11
Mutation Research 736 (2012) 82–92 Contents lists available at SciVerse ScienceDirect Mutation Research/Fundamental and Molecular Mechanisms of Mutagenesis journa l h o me pa g e: www.elsevier.com/locate/molmut Co mm unit y add re ss: www.elsevier.com/locate/mutres Review Oxidatively damaged DNA and its repair in colon carcinogenesis Barbara Tudek a,b , El ˙ zbieta Speina a,a Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawi´ nskiego 5a, 02-106 Warsaw, Poland b 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 Inflammation, high fat, high red meat and low fiber consumption have for long been known as the most important etiological factors of sporadic colorectal cancers (CRC). Colon cancer originates from neoplas- tic 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. Pro- gression 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 mecha- nisms 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 significantly increased; however, excision rate is regulated separately, being increased for 8- oxoGua, while decreased for lipid peroxidation derived ethenoadducts, Ade and Cyt; (iii) excision rate of Ade and Cyt in colon tumors is significantly 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. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 83 2. Histopathology of CRC and mutations in critical genes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 83 3. Oxidative stress in the development of colon cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 84 3.1. Inflammation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 84 3.2. Consumption of red meat and alcohol and tobacco smoking . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 84 3.3. DNA damage . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 85 4. Repair of oxidative DNA damage as a driving force to colon carcinogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 85 4.1. Repair of 8-oxo-7,8-dihydro-2 -deoxyguanosine in colon adenoma and carcinoma patients . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 86 4.2. Repair of etheno DNA adducts in colon carcinogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 87 5. Aberrant repair of oxidatively damaged DNA bases and mutations in critical genes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 88 6. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 89 Conflict of interest statement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 90 Funding . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 90 References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 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 poly- posis 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,N 6 -ethenoadenine; dA, 1,N 6 -etheno-2 -deoxyadenosine; Cyt, 3,N 4 -ethenocytosine; dC, 3,N 4 -etheno-2 -deoxycytidine; 1N 2 -dG, 1,N 2 - etheno-2 -deoxyguanosine; LPO, lipid peroxidation; MMR, mismatch repair. Corresponding author. Tel.: +48 22 592 3334; fax: +48 22 592 2190. E-mail address: [email protected] (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

DNA Repair MMR Colon Cancer

  • Upload
    mack22

  • View
    15

  • Download
    4

Embed Size (px)

DESCRIPTION

DNA Repair MMR Colon Cancer

Citation preview

  • Mutation Research 736 (2012) 82 92

    Contents lists available at SciVerse ScienceDirect

    Mutation Research/Fundamental and MolecularMechanisms of Mutagenesis

    journa l h o me pa g e: www.elsev ier .com/ locate /molmutCo mm uni t y add re ss : www.elsev ier .com/ locate /mutres

    Review

    Oxidatively damaged DNA and its repair in colon carcinogenesis

    Barbara Tudeka,b, Elzbieta Speinaa,

    a Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Pawinskiego 5a, 02-106 Warsaw, Polandb Institute of Ge

    a r t i c l

    Article history:Received 9 JanReceived in reAccepted 16 AAvailable onlin

    Keywords:Colorectal canOxidative streLipid peroxida8-Oxo-7,8-dihEtheno DNA adDNA repair patients is signicantly increased; however, excision rate is regulated separately, being increased for 8-

    oxoGua, while decreased for lipid peroxidation derived ethenoadducts, Ade and Cyt; (iii) excision rateof 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. Introd2. Histop3. Oxida

    3.1. 3.2. 3.3.

    4. Repai4.1. 4.2.

    5. Aberr6. Concl

    ConiFundiRefere

    Abbreviatiposis coli; FapBER, base excietheno-2-deo

    CorresponE-mail add

    0027-5107/$ http://dx.doi.ouction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 83athology of CRC and mutations in critical genes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 83tive stress in the development of colon cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 84Inammation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 84Consumption of red meat and alcohol and tobacco smoking. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 84DNA damage . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 85

    r of oxidative DNA damage as a driving force to colon carcinogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 85Repair of 8-oxo-7,8-dihydro-2-deoxyguanosine in colon adenoma and carcinoma patients . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 86Repair of etheno DNA adducts in colon carcinogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 87

    ant repair of oxidatively damaged DNA bases and mutations in critical genes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 88usions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 89ct of interest statement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 90ng. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 90nces . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 90

    ons: 8-oxodG, 8-oxo-7,8-dihydro-2-deoxyguanosine; 8-oxoGua, 8-oxo-7,8-dihydroguanine; AD, adenoma; CRC, colorectal cancer; APC, adenomatous poly-yGua, 2,6-diamino-4-hydroxy-5-formamidopyrimidine; APE1, apurinic site endonuclease 1; ROS, reactive oxygen species; RNS, reactive nitrogen species;sion 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-xyguanosine; LPO, lipid peroxidation; MMR, mismatch repair.ding author. Tel.: +48 22 592 3334; fax: +48 22 592 2190.ress: [email protected] (E. Speina).

    see front matter 2012 Elsevier B.V. All rights reserved.rg/10.1016/j.mrfmmm.2012.04.003netics and Biotechnology, Faculty of Biology, Warsaw University, Pawinskiego 5a, 02-106 Warsaw, Poland

    e i n f o

    uary 2012vised form 2 April 2012pril 2012e 25 April 2012

    cersstionydroguanineducts

    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 mostimportant etiological factors of sporadic colorectal cancers (CRC). Colon cancer originates from neoplas-tic transformation in a single layer of epithelial cells occupying colonic crypts, in which migration andapoptosis 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 abundantlyin the early phase of the process. This is followed by mutations in RAS, TP53, and other genes. Pro-gression 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, andin blood leukocytes and urine, already at the earliest stages of disease development. Defence mecha-nisms are deregulated in CRC patients: (i) antioxidative vitamins level in blood plasma declines withthe development of disease; (ii) mRNA level of base excision repair enzymes in blood leukocytes of CRC

  • B. Tudek, E. Speina / Mutation Research 736 (2012) 82 92 83

    1. Introduction

    Colorectal cancer (CRC) is one of the most frequent causes ofdeath in Western countries. The cases are roughly equally dis-tributed bepathogeneswhich incretions in crit

    Inammof reactive of nucleic amutagenic

    The geneciency in mphenotype instability colon carcibe associatposis coli) signaling pof APC prowell describlesions whilife. The prpolyps is cluated formage of onse1015% of quently, in a biallelic mMutY is a DDNA of ade(8-oxoGua)pathologicagenerative review focucolon carcitiation (genmolecular son the dereresponse an

    2. Histopa

    A singleand rectumtially increastem cells atypes, absolial cells. Thdifferentiatthe colonicfrom the crincreases una single probowel wall of colon cantions colonmain typesbut not by hyperplastimal morphthe basemetendency toplastic, has

    epithelial cells are observed; the nuclei of the epithelial cells arelarger than normal, and their position within the cells is often aber-rant. Crypts crowd together in kaleidoscopic pattern. As adenomasgrow in size, they become more dysplastic. They are also more likely

    tain cryps advely troun

    or ts ares; theyps a

    How obseina

    s ares, cae fouarcinmor

    secuing mand/omor enes tatedfrequ

    stagetic cnetiche ehe ea

    themas

    freqas as a

    syste wh

    ivatoiptio

    sevenes eP-7)/rowors, 1. Transcr

    [14]mulaons AS, Bl, an. K-Ras a

    d in growgene. Losr stimnoma

    coloro o

    ffect tween the sexes. Two main factors are considered inis of this cancer: (i) inammation and high fat diet [1],ases the number of lesions in DNA and induces muta-ical genes; and (ii) genetic predispositions.ation is associated with the release of large amountsoxygen and nitrogen species [2] leading to oxidationcids, proteins and lipids, and induction of several pro-DNA lesions.tic predisposition to CRC may be connected to the de-ismatch repair system (MMR), which causes mutatorof intestine cells (manifesting itself as microsatellitesMSI) and development of hereditary non-polyposis

    noma HNPCC (or Lynch syndrome). They may alsoed with the deciency of the APC (adenomatous poly-gene product, which functions are related to Wntathways and migration of intestine epithelium. Lacktein leads to familial adenomatous polyposis (FAP), aed syndrome characterized by multiple adenomatousch usually appear in the second and third decades ofobability of malignant transformation of one of theose to 100% [3]. A subset of patients present an atten-

    (AFAP) characterized by fewer lesions (

  • 84 B. Tudek, E. Speina / Mutation Research 736 (2012) 82 92

    Fig. 1. Aberra t opemethylene blu ); (B and eosin (ma

    extent of DNSection 5).

    3. Oxidativ

    The majcolon cancewhich is dewith insufdefence [15DNA damaggression of for the devconsumptiostimulate th(LPO) [1,19moderate cical processall compon

    3.1. Inamm

    Epidemiare linked of inammainammatioand RNS [2dation of pounsaturatedare relativechain reacti2-alkenals (2-alkenals ( HNE) andoxal, 4-oxois tremendoseveral of th

    Reactiveteins [22],

    tivesepox

    prorogeammlooxyserv

    tissuheliat in aultiary

    overatic ropotion nt crypt foci. (A) A view from the lumen site of intact mouse colon. The colon was cue. The mucosal side was observed under the light microscope (magnication 100gnication 1000).

    A damage, and potential of gaining new mutations (see

    e stress in the development of colon cancer

    ority of pathological factors favoring development ofr involve induction of oxidative stress in the colon,ned as overproduction of oxygen species combinedciency in protective mechanisms of anti-oxidative,16]. Oxidative stress is involved in the production ofe and mutations associated with the initiation or pro-human cancers [17,18]. The main pathological factorselopment of CRC are inammation, fat metabolism,n of meat and alcohol and tobacco smoking. All thesee release of ROS and RNS as well as lipid peroxidation]. These compounds play a dual role in cells. At low or

    derivaity of than toof hyding inas cycwas obcancerin epitbut noMin (mmammCOX-2enzymbeen ppromooncentrations they are mediators of specic physiolog-es, whereas high concentrations can be detrimental toents of the cell.

    ation

    ological studies suggest that inammatory processeswith CRC. This is true particularly in the associationtory bowel disease and colorectal cancer [20]. Duringn activated macrophages release large amounts of ROS

    ], which oxidize nucleic acids, proteins and lipids. Oxi-lyunsaturated fatty acids generates reactive aldehydes

    in , -positions which, in contrast to ROS and RNS,ly stable and can diffuse throughout the cell [19]. Shortve aldehydes are classied into the following families:e.g., acrolein, crotonaldehyde, 2-hexenal), 4-hydroxy-e.g., 4-hydroxy-2-hexenal HHE, 4-hydroxy-2-nonenal

    ketoaldehydes (e.g., malondialdehyde MDA, gly--2-nonenal ONE) [21]. Multiplicity of LPO productsusly augmented by the existence of stereoisomers ofese compounds.

    aldehydes either react directly with DNA and pro-or undergo further oxidation to more reactive epoxy

    suppressionsis [2931]metabolismtion of heptare highly pathways cmutations oof COX-2 byence appea[33,34].

    3.2. Consum

    Epidemihigh intakeincreased rsuggest thaof colonic heme oxyginhibitor, zia comet asstion [35]. TThe preincn along the longitudinal median axis, formalin xed and stained withand C) cross sections of mouse epithelium stained with hematoxylin

    , such as 2,3-epoxy-4-hydroxynonanal [23]. The afn-idized LPO products to nucleic acids is much higherteins. Epoxidation of enals may occur in the presencen peroxide, but is also catalysed by enzymes produc-atory mediators, prostaglandins and leukotrienes, suchgenase-2 (COX-2) and lipooxygenases (LOX) [24]. Ited that COX-2 is overexpressed in 80% of colorectales [25,26]. Increased levels of COX-2 were observedl cells, inammatory cells, as well as in stromal cells,djacent normal tissues [27]. COX-2 overexpression inple intestinal neoplasia) mice was sufcient to inducetumors [28]. The mechanism of tumor induction byexpression is complex, and involves the activity of itsend-products, prostanoids. A number of theories havesed to explain the role of tumor-derived prostanoids inof angiogenesis, induction of tumor cell proliferation, of immune response, and protection against apopto-. Additionally, it was shown that arachidonic acid (AA)

    mediated by COX-2 and 5-LOX results in the forma-anone-etheno (H)-DNA adducts. The H-DNA adductsmutagenic in mammalian cells suggesting that theseould be (at least in part) responsible for the somaticbserved in tumorigenesis [32]. That is why inhibition

    non-steroid anti-inammatory drugs or RNA interfer-red a successful approach in colon cancer prevention

    ption of red meat and alcohol and tobacco smoking

    ological and animal model studies have suggested that of heme, present in red meat, is associated with anisk of colon cancer. Recently elucidated mechanismst heme induces DNA damage and cell proliferationepithelial cells via hydrogen peroxide produced byenase. In Caco-2 cells the presence of heme oxygenasenc protoporphyrin, reduced DNA damage (measured byay) triggered by hemin as well as cell hyperprolifera-he effect of heme may be modied by LPO products.ubation of colon adenocarcinoma, SW480, cells with

  • B. Tudek, E. Speina / Mutation Research 736 (2012) 82 92 85

    hemoglobin (Hb) and its subsequent exposure to linoleic acidhydroperoxides (LAOOH) led to an increase in cell death, malon-dialdehyde formation, and DNA fragmentation and these effectswere related to the peroxide group and the heme present inHb. SW480level of thedeoxyguanowith Hb ledrelated witbowel bleedenhancer oquence con[36].

    Anotheris chronic contribute action of aalcohol metion of ethaand duringdeoxyguano[38]. Both tlipid peroxiZucker ratsthe liver DNwas correla[40]. Thus, odG, 1,N2-associated w

    The resuwith the riare controvAlthough inincreased tbase, 8-oxo8-oxoGua lwas observ

    3.3. DNA da

    Oxidativlogical chan[43,44]. DNneous groumolecules pA plethora genetic materties [45]. biomarker oesis [46]. T(8-oxodG) 8-OxodG isacids or capolymeraseoxidation a

    LPO prodextent as dear and cycDNA with g[1,2]purinear N6-(3-ooxopropenyM1dG is theand rodent to T and tra8-oxoGua [

    Exocyclic propano DNA adducts are formed by the attachmentof unsaturated reactive aldehyde to ring and extra-ring nitrogenatoms in DNA bases, which generates additional saturated, six-membered ring in a DNA base. These adducts are derived from the

    n of -dG

    ]. Ethed toknowtion into e or enosubs, deplic Dd exhe ine rel].ateddisead ul

    ]. Thiiolog

    air ocarci

    maj excied bmilyassicii) exal ofpatholyml DNagefromtly, 1ppinglly tunctsic (A(APE

    have3 toe. Th, 8-oxoGuorpo

    8-oua iTP todieons. DNA odG

    NA bohy-Oxod re

    corre cells treated with Hb and LAOOH presented a higher modied DNA bases 8-oxoGua and 1,N2-etheno-2-sine (1,N2-Gua) compared to the control. Incubations

    to an increase in intracellular iron levels, which cor-h DNA oxidation. Thus, Hb from either red meat oring caused by inammatory processes could act as anf fatty acid hydroperoxide genotoxicity, and in conse-tribute to the accumulation of DNA lesions in colon cells

    frequently suggested risk factor for CRC developmentalcohol consumption. One of the factors which mayto the development of alcohol-associated cancer is thecetaldehyde, the rst and most toxic metabolite oftabolism [37]. Acetaldehyde is produced by the oxida-nol by hepatic NAD-dependent alcohol dehydrogenase

    threonine catabolism. Acetaldehyde reacts with 2-sine in DNA to form 1,N2-propanodG and 1,N2-dGhese DNA lesions were described to derive also fromdation [39,36]. In hepatic cancer model, lean and obese

    higher levels of etheno DNA adducts were observed inA of alcohol-fed than in control animals. This increaseted with the induction of cytochrome P-450 isoform 2E1the formation of exocyclic DNA adducts, like propan-dG, dA and dC may be the important mechanismith acetaldehyde related cancer risk.

    lts of epidemiological studies also link tobacco smokingsk for CRC development. However, the obtained dataersial, and the molecular mechanism remains unclear.

    the leukocytes of lung cancer patients tobacco smokinghe level of one of the major oxidatively damaged DNAGua [41], in CRC patients no effect of tobacco smoking onevel in the leukocytes and 8-oxoGua urinary excretioned [42].

    mage

    ely damaged DNA has been blamed for the physio-ges associated with degenerative diseases and cancerA damage caused by free radicals is the most heteroge-p of DNA lesions due to the wide variety of deleteriousroduced in cells in the conditions of oxidative stress.

    of damaged DNA bases are formed upon ROS attack onerial, several of them reveal strong promutagenic prop-One of the major and best studied is 8-oxoGua, a typicalf oxidative stress, which may play a role in carcinogen-he presence of 8-oxo-7,8-dihydro-2-deoxyguanosineresidues in DNA leads to GCTA transversions [47].

    formed in DNA either via direct oxidation of nucleicn be incorporated from the nucleotide pool by DNAs, the latter process being an important source of DNAnd genome instability [48,49].ucts may contribute to DNA damage to nearly the sameirectly oxidized bases. There are distinct groups of lin-lic LPO specic DNA lesions. Malondialdehyde reacts inuanine, adenine and cytosine to form cyclic pyrimido-e-10(3H)-one-2-deoxyribose (M1dG) adduct and lin-xopropenyl)-2-deoxyadenosine (M1dA) and N4-(3-l)-2-deoxycytosine (M1dC) adducts, respectively [50].

    major MDA-DNA adduct and was detected in humantissues. In Escherichia coli cells, it induces transversionsnsitions to A with a frequency comparable with that of15].

    reactiodG, Cro[51,52attachis not exposiduced chloridand ethare unlengthexocycstituteWith tand ar[54,55

    Elevcolon ease an[20,56CRC et

    4. Repcolon

    Theis basedamagAlkB faIn a cltion, (removof the DNA porigina

    Damexcise Presenoverlanisticamonofan abaclease whichbonds enzymlesions

    8-Oits incexcises8-oxoGoxodGthis momutatidirect of 8-oxinto Dphosph[61]. 8dase andirect DNA with, e.g., acrolein, crotonaldehyde and HNE. Acr- and HNE-dG were found in rodent and human tissueseno DNA adducts posses ve-membered exocyclic rings

    DNA bases. The exact pathway of their formation in vivon, although it is clear that they are generated by the

    of DNA to lipid peroxides. Ethenoadducts are also intro-DNA by certain environmental carcinogens like vinylits metabolite, chloroacetaldehyde (CAA) [53]. Propano--type adducts that are found in native mammalian DNAtituted and substituted with side chains of differentending on the structure of reacting LPO product. AllNA adducts have strong miscoding potential. Unsub-ocyclic DNA adducts are more mutagenic than toxic.crease of the side chain the toxic properties increaseated to the inhibition of replication and transcription

    levels of dA and dC were found in pro-carcinogenicses, like inammatory bowel disease, Crohns dis-cerative colitis, and in addition in chronic pancreatitiss suggests that LPO-derived DNA lesions contribute toy.

    f oxidative DNA damage as a driving force tonogenesis

    or pathway for repair of oxidatively derived DNA lesionssion repair system (BER). Although several oxidativelyases are also repaired by mismatch repair system and

    dioxygenases, this review will focus on BER activity.al view BER consists of ve steps: (i) lesion recogni-cision of the damaged base, (iii) AP site incision and

    the chemical residues that could block further stepsway, (iv) incorporation of the proper nucleotide(s) byerases and (v) ligation of the DNA strand to restore theA molecule [5760].d bases are recognized by DNA glycosylases, which

    DNA a wide range of oxidized and alkylated bases.3 human DNA glycosylases with different, but partially

    substrate specicity have been identied. Mecha-wo types of DNA glycosylases can be distinguished,ional enzymes, that excise damaged base leaving behindP) site, which is subsequently cleaved by AP endonu-1) 5 to the AP site, and bifunctional DNA glycosylases,

    endowed AP lyase activity that incises phosphodiester the AP site created by the glycosylase activity of theis review will focus on repair of three oxidative DNAxoGua, Ade and Cyt.a is repaired by a three-step system, which (i) lim-

    ration of 8-oxodGTP into DNA during replication, (ii)xoGua from DNA, and (iii) corrects the base opposite ton DNA. Different DNA polymerases can incorporate 8-

    the DNA, and replicative DNA polymerases incorporated nucleotide usually opposite dA, generating A:T C:GApproximately a half of 8-oxodG in DNA arises from theoxidation, and the other half is a result of incorporationTP during DNA synthesis. Incorporation of 8-oxodGTPy DNA polymerases is limited by the activity of MTH1drolase, which hydrolyzes 8-oxodGTP to 8-oxodGMPdGMP is subsequently dephoshorylated by nucleoti-moved from the cell [62]. Many observations indicate alation between 8-oxodG formation and carcinogenesis

  • 86 B. Tudek, E. Speina / Mutation Research 736 (2012) 82 92

    in vivo [46,6366]. The second line of defence is glycosylase MutY(MYH) that removes adenine paired with 8-oxoGua. Activity of thisenzyme prevents mutations G:C T:A, when dATP is incorporatedopposite 8-oxoGua during replication [67]. After removal of Adeby MYH glycosylase, repair DNA polymerase incorporates dCMPopposite 8-ponent of thlyase is theDNA, but onactivity of tthe glycosyreplication which excitively [72].induced by is induced the pool of and OGG1 gto colon canof several m(exon 7) andA:8-oxodGquency. The[73].

    Several described, wphic Cys326activity, bocleotides [7from -irra[74,76]. It walleles mayryngeal cancancer wasa decreasehead and nrate and siin cellular the acquisiSuch an idpro-oxidancancer.

    Ade is ewhereas Cexcises thy5-methylcystranded uglycosylaseislands [88,

    ANPG gbesides Ahypoxanthimethylguanwith guaninext enzymTDG and A[9295].

    Ethenocon the levelbut also on primary subCyt, and aallow its exThis may edifferent hu[19].

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

    mR

    CRC

    1.2(0.7n = 12.(4.0n = 7.9(1.8n = 88.(42n = 86.(5.1n =

    fromvely.cts wpariso

    CRC lues a

    pair a an

    rderpair n caof diped rs of nd pease

    AD leveidant

    bloo to hd plaeasin

    by the fact that urinary level of 8-oxodG was augmented in AD patients, but 8-oxoGua level was enhanced only inut not in AD individuals [42]. Urinary 8-oxodG may reection of nucleotide pool, while 8-oxoGua reects rather oxi-

    of DNA. Since nucleotide pool is much more susceptible toion than dsDNA, 8-oxodG in urine may be a more sensitiveor of oxidative processes in the body, and shows that evenindividuals the oxidative processes are enhanced. The sug-

    mechanisms responsible for the oxidative stress in cancerts include [43]: (i) granulocyte activation with release of ROS;ulation of cytokines, of which some, e.g., tumor necrosis

    produce large amounts of ROS; (iii) production of hydrogende by malignant cells, which in advanced stages of cancere released into the blood stream and penetrate into other

    [2].restingly, we also observed an increase of mRNA levels ofenzymes, OGG1, APE1 and MTH1 in leukocytes of CRC andividuals (Table 1) [42]. Expression of APE1 and OGG1 genesown to be induced by ROS [97], and this could explain induc-

    repair enzymes transcription in leukocytes of CRC and ADuals. On the other hand induction of repair enzymes may beof a carcinogenic program, which might decrease apoptosisoxodG. Then 8-oxoGua can be excised by the third com-e system, OGG1 glycosylase [68]. OGG1 glycosylase/AP

    major enzyme catalyzing excision of 8-oxoGua fromly when paired with Cyt or Thy [69,70]. The AP lyasehe OGG1 protein is about 10-fold lower than that oflase [71]. When 8-oxodGTP is included into DNA duringit is probably removed by NEIL1 or NEIL2 glycosylases,ses 8-oxoGua paired with dC/G/T/A or dC/A, respec-

    This complex three-tier system prevents mutationsthe presence of 8-oxodG in DNA, both when this lesiondirectly in the DNA, and when it is incorporated fromnucleotides during replication. Polymorphisms of MYHlycosylases are related to modulation of susceptibilitycer. MYH variations signicantly increase CRC risk, andutations described, two are the most prevalent: Y165Cd G382D (exon 13). Mutated enzyme excises Ade from

    pair with a decreased rate, increasing mutation fre- association of MYH with CRC was rst reported in 2002

    polymorphic changes in the OGG1 gene have beenith the most common being Ser326Cys [74]. Polymor-

    OGG1 protein was found to have a lower enzymaticth when 8-oxoGua was excised from oligodeoxynu-5], and when 8-oxoGua and FapyGua were liberateddiated DNA by puried Cys326 or Ser326 OGG1 enzymeas suggested that the presence of two OGG1 326Cys

    confer an increased risk of lung, prostate and nasopha-cer [7779], but no association with the risk for colon

    found [80,81]. Our and other functional studies report in 8-oxoGua excision rate in lung [41,82,83] andeck cancer patients [84]. Such a decrease in excisionmultaneous increase in 8-oxodG and FapydG levelsDNA favors a pro-oxidant state and may acceleratetion of mutations in critical genes leading to cancer.ea is basically derived from the observation that at environment is characteristic for advanced stages of

    xcised by alkylpurine-DNA-N-glycosylase (ANPG) [85],yt by thymine-DNA glycosylase (TDG); the latter alsomine from G:T pairs, resulting from deamination oftosine [86]. Ethenocytosine is also excised by single-racil DNA glycosylase, SMUG1 [87] and by MBD4, although activity of the latter is strongly limited to CpG89].lycosylase has a wide substrate specicity, andde excises from DNA also 1,N2-ethenoguanine [90],ne and the alkylated bases, 3-methyladenine and 7-ine [85]. TDG excises Cyt, Thy and Ura from pairsne, and its activity is strongly stimulated by thee in the BER pathway, AP endonuclease, APE1 [91].PE1 also play the role of transcription regulators

    ytosine repair by the BER system is dependent not only and activity of TDG, MBD4 or SMUG1 DNA glycosylases,the presence and/or activity of ANPG glycosylase, whichstrate is Ade. ANPG binds strongly to DNA containinglthough does not excise this modied base, does notcision by the proper Cyt glycosylase, like TDG [96].xplain observation that the level of Cyt measured inman tissues was constantly higher than that of Ade

    OGG1

    ANPG

    TDG

    APE1

    MTH1

    AdaptedrespectiAll subje

    a Combetween

    b p va

    4.1. Readenom

    In oand reof colostages develomarkeDNA, a

    Incrand inoxodGantioxacid inparisonin bloois incrportedalreadyCRC, boxidatdationoxidatindicatin AD gestedpatien(ii) stimfactor,peroximay btissues

    Interepair AD indwas shtion ofindivida part NA level (arbitrary units)a p

    patients (C) AD patients (A) Controls (H)

    8 0.99 0.19

  • B. Tudek, E. Speina / Mutation Research 736 (2012) 82 92 87

    A

    50

    60

    70

    80

    90

    100

    * * *

    CRC patients

    Controls

    n = 68

    B

    0

    10

    20

    30

    40 n = 90

    8-o

    xo

    Gu

    a e

    xcis

    ion

    activity

    (p

    mo

    ls/h

    /mg

    pro

    tein

    )

    100

    110

    120

    *

    * * * CRC patients Controls

    1

    2

    3

    4

    5

    6

    7

    8

    90

    8-o

    xo

    Gu

    a e

    xcis

    ion

    activity

    (pm

    ols

    /h/m

    g p

    rote

    in)

    Fig. 2. The meof CRC patientdistinguished p < 0.005; *, p 0.0000 (C vs. H)33 (34), (18)d >0.079 (A vs. H)1 (1), (1)e >0.0000 (C vs. A)

    0.820.18

    e distribution analysis).

  • 88 B. Tudek, E. Speina / Mutation Research 736 (2012) 82 92

    40

    45

    50 * * * a

    ctivity

    g p

    rote

    in)

    CRC pati ents

    C ontrols

    AE

    xcis

    ion

    (pm

    ols

    /h/

    B

    Excis

    ion

    activity

    (pm

    ols

    /h/

    g p

    rote

    in)

    Fig. 3. (A) Thleukocyte DNA(MannWhitnexcision rate ip < 0.001 (Wilc

    Derived from [

    compared t(Fig. 3A) [1Ade and asymptomaCRC patientin controls,to be lowergests that thmultifactortissues.

    The lowpatients wagenes nor tTDG glycoscontrary, mhigher in PThus directby inammmore likelyshown to bgen peroxidAs shown fosistent oxidalready dur[111]. A nuof several robserved loous lower l

    may thus suggest that other mechanisms are up-regulated in CRCpatients and determine the level of etheno adducts in DNA. Otherrepair systems engaged in etheno adducts elimination from DNA

    e, e.g., oxidative demethylation by AlkB family proteins ortide ntereyt erounes ltudis thanive c,116]betw3) [er, dl typeA levent wer fordernd c colylasetissur APly alC geay inownN1 enPC ex25

    30

    35 n = 56

    54=

    0

    5

    10

    15

    20

    n = 56n = 54

    *

    Ade Cyt

    n

    14

    16

    n = 54

    * * * Tumor Normal colo n

    0

    2

    4

    6

    8

    10

    12

    n = 54

    n = 54

    n = 54

    * * *

    Ade Cyt

    e mean and standard deviation of Ade and Cyt excision rate in

    includnucleo

    Of iand Cthe sur23 timother stumorulceratsis [56found p = 0.01howevseverathe dconsistbe high

    In oAde atomatiglycoscolon only foprobabthe APtein mwas shand FEwith A of CRC patients and control individuals. ***, p < 0.001; *, p < 0.05ey U test). (B) The mean and standard deviation of Ade and Cytn colon tumor and normal surrounding tissues of CRC patients. ***,oxon rank sum test).

    105] with permission from Elsevier.

    o controls matched for age, sex and a smoking habit05]. However, in colon tissues, excision activities forCyt were signicantly higher in tumor than in adjacent,tic colon tissue (Fig. 3B) [105]. In leukocytes (PBL) ofs dA and 1,N2-dG level was unexpectedly lower than

    and the level of dA and dC also showed a tendency in tumor than in normal colon tissue [105]. This sug-e regulation of etheno adduct level and repair may be

    ial and may differ in target (colon) and surrogate (PBL)

    er excision rate for Ade and Cyt in leukocytes of CRCs not linked to mutations in exons of ANPG and TDGo down regulation of mRNA expression of ANPG andylases or its BER activator, APE1 endonuclease; in theRNA levels of these repair enzymes were signicantlyBL of CRC than in healthy controls (Table 1) [42,105].

    inactivation/inhibition of repair enzymes activitiesatory mediators released in the blood stream is a

    explanation. Indeed, several DNA repair enzymes weree directly inhibited by nitric oxide [106108], hydro-e [109] and LPO product, 4-hydroxynonenal [55,110].r several human cancer sites and animal tumors, per-ative stress and LPO-induced DNA damage increaseding premalignant stages in a time-dependent mannermber of studies demonstrated increase of mRNA levelepair enzymes upon oxidative stress [97,112,113]. Thewer Ade and Cyt excision (Fig. 3A) and simultane-evel of etheno adducts in PBL of cancer patients [105]

    In conclactivity of compared tings, that tPBL from Cmechanistiticularly Cstages in thin critical gsylase remoinduction odeaminatio

    5. Aberranmutations

    Activity naling moleAPC and TP5repair rate events withBER proteinlation of ge

    Mutatioesis, and areThe most frpart of the with DNA psuggested, Aproteins onBER (Fig. 4)protein an aactivity of Aexcision repair [114,115].st are the ndings in tumorous colon tissue. The Adexcising activities were much higher in tumor than inding normal colon (Fig. 3B) and was consistent with aower dA and dC adduct levels [105]. Also in severales lower level of dA and dC was observed in colon

    in colon tissues of cancer prone patients suffering fromolitis, Crohns disease or familial adenomatous polypo-. In our study in normal colon a negative correlation waseen dC level in DNA and its excision rate ( = 0.64,105]. This supports a role of BER in Cyt-elimination;oes not exclude other etheno adduct control systems. Ins of non tumorous human tissues previously analyzed,el in DNA was found to be lower than that of dC [111],ith our observation that excision activity was found tor Ade than for Cyt (Fig. 3) [105].

    to understand a huge increase of excision activity ofCyt in colon tumor tissue in comparison to asymp-on margin, we measured mRNA level of ANPG and TDGs, as well as APE1 endonuclease in tumor and normales. Higher mRNA level in tumor tissue was observedE1 [105]. APE1 stimulates Cyt excision [91], and mostso Ade. Another possibility are frequent mutations inne in colon tumors [117]. APC tumor suppressor pro-hibit BER engaged in repair of Ade and Cyt since it

    that it can directly interact with DNA polymerase donuclease [118]. BER activity was inversely associatedpression in several breast cancer cell lines [119].usion, we have shown, for the rst time, that excisionAde and Cyt is lower in PBL of CRC patients wheno healthy individuals. However, our unexpected nd-he level of dA and 1,N2-dG was lower in DNA ofRC patients than in healthy controls currently remainscally difcult to explain. If such etheno adducts, par-yt excision deciency also occurs during premalignante target organ, still may favor acquisition of mutationsenes leading to neoplastic transformation. TDG glyco-ves Thy and Ura paired with guanine, and may preventf mutations caused by cytosine and 5-methylcytosinen [120].

    t repair of oxidatively damaged DNA bases andin critical genes

    and expression of BER proteins is modulated by two sig-cules, which are frequently mutated in colon cancer, the3 gene products. Thus, mutations in these genes affectof oxidatively damaged DNA, and favor pro-mutagenicin colon epithelial cells. On the other hand at least threes, namely, APE1, TDG and OGG1 take part in the regu-ne expression.ns in the APC gene are early events in colon carcinogen-

    found in about 80% of colon adenomas and carcinomas.equent mutation results in truncation of the C-terminalAPC protein. Wild type APC protein directly interactsolymerase , FEN1 endonuclease [118], and as recentlyPE1 endonuclease [121] inhibiting the assembly of BER

    damaged DNA and the activity of short and long patch. In LoVo, colon cancer cells expressing truncated APCccelerated assembly of BER proteins, as well as higherPE1, FEN1 and Pol were observed [121]. Activation of

  • B. Tudek, E. Speina / Mutation Research 736 (2012) 82 92 89

    APE1/

    ANPG

    +

    Genome

    instability Progression

    +

    Genome

    instabilityProgression

    Fig. 4. The in me stactivity and tr nced BAPE1 activity APE1,interacts with scripregulated by c erationand may favor ted anin circle denot e ; Falkyladenine D sylasedeaminase; HI

    APE1 may which are about 50% oof APE1 tra[122]. On tby TP53 inh[123]. ThusAPE1 amou

    Increasepathway, asion: in inpatients (atDNA adducincreased, arelated witSimilarly, inAPE1 was a

    APE1, beregulatory participatesstages of Caccumulatiinstability wgrowth (Fig

    IncreaseDNA glycossusceptiblesites, and ADNA [91,12tion of promto hypomeinduction odeaminase Methylcytoby AID to tsubsequentfrom dT:dGreconstitutirecruiting pis engaged

    pmelso d-uoeatmf mo

    rom S-ph, exci

    G1 ayc tationhylatdemehylat

    the G, OlymPol FEN1

    APE1

    +TP53

    APC

    mutated +

    +

    TDG/MBD4

    OGG1

    mutated +

    ++

    HIF-1Angiogenesis

    Erythropoesi s

    uence of APC and TP53 genes mutations on the activity of BER enzymes and genoanscription, respectively, and up-regulate BER. Increased APE1, ANPG and unbalamay also accelerate the excision rate of DNA glycosylases, OGG1 and TDG/MBD4.

    the deaminase AID and may favor demethylation of promoters and induction of tran-Myc and estrogen receptors, and thus may contribute to acceleration of cell prolif

    genomic instability and cancer progression. APE1 also participates in HIF-1 mediaes stimulation or increase; APC, adenomatous polyposis coli; Pol , DNA polymerasNA N-glycosylase; OGG1, 8-oxoGua DNA N-glycosylase; TDG, thymine DNA N-glycoF-1, hypoxia-inducible factor 1 subunit.

    also occur as a consequence of TP53 gene mutations,late, but frequent events in colon carcinogenesis (inf cancers). TP53 protein participates in downregulationnscription in cells treated with DNA damaging agentshe other hand TP53 mediates ubiquitination of APE1ibitor, MDM2, and in consequence APE1 degradation, mutations in TP53 gene should additionally increasent in cancer cells.d APE1 activity may result in unbalancing of BERnd may favor genomic instability and cancer progres-amed non-cancerous colon tissues of ulcerative colitis

    high risk for colon cancer, that accumulate high ethenots [124]), ANPG and APE1 activities were signicantlynd microsatellite instability (MSI) was positively cor-h their imbalanced activities of repair enzymes [125].

    yeast and human cells over-expression of ANPG and/orssociated with frameshift mutations and MSI [125].sides DNA repair function, also plays a role of redox

    develomay awith 5CRC trtance o5-FU fdelaysHence[129].

    OGby c-MaccelerdemetLSD1 demetaround8-OxodRNA poprotein for several transcription factors. For example, in HIF-1 mediated angiogenesis [126]. Thus at lateRC development, mutations in TP53 gene will lead toon of APE1 protein, and in addition to increasing genomeill also favor angiogenesis and in consequence tumor

    . 4).d APE1 activity may also accelerate the excision rate ofylases. TDG and OGG1 were shown to be particularly

    to induction by APE1, since they bind strongly to APPE1 increases turnover of these enzymes on damaged7]. Increased activity of TDG may favor demethyla-oters of several genes and in consequence contribute

    thylation of DNA observed in colon cancer [13] andf transcription of several genes. TDG interacts with theAID and the damage response protein GADD45a. 5-sine and 5-hydroxymethylcytosine are rst deaminatedhymine and 5-hydroxymethyluracil, respectively, andly TDG removes thymine and 5-hydroxymethyluracil

    or 5OHMedU:dG pairs, which initiates BER and enableson of G:C base pairs [128]. TDG is also necessary for300 to retinoic acid (RA)-regulated promoters, andin transcriptional control of genes during embryonic

    gets and stiOGG1 or Athat regulaing loops thloci [132]. Fin histone the downstget chromachromatin bthat bring tregulation o

    6. Conclus

    Oxidativcarcinogenand AD paexcision retranscriptiosues. Activinetwork, winactivationAID-media ted

    demethyla tion of

    gene promoter s

    Transcription of

    several gene s

    Transcriptio n of

    Myc-regulated

    genes

    Prolifera tio n

    Progression

    ability. Mutations in APC and TP53 proteins lead to increase in APE1ER pathway may favor genomic instability and cancer progression.

    TDG and OGG1 take part in the regulation of gene expression. TDGtion of several genes. OGG1 and APE1 stimulate transcription of genes. Increased APE1 activity may result in unbalancing of BER pathway,giogenesis and erythropoesis and may favor tumor growth. + symbolEN 1, ap endonuclease 1; APE1, apurinic site endonuclease 1; ANPG,; MBD4, methyl-CpG binding domain protein; AID, activation-induced

    nt. Its deciency is embrionaly lethal [120]. TDG statusetermine sensitivity of CRC patients to chemotherapyrouracil (5-FU), a chemotherapeutic routinely used inent. Inactivation of TDG signicantly increases resis-use and human cancer cells toward 5-FU. Excision ofDNA by TDG generates persistent DNA strand breaks,ase progression, and activates DNA damage signaling.sion of 5-FU by TDG mediates DNA-directed cytotoxicity

    nd APE1 stimulate transcription of genes regulatedranscription factor [130], and thus may contribute to

    of cell proliferation (Fig. 4). The mechanism involvesion of histone H3 by specic FAD-containing enzyme,thylase which generates hydrogen peroxide duringion [131]. In consequence a local oxidation of DNAE-boxes and TSS region of Myc target genes is triggered.GG1 and APE1 were found to be engaged on the sameerase II and Myc occupied chromatin regions of Myc tar-

    mulate transcription of these genes. Silencing of eitherPE1 inhibits Myc mediated transcription. It is possibletory regions in a gene interact with each other form-at often bridge considerable distances on the genomicollowing Myc binding local demethylation of lysine 4H3 (H3K4me2) triggers recruitment of the OGG1 andream BER enzymes thus producing nicks on Myc tar-tin. This may relax the DNA strands and could favorending necessary for the formation of chromatin loopsogether RNAPII and Myc. OGG1 is also engaged in thef transcription from estrogen promoters [133].

    ions

    e stress is induced at the early, adenoma, stage of colonesis, and is increasing during disease progression. CRCtients reveal a huge increase of mRNA level of basepair enzymes in blood leukocytes. However, increasedn is not always correlated with repair capacity of tis-ties of BER enzymes remain under a complex regulativehich besides stimulation of transcription includes direct

    by oxidation stress products, interaction with other

  • 90 B. Tudek, E. Speina / Mutation Research 736 (2012) 82 92

    BER proteins, and with signaling molecules, which are frequentlymutated in colon cancer, that is APC and TP53 proteins. Mutatedproteins do not inhibit transcription and/or activity of base exci-sion repair enzymes. This increases the amount of repair enzymeswithin cellsDNA damagand may be

    Conict of

    The auth

    Funding

    Funded We thank al

    References

    [1] F. Levi, colorect

    [2] M. Dizdbases inphonuc

    [3] G. BougSiproudciated wColon R

    [4] S. Jonesson, J.P.colorect(2002) 2

    [5] V. GismFornasaP.L. Brumutatiocoli and

    [6] B.R. Berspectru

    [7] B. Tudekof oxidaJ. Transl

    [8] M.S. Coto DNA

    [9] S. Maynexcisionaging, C

    [10] S.E. Kentwo cas

    [11] B. Tudeand rats12361

    [12] B.A. Magpredict 44994

    [13] E.R. Fea(2011) 4

    [14] M. van K. van dMoerer,The betcolorect

    [15] M. Valkand ant(2006) 1

    [16] M. Valkand antBiochem

    [17] L.A. Loeb2556.

    [18] K.B. Bec19633

    [19] H. Bartslesions 38539

    [20] J. PotackLiver 2 (

    [21] K. UchidProg. Lip

    [22] F.L. Chung, R.G. Nath, M. Nagao, A. Nishikawa, G.D. Zhou, K. Randerath,Endogenous formation and signicance of 1,N2-propanodeoxyguanosineadducts, Mutat. Res. 424 (1999) 7181.

    [23] F.L. Chung, J. Pan, S. Choudhury, R. Roy, W. Hu, M.S. Tang, Formation of trans-4-hydroxy-2-nonenal- and other enal-derived cyclic DNA adducts from omega-and ouman .J. Cheid hy631

    .E. EbeuBois,lorec831. Sano. Katolorec.J. Thutican

    ancer .H. LiuF. Lanmorig. Wan.A. Inycloox(2003. Greeraske

    canc (200. Speee, Can.S. Redcloox516

    . StrilNAi-biotech

    Ishikae and

    roducencer, . Angi, Masamage.K. Sei

    (38.C. Gari Mascation 3 (20

    Ederopanohydronow. Wan.K. SeiNA les. Gackciore

    possi994

    Obtulnik, B. Szpilhanc347

    . OlinsNA daevelop. Olinsxidatisis, an002) 1.D. Evductio

    JarugNA baBS Le.C. Cheundations,.T. Ruruich

    eoxyn and introduces imbalance in the repair of oxidativee. Such imbalance causes increased genome instability

    a driving force in the disease progression.

    interest statement

    ors declare that there are no conicts of interest.

    by the Polish-French grant project 346/N-INCA/2008/0.l patients and volunteers who participated in this study.

    C. Pasche, C. La Vecchia, F. Lucchini, S. Franceschi, Food groups andal cancer risk, Br. J. Cancer 79 (1999) 12831287.aroglu, R. Olinski, J.H. Doroshow, S.A. Akman, Modication of DNA

    chromatin of intact target human cells by activated human polymor-lear leukocytes, Cancer Res. 53 (1993) 12691272.uen, S. Manfredi, M. Blayau, C. Dugast, B. Buecher, D. Bonneau, L.his, V. David, J.F. Bretagne, Colorectal adenomatous polyposis asso-ith MYH mutations: genotype and phenotype characteristics, Dis.

    ectum 50 (2007) 16121617., P. Emmerson, J. Maynard, J.M. Best, S. Jordan, G.T. Williams, J.R. Samp-

    Cheadle, Biallelic germline mutations in MYH predispose to multipleal adenoma and somatic G:C T:A mutations, Hum. Mol. Genet. 119612967.ondi, M. Meta, L. Bonelli, P. Radice, P. Sala, L. Bertario, A. Viel, M.rig, A. Arrigoni, M. Gentile, M. Ponz de Leon, L. Anselmi, C. Mareni,zzi, Varesco prevalence of the Y165C, G382D and 1395GGA germlinens of the MYH gene in Italian patients with adenomatous polyposis

    colorectal adenomas, Int. J. Cancer 109 (2004) 680684.quist, D.M. Wilson, 3rd pathways for repairing and tolerating them of oxidative DNA lesions, Cancer Lett. (2012)., A. Winczura, J. Janik, A. Siomek, M. Foksinski, R. Olinski, Involvementtively damaged DNA and repair in cancer development and aging, Am.. Res. 2 (2010) 254284.oke, R. Olinski, M.D. Evans, Does measurement of oxidative damagehave clinical signicance? Clin. Chim. Acta 365 (2006) 3049.ard, S.H. Schurman, C. Harboe, N.C. de Souza-Pinto, V.A. Bohr, Base

    repair of oxidative DNA damage and association with cancer andarcinogenesis 30 (2009) 210.t, B. Majumdar, Metastatic tumours in the maxillary sinus. A report ofes and a review of the literature, J. Laryngol. Otol. 99 (1985) 459462.k, R.P. Bird, W.R. Bruce, Foci of aberrant crypts in the colons of mice

    exposed to carcinogens associated with foods, Cancer Res. 49 (1989)240.nuson, I. Carr, R.P. Bird, Ability of aberrant crypt foci characteristics tocolonic tumor incidence in rats fed cholic acid, Cancer Res. 53 (1993)504.ron, Molecular genetics of colorectal cancer, Annu. Rev. Pathol. 679507.

    de Wetering, E. Sancho, C. Verweij, W. de Lau, I. Oving, A. Hurlstone,er Horn, E. Batlle, D. Coudreuse, A.P. Haramis, M. Tjon-Pon-Fong, P.

    M. van den Born, G. Soete, S. Pals, M. Eilers, R. Medema, H. Clevers,a-catenin/TCF-4 complex imposes a crypt progenitor phenotype onal cancer cells, Cell 111 (2002) 241250.o, C.J. Rhodes, J. Moncol, M. Izakovic, M. Mazur, Free radicals, metalsioxidants in oxidative stress-induced cancer, Chem. Biol. Interact. 16040.o, D. Leibfritz, J. Moncol, M.T. Cronin, M. Mazur, J. Telser, Free radicalsioxidants in normal physiological functions and human disease, Int. J.. Cell Biol. 39 (2007) 4484., Cancer cells exhibit a mutator phenotype, Adv. Cancer Res. 72 (1998)

    kman, B.N. Ames, Oxidative decay of DNA, J. Biol. Chem. 272 (1997)19636.ch, J. Nair, Oxidative stress and lipid peroxidation-derived DNA-in inammation driven carcinogenesis, Cancer Detect. Prev. 28 (2004)1., S.H. Itzkowitz, Colorectal cancer in inammatory bowel disease, Gut2008) 6173.a, 4-Hydroxy-2-nonenal: a product and mediator of oxidative stress,id Res. 42 (2003) 318343.

    3 h

    [24] Hac30

    [25] CDco11

    [26] HHco

    [27] ManC

    [28] CT.tu

    [29] D[30] M

    C9

    [31] APaof30

    [32] Nag

    [33] Bcy15

    [34] ARB

    [35] S.agpca

    [36] J.PDd

    [37] H30

    [38] CDm13

    [39] E.p4-su

    [40] YHD

    [41] DPaas48

    [42] T.JaAen46

    [43] RDd

    [44] ROro(2

    [45] Min

    [46] P.DFE

    [47] Kabtu

    [48] MFudmega-6 polyunsaturated fatty acids and their roles in DNA repair andp53 gene mutation, Mutat. Res. 531 (2003) 2536.n, F.L. Chung, Epoxidation of trans-4-hydroxy-2-nonenal by fattydroperoxides and hydrogen peroxide, Chem. Res. Toxicol. 9 (1996)2.rhart, R.J. Coffey, A. Radhika, F.M. Giardiello, S. Ferrenbach, R.N.

    Up-regulation of cyclooxygenase 2 gene expression in humantal adenomas and adenocarcinomas, Gastroenterology 107 (1994)188., Y. Kawahito, R.L. Wilder, A. Hashiramoto, S. Mukai, K. Asai, S. Kimura,, M. Kondo, T. Hla, Expression of cyclooxygenase-1 and -2 in humantal cancer, Cancer Res. 55 (1995) 37853789.n, S.J. Henley, C. Patrono, Nonsteroidal anti-inammatory drugs ascer agents: mechanistic, pharmacologic, and clinical issues, J. Natl.Inst. 94 (2002) 252266., S.H. Chang, K. Narko, O.C. Trifan, M.T. Wu, E. Smith, C. Haudenschild,e, T. Hla, Overexpression of cyclooxygenase-2 is sufcient to induceenesis in transgenic mice, J. Biol. Chem. 276 (2001) 1856318569.g, R.N. Dubois, Prostaglandins and cancer, Gut 55 (2006) 115122.iguez, A. Rodriguez, O.V. Volpert, M. Fresno, J.M. Redondo,ygenase-2: a therapeutic target in angiogenesis, Trends Mol. Med.) 7378.nhough, H.J. Smartt, A.E. Moore, H.R. Roberts, A.C. Williams, C.va, A. Kaidi, The COX-2/PGE2 pathway: key roles in the hallmarkser and adaptation to the tumour microenvironment, Carcinogenesis9) 377386.d, I.A. Blair, Cyclooxygenase- and lipoxygenase-mediated DNA dam-cer Metastasis Rev. 30 (2011) 437447.dy, C.V. Rao, Novel approaches for colon cancer prevention byygenase-2 inhibitors, J. Environ. Pathol. Toxicol. Oncol. 21 (2002)4.lacci, C. Griffoni, M.C. Valerii, G. Lazzarini, V. Tomasi, E. Spisni,ased strategies for cyclooxygenase-2 inhibition in cancer, J. Biomed.nol. (2010) 828045.wa, S. Tamaki, M. Ohata, K. Arihara, M. Itoh, Heme induces DNA dam-

    hyperproliferation of colonic epithelial cells via hydrogen peroxided by heme oxygenase: a possible mechanism of heme-induced colonMol. Nutr. Food Res. 54 (2010) 11821191.eli, C.C. Garcia, F. Sena, F.P. Freitas, S. Miyamoto, M.H. Medeiros, P.cio lipid hydroperoxide-induced and hemoglobin-enhanced oxidative

    to colon cancer cells, Free Radic. Biol. Med. 51 (2011) 503515.tz, P. Becker, Alcohol metabolism and cancer risk, Alcohol Res. Health41) (2007) 3744.cia, J.P. Angeli, F.P. Freitas, O.F. Gomes, T.F. de Oliveira, A.P. Loureiro, P.io, M.H. Medeiros, [13C2]-Acetaldehyde promotes unequivocal for-of 1,N2-propano-2-deoxyguanosine in human cells, J. Am. Chem. Soc.11) 91409143.r, M. Wacker, P. Wanek, Lipid peroxidation-related 1,N2-deoxyguanosine-DNA adducts induced by endogenously formedxy-2-nonenal in organs of female rats fed diets supplemented wither, rapeseed, olive or coconut oil, Mutat. Res. 654 (2008) 101107.g, G. Millonig, J. Nair, E. Patsenker, F. Stickel, S. Mueller, H. Bartsch,tz, Ethanol-induced cytochrome P4502E1 causes carcinogenic etheno-ions in alcoholic liver disease, Hepatology 50 (2009) 453461.owski, E. Speina, M. Zielinska, J. Kowalewski, R. Rozalski, A. Siomek, T.k, B. Tudek, R. Olinski, Products of oxidative DNA damage and repairble biomarkers of susceptibility to lung cancer, Cancer Res. 63 (2003)902.owicz, M. Swoboda, E. Speina, D. Gackowski, R. Rozalski, A. Siomek, J.. Janowska, J.M. Ciesla, A. Jawien, Z. Banaszkiewicz, J. Guz, T. Dziaman,a, R. Olinski, B. Tudek, Oxidative stress and 8-oxoguanine repair areed in colon adenoma and carcinoma patients, Mutagenesis 25 (2010)1.ki, D. Gackowski, R. Rozalski, M. Foksinski, K. Bialkowski, Oxidativemage in cancer patients: a cause or a consequence of the diseasement? Mutat. Res. 531 (2003) 177190.ki, D. Gackowski, M. Foksinski, R. Rozalski, K. Roszkowski, P. Jaruga,ve DNA damage: assessment of the role in carcinogenesis, atheroscle-d acquired immunodeciency syndrome, Free Radic. Biol. Med. 3392200.ans, M. Dizdaroglu, M.S. Cooke, Oxidative DNA damage and disease:n, repair and signicance, Mutat. Res. 567 (2004) 161.a, T.H. Zastawny, J. Skokowski, M. Dizdaroglu, R. Olinski, Oxidativese damage and antioxidant enzyme activities in human lung cancer,tt. 341 (1994) 5964.ng, D.S. Cahill, H. Kasai, S. Nishimura, L.A. Loeb, 8-Hydroxyguanine annt form of oxidative DNA damage, causes G- - - -T and A- - - -C substi-

    J. Biol. Chem. 267 (1992) 166172.sso, M.F. Blasi, F. Chiera, P. Fortini, P. Degan, P. Macpherson, M.i, Y. Nakabeppu, P. Karran, G. Aquilina, M. Bignami, The oxidizeducleoside triphosphate pool is a signicant contributor to genetic

  • B. Tudek, E. Speina / Mutation Research 736 (2012) 82 92 91

    instability in mismatch repair-decient cells, Mol. Cell. Biol. 24 (2004)465474.

    [49] E. Speina, K.D. Arczewska, D. Gackowski, M. Zielinska, A. Siomek, J.Kowalewski, R. Olinski, B. Tudek, J.T. Kusmierek, Contribution of hMTH1 tothe maintenance of 8-oxoguanine levels in lung DNA of non-small-cell lungcancer p

    [50] L.J. MarRes. 424

    [51] A. ChedeoxyuChem. R

    [52] A. Chenformed23612

    [53] A. Barbities of 1of vinylRes. 9 (1

    [54] P. Kowaadductsbase su(2004) 3

    [55] L. Maddicz, S. KKusmieproteintrans-4-

    [56] J. Nair, FDNA adulcerati10031

    [57] P. Fortmechanpathwa

    [58] K.H. AlmdependDNA Re

    [59] M.L. Hestrand i2747.

    [60] A.B. Robcells: ba98199

    [61] C. Colusran, M. pathwapool, Cu

    [62] H. Hayaand elimmutage(1995) 8

    [63] T. TsuzuKawateIshikawMTH1 g11456

    [64] K. SakuNakabesion by

    [65] K. SakamA. EgasTsuzukistress in

    [66] Y. Xie, LindahlOgg1 reK-ras on

    [67] M.M. Slusion of Bacterio

    [68] G. Dianoof 8-oxo33811

    [69] J.P. Raditerizatiocerevisi

    [70] K. Shinmtively dRedox S

    [71] N.A. KuFedorovoxogua

    [72] S. Bjellainduced

    [73] N. Al-TWilliam

    Inherited variants of MYH associated with somatic G:C T:A mutations incolorectal tumors, Nat. Genet. 30 (2002) 227232.

    [74] V.S. Sidorenko, A.P. Grollman, P. Jaruga, M. Dizdaroglu, D.O. Zharkov, Sub-strate specicity and excision kinetics of natural polymorphic variants andphosphomimetic mutants of human 8-oxoguanine-DNA glycosylase, FEBS J.76 (20

    . Hilpairm006) . Dheramagehic alpopulat. Goommaoradi

    Chen,hism o70 (20.Y. Cho.Y. Liuoma aancer . Hansthe, Ed OGenom

    Sliwinacki, Jot assoExp. M

    Paz-ENA repst. 95Janik,. Olinsng tisnes, M

    Paz-Eani, E

    -oxogu006) . Sapad humhen p. Saparimaryd hum.S.A. 9. Kavli

    Hager remrande002) . Bellaell. PhJiricnell 135. Sap.H. Eldrimaryse an6987.V. Prirnove,N(4)-. Dempalian

    Um, Mceptoymin. Tini,f CBP/anscri. Missycosyid tra3569

    Gros, f the heroxid. Tudeol. As. Winnvril, P. Sarasatients, J. Natl. Cancer Inst. 97 (2005) 384395.nett, Lipid peroxidation-DNA damage by malondialdehyde, Mutat.

    (1999) 8395.nna, C.R. Iden, Characterization of 2-deoxycytidine and 2-ridine adducts formed in reactions with acrolein and 2-bromoacrolein,es. Toxicol. 6 (1993) 261268.na, R.A. Rieger, C.R. Iden, Characterization of thymidine adducts

    by acrolein and 2-bromoacrolein, Carcinogenesis 13 (1992)365.n, H. Bartsch, P. Leconte, M. Radman, Studies on the miscoding proper-,N6-ethenoadenine and 3,N4-ethenocytosine, DNA reaction products

    chloride metabolites, during in vitro DNA synthesis, Nucleic Acids981) 375387.lczyk, J.M. Ciesla, M. Komisarski, J.T. Kusmierek, B. Tudek, Long-chain

    of trans-4-hydroxy-2-nonenal to DNA bases cause recombination,bstitutions and frameshift mutations in M13 phage, Mutat. Res. 550348.ukuri, E. Speina, M. Christiansen, D. Dudzinska, J. Zaim, T. Obtulow-abaczyk, M. Komisarski, Z. Bukowy, J. Szczegielniak, A. Wojcik, J.T.rek, T. Stevnsner, V.A. Bohr, B. Tudek, Cockayne syndrome group B

    is engaged in processing of DNA adducts of lipid peroxidation producthydroxy-2-nonenal, Mutat. Res. 666 (2009) 2331.. Gansauge, H. Beger, P. Dolara, G. Winde, H. Bartsch, Increased etheno-ducts in affected tissues of patients suffering from Crohns disease,ve colitis, and chronic pancreatitis, Antioxid. Redox Signal. 8 (2006)010.ini, E. Dogliotti, Base damage and single-strand break repair:isms and functional signicance of short- and long-patch repair sub-ys, DNA Repair (Amst.) 6 (2007) 398409.eida, R.W. Sobol, A unied view of base excision repair: lesion-

    ent protein complexes regulated by post-translational modication,pair (Amst.) 6 (2007) 695711.gde, T.K. Hazra, S. Mitra, Early steps in the DNA base excision/single-nterruption repair pathway in mammalian cells, Cell Res. 18 (2008)

    ertson, A. Klungland, T. Rognes, I. Leiros, DNA repair in mammalianse excision repair: the long and short of it, Cell. Mol. Life Sci. 66 (2009)3.si, E. Parlanti, P. Degan, G. Aquilina, D. Barnes, P. Macpherson, P. Kar-Crescenzi, E. Dogliotti, M. Bignami, The mammalian mismatch repairy removes DNA 8-oxodGMP incorporated from the oxidized dNTPrr. Biol. 12 (2002) 912918.kawa, A. Taketomi, K. Sakumi, M. Kuwano, M. Sekiguchi, Generationination of 8-oxo-7,8-dihydro-2-deoxyguanosine 5-triphosphate, a

    nic substrate for DNA synthesis, in human cells, Biochemistry 34995.ki, A. Egashira, H. Igarashi, T. Iwakuma, Y. Nakatsuru, Y. Tominaga, H., K. Nakao, K. Nakamura, F. Ide, S. Kura, Y. Nakabeppu, M. Katsuki, T.a, M. Sekiguchi, Spontaneous tumorigenesis in mice defective in theene encoding 8-oxo-dGTPase, Proc. Natl. Acad. Sci. U.S.A. 98 (2001)11461.mi, Y. Tominaga, M. Furuichi, P. Xu, T. Tsuzuki, M. Sekiguchi, Y.ppu, Ogg1 knockout-associated lung tumorigenesis and its suppres-Mth1 gene disruption, Cancer Res. 63 (2003) 902905.oto, Y. Tominaga, K. Yamauchi, Y. Nakatsu, K. Sakumi, K. Yoshiyama,

    hira, S. Kura, T. Yao, M. Tsuneyoshi, H. Maki, Y. Nakabeppu, T., MUTYH-null mice are susceptible to spontaneous and oxidativeduced intestinal tumorigenesis, Cancer Res. 67 (2007) 65996604.H. Yang, C. Cunanan, K. Okamoto, D. Shibata, J. Pan, D.E. Barnes, T., M. McIlhatton, R. Fishel, J.H. Miller, Deciencies in mouse Myh andsult in tumor predisposition and G to T mutations in codon 12 of thecogene in lung tumors, Cancer Res. 64 (2004) 30963102.pska, W.M. Luther, J.H. Chiang, H. Yang, J.H. Miller, Functional expres-hMYH, a human homolog of the Escherichia coli MutY protein, J.l. 181 (1999) 62106213.v, C. Bischoff, J. Piotrowski, V.A. Bohr, Repair pathways for processingguanine in DNA by mammalian cell extracts, J. Biol. Chem. 273 (1998)33816.cella, C. Dherin, C. Desmaze, M.S. Fox, S. Boiteux, Cloning and charac-n of hOGG1, a human homolog of the OGG1 gene of Saccharomycesae, Proc. Natl. Acad. Sci. U.S.A. 94 (1997) 80108015.ura, J. Yokota, The OGG1 gene encodes a repair enzyme for oxida-

    amaged DNA and is involved in human carcinogenesis, Antioxid.ignal. 3 (2001) 597609.znetsov, V.V. Koval, D.O. Zharkov, G.A. Nevinsky, K.T. Douglas, O.S.a, Kinetics of substrate recognition and cleavage by human 8-nine-DNA glycosylase, Nucleic Acids Res. 33 (2005) 39193931.nd, E. Seeberg, Mutagenicity, toxicity and repair of DNA base damage

    by oxidation, Mutat. Res. 531 (2003) 3780.assan, N.H. Chmiel, J. Maynard, N. Fleming, A.L. Livingston, G.T.s, A.K. Hodges, D.R. Davies, S.S. David, J.R. Sampson, J.P. Cheadle,

    2[75] J.W

    im(2

    [76] Cdpp

    [77] Msusp

    [78] L.p1

    [79] EMnC

    [80] RLoanad

    [81] T.jnnJ.

    [82] T.DIn

    [83] J. Rluge

    [84] T.Sh8(2

    [85] Manw

    [86] MpanU

    [87] BL.fost(2

    [88] AC

    [89] J. C

    [90] MRpla2

    [91] Ctu3

    [92] Bm

    [93] S.reth

    [94] Motr

    [95] Cglro3

    [96] L.op

    [97] BM

    [98] VAA09) 51495162.l, M.K. Evans, Dimerization and opposite base-dependent catalyticent of polymorphic S326C OGG1 glycosylase, Nucleic Acids Res. 34

    16201632.in, J.P. Radicella, M. Dizdaroglu, S. Boiteux, Excision of oxidativelyd DNA bases by the human alpha-hOgg1 protein and the polymor-ha-hOgg1(Ser326Cys) protein which is frequently found in humanions, Nucleic Acids Res. 27 (1999) 40014007.dman, R.M. Bostick, C. Dash, P. Terry, W.D. Flanders, J. Mandel, Ary measure of pro- and anti-oxidant exposures and risk of incidentc, colorectal adenomas, Cancer Causes Control 19 (2008) 10511064.

    A. Elahi, J. Pow-Sang, P.J. Lazarus, Park Association between polymor-f human oxoguanine glycosylase 1 and risk of prostate cancer, J. Urol.03) 24712474., A. Hildesheim, C.J. Chen, M.M. Hsu, I.H. Chen, B.F. Mittl, P.H. Levine,, J.Y. Chen, L.A. Brinton, Y.J. Cheng, C.S. Yang, Nasopharyngeal carci-nd genetic polymorphisms of DNA repair enzymes XRCC1 and hOGG1,Epidemiol. Biomarkers Prev. 12 (2003) 11001104.en, M. Saebo, C.F. Skjelbred, B.A. Nexo, P.C. Hagen, G. Bock, I.M. Bowitz. Johnson, S. Aase, I.L. Hansteen, U. Vogel, E.H. Kure, GPX Pro198LeuG1 Ser326Cys polymorphisms and risk of development of colorectalas and colorectal cancer, Cancer Lett. 229 (2005) 8591.ski, R. Krupa, M. Wisniewska-Jarosinska, E. Pawlowska, J. Lech, J. Cho-. Blasiak, Common polymorphisms in the XPD and hOGG1 genes areciated with the risk of colorectal cancer in a Polish population, Tohokued. 218 (2009) 185191.lizur, M. Krupsky, S. Blumenstein, D. Elinger, E. Schechtman, Z. Livneh,air activity for oxidative damage and risk of lung cancer, J. Natl. Cancer

    (2003) 13121319. M. Swoboda, B. Janowska, J.M. Ciesla, D. Gackowski, J. Kowalewski,ki, B. Tudek, E. Speina, 8-Oxoguanine incision activity is impaired insues of NSCLC patients with the polymorphism of OGG1 and XRCC1utat. Res. 709710 (2011) 2131.lizur, R. Ben-Yosef, D. Elinger, A. Vexler, M. Krupsky, A. Berrebi, A.. Schechtman, L. Freedman, Z. Livneh, Reduced repair of the oxidativeanine DNA damage and risk of head and neck cancer, Cancer Res. 66

    1168311689.rbaev, K. Kleibl, J. Laval, Escherichia coli, Saccharomyces cerevisiae, ratan 3-methyladenine DNA glycosylases repair 1,N6-ethenoadenine

    resent in DNA, Nucleic Acids Res. 23 (1995) 37503755.rbaev, J. Laval, 3,N4-ethenocytosine, a highly mutagenic adduct, is a

    substrate for Escherichia coli double-stranded uracil-DNA glycosylasean mismatch-specic thymine-DNA glycosylase, Proc. Natl. Acad. Sci.

    5 (1998) 85088513., O. Sundheim, M. Akbari, M. Otterlei, H. Nilsen, F. Skorpen, P.A. Aas,n, H.E. Krokan, G. Slupphaug, hUNG2 is the major repair enzymeoval of uracil from U:A matches, U:G mismatches, and U in single-d DNA, with hSMUG1 as a broad specicity backup, J. Biol. Chem. 2773992639936.cosa, Role of MED1 (MBD4) Gene in DNA repair and human cancer, J.ysiol. 187 (2001) 137144.y, M. Menigatti, DNA cytosine demethylation: are we getting close?

    (2008) 11671169.arbaev, S. Langouet, C.V. Privezentzev, F.P. Guengerich, H. Cai,er, J. Laval, 1,N(2)-ethenoguanine, a mutagenic DNA adduct, is a

    substrate of Escherichia coli mismatch-specic uracil-DNA glycosy-d human alkylpurine-DNA-N-glycosylase, J. Biol. Chem. 277 (2002)26993.vezentzev, M. Saparbaev, J. Laval, The HAP1 protein stimulates ther of human mismatch-specic thymine-DNA-glycosylase to processethenocytosine residues, Mutat. Res. 480481 (2001) 277284.le, J.S. Sung, Molecular and biological roles of Ape1 protein in mam-base excision repair, DNA Repair (Amst.) 4 (2005) 14421449.. Harbers, A. Benecke, B. Pierrat, R. Losson, P. Chambon, Retinoic acid

    rs interact physically and functionally with the T:G mismatch-specice-DNA glycosylase, J. Biol. Chem. 273 (1998) 2072820736.

    A. Benecke, S.J. Um, J. Torchia, R.M. Evans, P. Chambon, Associationp300 acetylase and thymine DNA glycosylase links DNA repair andption, Mol. Cell 9 (2002) 265277.ero, M.T. Pirro, S. Simeone, M. Pischetola, R. Di Lauro, The DNAlase T:G mismatch-specic thymine DNA glycosylase represses thy-nscription factor-1-activated transcription, J. Biol. Chem. 276 (2001)33575.A.V. Maksimenko, C.V. Privezentzev, J. Laval, M.K. Saparbaev, Hijackinguman alkyl-N-purine-DNA glycosylase by 3,N4-ethenocytosine, a lipidation-induced DNA adduct, J. Biol. Chem. 279 (2004) 1772317730.k, Base excision repair modulation as a risk factor for human cancers,pects Med. 28 (2007) 258275.epenninckx, V. Lazar, S. Michiels, P. Dessen, M. Stas, S.R. Alonso, M.F..L. Ortiz Romero, T. Robert, O. Balacescu, A.M. Eggermont, G. Lenoir,in, T. Tursz, J.J. van den Oord, A. Spatz, Gene expression proling of

  • 92 B. Tudek, E. Speina / Mutation Research 736 (2012) 82 92

    primary cutaneous melanoma and clinical outcome, J. Natl. Cancer Inst. 98(2006) 472482.

    [99] K. Janssen, K. Schlink, W. Gotte, B. Hippler, B. Kaina, F. Oesch, DNA repairactivity of 8-oxoguanine DNA glycosylase 1 (OGG1) in human lymphocytesis not dependent on genetic polymorphism Ser326/Cys326, Mutat. Res. 486(2001) 207216.

    [100] A. Jensen, M. Lohr, L. Eriksen, M. Gronbaek, E. Dorry, S. Loft, P. Moller, Inu-ence of the OGG1 Ser326Cys polymorphism on oxidatively damaged DNA andrepair activity, Free Radic. Biol. Med. 52 (2012) 118125.

    [101] D.J. Smart, J.K. Chipman, N.J. Hodges, Activity of OGG1 variants in the repair ofpro-oxidant-induced 8-oxo-2-deoxyguanosine, DNA Repair (Amst.) 5 (2006)13371345.

    [102] A. Bravard, M. Vacher, E. Moritz, L. Vaslin, J. Hall, B. Epe, J.P. Radicella, Oxidationstatus of human OGG1-S326C polymorphic variant determines cellular DNArepair capacity, Cancer Res. 69 (2009) 36423649.

    [103] 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 damagedDNA lesions in urine, Mutat. Res. 574 (2005) 5866.

    [104] 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 asbiomarker of oxidative damage to DNA, Arch. Biochem. Biophys. 518 (2012)142150.

    [105] 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 andcolon tissue of colon cancer patients, Free Radic. Biol. Med. 49 (2010)10641071.

    [106] M. Graziewicz, D.A. Wink, F. Laval, Nitric oxide inhibits DNA ligase activ-ity: potential mechanisms for NO-mediated DNA damage, Carcinogenesis 17(1996) 25012505.

    [107] D.A. Wink, J. Laval, The Fpg protein a DNA repair enzyme, is inhibited bythe biom21252

    [108] M. Jaiswa proteiCancer

    [109] M.A. Graas a targ

    [110] Z. Fengexcisionperoxid85988

    [111] H. Bartspotentianancies

    [112] S.H. Choxoguanexposur(2002) 2

    [113] A. Das, oxidizedBiol. Ch

    [114] B. Hang(2004) 1

    [115] J. RingvoDedon, 2-media68 (200

    [116] K. Schmid, J. Nair, G. Winde, I. Velic, H. Bartsch, Increased levels of promuta-genic 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 inAPC CTNNB1 and K-ras genes and expression of hMLH1 in sporadic colorectalcarcinomas 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 ade-nomatous polyposis coli (APC)-mediated blockage of long-patch base excisionrepair, Biochemistry 45 (2006) 1590315914.

    [119] S. Narayan, A.S. Jaiswal, R. Balusu, Tumor suppressor APC blocks DNA poly-merase beta-dependent strand displacement synthesis during long patch butnot short patch base excision repair and increases sensitivity to methyl-methane sulfonate, J. Biol. Chem. 280 (2005) 69426949.

    [120] D. Cortazar, C. Kunz, Y. Saito, R. Steinacher, P. Schar, The enigmatic thymineDNA glycosylase, DNA Repair (Amst.) 6 (2007) 489504.

    [121] A.S. Jaiswal, S. Narayan, Assembly of the base excision repair complex onabasic 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) expressionby 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 endonu-clease (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 etheno-DNA adducts in affected tissues of patients suffering from Crohns diseaseulcerative colitis, and chronic pancreatitis, Antioxid. Redox Signal. 8 (2006)10031010.

    [125] L.J. Hofseth, The adaptive imbalance to genotoxic stress: genome guardiansrear their ugly heads, Carcinogenesis 25 (2004) 17871793.. Singhillmany iso. Hil

    NA gleps in

    Corte. Devaambowmotoycosyse ex

    . Kunzcisionuoro

    Amenediate

    AmeD1-manscri

    Sextod gen

    (200. Perill. Malo3K9m9 (20ediator nitric oxide in vitro and in vivo, Carcinogenesis 15 (1994)129.al, N.F. LaRusso, N. Nishioka, Y. Nakabeppu, G.J. Gores, Human Ogg1n involved in the repair of 8-oxoguanine, is inhibited by nitric oxide,Res. 61 (2001) 63886393.ziewicz, B.J. Day, W.C. Copeland, The mitochondrial DNA polymeraseet of oxidative damage, Nucleic Acids Res. 30 (2002) 28172824., W. Hu, M.S. Tang, Trans-4-hydroxy-2-nonenal inhibits nucleotide

    repair in human cells: a possible mechanism for lipidation-induced carcinogenesis, Proc. Natl. Acad. Sci. U.S.A. 101 (2004)602.ch, J. Nair, Accumulation of lipid peroxidation-derived DNA lesions:l lead markers for chemoprevention of inammation-driven malig-, Mutat. Res. 591 (2005) 3444.erng, K.H. Huang, S.C. Yang, T.C. Wu, J.L. Yang, H. Lee, Human 8-ine DNA glycosylase 1 mRNA expression as an oxidative stresse biomarker of cooking oil fumes, J. Toxicol. Environ. Health A 6565278.T.K. Hazra, I. Boldogh, S. Mitra, K.K. Bhakat, Induction of the human

    base-specic DNA glycosylase NEIL1 by reactive oxygen species, J.em. 280 (2005) 3527235280., Repair of exocyclic DNA adducts: rings of complexity, Bioessays 261951208.ll, M.N. Moen, L.M. Nordstrand, L.B. Meira, B. Pang, A. Bekkelund, P.C.S. Bjelland, L.D. Samson, P.O. Falnes, A. Klungland, AlkB homologueted repair of ethenoadenine lesions in mammalian DNA, Cancer Res.8) 41424149.

    [126] VHso

    [127] J.WDst

    [128] S.KRsuglba

    [129] Cex5-

    [130] S.m

    [131] S.LStr

    [132] T.an20

    [133] BAH31-Gupta, H. Zhang, S. Banerjee, D. Kong, J.J. Raffoul, F.H. Sarkar, G.G., Radiation-induced HIF-1alpha cell survival pathway is inhibited byavones in prostate cancer cells, Int. J. Cancer 124 (2009) 16751684.l, T.K. Hazra, T. Izumi, S. Mitra, Stimulation of human 8-oxoguanine-ycosylase by AP-endonuclease: potential coordination of the initial

    base excision repair, Nucleic Acids Res. 29 (2001) 430438.llino, J. Xu, M. Sannai, R. Moore, E. Caretti, A. Cigliano, M. Le Coz,rajan, A. Wessels, D. Soprano, L.K. Abramowitz, M.S. Bartolomei, F., M.R. Bassi, T. Bruno, M. Fanciulli, C. Renner, A.J. Klein-Szanto, Y. Mat-

    , D. Kobi, I. Davidson, C. Alberti, L. Larue, A. Bellacosa, Thymine DNAlase is essential for active DNA demethylation by linked deamination-cision repair, Cell 146 (2011) 6779., F. Focke, Y. Saito, D. Schuermann, T. Lettieri, J. Selfridge, P. Schar, Base

    by thymine DNA glycosylase mediates DNA-directed cytotoxicity ofuracil, PLoS Biol. 7 (2009) e91.te, L. Lania, E.V. Avvedimento, B. Majello, DNA oxidation drives Mycd transcription, Cell Cycle 9 (2010) 30023004.nte, A. Bertoni, A. Morano, L. Lania, E.V. Avvedimento, B. Majello,ediated demethylation of histone H3 lysine 4 triggers Myc-inducedption, Oncogene 29 (2010) 36913702.n, F. Bantignies, G. Cavalli, Genomic interactions: chromatin loopse meeting points in transcriptional regulation, Semin. Cell Dev. Biol.9) 849855.o, M.N. Ombra, A. Bertoni, C. Cuozzo, S. Sacchetti, A. Sasso, L. Chiariotti,rni, C. Abbondanza, E.V. Avvedimento, DNA oxidation as triggered bye2 demethylation drives estrogen-induced gene expression, Science08) 202206.

    Oxidatively damaged DNA and its repair in colon carcinogenesis1 Introduction2 Histopathology of CRC and mutations in critical genes3 Oxidative stress in the development of colon cancer3.1 Inflammation3.2 Consumption of red meat and alcohol and tobacco smoking3.3 DNA damage

    4 Repair of oxidative DNA damage as a driving force to colon carcinogenesis4.1 Repair of 8-oxo-7,8-dihydro-2-deoxyguanosine in colon adenoma and carcinoma patients4.2 Repair of etheno DNA adducts in colon carcinogenesis

    5 Aberrant repair of oxidatively damaged DNA bases and mutations in critical genes6 ConclusionsConflict of interest statementFundingReferences