincoln (Dr. Franco); a Marie Curie International Rein- and methionine sulfoxide triggered by hydroxyl radical and implications for
tegration Grant within the 6th European Community Framework epigenetic modifications, Bioorg. Med. Chem. Lett. 20 (2010) 260–265.
Program (MIRG-CT-2006-036585) (Dr. Pappa); and, in part, by the [30] D. Ziech, R. Franco, A. Pappa, V.N. Mitsi, S. Georgakila, A. Georgakilas, M.
Panayiotidis, The role of epigenetics in environmental and occupational car-
School of Community Health Sciences, a Junior Faculty Research cinogenesis, Chem. Biol. Interact. 188 (2010) 340–349.
Award from the University of Nevada-Reno and a Marie Curie Inter- [31] M.J. Hitchler, F.E. Domann, An epigenetic perspective on the free radical theory
national Reintegration Grant within the 7th European Community of development, Free Radic. Biol. Med. 43 (2007) 1023–1036.
[32] F.E. Ahmed, Colorectal cancer epigenetics: the role of environmental factors
Framework Program (PIRG05-GA-2009-249315) (Dr. Panayiotidis). and the search for molecular biomarkers, J. Environ. Sci. Health C 25 (2007)
101–154.
[33] P.W. Laird, Cancer epigenetics, Hum. Mol. Genet. 14 (2005) R65–R76.
References [34] H. Bartsch, J. Nair, Chronic inflammation and oxidative stress in the genesis
and perpetuation of cancer: role of lipid peroxidation, DNA damage and repair,
[1] G.J. Kim, K. Chandrasekaran, W.F. Morgan, Mitochondrial dysfunction, per- Langenbecks Arch Surg. 391 (2006) 499–510.
sistently elevated levels of reactive oxygen species and radiation-induced [35] S. Mena, A. Ortega, J.M. Estrela, Oxidative stress in environmental-induced
genomic instability: a review, Mutagenesis 21 (2008) 361–368. carcinogenesis, Mutat. Res. 674 (2009) 36–44.
[2] M. Valko, C.J. Rhodes, J. Moncol, M. Izakovic, M. Mazur, Free radicals, metals [36] S.J. Schonfeld, P. Bhatti, E.E. Brown, M.S. Linet, S.L. Simon, R.M. Weinstock,
and antioxidants in oxidative stress-induced cancer, Chem. Biol. Interact. 160 A.A. Hutchinson, M. Stovall, D.L. Preston, B.H. Alexander, M.M. Doddy, A.J. Sig-
(2006) 1–40. urdson, Polymorphisms in oxidative stress and inflammation pathway genes,
[3] A.C.E. Campos, F. Molognoni, F.H.M. Melo, L.C. Galdier, C.R.W. Carneiro, V. low-dose ionizing radiation, and the risk of breast cancer among US radiologic
D’Almeida, M. Correa, M. Jasiulionis, Oxidative stress modulate DNA methy- technologists, Cancer Causes Control 21 (2010) 1857–1866.
lation during melanocyte anchorage blockade associated with malignant [37] D.C. Francisco, P. Peddi, J.M. Hair, B.A. Flood, A.M. Cecil, P.T. Kalogerinis, G.
transformation, Neoplasia 9 (2007) 1111–1121. Sigounas, A.G. Georgakilas, Induction and processing of complex DNA dam-
D. Ziech et al. / Mutation Research 711 (2011) 167–173 173

age in human breast cancer cells MCF-7 and nonmalignant MCF-10A cells, Free [55] A.G. Georgakilas, K. Aziz, D. Ziech, S. Georgakila, M.I. Panayiotidis, BRCA1
Radic. Biol. Med. 44 (2009) 558–569. involvement in toxicological responses and human cancer etiology, Toxicol.
[38] H. Zhao, A.P. Albino, E. Jorgensen, F. Traganos, Z. Darzynkiewicz, DNA damage Lett. 188 (2) (2009) 77–83.
response induced by tobacco smoke in normal human bronchial epithelial and [56] S.A. Weitzman, P.W. Turk, D.H. Milkowski, K. Kozlowski, Free radical adducts
A549 pulmonary adenocarcinoma cells assessed by laser scanning cytometry, induce alterations in DNA cytosine methylation, Proc. Natl. Acad. Sci. U.S.A. 91
Cytometry Part A 75A (2009) 840–847. (1994) 1261–1264.
[39] A. Katafuchi, T. Nohmi, DNA polymerases involved in the incorporation of [57] P.W. Turk, A. Laayoun, S.S. Smith, S.A. Weitzman, DNA adduct 8-hydroxyl-
oxidized nucleotides into DNA: their efficiency and template base prefer- 2-deoxyguanosine (8-hydroxyguanine) affects function of human DNA
ence, Mutat. Res. (2010), doi10.1016/j.mrgentox.2010.06.004 [Epub ahead of methyltransferase, Carcinogenesis 16 (1995) 1253–1255.
print]. [58] P.W. Turk, S.A. Weitzman, Free radical DNA adduct 8-OH-deoxyguanosine
[40] M.A. Lebedeva, J.S. Eaton, G.S. Shadel, Loss of p53 causes mitochondrial DNA affects activity of HPA II and MSP I restriction endonucleases, Free Radic. Res.
depletion and altered mitochondrial reactive oxygen species homeostasis, 23 (1995) 255–258.
Biochim. Biophys. Acta. 1787 (2009) 328–334. [59] Y. Kuchino, F. Mori, H. Kasai, H. Inoue, S. Iwai, K. Miura, E. Ohtsuka, S. Nishimura,
[41] S.J. Ralph, S.R. Enríquez, J. Neuzil, E. Saavedra, R.M. Sánchez, The causes of cancer Misreading of DNA templates containing 8-hydroxydeoxyguanosine at the
revisited: “mitochondrial malignancy” and ROS-induced oncogenic transfor- modified base and at adjacent residues, Nature 327 (1987) 77–79.
mation – Why mitochondria are targets for cancer therapy, Mol. Aspects Med. [60] P.A. Hepburn, G.P. Margison, M.J. Tisdale, Enzymatic methylation of cytosine in
31 (2010) 145–170. DNA is prevented by adjacent O6 -methylguanine residues, J. Biol. Chem. 266
[42] J.P. Fruehaug, F.L. Meyskens, Reactive oxygen species: a breath of life of death? (1991) 7985–7987.
Clin. Cancer Res. 13 (2007) 789–794. [61] N.W. Tan, B.F.L. Li, Interaction of oligonucleotides containing 6-O-
[43] P. Peddi, D.C. Francisco, A.M. Cecil, J.M. Hair, M.I. Panayiotidis, A.G. Georgakilas, methylguanine with human DNA (cytosine-5-)-methyltransferase,
Processing of clustered DNA damage in human breast cancer cells MCF-7 with Biochemistry 29 (1990) 9234–9240.
partial DNA-PKcs deficiency, Cancer Lett. 269 (2008) 174–183. [62] J.K. Christman, G. Sheikhnejad, C.J. Marasco, R.J. Sufrin, 5-Methyl-2-
[44] S.M. Holt, A.G. Georgakilas, Detection of complex DNA damage in ␥-irradiated deoxycytidine in single stranded DNA can act in cis to signal de novo DNA
acute lymphoblastic leukemia pre-B NALM-6 cells, Radiat. Res. 168 (2007) methylation, Proc. Natl. Acad. Sci. U.S.A. 92 (1995) 7347–7351.
527–534. [63] M.F. Kane, M. Loda, G.M. Gaida, J. Lipman, R. Mishra, H. Goldman, J.M. Jessup, R.
[45] M. Panayiotidis, Reactive Oxygen Species (ROS) in multistage carcinogenesis, Kolodner, Methylation of the hMLH1 promoter correlates with lack of expres-
Cancer Lett. 266 (2008) 3–5. sion of hMLH1 in sporadic colon tumors and mismatch-repair-defective human
[46] A. Pappa, R. Franco, O. Schoneveld, A. Galanis, R. Sandaltzopoulos, M.I. Panayi- tumor cell lines, Cancer Res. 60 (2000) 4366–4371.
otidis, Sulfur-containing compounds in protecting against oxidant-mediated [64] J.G. Herman, A. Umar, K. Polyak, J.R. Graff, N. Ahuja, J-P.J. Issa, S. Markowitz, J.K.V.
lung diseases, Curr. Med. Chem. 14 (2007) 2590–2596. Wilson, S.R. Hamilton, K.W. Kinzler, M.F. Kane, R.D. Kolodner, B. Vogelstein, T.A.
[47] R. Franco, M.I. Panayiotidis, Environmental toxicity, oxidative stress, human Kunkel, S.B. Baylin, Incidence and functional consequences of hMLH1 promoter
disease and the “black box” of their synergism: how much have we revealed? hypermethylation in colorectal carcinoma, Proc. Natl. Acad. Sci. U.S.A. 95 (1998)
Mutat. Res. 674 (1–2) (2009) 1–2. 6870–6875.
[48] D. Ziech, R. Franco, A.G. Georgakilas, S. Georgakila, V. Malamou-Mitsi, O. Schon- [65] M. Esteller, R.A. Risques, M. Toyota, G. Capella, V. Moreno, M.A. Peinado, S.B.
eveld, A. Pappa, M.I. Panayiotidis, The role of reactive oxygen species and Baylin, J.G. Herman, Promoter hypermethylation of the DNA repair gene O(6)-
oxidative stress in environmental carcinogenesis and biomarker development, methylguanine-DNA methyltransferase is associated with the presence of G:C
Chem. Biol. Interact. 188 (2) (2010) 334–339. to A:T transition mutations in p53 in human colorectal tumorigenesis, Cancer
[49] R. Franco, R. Sanchez-Olea, E.M. Reyes-Reyes, M.I. Panayiotidis, Environmental Res. 61 (2001) 4689–4692.
toxicity, oxidative stress and apoptosis: ménage a trios, Mutat. Res. 674 (1–2) [66] C.L. Peterson, J. Cote, Cellular machineries for chromosomal DNA repair, Genes.
(2009) 3–22. Dev. 18 (2004) 602–616.
[50] R. Franco, M.I. Panayiotidis, Cell death or survival: the double-edged sword of [67] A. Jazayeri, A.D. McAinsh, S.P. Jackson, Saccharomyces cerevisiae Sin3p facili-
environmental and occupational toxicity, Chem. Biol. Interact. 188 (2) (2010) tates DNA double-strand break repair, Proc. Natl. Acad. Sci. U.S.A. 101 (2004)
265–266. 1644–1649.
[51] M. Hada, A.G. Georgakilas, Formation of clustered DNA damage after high-LET [68] R. Murr, J.I. Loizou, Y.G. Yang, C. Cuenin, H. Li, Z.Q. Wang, Z. Herceg, Histone
irradiation: a review, J. Radiat. Res. (Tokyo) 49 (3) (2008) 203–210. acetylation by Trrap-Tip60 modulates loading of repair proteins and repair of
[52] A.G. Georgakilas, Processing of DNA damage clusters in human cells: current DNA double-strand breaks, Nat. Cell. Biol. 8 (2006) 91–99.
status of knowledge, Mol. Biosyst. 4 (1) (2008) 30–35. [69] J.A. Downs, S. Allard, O. Jobin-Robitaille, A. Javaheri, A. Auger, N. Bouchard,
[53] S. Nowsheen, R.L. Wukovich, K. Aziz, P.T. Kalogerinis, C.C. Richardson, M.I. S.J. Kron, S.P. Jackson, J. Cote, Binding of chromatin-modifying activities
Panayiotidis, W.M. Bonner, O.A. Sedelnikova, A.G. Georgakilas, Accumulation to phosphorylated histone H2A at DNA damage sites, Mol. Cell. 16 (2004)
of oxidatively induced clustered DNA lesions in human tumor tissues, Mutat. 979–990.
Res. 674 (1–2) (2009) 131–136. [70] B.A. Tamburini, J.K. Tyler, Localized histone acetylation and deacetylation trig-
[54] D. Ziech, R. Franco, A. Pappa, V. Malamou-Mitsi, S. Georgakila, A.G. Georgakilas, gered by the homologous recombination pathway of double-strand DNA repair,
M.I. Panayiotidis, The role of epigenetics in environmental and occupational Mol. Cell. Biol. 25 (2005) 4903–4913.
carcinogenesis, Chem. Biol. Interact. 188 (2) (2010) 340–349.

You might also like