You are on page 1of 11

Mutation Research 736 (2012) 8292

Contents lists available at SciVerse ScienceDirect

Mutation Research/Fundamental and Molecular


Mechanisms of Mutagenesis
journal homepage: www.elsevier.com/locate/molmut
Community address: www.elsevier.com/locate/mutres

Review

Oxidatively damaged DNA and its repair in colon carcinogenesis

Barbara Tudek a,b , Elzbieta


Speina a,
a
b

Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawi


nskiego 5a, 02-106 Warsaw, Poland
Institute of Genetics and Biotechnology, Faculty of Biology, Warsaw University, Pawi
nskiego 5a, 02-106 Warsaw, Poland

a r t i c l e

i n f o

Article history:
Received 9 January 2012
Received in revised form 2 April 2012
Accepted 16 April 2012
Available online 25 April 2012
Keywords:
Colorectal cancer
Oxidative stress
Lipid peroxidation
8-Oxo-7,8-dihydroguanine
Etheno DNA adducts
DNA repair

a b s t r a c t
Inammation, high fat, high red meat and low ber consumption have for long been known as the most
important etiological factors of sporadic colorectal cancers (CRC). Colon cancer originates from neoplastic transformation in a single layer of epithelial cells occupying colonic crypts, in which migration and
apoptosis program becomes disrupted. This results in the formation of polyps and metastatic cancers.
Mutational program in sporadic cancers involves APC gene, in which mutations occur most abundantly
in the early phase of the process. This is followed by mutations in RAS, TP53, and other genes. Progression of carcinogenic process in the colon is accompanied by augmentation of the oxidative stress,
which manifests in the increased level of oxidatively damaged DNA both in the colon epithelium, and
in blood leukocytes and urine, already at the earliest stages of disease development. Defence mechanisms are deregulated in CRC patients: (i) antioxidative vitamins level in blood plasma declines with
the development of disease; (ii) mRNA level of base excision repair enzymes in blood leukocytes of CRC
patients is signicantly increased; however, excision rate is regulated separately, being increased for 8oxoGua, while decreased for lipid peroxidation derived ethenoadducts, Ade and Cyt; (iii) excision rate
of Ade and Cyt in colon tumors is signicantly increased in comparison to asymptomatic colon margin,
and ethenoadducts level is decreased. This review highlights mechanisms underlying such deregulation,
which is the driving force to colon carcinogenesis.
2012 Elsevier B.V. All rights reserved.

Contents
1.
2.
3.

4.

5.
6.

Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Histopathology of CRC and mutations in critical genes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Oxidative stress in the development of colon cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
3.1.
Inammation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
3.2.
Consumption of red meat and alcohol and tobacco smoking. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
3.3.
DNA damage . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Repair of oxidative DNA damage as a driving force to colon carcinogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
4.1.
Repair of 8-oxo-7,8-dihydro-2 -deoxyguanosine in colon adenoma and carcinoma patients . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
4.2.
Repair of etheno DNA adducts in colon carcinogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Aberrant repair of oxidatively damaged DNA bases and mutations in critical genes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Conict of interest statement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Funding . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

83
83
84
84
84
85
85
86
87
88
89
90
90
90

Abbreviations: 8-oxodG, 8-oxo-7,8-dihydro-2 -deoxyguanosine; 8-oxoGua, 8-oxo-7,8-dihydroguanine; AD, adenoma; CRC, colorectal cancer; APC, adenomatous polyposis coli; FapyGua, 2,6-diamino-4-hydroxy-5-formamidopyrimidine; APE1, apurinic site endonuclease 1; ROS, reactive oxygen species; RNS, reactive nitrogen species;
BER, base excision repair; Ade, 1,N6 -ethenoadenine; dA, 1,N6 -etheno-2 -deoxyadenosine; Cyt, 3,N4 -ethenocytosine; dC, 3,N4 -etheno-2 -deoxycytidine; 1N2 -dG, 1,N2 etheno-2 -deoxyguanosine; LPO, lipid peroxidation; MMR, mismatch repair.
Corresponding author. Tel.: +48 22 592 3334; fax: +48 22 592 2190.
E-mail address: elasp@ibb.waw.pl (E. Speina).
0027-5107/$ see front matter 2012 Elsevier B.V. All rights reserved.
http://dx.doi.org/10.1016/j.mrfmmm.2012.04.003

B. Tudek, E. Speina / Mutation Research 736 (2012) 8292

1. Introduction
Colorectal cancer (CRC) is one of the most frequent causes of
death in Western countries. The cases are roughly equally distributed between the sexes. Two main factors are considered in
pathogenesis of this cancer: (i) inammation and high fat diet [1],
which increases the number of lesions in DNA and induces mutations in critical genes; and (ii) genetic predispositions.
Inammation is associated with the release of large amounts
of reactive oxygen and nitrogen species [2] leading to oxidation
of nucleic acids, proteins and lipids, and induction of several promutagenic DNA lesions.
The genetic predisposition to CRC may be connected to the deciency in mismatch repair system (MMR), which causes mutator
phenotype of intestine cells (manifesting itself as microsatellites
instability MSI) and development of hereditary non-polyposis
colon carcinoma HNPCC (or Lynch syndrome). They may also
be associated with the deciency of the APC (adenomatous polyposis coli) gene product, which functions are related to Wnt
signaling pathways and migration of intestine epithelium. Lack
of APC protein leads to familial adenomatous polyposis (FAP), a
well described syndrome characterized by multiple adenomatous
lesions which usually appear in the second and third decades of
life. The probability of malignant transformation of one of the
polyps is close to 100% [3]. A subset of patients present an attenuated form (AFAP) characterized by fewer lesions (<100), a later
age of onset, and a predilection for the proximal colon. About
1015% of AFAP cases reveal mutations in APC gene. More frequently, in 2030% of cases [4,5], AFAP has been correlated with
a biallelic mutation of the MutY human homologue gene (MYH).
MutY is a DNA glycosylase, which is involved in elimination from
DNA of adenine incorrectly paired with 8-oxo-7,8-dihydroguanine
(8-oxoGua). Oxidative DNA damage has been linked to various
pathological conditions, such as aging, carcinogenesis, neurodegenerative and cardiovascular disease (reviewed by [69]). This
review focuses on the role of oxidatively induced DNA lesions in
colon carcinogenesis. We discuss the mechanisms of tumor initiation (generation of DNA damage and mutations) and multiple
molecular steps in the colon cancer progression with special focus
on the deregulation of defense mechanisms, such as anti-oxidant
response and DNA repair.

2. Histopathology of CRC and mutations in critical genes


A single layer of epithelial cells lines the crypts of the colon
and rectum. Throughout the digestive tract, these crypts substantially increase the surface occupied by the epithelium. Four to six
stem cells at the base of each crypt give rise to three epithelial cell
types, absorptive cells, mucus-secreting goblet cells and epithelial cells. The cells multiply in the lower third of the crypts and
differentiate in the upper two-thirds. Normally, the birth rate of
the colonic epithelial cells precisely equals the rate of cell loss
from the crypts apex to the lumen of the colon. The birth/loss ratio
increases under neoplastic transformation of cells originating from
a single progenitor cell. Polyp, a mass of cells protruding from the
bowel wall is often the rst observed syndrome of the development
of colon cancer. However, according to ofcial medical classications colonic polyps are not recognized as cancer. There are two
main types of polyps, which can be distinguished histologically
but not by their external appearance [10]. The non-dysplastic or
hyperplastic type contains large number of cells that have a normal morphology. Cell epithelium is lined up in a single row along
the basement membrane, and these polyps apparently have little
tendency to become neoplastic. Adenomatous polyp type is dysplastic, has abnormal intercellular organization. Several layers of

83

epithelial cells are observed; the nuclei of the epithelial cells are
larger than normal, and their position within the cells is often aberrant. Crypts crowd together in kaleidoscopic pattern. As adenomas
grow in size, they become more dysplastic. They are also more likely
to contain villous components, i.e., ngerlike projections of dysplastic crypts that can be distinguished from the smooth contour of
the less advanced tubular adenomas. As adenomas progress, they
are likely to become malignant, dened as the ability for invading surrounding tissue and travel to distant organs through direct
spread or transport by blood and lymphatic vessels. Malignant
tumors are not necessarily larger or more dysplastic than benign
tumors; their sole determining feature is invasiveness.
Polyps are the earliest clinical manifestation of colorectal neoplasia. However, the earliest pre-carcinogenic changes in the colon
can be observed under microscope after methylene blue staining
or examination of the colonic mucosa cross sections (Fig. 1). These
changes are termed aberrant crypt foci (ACF). Like their larger counterparts, can be either dysplastic or non-dysplastic. In mice or rats
ACF are found as soon as 23 weeks following administration of
colon carcinogens [11], they grow with time giving rise to malignant tumors 6 months after animal intoxication with carcinogen
[12].
Consecutive stages of colon carcinogenesis are accompanied by
acquiring mutations in critical genes, which regulate cell proliferation, and/or apoptosis. A relatively limited number of oncogenes
and tumor suppressor genes, most prominently the APC, KRAS, and
TP53 genes are mutated in CRCs, and a larger collection of genes that
are mutated in subsets of CRC have begun to be dened. On the basis
of the frequency of mutations occurring in different genes at particular stages of adenoma to carcinoma progression, the sequence
of genetic changes was proposed [13]. We will briey summarize
the genetic events in order to shed light how these changes may
affect the extent of oxidative DNA damage, and modulate its repair
rate. The earliest genetic change in colon carcinogenesis is mutation in the APC gene. About 7080% of sporadic adenomas and
carcinomas have somatic mutation that inactivate the APC gene,
and its frequency is similar in the earliest lesions, like microscopic
adenomas composed of few dysplastic glands, and in advanced
adenomas and carcinomas, independently of the efciency of the
MMR system. APC protein functions in the Wnt signaling pathway, in which stimulates proteasomal degradation of -catenin,
an activator of TCF/LET (T cell factor/lymphoid enhancer family)
transcription factors family. In consequence APC inhibits transcription of several genes, such as proto-oncogenes, e.g., c-MYC or cyclin
D1, genes encoding membrane factors matrix metalloproteinase
7 (MMP-7)/Matrilysin, membrane type 1 MMP, laminin-5 2 chain,
CD44, growth factors, e.g., FGF20, FGF9, and Wnt pathway feedback
inhibitors, such as AXIN2, Naked-1/-2, Dickkopf-1, WNT inhibitory
factor 1. Transcriptional program induced by -catenin resembles
the transcriptional program in stem cells at the base of the colon
crypts [14]. Thus, activation of -catenin caused by APC mutation
will stimulate proliferation of epithelial cells in colonic crypts. Next,
mutations in genes functioning in EGFR signaling pathway, such
as K-RAS, BREF or PTEN will further enhance cell proliferation and
survival, and drive progression of early adenomas to intermediate
lesions. K-RAS is mutated in almost 40% of intermediate and late
adenomas and carcinomas. Inactivation of TGF- response, e.g., DCC
(deleted in colorectal cancer) or TGF- receptor mutations cause
loss of growth inhibitory effect of TGF- and suppress apoptosis.
These genetic changes are frequently observed in late adenoma
lesions. Loss of TP53 function is the late genetic change, which
further stimulates proliferation and survival promoting the shift
of adenoma to carcinoma. TP53 mutations are observed in about
50% of colorectal cancers, but much less frequently in adenomas. At
least two of these genetic changes, loss of APC and TP53 functions
may affect DNA repair in colonic crypts, and in consequence the

84

B. Tudek, E. Speina / Mutation Research 736 (2012) 8292

Fig. 1. Aberrant crypt foci. (A) A view from the lumen site of intact mouse colon. The colon was cut open along the longitudinal median axis, formalin xed and stained with
methylene blue. The mucosal side was observed under the light microscope (magnication 100); (B and C) cross sections of mouse epithelium stained with hematoxylin
and eosin (magnication 1000).

extent of DNA damage, and potential of gaining new mutations (see


Section 5).
3. Oxidative stress in the development of colon cancer
The majority of pathological factors favoring development of
colon cancer involve induction of oxidative stress in the colon,
which is dened as overproduction of oxygen species combined
with insufciency in protective mechanisms of anti-oxidative
defence [15,16]. Oxidative stress is involved in the production of
DNA damage and mutations associated with the initiation or progression of human cancers [17,18]. The main pathological factors
for the development of CRC are inammation, fat metabolism,
consumption of meat and alcohol and tobacco smoking. All these
stimulate the release of ROS and RNS as well as lipid peroxidation
(LPO) [1,19]. These compounds play a dual role in cells. At low or
moderate concentrations they are mediators of specic physiological processes, whereas high concentrations can be detrimental to
all components of the cell.
3.1. Inammation
Epidemiological studies suggest that inammatory processes
are linked with CRC. This is true particularly in the association
of inammatory bowel disease and colorectal cancer [20]. During
inammation activated macrophages release large amounts of ROS
and RNS [2], which oxidize nucleic acids, proteins and lipids. Oxidation of polyunsaturated fatty acids generates reactive aldehydes
unsaturated in , -positions which, in contrast to ROS and RNS,
are relatively stable and can diffuse throughout the cell [19]. Short
chain reactive aldehydes are classied into the following families:
2-alkenals (e.g., acrolein, crotonaldehyde, 2-hexenal), 4-hydroxy2-alkenals (e.g., 4-hydroxy-2-hexenal HHE, 4-hydroxy-2-nonenal
HNE) and ketoaldehydes (e.g., malondialdehyde MDA, glyoxal, 4-oxo-2-nonenal ONE) [21]. Multiplicity of LPO products
is tremendously augmented by the existence of stereoisomers of
several of these compounds.
Reactive aldehydes either react directly with DNA and proteins [22], or undergo further oxidation to more reactive epoxy

derivatives, such as 2,3-epoxy-4-hydroxynonanal [23]. The afnity of epoxidized LPO products to nucleic acids is much higher
than to proteins. Epoxidation of enals may occur in the presence
of hydrogen peroxide, but is also catalysed by enzymes producing inammatory mediators, prostaglandins and leukotrienes, such
as cyclooxygenase-2 (COX-2) and lipooxygenases (LOX) [24]. It
was observed that COX-2 is overexpressed in 80% of colorectal
cancer tissues [25,26]. Increased levels of COX-2 were observed
in epithelial cells, inammatory cells, as well as in stromal cells,
but not in adjacent normal tissues [27]. COX-2 overexpression in
Min (multiple intestinal neoplasia) mice was sufcient to induce
mammary tumors [28]. The mechanism of tumor induction by
COX-2 overexpression is complex, and involves the activity of its
enzymatic end-products, prostanoids. A number of theories have
been proposed to explain the role of tumor-derived prostanoids in
promotion of angiogenesis, induction of tumor cell proliferation,
suppression of immune response, and protection against apoptosis [2931]. Additionally, it was shown that arachidonic acid (AA)
metabolism mediated by COX-2 and 5-LOX results in the formation of heptanone-etheno (H)-DNA adducts. The H-DNA adducts
are highly mutagenic in mammalian cells suggesting that these
pathways could be (at least in part) responsible for the somatic
mutations observed in tumorigenesis [32]. That is why inhibition
of COX-2 by non-steroid anti-inammatory drugs or RNA interference appeared a successful approach in colon cancer prevention
[33,34].
3.2. Consumption of red meat and alcohol and tobacco smoking
Epidemiological and animal model studies have suggested that
high intake of heme, present in red meat, is associated with an
increased risk of colon cancer. Recently elucidated mechanisms
suggest that heme induces DNA damage and cell proliferation
of colonic epithelial cells via hydrogen peroxide produced by
heme oxygenase. In Caco-2 cells the presence of heme oxygenase
inhibitor, zinc protoporphyrin, reduced DNA damage (measured by
a comet assay) triggered by hemin as well as cell hyperproliferation [35]. The effect of heme may be modied by LPO products.
The preincubation of colon adenocarcinoma, SW480, cells with

B. Tudek, E. Speina / Mutation Research 736 (2012) 8292

hemoglobin (Hb) and its subsequent exposure to linoleic acid


hydroperoxides (LAOOH) led to an increase in cell death, malondialdehyde formation, and DNA fragmentation and these effects
were related to the peroxide group and the heme present in
Hb. SW480 cells treated with Hb and LAOOH presented a higher
level of the modied DNA bases 8-oxoGua and 1,N2 -etheno-2 deoxyguanosine (1,N2 -Gua) compared to the control. Incubations
with Hb led to an increase in intracellular iron levels, which correlated with DNA oxidation. Thus, Hb from either red meat or
bowel bleeding caused by inammatory processes could act as an
enhancer of fatty acid hydroperoxide genotoxicity, and in consequence contribute to the accumulation of DNA lesions in colon cells
[36].
Another frequently suggested risk factor for CRC development
is chronic alcohol consumption. One of the factors which may
contribute to the development of alcohol-associated cancer is the
action of acetaldehyde, the rst and most toxic metabolite of
alcohol metabolism [37]. Acetaldehyde is produced by the oxidation of ethanol by hepatic NAD-dependent alcohol dehydrogenase
and during threonine catabolism. Acetaldehyde reacts with 2 deoxyguanosine in DNA to form 1,N2 -propanodG and 1,N2 -dG
[38]. Both these DNA lesions were described to derive also from
lipid peroxidation [39,36]. In hepatic cancer model, lean and obese
Zucker rats higher levels of etheno DNA adducts were observed in
the liver DNA of alcohol-fed than in control animals. This increase
was correlated with the induction of cytochrome P-450 isoform 2E1
[40]. Thus, the formation of exocyclic DNA adducts, like propanodG, 1,N2 -dG, dA and dC may be the important mechanism
associated with acetaldehyde related cancer risk.
The results of epidemiological studies also link tobacco smoking
with the risk for CRC development. However, the obtained data
are controversial, and the molecular mechanism remains unclear.
Although in the leukocytes of lung cancer patients tobacco smoking
increased the level of one of the major oxidatively damaged DNA
base, 8-oxoGua [41], in CRC patients no effect of tobacco smoking on
8-oxoGua level in the leukocytes and 8-oxoGua urinary excretion
was observed [42].
3.3. DNA damage
Oxidatively damaged DNA has been blamed for the physiological changes associated with degenerative diseases and cancer
[43,44]. DNA damage caused by free radicals is the most heterogeneous group of DNA lesions due to the wide variety of deleterious
molecules produced in cells in the conditions of oxidative stress.
A plethora of damaged DNA bases are formed upon ROS attack on
genetic material, several of them reveal strong promutagenic properties [45]. One of the major and best studied is 8-oxoGua, a typical
biomarker of oxidative stress, which may play a role in carcinogenesis [46]. The presence of 8-oxo-7,8-dihydro-2 -deoxyguanosine
(8-oxodG) residues in DNA leads to GCTA transversions [47].
8-OxodG is formed in DNA either via direct oxidation of nucleic
acids or can be incorporated from the nucleotide pool by DNA
polymerases, the latter process being an important source of DNA
oxidation and genome instability [48,49].
LPO products may contribute to DNA damage to nearly the same
extent as directly oxidized bases. There are distinct groups of linear and cyclic LPO specic DNA lesions. Malondialdehyde reacts in
DNA with guanine, adenine and cytosine to form cyclic pyrimido[1,2]purine-10(3H)-one-2 -deoxyribose (M1 dG) adduct and linear N6 -(3-oxopropenyl)-2 -deoxyadenosine (M1 dA) and N4-(3oxopropenyl)-2 -deoxycytosine (M1 dC) adducts, respectively [50].
M1 dG is the major MDA-DNA adduct and was detected in human
and rodent tissues. In Escherichia coli cells, it induces transversions
to T and transitions to A with a frequency comparable with that of
8-oxoGua [15].

85

Exocyclic propano DNA adducts are formed by the attachment


of unsaturated reactive aldehyde to ring and extra-ring nitrogen
atoms in DNA bases, which generates additional saturated, sixmembered ring in a DNA base. These adducts are derived from the
reaction of DNA with, e.g., acrolein, crotonaldehyde and HNE. AcrdG, Cro-dG and HNE-dG were found in rodent and human tissues
[51,52]. Etheno DNA adducts posses ve-membered exocyclic rings
attached to DNA bases. The exact pathway of their formation in vivo
is not known, although it is clear that they are generated by the
exposition of DNA to lipid peroxides. Ethenoadducts are also introduced into DNA by certain environmental carcinogens like vinyl
chloride or its metabolite, chloroacetaldehyde (CAA) [53]. Propanoand etheno-type adducts that are found in native mammalian DNA
are unsubstituted and substituted with side chains of different
length, depending on the structure of reacting LPO product. All
exocyclic DNA adducts have strong miscoding potential. Unsubstituted exocyclic DNA adducts are more mutagenic than toxic.
With the increase of the side chain the toxic properties increase
and are related to the inhibition of replication and transcription
[54,55].
Elevated levels of dA and dC were found in pro-carcinogenic
colon diseases, like inammatory bowel disease, Crohns disease and ulcerative colitis, and in addition in chronic pancreatitis
[20,56]. This suggests that LPO-derived DNA lesions contribute to
CRC etiology.

4. Repair of oxidative DNA damage as a driving force to


colon carcinogenesis
The major pathway for repair of oxidatively derived DNA lesions
is base excision repair system (BER). Although several oxidatively
damaged bases are also repaired by mismatch repair system and
AlkB family dioxygenases, this review will focus on BER activity.
In a classical view BER consists of ve steps: (i) lesion recognition, (ii) excision of the damaged base, (iii) AP site incision and
removal of the chemical residues that could block further steps
of the pathway, (iv) incorporation of the proper nucleotide(s) by
DNA polymerases and (v) ligation of the DNA strand to restore the
original DNA molecule [5760].
Damaged bases are recognized by DNA glycosylases, which
excise from DNA a wide range of oxidized and alkylated bases.
Presently, 13 human DNA glycosylases with different, but partially
overlapping substrate specicity have been identied. Mechanistically two types of DNA glycosylases can be distinguished,
monofunctional enzymes, that excise damaged base leaving behind
an abasic (AP) site, which is subsequently cleaved by AP endonuclease (APE1) 5 to the AP site, and bifunctional DNA glycosylases,
which have endowed AP lyase activity that incises phosphodiester
bonds 3 to the AP site created by the glycosylase activity of the
enzyme. This review will focus on repair of three oxidative DNA
lesions, 8-oxoGua, Ade and Cyt.
8-OxoGua is repaired by a three-step system, which (i) limits incorporation of 8-oxodGTP into DNA during replication, (ii)
excises 8-oxoGua from DNA, and (iii) corrects the base opposite to
8-oxoGua in DNA. Different DNA polymerases can incorporate 8oxodGTP to the DNA, and replicative DNA polymerases incorporate
this modied nucleotide usually opposite dA, generating A:T C:G
mutations. Approximately a half of 8-oxodG in DNA arises from the
direct DNA oxidation, and the other half is a result of incorporation
of 8-oxodGTP during DNA synthesis. Incorporation of 8-oxodGTP
into DNA by DNA polymerases is limited by the activity of MTH1
phosphohydrolase, which hydrolyzes 8-oxodGTP to 8-oxodGMP
[61]. 8-OxodGMP is subsequently dephoshorylated by nucleotidase and removed from the cell [62]. Many observations indicate a
direct correlation between 8-oxodG formation and carcinogenesis

86

B. Tudek, E. Speina / Mutation Research 736 (2012) 8292

in vivo [46,6366]. The second line of defence is glycosylase MutY


(MYH) that removes adenine paired with 8-oxoGua. Activity of this
enzyme prevents mutations G:C T:A, when dATP is incorporated
opposite 8-oxoGua during replication [67]. After removal of Ade
by MYH glycosylase, repair DNA polymerase incorporates dCMP
opposite 8-oxodG. Then 8-oxoGua can be excised by the third component of the system, OGG1 glycosylase [68]. OGG1 glycosylase/AP
lyase is the major enzyme catalyzing excision of 8-oxoGua from
DNA, but only when paired with Cyt or Thy [69,70]. The AP lyase
activity of the OGG1 protein is about 10-fold lower than that of
the glycosylase [71]. When 8-oxodGTP is included into DNA during
replication it is probably removed by NEIL1 or NEIL2 glycosylases,
which excises 8-oxoGua paired with dC/G/T/A or dC/A, respectively [72]. This complex three-tier system prevents mutations
induced by the presence of 8-oxodG in DNA, both when this lesion
is induced directly in the DNA, and when it is incorporated from
the pool of nucleotides during replication. Polymorphisms of MYH
and OGG1 glycosylases are related to modulation of susceptibility
to colon cancer. MYH variations signicantly increase CRC risk, and
of several mutations described, two are the most prevalent: Y165C
(exon 7) and G382D (exon 13). Mutated enzyme excises Ade from
dA:8-oxodG pair with a decreased rate, increasing mutation frequency. The association of MYH with CRC was rst reported in 2002
[73].
Several polymorphic changes in the OGG1 gene have been
described, with the most common being Ser326Cys [74]. Polymorphic Cys326 OGG1 protein was found to have a lower enzymatic
activity, both when 8-oxoGua was excised from oligodeoxynucleotides [75], and when 8-oxoGua and FapyGua were liberated
from -irradiated DNA by puried Cys326 or Ser326 OGG1 enzyme
[74,76]. It was suggested that the presence of two OGG1 326Cys
alleles may confer an increased risk of lung, prostate and nasopharyngeal cancer [7779], but no association with the risk for colon
cancer was found [80,81]. Our and other functional studies report
a decrease in 8-oxoGua excision rate in lung [41,82,83] and
head and neck cancer patients [84]. Such a decrease in excision
rate and simultaneous increase in 8-oxodG and FapydG levels
in cellular DNA favors a pro-oxidant state and may accelerate
the acquisition of mutations in critical genes leading to cancer.
Such an idea is basically derived from the observation that a
pro-oxidant environment is characteristic for advanced stages of
cancer.
Ade is excised by alkylpurine-DNA-N-glycosylase (ANPG) [85],
whereas Cyt by thymine-DNA glycosylase (TDG); the latter also
excises thymine from G:T pairs, resulting from deamination of
5-methylcytosine [86]. Ethenocytosine is also excised by singlestranded uracil DNA glycosylase, SMUG1 [87] and by MBD4
glycosylase, although activity of the latter is strongly limited to CpG
islands [88,89].
ANPG glycosylase has a wide substrate specicity, and
besides Ade excises from DNA also 1,N2 -ethenoguanine [90],
hypoxanthine and the alkylated bases, 3-methyladenine and 7methylguanine [85]. TDG excises Cyt, Thy and Ura from pairs
with guanine, and its activity is strongly stimulated by the
next enzyme in the BER pathway, AP endonuclease, APE1 [91].
TDG and APE1 also play the role of transcription regulators
[9295].
Ethenocytosine repair by the BER system is dependent not only
on the level and activity of TDG, MBD4 or SMUG1 DNA glycosylases,
but also on the presence and/or activity of ANPG glycosylase, which
primary substrate is Ade. ANPG binds strongly to DNA containing
Cyt, and although does not excise this modied base, does not
allow its excision by the proper Cyt glycosylase, like TDG [96].
This may explain observation that the level of Cyt measured in
different human tissues was constantly higher than that of Ade
[19].

Table 1
mRNA level of DNA glycosylases (OGG1, ANPG and TDG) and APE endonuclease
(APE1), and MTH1 phosphohydrolase in relation to 18S rRNA (106 ) in leukocytes
of CRC and AD patients and healthy controls.
mRNA level (arbitrary units)a

OGG1

ANPG

TDG

APE1

MTH1

CRC patients (C)

AD patients (A)

Controls (H)

1.28
(0.715.58)
n = 44
12.14
(4.0637.07)
n = 44
7.97
(1.8721.03)
n = 44
88.25
(42.88362.51)
n = 44
86.82
(5.19728.53)
n = 37

0.99
(0.454.91)
n = 38

0.19
(0.060.54)
n = 40
1.08
(0.314.75)
n = 39
0.81
(0.261.84)
n = 39
13.87
(2.3243.04)
n = 40
5.26
(0.3311.69)
n = 19

113.83
(48.74292.37)
n = 38

<0.001 (C vs. H)b


<0.001 (A vs. H)b
0.46 (C vs. A)b
<0.001(C vs. H)

<0.001 (C vs. H)

<0.001 (C vs. H)b


<0.001 (A vs. H)b
0.5 (C vs. A)b
0.0012 (C vs. H)

Adapted from [42,105] with permission from Oxford University Press and Elsevier,
respectively.
All subjects were Caucasians.
a
Comparison of mRNA levels, expressed as median and interquartile range,
between CRC and AD patients and controls (MannWhitney U test).
b
p values are after Bonferroni correction.

4.1. Repair of 8-oxo-7,8-dihydro-2 -deoxyguanosine in colon


adenoma and carcinoma patients
In order to get an insight into the importance of oxidative stress
and repair of oxidatively damaged DNA bases in the pathology
of colon cancer, we examined groups of individuals at different
stages of disease development, namely CRC patients, patients who
developed benign adenomas (AD) and healthy individuals for the
markers of oxidative stress, repair capacity of oxidatively damaged
DNA, and polymorphism of DNA repair genes.
Increased oxidative stress was observed both in CRC patients
and in AD individuals. This manifested in an elevation of the 8oxodG level in leukocyte DNA and in urine as well as depletion of
antioxidant vitamins (ascorbic acid, -tocopherol, retinol) and uric
acid in blood plasma of CRC patients and AD individuals in comparison to healthy controls [42]. Depletion of antioxidant vitamins
in blood plasma was gradual, which suggests that oxidative stress
is increasing gradually with the disease progression. This is supported by the fact that urinary level of 8-oxodG was augmented
already in AD patients, but 8-oxoGua level was enhanced only in
CRC, but not in AD individuals [42]. Urinary 8-oxodG may reect
oxidation of nucleotide pool, while 8-oxoGua reects rather oxidation of DNA. Since nucleotide pool is much more susceptible to
oxidation than dsDNA, 8-oxodG in urine may be a more sensitive
indicator of oxidative processes in the body, and shows that even
in AD individuals the oxidative processes are enhanced. The suggested mechanisms responsible for the oxidative stress in cancer
patients include [43]: (i) granulocyte activation with release of ROS;
(ii) stimulation of cytokines, of which some, e.g., tumor necrosis
factor, produce large amounts of ROS; (iii) production of hydrogen
peroxide by malignant cells, which in advanced stages of cancer
may be released into the blood stream and penetrate into other
tissues [2].
Interestingly, we also observed an increase of mRNA levels of
repair enzymes, OGG1, APE1 and MTH1 in leukocytes of CRC and
AD individuals (Table 1) [42]. Expression of APE1 and OGG1 genes
was shown to be induced by ROS [97], and this could explain induction of repair enzymes transcription in leukocytes of CRC and AD
individuals. On the other hand induction of repair enzymes may be
a part of a carcinogenic program, which might decrease apoptosis

B. Tudek, E. Speina / Mutation Research 736 (2012) 8292

100

CRC patients
Controls

8-oxoGua excision activity


(pmols/h/mg protein)

90
80

***

70
60

n = 68

50
40

n = 90

30
20
10
0

120

8-oxoGua excision activity


(pmols/h/mg protein)

110

***

CRC patients
Controls

100

*
**

90
80
70

n = 34

60

n = 14

50
40

n = 53

30

n = 13

n = 32

20
10
0
Ser/Ser

Ser/Cys

Cys/Cys

OGG1 Ser326Cys polymorphism


Fig. 2. The mean and standard deviation of 8-oxoGua excision activity in leukocytes
of CRC patients and control individuals, in the entire groups (A) and in the groups
distinguished on the basis of OGG1 Ser326Cys polymorphism (B). ***, p < 0.001; **,
p < 0.005; *, p < 0.05 (MannWhitney U test).
Adapted from [42] with permission from Oxford University Press.

in aberrant cells and/or favor genomic instability. Our results corroborate the ndings of Winnepenninckx and coworkers [98] that
induction of most of the DNA repair genes occurs very early in the
carcinogenic process, e.g., four years before clinical manifestation
of malignant skin melanoma.
Increase of mRNA level of OGG1 and APE1 repair enzymes
was accompanied by the augmentation of 8-oxoGua excision rate
in leukocytes of CRC patients (Fig. 2A) [42]. The higher activity
observed in the cell extracts from the cancer patients is even more
meaningful in view of the nding that homozygous OGG1 326Cys
variants, which are generally characterized by lower OGG1 activity,

87

were much more frequent among the cancer patients than in the
control and adenoma groups (Table 2) [42]. In our study the effect of
the OGG1 Ser326Cys polymorphism on 8-oxoGua excision rate was
clearly seen in the CRC patient group, in which Cys homozygotes
were found to have a decreased 8-oxoGua excision rate in comparison with Ser homozygotes (Fig. 2B) [42]. Thus, the observed
increase in 8-oxoGua repair capacity in CRC patients in comparison to healthy individuals might be due to increased transcription
from OGG1 and APE1 genes, which was observed in AD and CRC individuals (Table 1) [42]. Interestingly, the OGG1 genotype exerted a
limited effect on 8-oxodG level in leukocytes and urinary excretion
of 8-oxoGua [42]. In contrast to our studies, Janssen et al. [99] and
Jensen et al. [100] reported that 8-oxoGua repair incision in human
lymphocytes was unaffected by the polymorphic status at codon
326 of the OGG1 gene. These studies were performed on the population of healthy subjects only. It was shown that OGG1 Cys326
variant forms dimers with lower enzymatic activity only under
conditions of oxidative stress probably due to OGG1 cysteine 326
oxidation [101]. In parallel, Bravard et al. [102] reported recently
that extracts from lymphoblastoid cell lines established from individuals carrying the Cys326Cys genotype have almost 2-fold lower
basal 8-oxoGua DNA glycosylase activity than the Ser326Ser variant
and were more sensitive to inactivation by oxidizing agents than
that of the Ser/Ser cells. Analysis of redox status of the OGG1 protein in cells conrmed that the lower activity of OGG1-Cys326 was
associated with the oxidation of Cys326 to form a disulde bond.
Thus different degrees of oxidative stress in different studies might
partially explain the controversies in the literature.
In the group of healthy subjects there was signicant positive
correlation between mRNA levels of the main enzymes involved in
8-oxoGua removal (OGG1 and APE1), and the amount of 8-oxoGua
in urine [42]. These ndings suggested that the amount of 8-oxoGua
in urine might be attributed to DNA repair. It is in parallel with
previous results, which also showed that DNA repair was responsible for the presence of oxidatively damaged DNA lesions in urine
[103]. However, we did not nd such correlation in the groups of
adenoma individuals and carcinoma patients. The main reason for
this inconsistency may be aberrant DNA oxidation in cancerous and
precancerous conditions which may overshadow the subtle relationship observed in healthy subjects. Alternatively, the excretion
of 8-oxoGua might reect the level of oxidative stress, independent
of the activities of the repair enzymes [104].
Also, only a weak positive correlation was found between 8oxoGua level in urine and the stage of disease [42].
4.2. Repair of etheno DNA adducts in colon carcinogenesis
In contrast to cleavage of 8-oxoGua, leukocytes excision activity of Ade and Cyt was signicantly lower in CRC patients when

Table 2
Genotypes and allelic frequencies of the OGG1 Ser326Cys polymorphism among CRC patients, individuals with AD and healthy controls.
OGG1 polymorphism
Ser326Cys
Ser/Ser
Ser/Cys
Cys/Cys
Allelic frequency
Ser (C)
Cys (G)

CRC patients (C)(n = 74)a n (%)

AD patients (A)(n = 76)a n (%)

Controls (H)(n = 97)a n (%), (%)b

38 (51.3)
19 (25.7)
17 (23)

46 (60.5)
29 (38.2)
1 (1.3)

63 (65), (81)c
33 (34), (18)d
1 (1), (1)e

>0.0000 (C vs. H)
>0.079 (A vs. H)
>0.0000 (C vs. A)

0.64
0.36

0.8
0.2

0.82
0.18

Adapted from [42] with permission from Oxford University Press.


All subjects were Caucasians.
a
Comparison of OGG1 polymorphic frequency between CRC and AD patients and controls (Chi-square distribution analysis).
b
Results from HardyWeinberg equilibrium.
c
Homozygous OGG1 Ser326Ser individuals.
d
Heterozygous OGG1 Ser326Cys individuals.
e
Homozygous OGG1 Cys326Cys individuals.

88

B. Tudek, E. Speina / Mutation Research 736 (2012) 8292

50
45

***

CRC patients
Controls

Excision activity
(pmols/h/g protein)

40

n = 56

35
30
25

n = 54

20
15

10

n = 54

n = 56

Excision activity
(pmols/h/g protein)

Ade

Cyt

16

***

14

Tumor
Normal colon

n = 54

12
10

***

n = 54

n = 54

n = 54
2
0

Ade

Cyt

Fig. 3. (A) The mean and standard deviation of Ade and Cyt excision rate in
leukocyte DNA of CRC patients and control individuals. ***, p < 0.001; *, p < 0.05
(MannWhitney U test). (B) The mean and standard deviation of Ade and Cyt
excision rate in colon tumor and normal surrounding tissues of CRC patients. ***,
p < 0.001 (Wilcoxon rank sum test).
Derived from [105] with permission from Elsevier.

compared to controls matched for age, sex and a smoking habit


(Fig. 3A) [105]. However, in colon tissues, excision activities for
Ade and Cyt were signicantly higher in tumor than in adjacent,
asymptomatic colon tissue (Fig. 3B) [105]. In leukocytes (PBL) of
CRC patients dA and 1,N2 -dG level was unexpectedly lower than
in controls, and the level of dA and dC also showed a tendency
to be lower in tumor than in normal colon tissue [105]. This suggests that the regulation of etheno adduct level and repair may be
multifactorial and may differ in target (colon) and surrogate (PBL)
tissues.
The lower excision rate for Ade and Cyt in leukocytes of CRC
patients was not linked to mutations in exons of ANPG and TDG
genes nor to down regulation of mRNA expression of ANPG and
TDG glycosylases or its BER activator, APE1 endonuclease; in the
contrary, mRNA levels of these repair enzymes were signicantly
higher in PBL of CRC than in healthy controls (Table 1) [42,105].
Thus direct inactivation/inhibition of repair enzymes activities
by inammatory mediators released in the blood stream is a
more likely explanation. Indeed, several DNA repair enzymes were
shown to be directly inhibited by nitric oxide [106108], hydrogen peroxide [109] and LPO product, 4-hydroxynonenal [55,110].
As shown for several human cancer sites and animal tumors, persistent oxidative stress and LPO-induced DNA damage increased
already during premalignant stages in a time-dependent manner
[111]. A number of studies demonstrated increase of mRNA level
of several repair enzymes upon oxidative stress [97,112,113]. The
observed lower Ade and Cyt excision (Fig. 3A) and simultaneous lower level of etheno adducts in PBL of cancer patients [105]

may thus suggest that other mechanisms are up-regulated in CRC


patients and determine the level of etheno adducts in DNA. Other
repair systems engaged in etheno adducts elimination from DNA
include, e.g., oxidative demethylation by AlkB family proteins or
nucleotide excision repair [114,115].
Of interest are the ndings in tumorous colon tissue. The Ade
and Cyt excising activities were much higher in tumor than in
the surrounding normal colon (Fig. 3B) and was consistent with a
23 times lower dA and dC adduct levels [105]. Also in several
other studies lower level of dA and dC was observed in colon
tumors than in colon tissues of cancer prone patients suffering from
ulcerative colitis, Crohns disease or familial adenomatous polyposis [56,116]. In our study in normal colon a negative correlation was
found between dC level in DNA and its excision rate ( = 0.64,
p = 0.013) [105]. This supports a role of BER in Cyt-elimination;
however, does not exclude other etheno adduct control systems. In
several types of non tumorous human tissues previously analyzed,
the dA level in DNA was found to be lower than that of dC [111],
consistent with our observation that excision activity was found to
be higher for Ade than for Cyt (Fig. 3) [105].
In order to understand a huge increase of excision activity of
Ade and Cyt in colon tumor tissue in comparison to asymptomatic colon margin, we measured mRNA level of ANPG and TDG
glycosylases, as well as APE1 endonuclease in tumor and normal
colon tissues. Higher mRNA level in tumor tissue was observed
only for APE1 [105]. APE1 stimulates Cyt excision [91], and most
probably also Ade. Another possibility are frequent mutations in
the APC gene in colon tumors [117]. APC tumor suppressor protein may inhibit BER engaged in repair of Ade and Cyt since it
was shown that it can directly interact with DNA polymerase
and FEN1 endonuclease [118]. BER activity was inversely associated
with APC expression in several breast cancer cell lines [119].
In conclusion, we have shown, for the rst time, that excision
activity of Ade and Cyt is lower in PBL of CRC patients when
compared to healthy individuals. However, our unexpected ndings, that the level of dA and 1,N2 -dG was lower in DNA of
PBL from CRC patients than in healthy controls currently remains
mechanistically difcult to explain. If such etheno adducts, particularly Cyt excision deciency also occurs during premalignant
stages in the target organ, still may favor acquisition of mutations
in critical genes leading to neoplastic transformation. TDG glycosylase removes Thy and Ura paired with guanine, and may prevent
induction of mutations caused by cytosine and 5-methylcytosine
deamination [120].

5. Aberrant repair of oxidatively damaged DNA bases and


mutations in critical genes
Activity and expression of BER proteins is modulated by two signaling molecules, which are frequently mutated in colon cancer, the
APC and TP53 gene products. Thus, mutations in these genes affect
repair rate of oxidatively damaged DNA, and favor pro-mutagenic
events within colon epithelial cells. On the other hand at least three
BER proteins, namely, APE1, TDG and OGG1 take part in the regulation of gene expression.
Mutations in the APC gene are early events in colon carcinogenesis, and are found in about 80% of colon adenomas and carcinomas.
The most frequent mutation results in truncation of the C-terminal
part of the APC protein. Wild type APC protein directly interacts
with DNA polymerase , FEN1 endonuclease [118], and as recently
suggested, APE1 endonuclease [121] inhibiting the assembly of BER
proteins on damaged DNA and the activity of short and long patch
BER (Fig. 4). In LoVo, colon cancer cells expressing truncated APC
protein an accelerated assembly of BER proteins, as well as higher
activity of APE1, FEN1 and Pol were observed [121]. Activation of

B. Tudek, E. Speina / Mutation Research 736 (2012) 8292

Progression

Genome
instability

APE1/
ANPG

+
APC
mutated

TDG/MBD4

AID-mediated
demethylation of
gene promoters

Transcription of
several genes

+
+

Angiogenesis
Erythropoesis

Progression

Pol
FEN1
APE1

TP53
mutated

Genome
instability

89

HIF-1

OGG1

Transcription of
Myc-regulated
genes

Proliferation
Progression

Fig. 4. The inuence of APC and TP53 genes mutations on the activity of BER enzymes and genome stability. Mutations in APC and TP53 proteins lead to increase in APE1
activity and transcription, respectively, and up-regulate BER. Increased APE1, ANPG and unbalanced BER pathway may favor genomic instability and cancer progression.
APE1 activity may also accelerate the excision rate of DNA glycosylases, OGG1 and TDG/MBD4. APE1, TDG and OGG1 take part in the regulation of gene expression. TDG
interacts with the deaminase AID and may favor demethylation of promoters and induction of transcription of several genes. OGG1 and APE1 stimulate transcription of genes
regulated by c-Myc and estrogen receptors, and thus may contribute to acceleration of cell proliferation. Increased APE1 activity may result in unbalancing of BER pathway,
and may favor genomic instability and cancer progression. APE1 also participates in HIF-1 mediated angiogenesis and erythropoesis and may favor tumor growth. + symbol
in circle denotes stimulation or increase; APC, adenomatous polyposis coli; Pol , DNA polymerase ; FEN 1, ap endonuclease 1; APE1, apurinic site endonuclease 1; ANPG,
alkyladenine DNA N-glycosylase; OGG1, 8-oxoGua DNA N-glycosylase; TDG, thymine DNA N-glycosylase; MBD4, methyl-CpG binding domain protein; AID, activation-induced
deaminase; HIF-1, hypoxia-inducible factor 1 subunit.

APE1 may also occur as a consequence of TP53 gene mutations,


which are late, but frequent events in colon carcinogenesis (in
about 50% of cancers). TP53 protein participates in downregulation
of APE1 transcription in cells treated with DNA damaging agents
[122]. On the other hand TP53 mediates ubiquitination of APE1
by TP53 inhibitor, MDM2, and in consequence APE1 degradation
[123]. Thus, mutations in TP53 gene should additionally increase
APE1 amount in cancer cells.
Increased APE1 activity may result in unbalancing of BER
pathway, and may favor genomic instability and cancer progression: in inamed non-cancerous colon tissues of ulcerative colitis
patients (at high risk for colon cancer, that accumulate high etheno
DNA adducts [124]), ANPG and APE1 activities were signicantly
increased, and microsatellite instability (MSI) was positively correlated with their imbalanced activities of repair enzymes [125].
Similarly, in yeast and human cells over-expression of ANPG and/or
APE1 was associated with frameshift mutations and MSI [125].
APE1, besides DNA repair function, also plays a role of redox
regulatory protein for several transcription factors. For example,
participates in HIF-1 mediated angiogenesis [126]. Thus at late
stages of CRC development, mutations in TP53 gene will lead to
accumulation of APE1 protein, and in addition to increasing genome
instability will also favor angiogenesis and in consequence tumor
growth (Fig. 4).
Increased APE1 activity may also accelerate the excision rate of
DNA glycosylases. TDG and OGG1 were shown to be particularly
susceptible to induction by APE1, since they bind strongly to AP
sites, and APE1 increases turnover of these enzymes on damaged
DNA [91,127]. Increased activity of TDG may favor demethylation of promoters of several genes and in consequence contribute
to hypomethylation of DNA observed in colon cancer [13] and
induction of transcription of several genes. TDG interacts with the
deaminase AID and the damage response protein GADD45a. 5Methylcytosine and 5-hydroxymethylcytosine are rst deaminated
by AID to thymine and 5-hydroxymethyluracil, respectively, and
subsequently TDG removes thymine and 5-hydroxymethyluracil
from dT:dG or 5OHMedU:dG pairs, which initiates BER and enables
reconstitution of G:C base pairs [128]. TDG is also necessary for
recruiting p300 to retinoic acid (RA)-regulated promoters, and
is engaged in transcriptional control of genes during embryonic

development. Its deciency is embrionaly lethal [120]. TDG status


may also determine sensitivity of CRC patients to chemotherapy
with 5-uorouracil (5-FU), a chemotherapeutic routinely used in
CRC treatment. Inactivation of TDG signicantly increases resistance of mouse and human cancer cells toward 5-FU. Excision of
5-FU from DNA by TDG generates persistent DNA strand breaks,
delays S-phase progression, and activates DNA damage signaling.
Hence, excision of 5-FU by TDG mediates DNA-directed cytotoxicity
[129].
OGG1 and APE1 stimulate transcription of genes regulated
by c-Myc transcription factor [130], and thus may contribute to
acceleration of cell proliferation (Fig. 4). The mechanism involves
demethylation of histone H3 by specic FAD-containing enzyme,
LSD1 demethylase which generates hydrogen peroxide during
demethylation [131]. In consequence a local oxidation of DNA
around the E-boxes and TSS region of Myc target genes is triggered.
8-OxodG, OGG1 and APE1 were found to be engaged on the same
RNA polymerase II and Myc occupied chromatin regions of Myc targets and stimulate transcription of these genes. Silencing of either
OGG1 or APE1 inhibits Myc mediated transcription. It is possible
that regulatory regions in a gene interact with each other forming loops that often bridge considerable distances on the genomic
loci [132]. Following Myc binding local demethylation of lysine 4
in histone H3 (H3K4me2) triggers recruitment of the OGG1 and
the downstream BER enzymes thus producing nicks on Myc target chromatin. This may relax the DNA strands and could favor
chromatin bending necessary for the formation of chromatin loops
that bring together RNAPII and Myc. OGG1 is also engaged in the
regulation of transcription from estrogen promoters [133].
6. Conclusions
Oxidative stress is induced at the early, adenoma, stage of colon
carcinogenesis, and is increasing during disease progression. CRC
and AD patients reveal a huge increase of mRNA level of base
excision repair enzymes in blood leukocytes. However, increased
transcription is not always correlated with repair capacity of tissues. Activities of BER enzymes remain under a complex regulative
network, which besides stimulation of transcription includes direct
inactivation by oxidation stress products, interaction with other

90

B. Tudek, E. Speina / Mutation Research 736 (2012) 8292

BER proteins, and with signaling molecules, which are frequently


mutated in colon cancer, that is APC and TP53 proteins. Mutated
proteins do not inhibit transcription and/or activity of base excision repair enzymes. This increases the amount of repair enzymes
within cells and introduces imbalance in the repair of oxidative
DNA damage. Such imbalance causes increased genome instability
and may be a driving force in the disease progression.
Conict of interest statement
The authors declare that there are no conicts of interest.
Funding
Funded by the Polish-French grant project 346/N-INCA/2008/0.
We thank all patients and volunteers who participated in this study.
References
[1] F. Levi, C. Pasche, C. La Vecchia, F. Lucchini, S. Franceschi, Food groups and
colorectal cancer risk, Br. J. Cancer 79 (1999) 12831287.
[2] M. Dizdaroglu, R. Olinski, J.H. Doroshow, S.A. Akman, Modication of DNA
bases in chromatin of intact target human cells by activated human polymorphonuclear leukocytes, Cancer Res. 53 (1993) 12691272.
[3] G. Bouguen, S. Manfredi, M. Blayau, C. Dugast, B. Buecher, D. Bonneau, L.
Siproudhis, V. David, J.F. Bretagne, Colorectal adenomatous polyposis associated with MYH mutations: genotype and phenotype characteristics, Dis.
Colon Rectum 50 (2007) 16121617.
[4] S. Jones, P. Emmerson, J. Maynard, J.M. Best, S. Jordan, G.T. Williams, J.R. Sampson, J.P. Cheadle, Biallelic germline mutations in MYH predispose to multiple
colorectal adenoma and somatic G:C T:A mutations, Hum. Mol. Genet. 11
(2002) 29612967.
[5] V. Gismondi, M. Meta, L. Bonelli, P. Radice, P. Sala, L. Bertario, A. Viel, M.
Fornasarig, A. Arrigoni, M. Gentile, M. Ponz de Leon, L. Anselmi, C. Mareni,
P.L. Bruzzi, Varesco prevalence of the Y165C, G382D and 1395GGA germline
mutations of the MYH gene in Italian patients with adenomatous polyposis
coli and colorectal adenomas, Int. J. Cancer 109 (2004) 680684.
[6] B.R. Berquist, D.M. Wilson, 3rd pathways for repairing and tolerating the
spectrum of oxidative DNA lesions, Cancer Lett. (2012).
[7] B. Tudek, A. Winczura, J. Janik, A. Siomek, M. Foksinski, R. Olinski, Involvement
of oxidatively damaged DNA and repair in cancer development and aging, Am.
J. Transl. Res. 2 (2010) 254284.
[8] M.S. Cooke, R. Olinski, M.D. Evans, Does measurement of oxidative damage
to DNA have clinical signicance? Clin. Chim. Acta 365 (2006) 3049.
[9] S. Maynard, S.H. Schurman, C. Harboe, N.C. de Souza-Pinto, V.A. Bohr, Base
excision repair of oxidative DNA damage and association with cancer and
aging, Carcinogenesis 30 (2009) 210.
[10] S.E. Kent, B. Majumdar, Metastatic tumours in the maxillary sinus. A report of
two cases and a review of the literature, J. Laryngol. Otol. 99 (1985) 459462.
[11] B. Tudek, R.P. Bird, W.R. Bruce, Foci of aberrant crypts in the colons of mice
and rats exposed to carcinogens associated with foods, Cancer Res. 49 (1989)
12361240.
[12] B.A. Magnuson, I. Carr, R.P. Bird, Ability of aberrant crypt foci characteristics to
predict colonic tumor incidence in rats fed cholic acid, Cancer Res. 53 (1993)
44994504.
[13] E.R. Fearon, Molecular genetics of colorectal cancer, Annu. Rev. Pathol. 6
(2011) 479507.
[14] M. van de Wetering, E. Sancho, C. Verweij, W. de Lau, I. Oving, A. Hurlstone,
K. van der Horn, E. Batlle, D. Coudreuse, A.P. Haramis, M. Tjon-Pon-Fong, P.
Moerer, M. van den Born, G. Soete, S. Pals, M. Eilers, R. Medema, H. Clevers,
The beta-catenin/TCF-4 complex imposes a crypt progenitor phenotype on
colorectal cancer cells, Cell 111 (2002) 241250.
[15] M. Valko, C.J. Rhodes, J. Moncol, M. Izakovic, M. Mazur, Free radicals, metals
and antioxidants in oxidative stress-induced cancer, Chem. Biol. Interact. 160
(2006) 140.
[16] M. Valko, D. Leibfritz, J. Moncol, M.T. Cronin, M. Mazur, J. Telser, Free radicals
and antioxidants in normal physiological functions and human disease, Int. J.
Biochem. Cell Biol. 39 (2007) 4484.
[17] L.A. Loeb, Cancer cells exhibit a mutator phenotype, Adv. Cancer Res. 72 (1998)
2556.
[18] K.B. Beckman, B.N. Ames, Oxidative decay of DNA, J. Biol. Chem. 272 (1997)
1963319636.
[19] H. Bartsch, J. Nair, Oxidative stress and lipid peroxidation-derived DNAlesions in inammation driven carcinogenesis, Cancer Detect. Prev. 28 (2004)
385391.
[20] J. Potack, S.H. Itzkowitz, Colorectal cancer in inammatory bowel disease, Gut
Liver 2 (2008) 6173.
[21] K. Uchida, 4-Hydroxy-2-nonenal: a product and mediator of oxidative stress,
Prog. Lipid Res. 42 (2003) 318343.

[22] F.L. Chung, R.G. Nath, M. Nagao, A. Nishikawa, G.D. Zhou, K. Randerath,
Endogenous formation and signicance of 1,N2-propanodeoxyguanosine
adducts, Mutat. Res. 424 (1999) 7181.
[23] F.L. Chung, J. Pan, S. Choudhury, R. Roy, W. Hu, M.S. Tang, Formation of trans-4hydroxy-2-nonenal- and other enal-derived cyclic DNA adducts from omega3 and omega-6 polyunsaturated fatty acids and their roles in DNA repair and
human p53 gene mutation, Mutat. Res. 531 (2003) 2536.
[24] H.J. Chen, F.L. Chung, Epoxidation of trans-4-hydroxy-2-nonenal by fatty
acid hydroperoxides and hydrogen peroxide, Chem. Res. Toxicol. 9 (1996)
306312.
[25] C.E. Eberhart, R.J. Coffey, A. Radhika, F.M. Giardiello, S. Ferrenbach, R.N.
DuBois, Up-regulation of cyclooxygenase 2 gene expression in human
colorectal adenomas and adenocarcinomas, Gastroenterology 107 (1994)
11831188.
[26] H. Sano, Y. Kawahito, R.L. Wilder, A. Hashiramoto, S. Mukai, K. Asai, S. Kimura,
H. Kato, M. Kondo, T. Hla, Expression of cyclooxygenase-1 and -2 in human
colorectal cancer, Cancer Res. 55 (1995) 37853789.
[27] M.J. Thun, S.J. Henley, C. Patrono, Nonsteroidal anti-inammatory drugs as
anticancer agents: mechanistic, pharmacologic, and clinical issues, J. Natl.
Cancer Inst. 94 (2002) 252266.
[28] C.H. Liu, S.H. Chang, K. Narko, O.C. Trifan, M.T. Wu, E. Smith, C. Haudenschild,
T.F. Lane, T. Hla, Overexpression of cyclooxygenase-2 is sufcient to induce
tumorigenesis in transgenic mice, J. Biol. Chem. 276 (2001) 1856318569.
[29] D. Wang, R.N. Dubois, Prostaglandins and cancer, Gut 55 (2006) 115122.
[30] M.A. Iniguez, A. Rodriguez, O.V. Volpert, M. Fresno, J.M. Redondo,
Cyclooxygenase-2: a therapeutic target in angiogenesis, Trends Mol. Med.
9 (2003) 7378.
[31] A. Greenhough, H.J. Smartt, A.E. Moore, H.R. Roberts, A.C. Williams, C.
Paraskeva, A. Kaidi, The COX-2/PGE2 pathway: key roles in the hallmarks
of cancer and adaptation to the tumour microenvironment, Carcinogenesis
30 (2009) 377386.
[32] N. Speed, I.A. Blair, Cyclooxygenase- and lipoxygenase-mediated DNA damage, Cancer Metastasis Rev. 30 (2011) 437447.
[33] B.S. Reddy, C.V. Rao, Novel approaches for colon cancer prevention by
cyclooxygenase-2 inhibitors, J. Environ. Pathol. Toxicol. Oncol. 21 (2002)
155164.
[34] A. Strillacci, C. Griffoni, M.C. Valerii, G. Lazzarini, V. Tomasi, E. Spisni,
RNAi-based strategies for cyclooxygenase-2 inhibition in cancer, J. Biomed.
Biotechnol. (2010) 828045.
[35] S. Ishikawa, S. Tamaki, M. Ohata, K. Arihara, M. Itoh, Heme induces DNA damage and hyperproliferation of colonic epithelial cells via hydrogen peroxide
produced by heme oxygenase: a possible mechanism of heme-induced colon
cancer, Mol. Nutr. Food Res. 54 (2010) 11821191.
[36] J.P. Angeli, C.C. Garcia, F. Sena, F.P. Freitas, S. Miyamoto, M.H. Medeiros, P.
Di, Mascio lipid hydroperoxide-induced and hemoglobin-enhanced oxidative
damage to colon cancer cells, Free Radic. Biol. Med. 51 (2011) 503515.
[37] H.K. Seitz, P. Becker, Alcohol metabolism and cancer risk, Alcohol Res. Health
30 (3841) (2007) 3744.
[38] C.C. Garcia, J.P. Angeli, F.P. Freitas, O.F. Gomes, T.F. de Oliveira, A.P. Loureiro, P.
Di Mascio, M.H. Medeiros, [13C2]-Acetaldehyde promotes unequivocal formation of 1,N2-propano-2 -deoxyguanosine in human cells, J. Am. Chem. Soc.
133 (2011) 91409143.
[39] E. Eder, M. Wacker, P. Wanek, Lipid peroxidation-related 1,N2propanodeoxyguanosine-DNA adducts induced by endogenously formed
4-hydroxy-2-nonenal in organs of female rats fed diets supplemented with
sunower, rapeseed, olive or coconut oil, Mutat. Res. 654 (2008) 101107.
[40] Y. Wang, G. Millonig, J. Nair, E. Patsenker, F. Stickel, S. Mueller, H. Bartsch,
H.K. Seitz, Ethanol-induced cytochrome P4502E1 causes carcinogenic ethenoDNA lesions in alcoholic liver disease, Hepatology 50 (2009) 453461.
[41] D. Gackowski, E. Speina, M. Zielinska, J. Kowalewski, R. Rozalski, A. Siomek, T.
Paciorek, B. Tudek, R. Olinski, Products of oxidative DNA damage and repair
as possible biomarkers of susceptibility to lung cancer, Cancer Res. 63 (2003)
48994902.
[42] T. Obtulowicz, M. Swoboda, E. Speina, D. Gackowski, R. Rozalski, A. Siomek, J.
Janik, B. Janowska, J.M. Ciesla, A. Jawien, Z. Banaszkiewicz, J. Guz, T. Dziaman,
A. Szpila, R. Olinski, B. Tudek, Oxidative stress and 8-oxoguanine repair are
enhanced in colon adenoma and carcinoma patients, Mutagenesis 25 (2010)
463471.
[43] R. Olinski, D. Gackowski, R. Rozalski, M. Foksinski, K. Bialkowski, Oxidative
DNA damage in cancer patients: a cause or a consequence of the disease
development? Mutat. Res. 531 (2003) 177190.
[44] R. Olinski, D. Gackowski, M. Foksinski, R. Rozalski, K. Roszkowski, P. Jaruga,
Oxidative DNA damage: assessment of the role in carcinogenesis, atherosclerosis, and acquired immunodeciency syndrome, Free Radic. Biol. Med. 33
(2002) 192200.
[45] M.D. Evans, M. Dizdaroglu, M.S. Cooke, Oxidative DNA damage and disease:
induction, repair and signicance, Mutat. Res. 567 (2004) 161.
[46] P. Jaruga, T.H. Zastawny, J. Skokowski, M. Dizdaroglu, R. Olinski, Oxidative
DNA base damage and antioxidant enzyme activities in human lung cancer,
FEBS Lett. 341 (1994) 5964.
[47] K.C. Cheng, D.S. Cahill, H. Kasai, S. Nishimura, L.A. Loeb, 8-Hydroxyguanine an
abundant form of oxidative DNA damage, causes G- - - -T and A- - - -C substitutions, J. Biol. Chem. 267 (1992) 166172.
[48] M.T. Russo, M.F. Blasi, F. Chiera, P. Fortini, P. Degan, P. Macpherson, M.
Furuichi, Y. Nakabeppu, P. Karran, G. Aquilina, M. Bignami, The oxidized
deoxynucleoside triphosphate pool is a signicant contributor to genetic

B. Tudek, E. Speina / Mutation Research 736 (2012) 8292

[49]

[50]
[51]

[52]

[53]

[54]

[55]

[56]

[57]

[58]

[59]

[60]

[61]

[62]

[63]

[64]

[65]

[66]

[67]

[68]

[69]

[70]

[71]

[72]
[73]

instability in mismatch repair-decient cells, Mol. Cell. Biol. 24 (2004)


465474.
E. Speina, K.D. Arczewska, D. Gackowski, M. Zielinska, A. Siomek, J.
Kowalewski, R. Olinski, B. Tudek, J.T. Kusmierek, Contribution of hMTH1 to
the maintenance of 8-oxoguanine levels in lung DNA of non-small-cell lung
cancer patients, J. Natl. Cancer Inst. 97 (2005) 384395.
L.J. Marnett, Lipid peroxidation-DNA damage by malondialdehyde, Mutat.
Res. 424 (1999) 8395.
A. Chenna, C.R. Iden, Characterization of 2 -deoxycytidine and 2 deoxyuridine adducts formed in reactions with acrolein and 2-bromoacrolein,
Chem. Res. Toxicol. 6 (1993) 261268.
A. Chenna, R.A. Rieger, C.R. Iden, Characterization of thymidine adducts
formed by acrolein and 2-bromoacrolein, Carcinogenesis 13 (1992)
23612365.
A. Barbin, H. Bartsch, P. Leconte, M. Radman, Studies on the miscoding properties of 1,N6-ethenoadenine and 3,N4-ethenocytosine, DNA reaction products
of vinyl chloride metabolites, during in vitro DNA synthesis, Nucleic Acids
Res. 9 (1981) 375387.
P. Kowalczyk, J.M. Ciesla, M. Komisarski, J.T. Kusmierek, B. Tudek, Long-chain
adducts of trans-4-hydroxy-2-nonenal to DNA bases cause recombination,
base substitutions and frameshift mutations in M13 phage, Mutat. Res. 550
(2004) 3348.
L. Maddukuri, E. Speina, M. Christiansen, D. Dudzinska, J. Zaim, T. Obtulowicz, S. Kabaczyk, M. Komisarski, Z. Bukowy, J. Szczegielniak, A. Wojcik, J.T.
Kusmierek, T. Stevnsner, V.A. Bohr, B. Tudek, Cockayne syndrome group B
protein is engaged in processing of DNA adducts of lipid peroxidation product
trans-4-hydroxy-2-nonenal, Mutat. Res. 666 (2009) 2331.
J. Nair, F. Gansauge, H. Beger, P. Dolara, G. Winde, H. Bartsch, Increased ethenoDNA adducts in affected tissues of patients suffering from Crohns disease,
ulcerative colitis, and chronic pancreatitis, Antioxid. Redox Signal. 8 (2006)
10031010.
P. Fortini, E. Dogliotti, Base damage and single-strand break repair:
mechanisms and functional signicance of short- and long-patch repair subpathways, DNA Repair (Amst.) 6 (2007) 398409.
K.H. Almeida, R.W. Sobol, A unied view of base excision repair: lesiondependent protein complexes regulated by post-translational modication,
DNA Repair (Amst.) 6 (2007) 695711.
M.L. Hegde, T.K. Hazra, S. Mitra, Early steps in the DNA base excision/singlestrand interruption repair pathway in mammalian cells, Cell Res. 18 (2008)
2747.
A.B. Robertson, A. Klungland, T. Rognes, I. Leiros, DNA repair in mammalian
cells: base excision repair: the long and short of it, Cell. Mol. Life Sci. 66 (2009)
981993.
C. Colussi, E. Parlanti, P. Degan, G. Aquilina, D. Barnes, P. Macpherson, P. Karran, M. Crescenzi, E. Dogliotti, M. Bignami, The mammalian mismatch repair
pathway removes DNA 8-oxodGMP incorporated from the oxidized dNTP
pool, Curr. Biol. 12 (2002) 912918.
H. Hayakawa, A. Taketomi, K. Sakumi, M. Kuwano, M. Sekiguchi, Generation
and elimination of 8-oxo-7,8-dihydro-2 -deoxyguanosine 5 -triphosphate, a
mutagenic substrate for DNA synthesis, in human cells, Biochemistry 34
(1995) 8995.
T. Tsuzuki, A. Egashira, H. Igarashi, T. Iwakuma, Y. Nakatsuru, Y. Tominaga, H.
Kawate, K. Nakao, K. Nakamura, F. Ide, S. Kura, Y. Nakabeppu, M. Katsuki, T.
Ishikawa, M. Sekiguchi, Spontaneous tumorigenesis in mice defective in the
MTH1 gene encoding 8-oxo-dGTPase, Proc. Natl. Acad. Sci. U.S.A. 98 (2001)
1145611461.
K. Sakumi, Y. Tominaga, M. Furuichi, P. Xu, T. Tsuzuki, M. Sekiguchi, Y.
Nakabeppu, Ogg1 knockout-associated lung tumorigenesis and its suppression by Mth1 gene disruption, Cancer Res. 63 (2003) 902905.
K. Sakamoto, Y. Tominaga, K. Yamauchi, Y. Nakatsu, K. Sakumi, K. Yoshiyama,
A. Egashira, S. Kura, T. Yao, M. Tsuneyoshi, H. Maki, Y. Nakabeppu, T.
Tsuzuki, MUTYH-null mice are susceptible to spontaneous and oxidative
stress induced intestinal tumorigenesis, Cancer Res. 67 (2007) 65996604.
Y. Xie, H. Yang, C. Cunanan, K. Okamoto, D. Shibata, J. Pan, D.E. Barnes, T.
Lindahl, M. McIlhatton, R. Fishel, J.H. Miller, Deciencies in mouse Myh and
Ogg1 result in tumor predisposition and G to T mutations in codon 12 of the
K-ras oncogene in lung tumors, Cancer Res. 64 (2004) 30963102.
M.M. Slupska, W.M. Luther, J.H. Chiang, H. Yang, J.H. Miller, Functional expression of hMYH, a human homolog of the Escherichia coli MutY protein, J.
Bacteriol. 181 (1999) 62106213.
G. Dianov, C. Bischoff, J. Piotrowski, V.A. Bohr, Repair pathways for processing
of 8-oxoguanine in DNA by mammalian cell extracts, J. Biol. Chem. 273 (1998)
3381133816.
J.P. Radicella, C. Dherin, C. Desmaze, M.S. Fox, S. Boiteux, Cloning and characterization of hOGG1, a human homolog of the OGG1 gene of Saccharomyces
cerevisiae, Proc. Natl. Acad. Sci. U.S.A. 94 (1997) 80108015.
K. Shinmura, J. Yokota, The OGG1 gene encodes a repair enzyme for oxidatively damaged DNA and is involved in human carcinogenesis, Antioxid.
Redox Signal. 3 (2001) 597609.
N.A. Kuznetsov, V.V. Koval, D.O. Zharkov, G.A. Nevinsky, K.T. Douglas, O.S.
Fedorova, Kinetics of substrate recognition and cleavage by human 8oxoguanine-DNA glycosylase, Nucleic Acids Res. 33 (2005) 39193931.
S. Bjelland, E. Seeberg, Mutagenicity, toxicity and repair of DNA base damage
induced by oxidation, Mutat. Res. 531 (2003) 3780.
N. Al-Tassan, N.H. Chmiel, J. Maynard, N. Fleming, A.L. Livingston, G.T.
Williams, A.K. Hodges, D.R. Davies, S.S. David, J.R. Sampson, J.P. Cheadle,

[74]

[75]

[76]

[77]

[78]

[79]

[80]

[81]

[82]

[83]

[84]

[85]

[86]

[87]

[88]
[89]
[90]

[91]

[92]
[93]

[94]

[95]

[96]

[97]
[98]

91

Inherited variants of MYH associated with somatic G:C T:A mutations in


colorectal tumors, Nat. Genet. 30 (2002) 227232.
V.S. Sidorenko, A.P. Grollman, P. Jaruga, M. Dizdaroglu, D.O. Zharkov, Substrate specicity and excision kinetics of natural polymorphic variants and
phosphomimetic mutants of human 8-oxoguanine-DNA glycosylase, FEBS J.
276 (2009) 51495162.
J.W. Hill, M.K. Evans, Dimerization and opposite base-dependent catalytic
impairment of polymorphic S326C OGG1 glycosylase, Nucleic Acids Res. 34
(2006) 16201632.
C. Dherin, J.P. Radicella, M. Dizdaroglu, S. Boiteux, Excision of oxidatively
damaged DNA bases by the human alpha-hOgg1 protein and the polymorphic alpha-hOgg1(Ser326Cys) protein which is frequently found in human
populations, Nucleic Acids Res. 27 (1999) 40014007.
M. Goodman, R.M. Bostick, C. Dash, P. Terry, W.D. Flanders, J. Mandel, A
summary measure of pro- and anti-oxidant exposures and risk of incident
sporadic, colorectal adenomas, Cancer Causes Control 19 (2008) 10511064.
L. Chen, A. Elahi, J. Pow-Sang, P.J. Lazarus, Park Association between polymorphism of human oxoguanine glycosylase 1 and risk of prostate cancer, J. Urol.
170 (2003) 24712474.
E.Y. Cho, A. Hildesheim, C.J. Chen, M.M. Hsu, I.H. Chen, B.F. Mittl, P.H. Levine,
M.Y. Liu, J.Y. Chen, L.A. Brinton, Y.J. Cheng, C.S. Yang, Nasopharyngeal carcinoma and genetic polymorphisms of DNA repair enzymes XRCC1 and hOGG1,
Cancer Epidemiol. Biomarkers Prev. 12 (2003) 11001104.
R. Hansen, M. Saebo, C.F. Skjelbred, B.A. Nexo, P.C. Hagen, G. Bock, I.M. Bowitz
Lothe, E. Johnson, S. Aase, I.L. Hansteen, U. Vogel, E.H. Kure, GPX Pro198Leu
and OGG1 Ser326Cys polymorphisms and risk of development of colorectal
adenomas and colorectal cancer, Cancer Lett. 229 (2005) 8591.
T. Sliwinski, R. Krupa, M. Wisniewska-Jarosinska, E. Pawlowska, J. Lech, J. Chojnacki, J. Blasiak, Common polymorphisms in the XPD and hOGG1 genes are
not associated with the risk of colorectal cancer in a Polish population, Tohoku
J. Exp. Med. 218 (2009) 185191.
T. Paz-Elizur, M. Krupsky, S. Blumenstein, D. Elinger, E. Schechtman, Z. Livneh,
DNA repair activity for oxidative damage and risk of lung cancer, J. Natl. Cancer
Inst. 95 (2003) 13121319.
J. Janik, M. Swoboda, B. Janowska, J.M. Ciesla, D. Gackowski, J. Kowalewski,
R. Olinski, B. Tudek, E. Speina, 8-Oxoguanine incision activity is impaired in
lung tissues of NSCLC patients with the polymorphism of OGG1 and XRCC1
genes, Mutat. Res. 709710 (2011) 2131.
T. Paz-Elizur, R. Ben-Yosef, D. Elinger, A. Vexler, M. Krupsky, A. Berrebi, A.
Shani, E. Schechtman, L. Freedman, Z. Livneh, Reduced repair of the oxidative
8-oxoguanine DNA damage and risk of head and neck cancer, Cancer Res. 66
(2006) 1168311689.
M. Saparbaev, K. Kleibl, J. Laval, Escherichia coli, Saccharomyces cerevisiae, rat
and human 3-methyladenine DNA glycosylases repair 1,N6-ethenoadenine
when present in DNA, Nucleic Acids Res. 23 (1995) 37503755.
M. Saparbaev, J. Laval, 3,N4-ethenocytosine, a highly mutagenic adduct, is a
primary substrate for Escherichia coli double-stranded uracil-DNA glycosylase
and human mismatch-specic thymine-DNA glycosylase, Proc. Natl. Acad. Sci.
U.S.A. 95 (1998) 85088513.
B. Kavli, O. Sundheim, M. Akbari, M. Otterlei, H. Nilsen, F. Skorpen, P.A. Aas,
L. Hagen, H.E. Krokan, G. Slupphaug, hUNG2 is the major repair enzyme
for removal of uracil from U:A matches, U:G mismatches, and U in singlestranded DNA, with hSMUG1 as a broad specicity backup, J. Biol. Chem. 277
(2002) 3992639936.
A. Bellacosa, Role of MED1 (MBD4) Gene in DNA repair and human cancer, J.
Cell. Physiol. 187 (2001) 137144.
J. Jiricny, M. Menigatti, DNA cytosine demethylation: are we getting close?
Cell 135 (2008) 11671169.
M. Saparbaev, S. Langouet, C.V. Privezentzev, F.P. Guengerich, H. Cai,
R.H. Elder, J. Laval, 1,N(2)-ethenoguanine, a mutagenic DNA adduct, is a
primary substrate of Escherichia coli mismatch-specic uracil-DNA glycosylase and human alkylpurine-DNA-N-glycosylase, J. Biol. Chem. 277 (2002)
2698726993.
C.V. Privezentzev, M. Saparbaev, J. Laval, The HAP1 protein stimulates the
turnover of human mismatch-specic thymine-DNA-glycosylase to process
3,N(4)-ethenocytosine residues, Mutat. Res. 480481 (2001) 277284.
B. Demple, J.S. Sung, Molecular and biological roles of Ape1 protein in mammalian base excision repair, DNA Repair (Amst.) 4 (2005) 14421449.
S. Um, M. Harbers, A. Benecke, B. Pierrat, R. Losson, P. Chambon, Retinoic acid
receptors interact physically and functionally with the T:G mismatch-specic
thymine-DNA glycosylase, J. Biol. Chem. 273 (1998) 2072820736.
M. Tini, A. Benecke, S.J. Um, J. Torchia, R.M. Evans, P. Chambon, Association
of CBP/p300 acetylase and thymine DNA glycosylase links DNA repair and
transcription, Mol. Cell 9 (2002) 265277.
C. Missero, M.T. Pirro, S. Simeone, M. Pischetola, R. Di Lauro, The DNA
glycosylase T:G mismatch-specic thymine DNA glycosylase represses thyroid transcription factor-1-activated transcription, J. Biol. Chem. 276 (2001)
3356933575.
L. Gros, A.V. Maksimenko, C.V. Privezentzev, J. Laval, M.K. Saparbaev, Hijacking
of the human alkyl-N-purine-DNA glycosylase by 3,N4-ethenocytosine, a lipid
peroxidation-induced DNA adduct, J. Biol. Chem. 279 (2004) 1772317730.
B. Tudek, Base excision repair modulation as a risk factor for human cancers,
Mol. Aspects Med. 28 (2007) 258275.
V. Winnepenninckx, V. Lazar, S. Michiels, P. Dessen, M. Stas, S.R. Alonso, M.F.
Avril, P.L. Ortiz Romero, T. Robert, O. Balacescu, A.M. Eggermont, G. Lenoir,
A. Sarasin, T. Tursz, J.J. van den Oord, A. Spatz, Gene expression proling of

92

[99]

[100]

[101]

[102]

[103]

[104]

[105]

[106]

[107]

[108]

[109]
[110]

[111]

[112]

[113]

[114]
[115]

B. Tudek, E. Speina / Mutation Research 736 (2012) 8292


primary cutaneous melanoma and clinical outcome, J. Natl. Cancer Inst. 98
(2006) 472482.
K. Janssen, K. Schlink, W. Gotte, B. Hippler, B. Kaina, F. Oesch, DNA repair
activity of 8-oxoguanine DNA glycosylase 1 (OGG1) in human lymphocytes
is not dependent on genetic polymorphism Ser326/Cys326, Mutat. Res. 486
(2001) 207216.
A. Jensen, M. Lohr, L. Eriksen, M. Gronbaek, E. Dorry, S. Loft, P. Moller, Inuence of the OGG1 Ser326Cys polymorphism on oxidatively damaged DNA and
repair activity, Free Radic. Biol. Med. 52 (2012) 118125.
D.J. Smart, J.K. Chipman, N.J. Hodges, Activity of OGG1 variants in the repair of
pro-oxidant-induced 8-oxo-2 -deoxyguanosine, DNA Repair (Amst.) 5 (2006)
13371345.
A. Bravard, M. Vacher, E. Moritz, L. Vaslin, J. Hall, B. Epe, J.P. Radicella, Oxidation
status of human OGG1-S326C polymorphic variant determines cellular DNA
repair capacity, Cancer Res. 69 (2009) 36423649.
M.S. Cooke, M.D. Evans, R. Dove, R. Rozalski, D. Gackowski, A. Siomek, J. Lunec,
R. Olinski, DNA repair is responsible for the presence of oxidatively damaged
DNA lesions in urine, Mutat. Res. 574 (2005) 5866.
S. Loft, P. Danielsen, M. Lohr, K. Jantzen, J.G. Hemmingsen, M. Roursgaard,
D.G. Karotki, P. Moller, Urinary excretion of 8-oxo-7,8-dihydroguanine as
biomarker of oxidative damage to DNA, Arch. Biochem. Biophys. 518 (2012)
142150.
T. Obtulowicz, A. Winczura, E. Speina, M. Swoboda, J. Janik, B. Janowska,
J.M. Ciesla, P. Kowalczyk, A. Jawien, D. Gackowski, Z. Banaszkiewicz,
I. Krasnodebski, A. Chaber, R. Olinski, J. Nair, H. Bartsch, T. Douki, J.
Cadet, B. Tudek, Aberrant repair of etheno-DNA adducts in leukocytes and
colon tissue of colon cancer patients, Free Radic. Biol. Med. 49 (2010)
10641071.
M. Graziewicz, D.A. Wink, F. Laval, Nitric oxide inhibits DNA ligase activity: potential mechanisms for NO-mediated DNA damage, Carcinogenesis 17
(1996) 25012505.
D.A. Wink, J. Laval, The Fpg protein a DNA repair enzyme, is inhibited by
the biomediator nitric oxide in vitro and in vivo, Carcinogenesis 15 (1994)
21252129.
M. Jaiswal, N.F. LaRusso, N. Nishioka, Y. Nakabeppu, G.J. Gores, Human Ogg1
a protein involved in the repair of 8-oxoguanine, is inhibited by nitric oxide,
Cancer Res. 61 (2001) 63886393.
M.A. Graziewicz, B.J. Day, W.C. Copeland, The mitochondrial DNA polymerase
as a target of oxidative damage, Nucleic Acids Res. 30 (2002) 28172824.
Z. Feng, W. Hu, M.S. Tang, Trans-4-hydroxy-2-nonenal inhibits nucleotide
excision repair in human cells: a possible mechanism for lipid
peroxidation-induced carcinogenesis, Proc. Natl. Acad. Sci. U.S.A. 101 (2004)
85988602.
H. Bartsch, J. Nair, Accumulation of lipid peroxidation-derived DNA lesions:
potential lead markers for chemoprevention of inammation-driven malignancies, Mutat. Res. 591 (2005) 3444.
S.H. Cherng, K.H. Huang, S.C. Yang, T.C. Wu, J.L. Yang, H. Lee, Human 8oxoguanine DNA glycosylase 1 mRNA expression as an oxidative stress
exposure biomarker of cooking oil fumes, J. Toxicol. Environ. Health A 65
(2002) 265278.
A. Das, T.K. Hazra, I. Boldogh, S. Mitra, K.K. Bhakat, Induction of the human
oxidized base-specic DNA glycosylase NEIL1 by reactive oxygen species, J.
Biol. Chem. 280 (2005) 3527235280.
B. Hang, Repair of exocyclic DNA adducts: rings of complexity, Bioessays 26
(2004) 11951208.
J. Ringvoll, M.N. Moen, L.M. Nordstrand, L.B. Meira, B. Pang, A. Bekkelund, P.C.
Dedon, S. Bjelland, L.D. Samson, P.O. Falnes, A. Klungland, AlkB homologue
2-mediated repair of ethenoadenine lesions in mammalian DNA, Cancer Res.
68 (2008) 41424149.

[116] K. Schmid, J. Nair, G. Winde, I. Velic, H. Bartsch, Increased levels of promutagenic etheno-DNA adducts in colonic polyps of FAP patients, Int. J. Cancer 87
(2000) 14.
[117] M. Luchtenborg, M.P. Weijenberg, P.A. Wark, A.M. Saritas, G.M. Roemen, G.N.
van Muijen, A.P. de Bruine, P.A. van den Brandt, A.F. de, Goeij mutations in
APC CTNNB1 and K-ras genes and expression of hMLH1 in sporadic colorectal
carcinomas from the Netherlands Cohort Study, BMC Cancer 5 (2005) 160.
[118] A.S. Jaiswal, R. Balusu, M.L. Armas, C.N. Kundu, S. Narayan, Mechanism of adenomatous polyposis coli (APC)-mediated blockage of long-patch base excision
repair, Biochemistry 45 (2006) 1590315914.
[119] S. Narayan, A.S. Jaiswal, R. Balusu, Tumor suppressor APC blocks DNA polymerase beta-dependent strand displacement synthesis during long patch but
not short patch base excision repair and increases sensitivity to methylmethane sulfonate, J. Biol. Chem. 280 (2005) 69426949.
[120] D. Cortazar, C. Kunz, Y. Saito, R. Steinacher, P. Schar, The enigmatic thymine
DNA glycosylase, DNA Repair (Amst.) 6 (2007) 489504.
[121] A.S. Jaiswal, S. Narayan, Assembly of the base excision repair complex on
abasic DNA and role of adenomatous polyposis coli on its functional activity,
Biochemistry 50 (2011) 19011909.
[122] A. Zaky, C. Busso, T. Izumi, R. Chattopadhyay, A. Bassiouny, S. Mitra, K.K.
Bhakat, Regulation of the human AP-endonuclease (APE1/Ref-1) expression
by the tumor suppressor p53 in response to DNA damage, Nucleic Acids Res.
36 (2008) 15551566.
[123] C.S. Busso, T. Iwakuma, T. Izumi, Ubiquitination of mammalian AP endonuclease (APE1) regulated by the p53-MDM2 signaling pathway, Oncogene 28
(2009) 16161625.
[124] J. Nair, F. Gansauge, H. Beger, P. Dolara, G. Winde, H. Bartsch, Increased ethenoDNA adducts in affected tissues of patients suffering from Crohns disease
ulcerative colitis, and chronic pancreatitis, Antioxid. Redox Signal. 8 (2006)
10031010.
[125] L.J. Hofseth, The adaptive imbalance to genotoxic stress: genome guardians
rear their ugly heads, Carcinogenesis 25 (2004) 17871793.
[126] V. Singh-Gupta, H. Zhang, S. Banerjee, D. Kong, J.J. Raffoul, F.H. Sarkar, G.G.
Hillman, Radiation-induced HIF-1alpha cell survival pathway is inhibited by
soy isoavones in prostate cancer cells, Int. J. Cancer 124 (2009) 16751684.
[127] J.W. Hill, T.K. Hazra, T. Izumi, S. Mitra, Stimulation of human 8-oxoguanineDNA glycosylase by AP-endonuclease: potential coordination of the initial
steps in base excision repair, Nucleic Acids Res. 29 (2001) 430438.
[128] S. Cortellino, J. Xu, M. Sannai, R. Moore, E. Caretti, A. Cigliano, M. Le Coz,
K. Devarajan, A. Wessels, D. Soprano, L.K. Abramowitz, M.S. Bartolomei, F.
Rambow, M.R. Bassi, T. Bruno, M. Fanciulli, C. Renner, A.J. Klein-Szanto, Y. Matsumoto, D. Kobi, I. Davidson, C. Alberti, L. Larue, A. Bellacosa, Thymine DNA
glycosylase is essential for active DNA demethylation by linked deaminationbase excision repair, Cell 146 (2011) 6779.
[129] C. Kunz, F. Focke, Y. Saito, D. Schuermann, T. Lettieri, J. Selfridge, P. Schar, Base
excision by thymine DNA glycosylase mediates DNA-directed cytotoxicity of
5-uorouracil, PLoS Biol. 7 (2009) e91.
[130] S. Amente, L. Lania, E.V. Avvedimento, B. Majello, DNA oxidation drives Myc
mediated transcription, Cell Cycle 9 (2010) 30023004.
[131] S. Amente, A. Bertoni, A. Morano, L. Lania, E.V. Avvedimento, B. Majello,
LSD1-mediated demethylation of histone H3 lysine 4 triggers Myc-induced
transcription, Oncogene 29 (2010) 36913702.
[132] T. Sexton, F. Bantignies, G. Cavalli, Genomic interactions: chromatin loops
and gene meeting points in transcriptional regulation, Semin. Cell Dev. Biol.
20 (2009) 849855.
[133] B. Perillo, M.N. Ombra, A. Bertoni, C. Cuozzo, S. Sacchetti, A. Sasso, L. Chiariotti,
A. Malorni, C. Abbondanza, E.V. Avvedimento, DNA oxidation as triggered by
H3K9me2 demethylation drives estrogen-induced gene expression, Science
319 (2008) 202206.

You might also like