molecular pathways: involvement of helicobacter pylori...

8
Molecular Pathways Molecular Pathways: Involvement of Helicobacter pyloriTriggered Inammation in the Formation of an Epigenetic Field Defect, and Its Usefulness as Cancer Risk and Exposure Markers Toshikazu Ushijima and Naoko Hattori Abstract Infection-associated cancers account for a large proportion of human cancers, and gastric cancer, the vast majority of which is associated with Helicobacter pylori infection, is a typical example of such cancers. Epigenetic alterations are known to occur frequently in gastric cancers, and H. pylori infection has now been shown to induce aberrant DNA methylation in gastric mucosae. Accumulation of aberrant methylation in gastric mucosae produces a field for cancerization, and methylation levels correlate with gastric cancer risk. H. pylori infection induces methylation of specific genes, and such specificity is determined by the epigenetic status in normal cells, including the presence of H3K27me3 and RNA polymerase II (active or stalled). Specific types of inflammation, such as that induced by H. pylori infection, are important for methylation induction, and infiltration of monocytes appears to be involved. The presence of an epigenetic field defect is not limited to gastric cancers and is observed in various types of cancers. It provides translational opportunities for cancer risk diagnosis incorporating life history, assessment of past exposure to carcino- genic factors, and cancer prevention. Clin Cancer Res; 18(4); 923–9. Ó2011 AACR. Background Infection-associated cancers account for a large propor- tion of human cancers. These include gastric cancers induced by Helicobacter pylori (1), hepatocellular carcino- mas induced by hepatitis C virus (HCV) and hepatitis B virus [HBV (2–4)], cervical cancers induced by human papilloma virus [HPV (5, 6)], and lymphomas and naso- pharyngeal cancers associated with Epstein-Barr virus [EBV (7, 8)]. The carcinogenic mechanisms of these infection- associated cancers have been extensively investigated, and although multiple contributing mechanisms have been clarified, they are not yet completely understood. General mechanisms of infection-associated cancers Virus-associated cancers have complex mechanisms of car- cinogenesis. Viral oncogenes, such as E6 and E7 of HPV and X protein of HBV, can be integrated into host cells and produce aberrant growth signals and inactivate tumor-suppressor genes (6). Also, integration of virus genes into the host genome can alter the expression of nearby tumor-related genes and induce a genomic instability that will eventually contribute to cancer development (4). Even if the virus genes are not inte- grated, they can be persistently expressed and perturb impor- tant cellular signaling, such as cell proliferation, apoptosis, and cytokine expression, as in the case of HCV and EBV (7). Both bacterial and viral infections can be associated with severe tissue damage and resultant chronic inflammation (4, 6, 7). Tissue damage itself activates cell proliferation and increases the chance that mutations will occur. In addition, chronic inflammation is considered to be deeply involved in cancer development and progression by multiple mechanisms, such as increased production of active oxygen species, induction of inflammation-mediated cell prolifer- ation, and increased cytokine production (9, 10). In addi- tion to this, induction of epigenetic alterations is now recognized as one of the mechanisms underlying induction of cancer by chronic inflammation. H. pylori infection and epigenetic alterations in gastric cancers Gastric cancer is still the third-leading cause of death from cancer in men and the fifth-leading cause in women world- wide, although its incidence is gradually decreasing (11). The vast majority of gastric cancers are caused by H. pylori infection (12), which is a Gram-negative bacterium (13). A minor percentage (10%) of gastric cancers are associated with EBV infection (14). It is known that when H. pylori infects the human stomach, it induces severe inflammation, including ulcers, then chronic inflammation, and finally gastric cancers within tens of years. Investigators have main- ly discussed the carcinogenic mechanisms of H. pylori from Authors' Afliation: Division of Epigenomics, National Cancer Center Research Institute, Tokyo, Japan Corresponding Author: Toshikazu Ushijima, National Cancer Center Research Institute, 5-1-1-Tsukiji, Chuo-ku, Tokyo, Japan 104-0045. Phone: 81-3-3542-2511; Fax: 81-3-5565-1753; E-mail: [email protected] doi: 10.1158/1078-0432.CCR-11-2011 Ó2011 American Association for Cancer Research. Clinical Cancer Research www.aacrjournals.org 923 on May 11, 2019. © 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from Published OnlineFirst December 28, 2011; DOI: 10.1158/1078-0432.CCR-11-2011

Upload: trinhquynh

Post on 11-May-2019

222 views

Category:

Documents


0 download

TRANSCRIPT

Molecular Pathways

Molecular Pathways: Involvement of HelicobacterpyloriTriggered Inflammation in the Formation of anEpigenetic Field Defect, and Its Usefulness as CancerRisk and Exposure Markers

Toshikazu Ushijima and Naoko Hattori

AbstractInfection-associated cancers account for a large proportion of human cancers, and gastric cancer, the vast

majority of which is associated with Helicobacter pylori infection, is a typical example of such cancers.

Epigenetic alterations are known to occur frequently in gastric cancers, andH. pylori infection has now been

shown to induce aberrant DNA methylation in gastric mucosae. Accumulation of aberrant methylation in

gastric mucosae produces a field for cancerization, and methylation levels correlate with gastric cancer risk.

H. pylori infection inducesmethylation of specific genes, and such specificity is determined by the epigenetic

status in normal cells, including the presence of H3K27me3 and RNA polymerase II (active or stalled).

Specific types of inflammation, such as that induced by H. pylori infection, are important for methylation

induction, and infiltration ofmonocytes appears to be involved. The presence of an epigenetic field defect is

not limited to gastric cancers and is observed in various types of cancers. It provides translational

opportunities for cancer risk diagnosis incorporating life history, assessment of past exposure to carcino-

genic factors, and cancer prevention. Clin Cancer Res; 18(4); 9239. 2011 AACR.

Background

Infection-associated cancers account for a large propor-tion of human cancers. These include gastric cancersinduced by Helicobacter pylori (1), hepatocellular carcino-mas induced by hepatitis C virus (HCV) and hepatitis Bvirus [HBV (24)], cervical cancers induced by humanpapilloma virus [HPV (5, 6)], and lymphomas and naso-pharyngeal cancers associated with Epstein-Barr virus [EBV(7, 8)]. The carcinogenic mechanisms of these infection-associated cancers have been extensively investigated, andalthough multiple contributing mechanisms have beenclarified, they are not yet completely understood.

General mechanisms of infection-associated cancersVirus-associated cancers have complex mechanisms of car-

cinogenesis. Viral oncogenes, such as E6 and E7 of HPV and Xprotein of HBV, can be integrated into host cells and produceaberrant growth signals and inactivate tumor-suppressorgenes (6).Also, integrationof virusgenes into thehost genomecan alter the expression of nearby tumor-related genes andinduce a genomic instability that will eventually contribute to

cancer development (4). Even if the virus genes are not inte-grated, they can be persistently expressed and perturb impor-tant cellular signaling, suchas cell proliferation, apoptosis, andcytokine expression, as in the case of HCV and EBV (7).

Both bacterial and viral infections can be associated withsevere tissue damage and resultant chronic inflammation(4, 6, 7). Tissue damage itself activates cell proliferation andincreases the chance that mutations will occur. In addition,chronic inflammation is considered to be deeply involvedin cancer development and progression by multiplemechanisms, such as increased production of active oxygenspecies, induction of inflammation-mediated cell prolifer-ation, and increased cytokine production (9, 10). In addi-tion to this, induction of epigenetic alterations is nowrecognized as one of the mechanisms underlying inductionof cancer by chronic inflammation.

H. pylori infection and epigenetic alterations in gastriccancers

Gastric cancer is still the third-leading cause of death fromcancer in men and the fifth-leading cause in women world-wide, although its incidence is gradually decreasing (11).The vast majority of gastric cancers are caused by H. pyloriinfection (12), which is a Gram-negative bacterium (13). Aminor percentage (10%) of gastric cancers are associatedwith EBV infection (14). It is known that when H. pyloriinfects the human stomach, it induces severe inflammation,including ulcers, then chronic inflammation, and finallygastric cancers within tens of years. Investigators havemain-ly discussed the carcinogenic mechanisms of H. pylori from

Authors' Affiliation: Division of Epigenomics, National Cancer CenterResearch Institute, Tokyo, Japan

Corresponding Author: Toshikazu Ushijima, National Cancer CenterResearch Institute, 5-1-1-Tsukiji, Chuo-ku, Tokyo, Japan 104-0045. Phone:81-3-3542-2511; Fax: 81-3-5565-1753; E-mail: [email protected]

doi: 10.1158/1078-0432.CCR-11-2011

2011 American Association for Cancer Research.

ClinicalCancer

Research

www.aacrjournals.org 923

on May 11, 2019. 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 28, 2011; DOI: 10.1158/1078-0432.CCR-11-2011

http://clincancerres.aacrjournals.org/

the standpoint of induction of cell proliferation,mutations,and direct activation of cellular signaling (1517).

However, tumor-suppressor genes such as CDKN2A,CDH1,MLH1, and RUNX3 are inactivated more frequentlyby aberrant DNA methylation than by mutations, indicat-ing that gastric cancer is an epigenetic disease (18). Inaddition to methylation silencing of driver tumor-suppres-sor genes, recent genome-wide analyses have revealed thathundreds of passenger genes are also methylated in gastriccancers (19). The fact that H. pylori infection induces epi-genetic alterations provides the missing link between thecausal role ofH.pylori infection ingastric carcinogenesis andthe deep involvement of epigenetic alterations in gastriccancers. Gastric cancer is a typical example of a disease inwhich infection, chronic inflammation, and epigeneticalterations are interconnected.

Induction of epigenetic alterations byH. pylori and theformation of field defects

The first hint that the presence of aberrant DNA methyl-ationmight be associated withH. pylori infection came fromthe observation that promoter methylation of CDH1 wasdetected more frequently in the gastric mucosae of indivi-duals with H. pylori infection than in those without theinfection (20). By quantifying the methylation levels of 8marker CpG islands, Maekita and colleagues (21) convinc-ingly showed that individuals with H. pylori infection havemuchhighermethylation levels in their gastricmucosae (5.4-to 303-fold) than those without (P < 0.0001). In addition tothe 8 marker CpG islands associated with protein-codinggenes,CpG islandsofmicroRNAgenes are alsomethylated inassociation with H. pylori infection (22, 23).

In one study, patients with gastric cancer who had pre-viously had an H. pylori infection but were currently notinfected had lower methylation levels of the 8 marker CpGislands in the gastric mucosae compared with patients whowere currently infectedwithH. pylori (21). This suggests thatthe methylation level is very high when active H. pyloriinfection is present in the stomach and decreases to certainlevels when the infection is discontinued. In other studies,various degrees of decrease were observed in individualswho received eradication therapy forH. pylori (24, 25), andthe methylation level after the decrease was considered torepresent the degree of epigenomic damage to the individ-ual. This decrease ofmethylation could be due to a turnoverof gastric epithelial cells withmethylation or to the removalof 5-methylcytosine, which is present in individuals withactive H. pylori infection.

Of importance, among individuals without current H.pylori infection, the methylation levels of the 8 marker CpGislands in gastric mucosae were shown to correlate withgastric cancer risk (21, 26). Patients with gastric cancers had2.2- to 32-fold higher methylation levels in gastric mucosaecompared with healthy individuals (21), and patients withmultiple gastric cancers had significantly higher methyla-tion levels than those with a single gastric cancer (26). Thiscorrelation strongly supports the notion that the accumu-lation of aberrant methylation in gastric mucosae produces

anepigenetic field for cancerization, i.e., afielddefect (Fig. 1;ref. 27).

Epigenetic field for cancerizationIn the epigenetic field for gastric cancers, tumor-suppres-

sor genes that are causally involved in gastric cancer devel-opment (i.e., driver genes), such as CDKN2A, CDH1,MLH1, and RUNX3, are methylated only at very low levels,showing that such events are present only in a very smallfraction of cells (21). In contrast, many other genes that areunlikely to be causally involved in gastric carcinogenesis(i.e., passenger genes), such as HAND1 (a transcriptionfactor involved in heart morphogenesis), are methylatedat high levels, showing that their methylation is present in alarge fraction of cells. Most of the genes that are highlymethylated in gastric cancers are either unexpressed orexpressed only at low levels in normal cells (28). Generally,genes with low expression are susceptible to methylationinduction (29), and it is considered that most of the genesthat are methylated in the epigenetic field were methylatedas a consequence of gastric carcinogenesis. In addition toaccumulation of aberrant methylation, an epigenetic fieldinvolves hypomethylation of the Alu and Sata repeatsequences (30), which potentially can be involved in geno-mic instability.

Epigenetic field defects are present not only in gastriccancers but in other cancers as well (27). In the case ofhepatocellular carcinoma, aberrant DNA methylation wasfrequently observed in noncancerous tissues of cancerpatients compared with normal livers of patients withmetastatic liver tumors (31). A quantitative analysisrevealed increased methylation of multiple tumor-suppres-sor genes, such as SOCS1, RASSF1A, and CDH1, in HCV-infected, noncancerous liver tissues (32), suggesting theimportance of an epigenetic field for HCV-associated hepa-tocarcinogenesis. In the case of esophageal adenocarcino-ma, the presence of APC and CDKN2A methylation inBarretts metaplasia has been reported (33), and suchmeth-ylation was shown to be associated with progression ofBarretts metaplasia (34). Also in the case of esophagealsquamous cell carcinoma, methylation of specific genes,such as UCHL1 and HOXA9, in esophageal mucosae wasassociatedwith the riskof developing esophageal squamouscell carcinoma (35, 36). In ulcerative colitis, the driver geneCDKN2A and passenger genes such as MYOD and ESRwere shown to be methylated in colonic mucosae, whicharepredisposed to colon cancers (37, 38). In addition, in thecase of sporadic colorectal cancers, MGMT methylation incancer tissues was associated with high levels of MGMTmethylation in the background colonic mucosae (39). Thepresence of epigenetic field defects has also been indicatedfor breast (40), renal (41), and bladder cancers (42, 43).

Critical roles of specific types of inflammation inmethylation induction

The association between H. pylori infection and highlevels of DNA methylation in gastric mucosae in humansstrongly indicates that H. pylori infection induces aberrant

Ushijima and Hattori

Clin Cancer Res; 18(4) February 15, 2012 Clinical Cancer Research924

on May 11, 2019. 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 28, 2011; DOI: 10.1158/1078-0432.CCR-11-2011

http://clincancerres.aacrjournals.org/

DNA methylation. This cause-consequence relationshipwas shown with the use of Mongolian gerbils, in whichH. pylori infection-induced gastritis and gastric cancers canbe recapitulated (44). Gerbils infected with H. pylori devel-oped severe gastritis and had markedly increased methyl-ation levels, showing the causal role ofH. pylori infection inmethylation induction (45). The methylation levels wereclearly decreased after eradication of the H. pylori, in agree-

ment with the decreased methylation levels observed inpatients who received eradication therapy.

In the attempt to determine how H. pylori induces meth-ylation, investigators have considered both direct and indi-rect actions of H. pylori. First, because H. pylori possessesmultiple DNA methyltransferases (46) and a type IV secre-tion system [a syringe-like structure capable of deliveringbacterial materials into a host cell (47)],H. pylori itself may

2011 American Association for Cancer Research

Normalepithelium

Acuteinflammation

Chronicinflammation

Epigeneticfield for

cancerization

Clinical tumorformation

Suppression ofmethylation induction

Removal of aberrantmethylation

UnmethylatedMethylated

Gene1234

H. pylori infection

Figure 1. Formation of epigenetic field for cancerization by chronic inflammation triggered byH. pylori infection.H. pylori infection induces acute inflammation,followed by chronic inflammation, formation of an epigenetic field for cancerization, and development of gastric cancers. Aberrant methylation is induced indriver genes (schematically represented by gene 1) and passenger genes (genes 3 and 4). Specific genes are methylated in gastric mucosae withH.pylori infection, anddriver genesusually have very lowmethylation levels. On theother hand, passenger genes that have lowor noexpression in normal cellsusually have high methylation levels. The methylation level of some passenger genes reflects the degree of accumulation of epigenomic damage, andcorrelates with gastric cancer risk. Chronic inflammation triggered by H. pylori infection is critical for methylation induction, and if data from a mousecolitismodel are combined, the importanceofmonocytes canbespeculated. As translational targets,methylation levels of specificgenes innormal-appearingtissues can be used as a cancer risk marker that reflects a person's life. The methylation signature has potential as a marker for past exposure tospecific environmental factors. Suppression of induction of aberrant DNA methylation, and possibly removal of accumulated aberrant methylation canbe used for cancer prevention (shown in red).

Epigenetic Field by H. pylori Infection

www.aacrjournals.org Clin Cancer Res; 18(4) February 15, 2012 925

on May 11, 2019. 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 28, 2011; DOI: 10.1158/1078-0432.CCR-11-2011

http://clincancerres.aacrjournals.org/

induce methylation in epithelial cells by injecting its ownDNA methyltransferases. Alternatively, studies in patientswith ulcerative colitis showed that chronic inflammationplayed a role inmethylation induction (37, 38), and chron-ic inflammation triggered by H. pylori infection may havebeen responsible for the methylation induction. Niwa andcolleagues (45) addressed this issue by suppressing inflam-mation in gerbils with H. pylori infection using cyclosporinA, an immunosuppressant. Although colonization of H.pylori was not affected at all, methylation induction wasmarkedly suppressed. This clearly shows that it was theinflammation triggered by theH. pylori infection, not theH.pylori itself, that was involved in methylation induction. Atemporal analysis of the expression of inflammation-relatedgenes in gastric mucosae of infected gerbils showed that theexpression levels of Cxcl2, Il1b,Nos2, and Tnf paralleled themethylation levels.

Inflammation in the stomach can be induced not only byH. pylori infection but also by high concentrations of eth-anol (EtOH) or saturated sodium chloride (NaCl) solution.A methylation analysis of gastric mucosae exposed to thesekinds of inflammation showed that only inflammationtriggered by H. pylori infection was capable of inducingaberrant DNA methylation (48). Histologically, H. pyloriinfection induced chronic inflammation with prominentlymphocyte and macrophage infiltration, whereas EtOHand NaCl treatment induced persistent neutrophil infiltra-tion. Cell proliferation, which is known to be important formethylation induction (38), was most strongly induced inthe NaCl group and was shown to be insufficient formethylation induction. Among inflammation-relatedgenes, expression of Il1b, Nos2, and Tnf was increasedspecifically in gastric mucosae of gerbils with H. pyloriinfection. Therefore, it is considered that specific types ofinflammation are necessary for methylation induction.

Chronic inflammation is characterized by infiltration ofmononuclear cells, i.e., lymphocytes and monocytes. Toclarifywhich cell type(s) plays themajor role inmethylationinduction, Katsurano and colleagues (49) examined SCIDmice, which lack both B and T lymphocytes. Because H.pylori cannot infectmice efficiently, theyused a colitismodelinduced by dextran sulfate sodium (DSS). Even in SCIDmice,DNAmethylation and colon tumors couldbe inducedat the same levels as in wild-type mice. This shows thatlymphocytes are dispensable for methylation induction,and strongly suggests thatmonocytes are important. Expres-sion of Ifng, Il1b, and Nos2 was induced in both wild-typeand SCID mice by DSS treatment.

If we hypothesize that the same effectors are working ingerbil stomachs infected by H. pylori and mouse colonstreated with DSS, we can conclude that expression of Il1band Nos2 may be involved in methylation induction. Pro-moter polymorphisms of IL1B are reported to be associatedwith human gastric cancer susceptibility by increasing ordecreasing IL1b production in response to H. pylori infec-tion and thus the progression of gastric atrophy (50, 51).Increased production of NO in vitro is reported to increasethe enzyme activity of DNA methyltransferases without

changing their expression, and to induce DNAmethylationof specific genes (52). In the human and gerbil stomachsinfected byH. pylori andmouse colons treated with DSS, nochanges in the expression of DNAmethyltransferases 1, 3a,and 3bwere observed (28, 45, 49). It is possible that a signalfrom chronic inflammation, possibly IL1b, and elevation ofNO in epithelial cells lead to inappropriate localization ofderegulated DNA methyltransferase(s) to methylation-sus-ceptible CpG islands (see below) and induce aberrant DNAmethylation as an infrequent event.

Methylation fingerprints produced by H. pyloriinfection

Target genes for methylation induction byH. pylori infec-tion are present in gastric mucosae (28). Among 48 pro-moter CpG islands whose methylation was analyzed ingastric mucosae of individuals with and without H. pyloriinfection, somewere consistentlymethylated in individualswith current or past infection and others were not methyl-ated at all. Analysis of polyclonal tissues, unlike that ofcancers, canprovide information aboutmultiple events thathave taken place independently, and the presence of targetgenes was convincingly shown in gastric mucosae (29).Similarly, in the esophagus, specific genes were methylatedin association with smoking history (35), and again thepresence of target gene specificity formethylation inductionwas shown.

The target gene specificity is defined by epigenetic factorsin the cells wheremethylation is induced (29, 53, 54) and inthe genome architecture (55, 56). Epigenetic factors thatpromote methylation induction include low transcriptionand the presence of an H3K27me3 modification. In con-trast, the presence of histone acetylation and RNA polymer-ase II (active or stalled)protectsCpG islands frombecomingmethylated. A multivariate analysis revealed that the mostinfluential factors are the promoting effect of H3K27me3and the protective effect of RNA polymerase II (54). Agenomic factor that promotes methylation induction is adistant location from repetitive elements (55, 56). It iscurrently speculated that infection by H. pylori inducesH3K27me3 and removes RNA polymerase II at its targetgenes, and that these genes then become methylated.

Clinical-Translational Advances

Cancer risk marker that reflects life historyThe importance of predicting cancer risks has been

repeatedly emphasized because the ability to select high-risk individuals enables efficient cancer screening andreduces social costs (5759). To this end, a massive efforthas been made in association studies, and many cancer riskalleles for common cancers have been identified. Most ofthese risk alleles give odds ratios between 1.5 and 2.0(51, 58, 59), and can be used to estimate cancer risk whena person is born.

At the same time, a person is exposed to various envi-ronmental carcinogenic factors, and the cancer risk of anadult will differ depending on what sort of life he or she has

Ushijima and Hattori

Clin Cancer Res; 18(4) February 15, 2012 Clinical Cancer Research926

on May 11, 2019. 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 28, 2011; DOI: 10.1158/1078-0432.CCR-11-2011

http://clincancerres.aacrjournals.org/

spent. Therefore, a cancer risk marker that incorporatesinformation about life-to-date is important. At least someof a persons life history, such as smoking and infection byH. pylori, is imprinted on the epigenome and produces anepigenetic field for cancerization. The severity of the fieldcan be measured as methylation levels of specific markergenes, and correlates with cancer risk. The odds ratiosobtained by DNA methylation markers of gastric cancers,such as THBD, FLNc, andmiR-124a, range from 2.4 to 22.1[calculated based on our previous reports (21, 22)]. Pas-senger genes can be useful as marker genes because they areconsistentlymethylated andhavehighmethylation levels innoncancerous tissues (21, 27), whereas driver genes areonly stochastically methylated and have low methylationlevels (Fig. 1). Therefore, for evaluating the degree of epi-genomic damage that has been done in the past, methyl-ation of passenger genes is often superior to that of drivergenes.The presence of an epigenetic field defect is also known

for other types of cancers, as mentioned above (27). There-fore, investigators are now developing methods to estimateepigenetic cancer risk, taking life history into account, invarious types of cancer. For example, a multicenter studywas conducted to evaluate the validity of methylationmarkers to predict progression of Barretts esophagus, andmethylation ofHPP1,CDKN2A, andRUNX3were shown tobe informative (34).

Marker for past exposure to specific environmentalfactorsH. pylori infection is associated with methylation of a

specific set of genes, most of which are considered aspassengers, in gastric mucosae (28). A history of smokingis associated with methylation of UCHL1 and HOXA9,which are also considered to be passengers, in esophagealmucosae (35, 36). Once the specificity of methylationsignatures to various carcinogenic agents is clarified, pastexposure to such carcinogenic factors can be estimated bythe methylation signature. The methylation signature hasadvantages over other exposure markers because it persistsfor a long time and does not require any record by humans.For example, past exposure to H. pylori infection can beestimated by serum antibody, but it persists only up toseveral years afterH. pylori infection discontinues (60). Theability to estimate past exposure using a methylation sig-nature would be very helpful from an epidemiologicalviewpoint.

Epigenetic cancer preventionThe presence of an epigenetic field for cancerization

and the deep involvement of chronic inflammation inits formation provide targets for cancer prevention. Sup-pression of induction of aberrant DNA methylation isexpected to lead to a decreased incidence of cancers.This concept is supported by animal models for macro-scopic colon tumors (61, 62), lung tumors (63), andprostate cancers (64, 65). It was shown that in gerbilstomachs, administration of a demethylating agent,

5-aza-20-deoxycytidine (5-aza-dC), decreased the inci-dence of gastric cancers induced by H. pylori infectionand a mutagen, N-methyl-N-nitrosourea (unpublished).In addition to suppression of DNA methylation induc-tion, suppression of H3K27me3, a premarker for DNAmethylation induction, is also an attractive target. Thehistone methyltransferase that is responsible for thismodification, EZH2, is known to be overexpressed inaggressive tumors (66) and precancerous lesions (67),and therefore inhibitors of EZH2, such as 3-deazanepla-nocin A (66), may have preventive applications.

Anti-inflammatory drugs, especially nonsteroidal anti-inflammatory drugs (NSAIDs), are effective for preventionof at least some cancers, but their use is still limited toindividuals with high risk (68). The use of NSAIDS islimited in part because of possible side-effects, such aspeptic ulcer. To avoid such side-effects and suppress thepathways that are responsible for cancer development,researchers are actively investigating the mechanisms ofcancer induction by chronic inflammation (69). Becauseinduction of epigenetic alterations is one of these importantmechanisms, suppressionof specific components of inflam-mation that are responsible for induction of epigeneticalterations is expected to provide a good target for cancerprevention.

Lastly, DNA methylation is reversible by DNA demethy-lating agents, such as 5-aza-dC and 5-azacytidine (70).Currently available demethylating agents do not have ahigh specificity for aberrantly methylated genes, and candemethylate normally methylated sequences. Suchsequences include normally methylated CpG islands andrepetitive sequences originating from retrotransposons, andit is feared that DNA demethylating agents might inducedemethylation of these retrotransposons. Therefore, forcancer prevention using current demethylating agents, wemust carefully balance risk and benefit, and probably suchagents are not widely indicated. However, many epigeneticdrugs are beingdeveloped, and it is possible that someof thenew demethylating agents will have a specificity or prefer-ence for aberrantly methylated promoter CpG islands, andcan be used in a wider range of individuals in the future.

Conclusions

The fact that H. pylori infection induces aberrant DNAmethylation in gastric mucosae provides the missing linkbetween the major role of H. pylori infection in gastriccancers and the deep involvement of epigenetic alterationsin gastric cancers. The severity of infection correlates withgastric cancer risk and can provide a unique cancer riskmarker that reflects a persons life. H. pylori infection hasbeen shown to induce methylation of specific genes, andthere are underlying mechanisms. The methylation signa-ture has potential as a marker for past exposure to H. pyloriinfection. Specific types of inflammation, such as thatinduced by H. pylori infection, are capable of inducingaberrantmethylation, andmonocytes appear to be involvedin the induction. Suppression of methylation induction,

Epigenetic Field by H. pylori Infection

www.aacrjournals.org Clin Cancer Res; 18(4) February 15, 2012 927

on May 11, 2019. 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 28, 2011; DOI: 10.1158/1078-0432.CCR-11-2011

http://clincancerres.aacrjournals.org/

specific inflammatory processes, and reversal of epigeneticalterations are targets for cancer prevention.

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Grant Support

Third-Term Comprehensive Cancer Control Strategy, Ministry of Health,Labor and Welfare, Japan.

ReceivedAugust 16, 2011; revisedDecember 11, 2011; acceptedDecember13, 2011; published OnlineFirst December 28, 2011.

References1. Uemura N, Okamoto S, Yamamoto S, Matsumura N, Yamaguchi S,

Yamakido M, et al. Helicobacter pylori infection and the developmentof gastric cancer. N Engl J Med 2001;345:7849.

2. Kremsdorf D, Soussan P, Paterlini-Brechot P, Brechot C. Hepatitis Bvirus-related hepatocellular carcinoma: paradigms for viral-relatedhuman carcinogenesis. Oncogene 2006;25:382333.

3. Levrero M. Viral hepatitis and liver cancer: the case of hepatitis C.Oncogene 2006;25:383447.

4. Feitelson MA, Lee J. Hepatitis B virus integration, fragile sites, andhepatocarcinogenesis. Cancer Lett 2007;252:15770.

5. Whiteside MA, Siegel EM, Unger ER. Human papillomavirus andmolecular considerations for cancer risk. Cancer 2008;113[Suppl]:298194.

6. Martin D, Gutkind JS. Human tumor-associated viruses and newinsights into the molecular mechanisms of cancer. Oncogene2008;27[Suppl 2]:S3142.

7. Saha A, Robertson ES. Epstein-Barr virus-associated B-cell lympho-mas: pathogenesis and clinical outcomes. Clin Cancer Res 2011;17:305663.

8. Young LS, Rickinson AB. Epstein-Barr virus: 40 years on. Nat RevCancer 2004;4:75768.

9. Hussain SP, Harris CC. Inflammation and cancer: an ancient link withnovel potentials. Int J Cancer 2007;121:237380.

10. Grivennikov SI, Greten FR, Karin M. Immunity, inflammation, andcancer. Cell 2010;140:88399.

11. Jemal A,Bray F,CenterMM,Ferlay J,WardE, FormanD.Global cancerstatistics. CA Cancer J Clin 2011;61:6990.

12. Correa P. Bacterial infections as a cause of cancer. J Natl Cancer Inst2003;95:E3.

13. Asaka M, Kato M, Takahashi S, Fukuda Y, Sugiyama T, Ota H, et al.Japanese Society for Helicobacter Research. Guidelines for the man-agement ofHelicobacter pylori infection in Japan: 2009 revisededition.Helicobacter 2010;15:120.

14. Fukayama M. Epstein-Barr virus and gastric carcinoma. Pathol Int2010;60:33750.

15. Peek RM Jr, Blaser MJ. Helicobacter pylori and gastrointestinal tractadenocarcinomas. Nat Rev Cancer 2002;2:2837.

16. Matsumoto Y, Marusawa H, Kinoshita K, Endo Y, Kou T, Morisawa T,et al. Helicobacter pylori infection triggers aberrant expression ofactivation-induced cytidine deaminase in gastric epithelium. Nat Med2007;13:4706.

17. Hatakeyama M. Helicobacter pylori CagAa bacterial intruder con-spiring gastric carcinogenesis. Int J Cancer 2006;119:121723.

18. Ushijima T, Sasako M. Focus on gastric cancer. Cancer Cell 2004;5:1215.

19. Yamashita S, Tsujino Y, Moriguchi K, Tatematsu M, Ushijima T.Chemical genomic screening formethylation-silenced genes in gastriccancer cell lines using 5-aza-2-deoxycytidine treatment and oligo-nucleotide microarray. Cancer Sci 2006;97:6471.

20. Chan AO, Lam SK, Wong BC, Wong WM, Yuen MF, Yeung YH, et al.Promoter methylation of E-cadherin gene in gastric mucosa associ-ated with Helicobacter pylori infection and in gastric cancer. Gut2003;52:5026.

21. Maekita T, Nakazawa K, Mihara M, Nakajima T, Yanaoka K, Iguchi M,et al. High levels of aberrant DNA methylation in Helicobacter pylori-infected gastric mucosae and its possible association with gastriccancer risk. Clin Cancer Res 2006;12:98995.

22. Ando T, Yoshida T, Enomoto S, Asada K, Tatematsu M, Ichinose M,et al. DNA methylation of microRNA genes in gastric mucosae of

gastric cancer patients: its possible involvement in the formation ofepigenetic field defect. Int J Cancer 2009;124:236774.

23. Suzuki H, Yamamoto E, Nojima M, Kai M, Yamano HO, Yoshikawa K,et al. Methylation-associated silencing of microRNA-34b/c in gastriccancer and its involvement in an epigenetic field defect. Carcinogen-esis 2010;31:206673.

24. Miyazaki T, Murayama Y, Shinomura Y, Yamamoto T, Watabe K,Tsutsui S, et al. E-cadherin gene promoter hypermethylation in H.pylori-induced enlarged fold gastritis. Helicobacter 2007;12:52331.

25. Nakajima T, Enomoto S, Yamashita S, Ando T, Nakanishi Y, NakazawaK, et al. Persistence of a component of DNA methylation in gastricmucosae after Helicobacter pylori eradication. J Gastroenterol 2010;45:3744.

26. Nakajima T, Maekita T, Oda I, Gotoda T, Yamamoto S, Umemura S,et al. Higher methylation levels in gastric mucosae significantly cor-relatewith higher risk of gastric cancers. Cancer Epidemiol BiomarkersPrev 2006;15:231721.

27. Ushijima T. Epigenetic field for cancerization. J Biochem Mol Biol2007;40:14250.

28. Nakajima T, Yamashita S, Maekita T, Niwa T, Nakazawa K, Ushijima T.The presence of a methylation fingerprint of Helicobacter pylori infec-tion in human gastric mucosae. Int J Cancer 2009;124:90510.

29. Takeshima H, Ushijima T. Methylation destiny: Moira takes account ofhistones and RNA polymerase II. Epigenetics 2010;5:8995.

30. Yoshida T, Yamashita S, Takamura-Enya T, Niwa T, Ando T, EnomotoS, et al. Alu and Sata hypomethylation in Helicobacter pylori-infectedgastric mucosae. Int J Cancer 2011;128:339.

31. Kondo Y, Kanai Y, Sakamoto M, Mizokami M, Ueda R, Hirohashi S.Genetic instability and aberrant DNA methylation in chronic hepatitisand cirrhosisa comprehensive study of loss of heterozygosity andmicrosatellite instability at 39 loci andDNAhypermethylation on 8CpGislands inmicrodissected specimens from patients with hepatocellularcarcinoma. Hepatology 2000;32:9709.

32. NishidaN,NagasakaT,Nishimura T, Ikai I, BolandCR,Goel A.Aberrantmethylation of multiple tumor suppressor genes in aging liver, chronichepatitis, and hepatocellular carcinoma. Hepatology 2008;47:90818.

33. Eads CA, Lord RV, Kurumboor SK, Wickramasinghe K, Skinner ML,Long TI, et al. Fields of aberrant CpG island hypermethylation inBarrett's esophagus and associated adenocarcinoma. Cancer Res2000;60:50216.

34. Jin Z, Cheng Y, Gu W, Zheng Y, Sato F, Mori Y, et al. A multicenter,double-blinded validation study of methylation biomarkers for progres-sion prediction in Barrett's esophagus. Cancer Res 2009;69:41125.

35. Oka D, Yamashita S, Tomioka T, Nakanishi Y, Kato H, Kaminishi M,et al. The presence of aberrant DNA methylation in noncancerousesophageal mucosae in association with smoking history: a target forrisk diagnosis and prevention of esophageal cancers. Cancer2009;115:341226.

36. Lee YC, Wang HP, Wang CP, Ko JY, Lee JM, Chiu HM, et al. Revisit offield cancerization in squamous cell carcinoma of upper aerodigestivetract: better risk assessmentwith epigeneticmarkers. Cancer PrevRes2011;4:1982.

37. Hsieh CJ, Klump B, Holzmann K, Borchard F, Gregor M, Porschen R.Hypermethylation of the p16INK4a promoter in colectomy specimensof patients with long-standing and extensive ulcerative colitis. CancerRes 1998;58:39425.

38. Issa JP, Ahuja N, Toyota M, Bronner MP, Brentnall TA. Acceleratedage-related CpG island methylation in ulcerative colitis. Cancer Res2001;61:35737.

Ushijima and Hattori

Clin Cancer Res; 18(4) February 15, 2012 Clinical Cancer Research928

on May 11, 2019. 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 28, 2011; DOI: 10.1158/1078-0432.CCR-11-2011

http://clincancerres.aacrjournals.org/

39. Shen L,KondoY,RosnerGL, Xiao L, HernandezNS, Vilaythong J, et al.MGMT promoter methylation and field defect in sporadic colorectalcancer. J Natl Cancer Inst 2005;97:13308.

40. Yan PS, Venkataramu C, Ibrahim A, Liu JC, Shen RZ, Diaz NM, et al.Mapping geographic zones of cancer risk with epigenetic biomarkersin normal breast tissue. Clin Cancer Res 2006;12:662636.

41. Arai E, Kanai Y, Ushijima S, Fujimoto H,Mukai K, Hirohashi S. RegionalDNA hypermethylation and DNA methyltransferase (DNMT) 1 proteinoverexpression in both renal tumors and corresponding nontumorousrenal tissues. Int J Cancer 2006;119:28896.

42. Wolff EM, Chihara Y, Pan F, Weisenberger DJ, Siegmund KD, SuganoK, et al. Unique DNAmethylation patterns distinguish noninvasive andinvasive urothelial cancers and establish an epigenetic field defect inpremalignant tissue. Cancer Res 2010;70:816978.

43. Nishiyama N, Arai E, Chihara Y, Fujimoto H, Hosoda F, Shibata T, et al.Genome-wide DNA methylation profiles in urothelial carcinomas andurothelia at the precancerous stage. Cancer Sci 2010;101:23140.

44. Tatematsu M, Nozaki K, Tsukamoto T. Helicobacter pylori infectionand gastric carcinogenesis in animal models. Gastric Cancer 2003;6:17.

45. Niwa T, Tsukamoto T, Toyoda T, Mori A, Tanaka H, Maekita T, et al.Inflammatory processes triggered by Helicobacter pylori infectioncause aberrant DNA methylation in gastric epithelial cells. Cancer Res2010;70:143040.

46. Vitkute J, Stankevicius K, Tamulaitiene G, Maneliene Z, Timinskas A,Berg DE, et al. Specificities of eleven different DNAmethyltransferasesof Helicobacter pylori strain 26695. J Bacteriol 2001;183:44350.

47. Hatakeyama M. Helicobacter pylori and gastric carcinogenesis. JGastroenterol 2009;44:23948.

48. Hur K, Niwa T, Toyoda T, Tsukamoto T, Tatematsu M, Yang HK, et al.Insufficient role of cell proliferation in aberrant DNAmethylation induc-tion and involvement of specific typesof inflammation. Carcinogenesis2011;32:3541.

49. KatsuranoM,Niwa T, Yasui Y, ShigematsuY, Yamashita S, TakeshimaH, et al. Early-stage formation of an epigenetic field defect in a mousecolitis model, and non-essential roles of T- and B-cells in DNA meth-ylation induction. Oncogene 2011. Jun 20 [Epub ahead of print].

50. El-Omar EM, Carrington M, Chow WH, McColl KE, Bream JH, YoungHA, et al. Interleukin-1 polymorphisms associated with increased riskof gastric cancer. Nature 2000;404:398402.

51. LohM,KohKX, YeoBH, SongCM,Chia KS, Zhu F, et al.Meta-analysisof genetic polymorphisms and gastric cancer risk: variability in asso-ciations according to race. Eur J Cancer 2009;45:25628.

52. Hmadcha A, Bedoya FJ, Sobrino F, Pintado E.Methylation-dependentgene silencing induced by interleukin 1beta via nitric oxide production.J Exp Med 1999;190:1595604.

53. McCabe MT, Lee EK, Vertino PM. A multifactorial signature of DNAsequence and polycomb binding predicts aberrant CpG island meth-ylation. Cancer Res 2009;69:28291.

54. Takeshima H, Yamashita S, Shimazu T, Niwa T, Ushijima T. Thepresence of RNA polymerase II, active or stalled, predicts epigeneticfate of promoter CpG islands. Genome Res 2009;19:197482.

55. Estecio MR, Gallegos J, Vallot C, Castoro RJ, Chung W, Maegawa S,et al. Genome architecture marked by retrotransposons modulatespredisposition to DNA methylation in cancer. Genome Res 2010;20:136982.

56. Takeshima H, Yamashita S, Shimazu T, Ushijima T. Effects of genomearchitecture and epigenetic factors on susceptibility of promoter CpGislands to aberrant DNA methylation induction. Genomics 2011;98:1828.

57. Ponder BA. Cancer genetics. Nature 2001;411:33641.58. Amir E, FreedmanOC, Seruga B, Evans DG. Assessing women at high

risk of breast cancer: a review of risk assessment models. J NatlCancer Inst 2010;102:68091.

59. Kim ST, Cheng Y, Hsu FC, Jin T, Kader AK, Zheng SL, et al. Prostatecancer risk-associated variants reported from genome-wide associ-ation studies: meta-analysis and their contribution to genetic variation.Prostate 2010;70:172938.

60. EkstromAM,HeldM,Hansson LE, Engstrand L, NyrenO.Helicobacterpylori in gastric cancer establishedbyCagA immunoblot as amarker ofpast infection. Gastroenterology 2001;121:78491.

61. Eads CA, Nickel AE, Laird PW. Complete genetic suppression ofpolyp formation and reduction of CpG-island hypermethylation inApc(Min/) Dnmt1-hypomorphic Mice. Cancer Res 2002;62:12969.

62. Laird PW, Jackson-Grusby L, Fazeli A, Dickinson SL, Jung WE, Li E,et al. Suppression of intestinal neoplasia byDNAhypomethylation.Cell1995;81:197205.

63. Belinsky SA, Klinge DM, Stidley CA, Issa JP, Herman JG, March TH,et al. Inhibition of DNAmethylation and histone deacetylation preventsmurine lung cancer. Cancer Res 2003;63:708993.

64. McCabe MT, Low JA, Daignault S, Imperiale MJ, Wojno KJ, DayML. Inhibition of DNA methyltransferase activity prevents tumor-igenesis in a mouse model of prostate cancer. Cancer Res 2006;66:38592.

65. Zorn CS, Wojno KJ, McCabe MT, Kuefer R, Gschwend JE, Day ML. 5-aza-2-deoxycytidine delays androgen-independent disease andimproves survival in the transgenic adenocarcinoma of the mouseprostate mouse model of prostate cancer. Clin Cancer Res 2007;13:213643.

66. Chase A, Cross NC. Aberrations of EZH2 in cancer. Clin Cancer Res2011;17:26138.

67. Ding L, Erdmann C, Chinnaiyan AM, Merajver SD, Kleer CG. Iden-tification of EZH2 as a molecular marker for a precancerous statein morphologically normal breast tissues. Cancer Res 2006;66:40959.

68. Cuzick J, Otto F, Baron JA, Brown PH, Burn J, Greenwald P, et al.Aspirin and non-steroidal anti-inflammatory drugs for cancer preven-tion: an international consensus statement. Lancet Oncol 2009;10:5017.

69. Aggarwal BB, Vijayalekshmi RV, Sung B. Targeting inflammatory path-ways for prevention and therapy of cancer: short-term friend, long-term foe. Clin Cancer Res 2009;15:42530.

70. Issa JP, Kantarjian HM. Targeting DNA methylation. Clin Cancer Res2009;15:393846.

Epigenetic Field by H. pylori Infection

www.aacrjournals.org Clin Cancer Res; 18(4) February 15, 2012 929

on May 11, 2019. 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 28, 2011; DOI: 10.1158/1078-0432.CCR-11-2011

http://clincancerres.aacrjournals.org/

2012;18:923-929. Published OnlineFirst December 28, 2011.Clin Cancer Res Toshikazu Ushijima and Naoko Hattori Usefulness as Cancer Risk and Exposure Markers

ItsInflammation in the Formation of an Epigenetic Field Defect, and TriggeredHelicobacter pyloriMolecular Pathways: Involvement of

Updated version

10.1158/1078-0432.CCR-11-2011doi:

Access the most recent version of this article at:

Cited articles

http://clincancerres.aacrjournals.org/content/18/4/923.full#ref-list-1

This article cites 69 articles, 25 of which you can access for free at:

Citing articles

http://clincancerres.aacrjournals.org/content/18/4/923.full#related-urls

This article has been cited by 12 HighWire-hosted articles. Access the articles at:

E-mail alerts related to this article or journal.Sign up to receive free email-alerts

Subscriptions

Reprints and

[email protected]

To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at

Permissions

Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://clincancerres.aacrjournals.org/content/18/4/923To request permission to re-use all or part of this article, use this link

on May 11, 2019. 2012 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 28, 2011; DOI: 10.1158/1078-0432.CCR-11-2011

http://clincancerres.aacrjournals.org/lookup/doi/10.1158/1078-0432.CCR-11-2011http://clincancerres.aacrjournals.org/content/18/4/923.full#ref-list-1http://clincancerres.aacrjournals.org/content/18/4/923.full#related-urlshttp://clincancerres.aacrjournals.org/cgi/alertsmailto:[email protected]://clincancerres.aacrjournals.org/content/18/4/923http://clincancerres.aacrjournals.org/