Review Open Access
Copyright ©2013 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Gastroenterol. Feb 28, 2013; 19(8): 1182-1192
Published online Feb 28, 2013. doi: 10.3748/wjg.v19.i8.1182
DNA and histone methylation in gastric carcinogenesis
Danielle Queiroz Calcagno, Carolina Oliveira Gigek, Elizabeth Suchi Chen, Marília de Arruda Cardoso Smith, Department of Morphology and Genetics, Federal University of Sao Paulo, Sao Paulo, SP 04023-900, Brazil
Rommel Rodriguez Burbano, Laboratory of Human Cytogenetics, Institute of Biological Sciences, Federal University of Pará, Belém, PA 66075-110, Brazil
Author contributions: Calcagno DQ, Gigek CO, Chen ES, Burbano RR and Smith MAC contributed to the review design and wrote the manuscript.
Supported by Sao Paulo State Research Foundation (FAPESP), No. 2009/07145-9 and 2010/11174-1; National Counsel of Technological and Scientific Development (CNPq); Coordination for the Improvement of Higher Level Personnel (CAPES)
Correspondence to: Danielle Queiroz Calcagno, PhD, Division of Genetics, Department of Morphology and Genetics, Federal University of Sao Paulo, Rua Botucatu 740, Sao Paulo, SP 04023-900, Brazil. danicalcagno@gmail.com
Telephone: +55-11-55764260 Fax: +55-11-55764264
Received: February 29, 2012
Revised: June 13, 2012
Accepted: June 28, 2012
Published online: February 28, 2013

Abstract

Epigenetic alterations contribute significantly to the development and progression of gastric cancer, one of the leading causes of cancer death worldwide. Epigenetics refers to the number of modifications of the chromatin structure that affect gene expression without altering the primary sequence of DNA, and these changes lead to transcriptional activation or silencing of the gene. Over the years, the study of epigenetic processes has increased, and novel therapeutic approaches that target DNA methylation and histone modifications have emerged. A greater understanding of epigenetics and the therapeutic potential of manipulating these processes is necessary for gastric cancer treatment. Here, we review recent research on the effects of aberrant DNA and histone methylation on the onset and progression of gastric tumors and the development of compounds that target enzymes that regulate the epigenome.

Key Words: Epigenetic, DNA methylation, Histone methylation, Gastric cancer, Gastric carcinogenesis



INTRODUCTION

Gastric cancer (GC) is the fourth most frequent cancer and is the second leading cause of cancer-related death worldwide[1]. Histologically, gastric tumors are divided into intestinal and diffuse types according to the Lauren classification[2]. The intestinal type of GC mostly progresses through the successive steps of normal gastric mucosa, leading to acute and chronic gastritis, atrophic gastritis, intestinal metaplasia, dysplasia, and finally a gastric tumor[3]. In contrast, the sequence of events in the development of diffuse type GC is poorly understood, although a subset of diffuse type GC appears to develop independently of atrophic gastritis or intestinal metaplasia[4,5]. Differences in the clinicopathological characteristics between these two histological types indicate that development occurs through distinct molecular pathways[6-10]. Each histological type is a consequence of a progressive accumulation of different genetic and epigenetic alterations.

Epigenetics refers to a number of modifications in the chromatin structure that affect gene expression without altering the primary DNA sequence, and these changes lead to transcriptional activation or silencing of the gene. Interestingly, epigenetic modifications of DNA can also increases mutagenesis and influence the interactions between DNA and carcinogens and ultraviolet light[11]. Epigenetic modifications play a central role in gastric carcinogenesis[12]. Recent reports indicate that infection with Helicobacter pylori (H. pylori) or Epstein-Barr virus (EBV), pathogens with a substantial role in development of GC, are associated with elevated levels of aberrant DNA methylation in GC[13-16]. The study of epigenetic processes has increased in recent years, and novel therapeutic approaches that target DNA methylation and histone modifications have emerged. A greater understanding of epigenetics and the therapeutic potential of intervention into these processes is necessary to help GC treatment.

In this review, after a brief introduction to the methylation machinery, we focus on the roles that aberrant DNA and histone methylation play in the onset and progression of gastric tumors, and the development of compounds that target enzymes that regulate the epigenome.

METHYLATION MACHINERY

DNA methylation refers to the addition or subtraction of a methyl moiety at the 5 position of the cytosine ring within CpG dinucleotides that are usually located in CpG-rich regions or CpG islands and around the gene promoter. DNA methylation in gene promoter regions represses transcription of their downstream genes associated with the suppression of gene expression[17]. However, methylation in gene bodies does not block transcription and is sometimes associated with active transcription[18]. Methylation status is controlled by DNA methyltransferases (DNMT1, DNMT3A, and DNMT3B)[19]. DNMT1 maintains the existing methylation patterns following DNA replication, whereas DNMT3A and DNMT3B target unmethylated CpGs to initiate methylation and are highly expressed during embryogenesis and minimally expressed in adult tissues[20]. Another DNA methyltransferase family member, DNMT3L, interacts with DNMT3A and DNMT3B to facilitate methylation of retrotransposons[21]. Many studies have shown that overexpression of DNA methyltransferases is closely related to tumorigenesis, although the role of DNMT3L in cancer is still unclear (Table 1). In addition, H. pylori infection may increase DNA methyltransferase activity through upregulation of the epidermal growth factor and its receptor or via the release of inflammatory mediators, such as nitric oxide[22]. In particular, DNMT1 overexpression has been associated with EBV infection in GC[23-25].

Table 1 Methylation machinery in gastric cancer.
GeneFunctionAlteration in cancerRef.
DNMT1Maintenance of methylation Repression of transcriptionUpregulation MutationKanai et al[93] Fang et al[94] Ding et al[95] Yang et al[96] Mutze et al[97]
DNMT3ADe novo methylation during embryogenesis Imprint establishment RepressionUpregulation MutationDing et al[95] Fan et al[98] Yang et al[96]
DNMT3BDe novo methylation during embryogenesis Repeat methylation RepressionUpregulation MutationDing et al[95] Su et al[99] Hu et al[100] Yang et al[96]
MeCP2Transcription repressionUpregulation MutationWada et al[101]
MBD1Transcription repressionUpregulation Mutation-
MBD2Transcription repression DNA demethylaseDownregulation MutationKanai et al[102]
MBD3Transcription repression, but requires MBD2 to recruit it to methylated DNAUpregulation Mutation-
MBD4Transcription repression DNA repair Glycosylase domain, repair of deaminated 5-methyl CDownregulation MutationPinto et al[38] D'Errico et al[37]
KaisoTranscription repressionUpregulationOgden et al[103]
G9aHistone methyltransferaseGene RepressionLee et al[104]
RIZ1Histone methyltransferaseUnderexpressionOshimo et al[105]
PRDM2MutationPan et al[106]
SUZ12Histone methyltransferaseUpregulationYoo et al[107]
BMI1Histone methyltransferaseUpreguletionLiu et al[108] Xiao et al[109] Lu et al[110] Zhang et al[111] Li et al[112]
EVI1Histone methyltransferaseChromosomal rearrangementTakahata et al[113]
EZH2Histone methyltransferaseAmplification Upregulation MutationMattioli et al[114] Varambally et al[115] Fujii et al[48] Cai et al[47] Choi et al[46] Zhou et al[116]
NSD2/MMMSETHistone methyltransferaseUpregulation TranslocationHudlebusch et al[117]
SUV39H1 -2Histone methyltransferasePolymorphismLi et al[84]
LSD1/BHC110Histone demethylaseDownregulationMagerl et al[118]
JARID1A-DHistone demethylaseUpregulation InactivationZeng et al[51]
JMJD2AHistone demethylaseMutation UpregulationLi et al[119]
JHDM3A
JMJD1A-CHistone demethylaseDownregulationKatoh et al[120]

DNA methylation has also been implicated in the regulation of higher order chromatin structure, the maintenance of genome integrity, and stable patterns of gene expression. These biological effects of DNA methylation are, at least in part, mediated by proteins that preferentially bind to methylated DNA[26]. Methylated DNA is specifically recognized by a set of proteins called methyl-CpG-binding proteins (MBPs), which belong to three different structural families: methyl-CpG binding domain proteins (MBDs), Kaiso domain proteins, and SET and RING finger-associated domain (SRA) domain proteins[27,28]. MBD family proteins (MeCP2, MBD1, MBD2, MBD3 and MBD4) bind methylated CpG (5mCpG) through a conserved protein motif called the methyl-CpG binding domain[29,30]. Over the last decade, proteins that utilize different structures to recognize and bind DNA or its components have been identified. In 2001, Prokhortchouk et al[31] identified Kaiso proteins, which bind methylated DNA through a zinc finger motif. Other MBPs including UHRF1 and UHRF2 were identified, and these proteins use the SRA to bind 5mCpG[32,33].

In cancer, the roles of MBPs are related to their functions as transcriptional repressors or chromatin remodelers (Table 1)[34-36]. However, a few studies have reported MBPs in GC (Table 1). Mutations in MBD4 have been found in gastric tumors in association with microsatellite instability[37,38]. MBD4 encodes a protein that interacts with the mismatch repair protein hMLH1. Therefore, it has been postulated that mutations in MBD4 may result in mismatch repair deficiency[30].

The processes of DNA methylation and histone modification often involve dynamic interactions that either reinforce or inhibit epigenetic changes. Thus, histone modification can also alter chromatin remodeling, and this is a possible mechanism for decreased gene expression[39-41].

The nature of the interaction between DNA and histones, which are composed of pairs of the four core proteins H2A, H2B, H3, and H4, alters the accessibility of DNA transcription sites to RNA polymerase II and other transcription factors. The interaction between histones and DNA is thought to be under epigenetic control, because specific amino acid residues on specific histone core proteins are subjected to post-translational modifications, such as acetylation, methylation, phosphorylation, ubiquitination, sumoylation, proline isomerization, and ADP ribosylation[42,43]. Histone acetylation and methylation are the only modifications that have been clinically associated with pathological epigenetic disruption in cancer cells[44]. In this review, we focus on histone methylation modifications.

Histones can be mono-, di-, or trimethylated at lysine and arginine residues by histone methyltransferases (HMTs) or demethylated by histone demethylases (HDTs). Depending on the residue and the level of methylation, the chromatin may be transcriptionally active or inactive. In general, trimethylation at H3K4 and H3K36 or monomethylation at H3K27, H3K9, H4K20, H3K79, and H2BK5 is associated with transcriptional activation. In contrast, trimethylation at H3K27, H3K9, and H4K20 or monomethylation at H3K27, H3K9, H4K20, H3K79, and H2BK5 is associated with transcriptional repression[44].

A growing number of studies have analyzed the HMTs and HDMs in tumor cells, whereas few genes involved in histone methylation activity have been described for GC (Table 1). EZH2, an HMT that plays a role in trimethylation of H3K27 and leads to silencing of important genes in carcinogenesis, is overexpressed in several types of cancer, including GC[45,46]. Cai et al[47] reported that EZH2 plays an important role in the multi-step process of intestinal-type GC. In addition, Fujii et al[48] demonstrated that silencing of EZH2 by siRNA resulted in a lower H3K27me3 protein level in GC cells.

Among the HDTs, RBP2 is a newly identified member of the JARID family of proteins, and RBP2 specifically targets tri- and dimethylated H3K4 for demethylation in cancer[49,50]. Zeng et al[51] reported that RBP2 is overexpressed in GC and suggested that HDT inhibition by targeting RBP2 may be an anticancer strategy.

DNA METHYLATION

DNA methylation contributes to cancer mainly through DNA hypo- or hypermethylation. DNA hypomethylation, which refers to the loss of DNA methylation, affects chromosomal stability and increases aneuploidy[52]. DNA hypermethylation, which refers to the gain of methylation at a locus originally unmethylated, usually results in stable transcriptional silencing, which functions in regulating gene expression[53,54].

Global DNA hypomethylation is usually considered one of the hallmarks of cancer cells, because aberrant hypermethylation-vulnerable genes are overlapped by genes targeted by hypomethylation[55,56]. Compare et al[57] suggested that global DNA hypomethylation may be implicated in GC associated with H. pylori infection at an early stage. At the individual gene level, DNA hypomethylation is often associated with activation of proto-oncogenes.

In GC, few studies have shown promoter hypomethylation associated with the activation of proto-oncogenes (Table 2). In particular, Shin et al[58] reported that the hypomethylation of the MOS promoter in GC was associated with tumor invasion, lymph node metastasis, and the diffuse type. A number of genes involved in cell cycle regulation, tumor cell invasion, DNA repair, chromatin remodeling, cell signaling, transcription, and apoptosis are known to be silenced by hypermethylation in GC (Table 2).

Table 2 Aberrant DNA methylation in gastric cancer.
GeneRoleAberrant methylationRef.
ABCB1Multidrug resistanceHyperPoplawski et al[121], Tahara et al[122], Lee et al[123]
ADAM23Tissue cell invasion and metastasisHyperTakada et al[124], Watanabe et al[125], Kim et al[126]
ALDH2Oxidative pathway of alcohol metabolismHypoBalassiano et al[127]
APCTissue cell invasion and metastasis Signal transductionHyperBernal et al[128], Ksiaa et al[63], Shin et al[69], Geddert et al[129]
ARPC1B (p41ARC)Cell morphologyHyperMaekita et al[130], Shin et al[69]
BNIP3ApoptosisHyperMurai et al[131], Hiraki et al[132], Sugita et al[133]
BRCA1DNA repairHyperBernal et al[128], Ryan et al[134]
CAV1Tissue cell invasion and metastasisHyperYamashida et al[135]
CDH1Tissue invasion and metastasisHyperLeal et al[136], Bernal et al[136], Borges et al[61], Tahara et al[122], Al-Moundhri et al[137], Balassiano et al[127]
CHFRCell cycle regulationHyperOki et al[138], Hiraki et al[139], Hu et al[140]
DAPKApoptosisHyperBernal et al[128], Zou et al[74], Hu et al[140], Tahara et al[122], Sugita et al[133]
FHITApoptosisHyperLeal et al[136], Bernal et al[128]
FLNCCell morphologyHyperKim et al[126], Shi et al[141]
GATA4/5Transcriptional factorHyperAkiyama et al[142], Wen et al[143],
HAND1Cell differentiationHyperMaekita et al[130], Shin et al[69], Shi et al[141]
HRASSignal transductionHypoFang et al[144], Luo et al[145]
IGFBP3Cell cycle regulationHyperGigek et al[146], Ryan et al[134], Chen et al[147]
LOXTissue cell invasion and adhesionHyperMaekita et al[130], Shin et al[69], Tamura et al[148]
MGMTDNA repairHyperBernal et al[128], Hibi et al[149], Ksiaa et al[63]; Zou et al[74], Schneider et al[14], Hiraki et al[139], Balassiano et al[127], Shi et al[141]
MLF1Cell differentiationHyperWatanabe et al[125], Shi et al[141], Yamashita et al[135]
MLH1DNA repairHyperBernal et al[128], Poplawski et al[121], Hiraki et al[139], Kim et al[150], Shin et al[58]
MOSCell cycle regulationHypoShin et al[58]
MTHFRDNA synthesis DNA repair DNA methylationHypoBalassiano et al[127]
MYCCell cycle regulationHypoFang et al[144], Luo et al[145]
P14ARFCell cycle regulation Apoptosis Cell differentiationHyperBalassiano et al[127], Geddert et al[129]
P16Cell cycle regulationHyperKsiaa et al[63], Dong et al[151], Zou et al[74], Shin et al[69], Hu et al[140], Ryan et al[134], Geddert et al[129], Balassiano et al[124], Al-Moundhri et al[137], Shin et al[58]
PRDM5Cell differentiationHyperWatanabe et al[125], Shu et al[152]
RAR-beta 2DNA binding Activation transcriptionHyperBernal et al[128], Ksiaa et al[63]
RASSF1A/ RASSF2DNA repair Cell cycle regulationHyperZou et al[74], Guo et al[153], Shin et al[58]
RORACell differentiationHyperWatanabe et al[125], Yamashida et al[131]
RPRMCell cycle regulationHyperBernal et al[128], Schneider et al[14]
RUNX3Signal transductionHyperBernal et al[128], Sakakura et al[154], Lee et al[104], Zou et al[74], Hiraki et al[139], Tamura et al[148], Hu et al[140], Fan et al[155], Al-Moundhri et al[137]
SHP1Signal transductionHyperBernal et al[128], Ksiaa et al[63],
TERTCell senescenceHyperKang et al[67], Wang et al[75], Gigek et al[77]
TFF1Repair geneHyperCarvalho et al[156], Ryan et al[134]
THBDInflammation responseHyperMaekita et al[130]; Shin et al[69]
TWIST1Cell differentiationHyperKang et al[67], Schneider et al[14]

Multiple reports have been published regarding gene hypermethylation in both intestinal and diffuse types of GC. Interestingly, the methylation profile differs between the intestinal and diffuse types of GC[54].

The epithelial cadherin gene CDH1, which is a well-studied gene involved in cancer, is downregulated in gastric tumors and is hypermethylated more frequently in the diffuse type than in the intestinal type of GC. Loss of CDH1 during tumor progression has led to the notion that this is a tumor suppressor gene[59,60]. In addition, mapping of the CDH1 promoter has revealed a positive association between hypermethylation and older age, as well as a significant correlation between DNA hypermethylation and the A allele of the -160 C→A polymorphism. The A allele has been described to increase the risk of developing GC in association with the methylation status[61]. Unlike the CDH1 gene, the P16 gene is hypermethylated mainly in the intestinal type of GC[54,62,63]. This epigenetic mark was recently associated with tumor location and H. pylori infection in GC[64].

Other studies have also described a number of genes that are silenced by hypermethylation in association with H. pylori or EBV infection: APC, SHP1, p14, and CDH1[63,65-67]. According to Chan et al[68], the eradication of H. pylori infection significantly reduces the methylation index of the CDH1 promoter. In contrast, it has been shown that a portion of the aberrant DNA methylation induced by H. pylori infection may persist even after the infection has disappeared[69,70]. Shin et al[58] observed that the methylation levels in MOS remained significantly increased in patients with previous H. pylori infection compared with H. pylori-negative subjects.

Moreover, hypermethylation of several gene promoters has also been observed in the premalignant stages of GC, suggesting that aberrant methylation occurs early during gastric carcinogenesis[59,71-74]. For example, the methylation levels of the catalytic subunit of the telomerase gene (hTERT) promoter are increased during gastric carcinogenesis. Wang et al[75] reported that the hTERT promoter was more methylated in GC than in precancerous lesions and non-neoplastic gastric tissues. Therefore, it has been suggested that the degree of methylation of the hTERT promoter may be useful in the early diagnosis of GC and/or may have an impact on the anti-telomerase strategy for cancer therapy. Other studies, however, showed that methylation of the hTERT promoter and resultant gene expression were opposite to the general model of regulation by DNA methylation, which is usually dependent on the CpG islands studied[76,77].

Recently, aberrant hypermethylation of the newly associated metastatic suppressor gene RECK was found to be associated with GC development and may also be useful for early diagnosis and treatment[78]. These abovementioned findings lead to the possibilities that epigenetic alterations may also occur at different stages of gastric tumorigenesis.

HISTONE METHYLATION

Histone modifications leading to gene expression alterations have been described in several cancer types, but the methylation status of chromatin is still unclear for GC. Using the ChIP-on-chip technique, Zhang et al[79] identified candidate genes with significant differences in H3K27me3 in GC samples compared to adjacent non-neoplastic gastric tissues. These genes included oncogenes, tumor suppressor genes, cell cycle regulators, and genes involved in cell adhesion. Moreover, these investigators demonstrated that higher levels of H3K27me3 produce gene expression changes in MMP15, UNC5B, and SHH.

In 2011, Kwon et al[80] showed that LAMB3 and LAMC2 were overexpressed in GC samples in comparison with non-neoplastic adjacent tissue samples. Furthermore, these researchers demonstrated that overexpression of these genes was a result of the enrichment of H3K4me3 in the gene promoter. Using immunohistochemistry, Park et al[81] showed that higher levels of H3K9me3, which is a repressive mark, was associated with higher T stage, lymphovascular invasion, and recurrence in gastric tumors. They also observed that the level of H3K9me3 was correlated with patient survival, because stronger methylation corresponded to a worse prognosis and intermediate methylation to an intermediate prognosis.

Taken together with results from previous studies, these results have suggested that histone methylation results in a worse prognosis by inactivating certain tumor suppressor genes[82,83]. Moreover, Li et al[84] used GC cell lines to demonstrate that the PRC1 member CBX7 initiated trimethylation of H3K9 at the P16 locus through recruitment and/or activation of the HMT SUV39H2 to the target locus. This finding links two repressive epigenetic landmarks, H3K9me3 formation and PRC1 binding within the silenced domains in euchromatin, and builds up a full pathway for epigenetic inactivation of P16 by histone modifications.

Recently, Angrisano et al[85] reported that H. pylori infection is followed by activation of iNOS gene expression, chromatin changes at the iNOS promoter (including decreased H3K9 methylation and increased H3K4 methylation), and selective release of MBD2 from the iNOS promoter in a GC cell line.

METHYLATION INHIBITOR DRUGS

The silencing of cancer-related genes by DNA methylation and chromatin modification are reversible and may represent a viable epigenetic therapeutic target. In the last decade, drugs that modify chromatin or DNA methylation status have been used alone or in combination in order to affect therapeutic outcomes[86]. Specially, cytosine analogs (5-azacytidine and 5-aza-2’- deoxycytidine) are powerful mechanism-based inhibitors of DNA cytosine methylation. These cytosine analogs are incorporated into the DNA of replicating cells after the drugs have been metabolized to the appropriate dNTP. After incorporation into the DNA, the analogs interact with DNA methyltransferases to form covalent intermediates, and this interaction inhibits DNA methylation in subsequent rounds of DNA synthesis[87]. Both drugs have been approved by the US Food and Drug Administration for use in hematological malignancy treatment[88].

In GC, surgery remains the primary curative treatment for gastric tumors. Currently, adjuvant and neoadjuvant therapies are accepted[89]; however, so-called epigenetic therapy has not yet been used in treatment of GC patients.

In the past few years, epigenetic screening techniques using treatment with a demethylating agent have been developed to identify genes with epigenetic aberrations in GC cell lines. Zheng et al[90] treated a GC cell line with 5-aza-2’-deoxycytidine and performed DNA methylation array analysis of these cells with six normal mucosal samples from healthy patients. These results revealed 82 hypermethylated gene promoters. These authors investigated 15 candidate genes by methylation-specific PCR and confirmed five highly methylated promoters: BX141696, WT1, CYP26B1, KCNA4, and FAM84A. All of these, except FAM84A, also showed DNA hypermethylation in serum of GC patients, suggesting that serum DNA offers a readily accessible bioresource for methylation analysis.

A similar study conducted by Jee et al[91] described 11 selected genes and validated the genes in three GC cell lines and in non-neoplastic gastric tissue by bisulfate sequencing. Differential DNA hypermethylation was observed in GPX1, IGFBP6, IRF7, GPX3, TFPI2, and DMRT1 promoter regions in GC cells but not in non-neoplastic tissues. Moreover, a poor survival rate was observed in those individuals with higher methylation status at the TFPI2 gene. TFPI2 is a serine protease inhibitor, which negatively regulates the enzymatic activities of trypsin, plasmin, and a tissue factor complex. Therefore, it has been proposed that this gene inactivation may be implicated in human carcinogenesis and metastasis[92].

CONCLUSION

In summary, aberrant DNA methylation and histone modification play a crucial role in gastric carcinogenesis. Thus, the recognition of the methylation machinery, genes with aberrant methylation status, and histone methylation levels in gastric carcinogenesis exemplified in this review allow us to contemplate the possibility of dealing with the aforementioned oncological issue in a new way that may have a significant impact on the therapy and management of GC.

Footnotes

P- Reviewers Feo F, Shibata T S- Editor Gou SX L- Editor Kerr C E- Editor Zhang DN

References
1.  Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J Clin. 2011;61:69-90.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23762]  [Cited by in F6Publishing: 25182]  [Article Influence: 1937.1]  [Reference Citation Analysis (3)]
2.  Lauren P. The two histological main types of gastric carcinoma: diffuse and so-called intestinal-type carcinoma. an attempt at a histo-clinical classification. Acta Pathol Microbiol Scand. 1965;64:31-49.  [PubMed]  [DOI]  [Cited in This Article: ]
3.  Correa P, Haenszel W, Cuello C, Tannenbaum S, Archer M. A model for gastric cancer epidemiology. Lancet. 1975;2:58-60.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Humar B, Guilford P. Hereditary diffuse gastric cancer: a manifestation of lost cell polarity. Cancer Sci. 2009;100:1151-1157.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 69]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
5.  Carneiro F, Huntsman DG, Smyrk TC, Owen DA, Seruca R, Pharoah P, Caldas C, Sobrinho-Simões M. Model of the early development of diffuse gastric cancer in E-cadherin mutation carriers and its implications for patient screening. J Pathol. 2004;203:681-687.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 196]  [Cited by in F6Publishing: 208]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
6.  Tahara E. Genetic pathways of two types of gastric cancer. IARC Sci Publ. 2004;327-349.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Calcagno DQ, Guimarães AC, Leal MF, Seabra AD, Khayat AS, Pontes TB, Assumpção PP, De Arruda Cardoso Smith M, Burbano RR. MYC insertions in diffuse-type gastric adenocarcinoma. Anticancer Res. 2009;29:2479-2483.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Calcagno DQ, Leal MF, Assumpcao PP, Smith MA, Burbano RR. MYC and gastric adenocarcinoma carcinogenesis. World J Gastroenterol. 2008;14:5962-5968.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Calcagno DQ, Leal MF, Seabra AD, Khayat AS, Chen ES, Demachki S, Assumpção PP, Faria MH, Rabenhorst SH, Ferreira MV. Interrelationship between chromosome 8 aneuploidy, C-MYC amplification and increased expression in individuals from northern Brazil with gastric adenocarcinoma. World J Gastroenterol. 2006;12:6207-6211.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Calcagno DQ, Leal MF, Taken SS, Assumpção PP, Demachki S, Smith Mde A, Burbano RR. Aneuploidy of chromosome 8 and C-MYC amplification in individuals from northern Brazil with gastric adenocarcinoma. Anticancer Res. 2005;25:4069-4074.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Pfeifer GP, Tang M, Denissenko MF. Mutation hotspots and DNA methylation. Curr Top Microbiol Immunol. 2000;249:1-19.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Gigek CO, Chen ES, Calcagno DQ, Wisnieski F, Burbano RR, Smith MA. Epigenetic mechanisms in gastric cancer. Epigenomics. 2012;4:279-294.  [PubMed]  [DOI]  [Cited in This Article: ]
13.  Ferrasi AC, Pinheiro NA, Rabenhorst SH, Caballero OL, Rodrigues MA, de Carvalho F, Leite CV, Ferreira MV, Barros MA, Pardini MI. Helicobacter pylori and EBV in gastric carcinomas: methylation status and microsatellite instability. World J Gastroenterol. 2010;16:312-319.  [PubMed]  [DOI]  [Cited in This Article: ]
14.  Schneider BG, Peng DF, Camargo MC, Piazuelo MB, Sicinschi LA, Mera R, Romero-Gallo J, Delgado AG, Bravo LE, Wilson KT. Promoter DNA hypermethylation in gastric biopsies from subjects at high and low risk for gastric cancer. Int J Cancer. 2010;127:2588-2597.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 49]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
15.  Shin CM, Kim N, Jung Y, Park JH, Kang GH, Park WY, Kim JS, Jung HC, Song IS. Genome-wide DNA methylation profiles in noncancerous gastric mucosae with regard to Helicobacter pylori infection and the presence of gastric cancer. Helicobacter. 2011;16:179-188.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 34]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
16.  Matsusaka K, Kaneda A, Nagae G, Ushiku T, Kikuchi Y, Hino R, Uozaki H, Seto Y, Takada K, Aburatani H. Classification of Epstein-Barr virus-positive gastric cancers by definition of DNA methylation epigenotypes. Cancer Res. 2011;71:7187-7197.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 159]  [Cited by in F6Publishing: 178]  [Article Influence: 13.7]  [Reference Citation Analysis (0)]
17.  Jones PA, Takai D. The role of DNA methylation in mammalian epigenetics. Science. 2001;293:1068-1070.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1432]  [Cited by in F6Publishing: 1352]  [Article Influence: 58.8]  [Reference Citation Analysis (0)]
18.  Baylin SB, Ohm JE. Epigenetic gene silencing in cancer - a mechanism for early oncogenic pathway addiction? Nat Rev Cancer. 2006;6:107-116.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1182]  [Cited by in F6Publishing: 1155]  [Article Influence: 64.2]  [Reference Citation Analysis (0)]
19.  Kim H, Park J, Jung Y, Song SH, Han SW, Oh DY, Im SA, Bang YJ, Kim TY. DNA methyltransferase 3-like affects promoter methylation of thymine DNA glycosylase independently of DNMT1 and DNMT3B in cancer cells. Int J Oncol. 2010;36:1563-1572.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Okano M, Bell DW, Haber DA, Li E. DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell. 1999;99:247-257.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Wienholz BL, Kareta MS, Moarefi AH, Gordon CA, Ginno PA, Chédin F. DNMT3L modulates significant and distinct flanking sequence preference for DNA methylation by DNMT3A and DNMT3B in vivo. PLoS Genet. 2010;6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 105]  [Article Influence: 7.5]  [Reference Citation Analysis (0)]
22.  Wallasch C, Crabtree JE, Bevec D, Robinson PA, Wagner H, Ullrich A. Helicobacter pylori-stimulated EGF receptor transactivation requires metalloprotease cleavage of HB-EGF. Biochem Biophys Res Commun. 2002;295:695-701.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Etoh T, Kanai Y, Ushijima S, Nakagawa T, Nakanishi Y, Sasako M, Kitano S, Hirohashi S. Increased DNA methyltransferase 1 (DNMT1) protein expression correlates significantly with poorer tumor differentiation and frequent DNA hypermethylation of multiple CpG islands in gastric cancers. Am J Pathol. 2004;164:689-699.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 207]  [Cited by in F6Publishing: 215]  [Article Influence: 10.8]  [Reference Citation Analysis (0)]
24.  Hino R, Uozaki H, Murakami N, Ushiku T, Shinozaki A, Ishikawa S, Morikawa T, Nakaya T, Sakatani T, Takada K. Activation of DNA methyltransferase 1 by EBV latent membrane protein 2A leads to promoter hypermethylation of PTEN gene in gastric carcinoma. Cancer Res. 2009;69:2766-2774.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 258]  [Cited by in F6Publishing: 272]  [Article Influence: 18.1]  [Reference Citation Analysis (0)]
25.  Fukayama M. Epstein-Barr virus and gastric carcinoma. Pathol Int. 2010;60:337-350.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 102]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
26.  Bogdanović O, Veenstra GJ. DNA methylation and methyl-CpG binding proteins: developmental requirements and function. Chromosoma. 2009;118:549-565.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 320]  [Cited by in F6Publishing: 320]  [Article Influence: 21.3]  [Reference Citation Analysis (0)]
27.  Fournier A, Sasai N, Nakao M, Defossez PA. The role of methyl-binding proteins in chromatin organization and epigenome maintenance. Brief Funct Genomics. 2012;11:251-264.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 72]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
28.  Defossez PA, Stancheva I. Biological functions of methyl-CpG-binding proteins. Prog Mol Biol Transl Sci. 2011;101:377-398.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 75]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
29.  Hendrich B, Bird A. Identification and characterization of a family of mammalian methyl-CpG binding proteins. Mol Cell Biol. 1998;18:6538-6547.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Bellacosa A. Role of MED1 (MBD4) Gene in DNA repair and human cancer. J Cell Physiol. 2001;187:137-144.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 77]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
31.  Prokhortchouk A, Hendrich B, Jørgensen H, Ruzov A, Wilm M, Georgiev G, Bird A, Prokhortchouk E. The p120 catenin partner Kaiso is a DNA methylation-dependent transcriptional repressor. Genes Dev. 2001;15:1613-1618.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 371]  [Cited by in F6Publishing: 357]  [Article Influence: 15.5]  [Reference Citation Analysis (0)]
32.  Unoki M, Nishidate T, Nakamura Y. ICBP90, an E2F-1 target, recruits HDAC1 and binds to methyl-CpG through its SRA domain. Oncogene. 2004;23:7601-7610.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 219]  [Cited by in F6Publishing: 236]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
33.  Hashimoto H, Horton JR, Zhang X, Cheng X. UHRF1, a modular multi-domain protein, regulates replication-coupled crosstalk between DNA methylation and histone modifications. Epigenetics. 2009;4:8-14.  [PubMed]  [DOI]  [Cited in This Article: ]
34.  Sansom OJ, Maddison K, Clarke AR. Mechanisms of disease: methyl-binding domain proteins as potential therapeutic targets in cancer. Nat Clin Pract Oncol. 2007;4:305-315.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 87]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
35.  Lopez-Serra L, Ballestar E, Ropero S, Setien F, Billard LM, Fraga MF, Lopez-Nieva P, Alaminos M, Guerrero D, Dante R. Unmasking of epigenetically silenced candidate tumor suppressor genes by removal of methyl-CpG-binding domain proteins. Oncogene. 2008;27:3556-3566.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 48]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
36.  Parry L, Clarke AR. The Roles of the Methyl-CpG Binding Proteins in Cancer. Genes Cancer. 2011;2:618-630.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 85]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
37.  D’Errico M, de Rinaldis E, Blasi MF, Viti V, Falchetti M, Calcagnile A, Sera F, Saieva C, Ottini L, Palli D. Genome-wide expression profile of sporadic gastric cancers with microsatellite instability. Eur J Cancer. 2009;45:461-469.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 212]  [Cited by in F6Publishing: 250]  [Article Influence: 15.6]  [Reference Citation Analysis (0)]
38.  Pinto M, Wu Y, Suriano G, Mensink RG, Duval A, Oliveira C, Carvalho B, Hamelin R, Seruca R, Hofstra RM. MBD4 mutations are rare in gastric carcinomas with microsatellite instability. Cancer Genet Cytogenet. 2003;145:103-107.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Shilatifard A. Chromatin modifications by methylation and ubiquitination: implications in the regulation of gene expression. Annu Rev Biochem. 2006;75:243-269.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 836]  [Cited by in F6Publishing: 809]  [Article Influence: 47.6]  [Reference Citation Analysis (0)]
40.  Weisbrod S. Active chromatin. Nature. 1982;297:289-295.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Richards EJ, Elgin SC. Epigenetic codes for heterochromatin formation and silencing: rounding up the usual suspects. Cell. 2002;108:489-500.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Zhang Y, Reinberg D. Transcription regulation by histone methylation: interplay between different covalent modifications of the core histone tails. Genes Dev. 2001;15:2343-2360.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1142]  [Cited by in F6Publishing: 1089]  [Article Influence: 47.3]  [Reference Citation Analysis (0)]
43.  Sawan C, Herceg Z. Histone modifications and cancer. Adv Genet. 2010;70:57-85.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 141]  [Cited by in F6Publishing: 155]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
44.  Ellis L, Atadja PW, Johnstone RW. Epigenetics in cancer: targeting chromatin modifications. Mol Cancer Ther. 2009;8:1409-1420.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 339]  [Cited by in F6Publishing: 323]  [Article Influence: 21.5]  [Reference Citation Analysis (0)]
45.  Matsukawa Y, Semba S, Kato H, Ito A, Yanagihara K, Yokozaki H. Expression of the enhancer of zeste homolog 2 is correlated with poor prognosis in human gastric cancer. Cancer Sci. 2006;97:484-491.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 167]  [Cited by in F6Publishing: 177]  [Article Influence: 9.8]  [Reference Citation Analysis (0)]
46.  Choi JH, Li Y, Guo J, Pei L, Rauch TA, Kramer RS, Macmil SL, Wiley GB, Bennett LB, Schnabel JL. Genome-wide DNA methylation maps in follicular lymphoma cells determined by methylation-enriched bisulfite sequencing. PLoS One. 2010;5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 32]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
47.  Cai GH, Wang K, Miao Q, Peng YS, Chen XY. Expression of polycomb protein EZH2 in multi-stage tissues of gastric carcinogenesis. J Dig Dis. 2010;11:88-93.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 15]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
48.  Fujii S, Ito K, Ito Y, Ochiai A. Enhancer of zeste homologue 2 (EZH2) down-regulates RUNX3 by increasing histone H3 methylation. J Biol Chem. 2008;283:17324-17332.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 140]  [Cited by in F6Publishing: 151]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
49.  Christensen J, Agger K, Cloos PA, Pasini D, Rose S, Sennels L, Rappsilber J, Hansen KH, Salcini AE, Helin K. RBP2 belongs to a family of demethylases, specific for tri-and dimethylated lysine 4 on histone 3. Cell. 2007;128:1063-1076.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 400]  [Cited by in F6Publishing: 414]  [Article Influence: 24.4]  [Reference Citation Analysis (0)]
50.  Lopez-Bigas N, Kisiel TA, Dewaal DC, Holmes KB, Volkert TL, Gupta S, Love J, Murray HL, Young RA, Benevolenskaya EV. Genome-wide analysis of the H3K4 histone demethylase RBP2 reveals a transcriptional program controlling differentiation. Mol Cell. 2008;31:520-530.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 110]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
51.  Zeng J, Ge Z, Wang L, Li Q, Wang N, Björkholm M, Jia J, Xu D. The histone demethylase RBP2 Is overexpressed in gastric cancer and its inhibition triggers senescence of cancer cells. Gastroenterology. 2010;138:981-992.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Gaudet F, Hodgson JG, Eden A, Jackson-Grusby L, Dausman J, Gray JW, Leonhardt H, Jaenisch R. Induction of tumors in mice by genomic hypomethylation. Science. 2003;300:489-492.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1147]  [Cited by in F6Publishing: 1070]  [Article Influence: 51.0]  [Reference Citation Analysis (0)]
53.  Esteller M, Corn PG, Baylin SB, Herman JG. A gene hypermethylation profile of human cancer. Cancer Res. 2001;61:3225-3229.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Selaru FM, David S, Meltzer SJ, Hamilton JP. Epigenetic events in gastrointestinal cancer. Am J Gastroenterol. 2009;104:1910-1912.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 33]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
55.  Yoshida T, Yamashita S, Takamura-Enya T, Niwa T, Ando T, Enomoto S, Maekita T, Nakazawa K, Tatematsu M, Ichinose M. Alu and Satα hypomethylation in Helicobacter pylori-infected gastric mucosae. Int J Cancer. 2011;128:33-39.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 51]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
56.  Najjar Sadeghi R, Zojaji H, Mohebbi SR, Chiani M, Vahedi M, Mirsattari D, Molaei M, Mashayekhi R, Zali MR. Evaluation of global genome methylation in gastritis lesion and its correlation with clinicopatological findings. Oncol Res. 2009;17:549-558.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Compare D, Rocco A, Liguori E, D’Armiento FP, Persico G, Masone S, Coppola-Bottazzi E, Suriani R, Romano M, Nardone G. Global DNA hypomethylation is an early event in Helicobacter pylori-related gastric carcinogenesis. J Clin Pathol. 2011;64:677-682.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 42]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
58.  Shin CM, Kim N, Park JH, Kang GH, Kim JS, Jung HC, Song IS. Prediction of the risk for gastric cancer using candidate methylation markers in the non-neoplastic gastric mucosae. J Pathol. 2012;226:654-665.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 29]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
59.  Yamamoto E, Suzuki H, Takamaru H, Yamamoto H, Toyota M, Shinomura Y. Role of DNA methylation in the development of diffuse-type gastric cancer. Digestion. 2011;83:241-249.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 43]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
60.  Cavallaro U, Christofori G. Multitasking in tumor progression: signaling functions of cell adhesion molecules. Ann N Y Acad Sci. 2004;1014:58-66.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Borges Bdo N, Santos Eda S, Bastos CE, Pinto LC, Anselmo NP, Quaresma JA, Calcagno DQ, Burbano RM, Harada ML. Promoter polymorphisms and methylation of E-cadherin (CDH1) and KIT in gastric cancer patients from northern Brazil. Anticancer Res. 2010;30:2225-2233.  [PubMed]  [DOI]  [Cited in This Article: ]
62.  Lima EM, Leal MF, Burbano RR, Khayat AS, Assumpção PP, Bello MJ, Rey JA, Smith MA, Casartelli C. Methylation status of ANAPC1, CDKN2A and TP53 promoter genes in individuals with gastric cancer. Braz J Med Biol Res. 2008;41:539-543.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Ksiaa F, Ziadi S, Amara K, Korbi S, Trimeche M. Biological significance of promoter hypermethylation of tumor-related genes in patients with gastric carcinoma. Clin Chim Acta. 2009;404:128-133.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 62]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
64.  Alves MK, Lima VP, Ferrasi AC, Rodrigues MA, De Moura Campos Pardini MI, Rabenhorst SH. CDKN2A promoter methylation is related to the tumor location and histological subtype and associated with Helicobacter pylori flaA(+) strains in gastric adenocarcinomas. APMIS. 2010;118:297-307.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 14]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
65.  Ding SZ, Fischer W, Kaparakis-Liaskos M, Liechti G, Merrell DS, Grant PA, Ferrero RL, Crowe SE, Haas R, Hatakeyama M. Helicobacter pylori-induced histone modification, associated gene expression in gastric epithelial cells, and its implication in pathogenesis. PLoS One. 2010;5:e9875.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 71]  [Cited by in F6Publishing: 78]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
66.  Hu XT, He C. Recent progress in the study of methylated tumor suppressor genes in gastric cancer. Chin J Cancer. 2013;32:31-41.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 26]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
67.  Kang GH, Lee S, Cho NY, Gandamihardja T, Long TI, Weisenberger DJ, Campan M, Laird PW. DNA methylation profiles of gastric carcinoma characterized by quantitative DNA methylation analysis. Lab Invest. 2008;88:161-170.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 126]  [Cited by in F6Publishing: 134]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
68.  Chan AO. E-cadherin in gastric cancer. World J Gastroenterol. 2006;12:199-203.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Shin CM, Kim N, Yang HJ, Cho SI, Lee HS, Kim JS, Jung HC, Song IS. Stomach cancer risk in gastric cancer relatives: interaction between Helicobacter pylori infection and family history of gastric cancer for the risk of stomach cancer. J Clin Gastroenterol. 2010;44:e34-e39.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 93]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
70.  Nakajima T, Enomoto S, Yamashita S, Ando T, Nakanishi Y, Nakazawa K, Oda I, Gotoda T, Ushijima T. Persistence of a component of DNA methylation in gastric mucosae after Helicobacter pylori eradication. J Gastroenterol. 2010;45:37-44.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 98]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
71.  Kang GH, Shim YH, Jung HY, Kim WH, Ro JY, Rhyu MG. CpG island methylation in premalignant stages of gastric carcinoma. Cancer Res. 2001;61:2847-2851.  [PubMed]  [DOI]  [Cited in This Article: ]
72.  Kang GH, Lee S, Kim JS, Jung HY. Profile of aberrant CpG island methylation along the multistep pathway of gastric carcinogenesis. Lab Invest. 2003;83:635-641.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Jang BG, Kim WH. Molecular pathology of gastric carcinoma. Pathobiology. 2011;78:302-310.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 94]  [Article Influence: 7.2]  [Reference Citation Analysis (0)]
74.  Zou XP, Zhang B, Zhang XQ, Chen M, Cao J, Liu WJ. Promoter hypermethylation of multiple genes in early gastric adenocarcinoma and precancerous lesions. Hum Pathol. 2009;40:1534-1542.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 61]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
75.  Wang YC, Xu JH, Geng X, Zhang WM. [Preliminary study on the alternative splicing pattern of human telomerase reverse transcriptase gene during gastric carcinogenesis]. Zhonghua Yi Xue Yi Chuan Xue Zazhi. 2009;26:151-155.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
76.  Guilleret I, Yan P, Grange F, Braunschweig R, Bosman FT, Benhattar J. Hypermethylation of the human telomerase catalytic subunit (hTERT) gene correlates with telomerase activity. Int J Cancer. 2002;101:335-341.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 179]  [Cited by in F6Publishing: 183]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
77.  Gigek CO, Leal MF, Silva PN, Lisboa LC, Lima EM, Calcagno DQ, Assumpção PP, Burbano RR, Smith Mde A. hTERT methylation and expression in gastric cancer. Biomarkers. 2009;14:630-636.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 35]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
78.  Du YY, Dai DQ, Yang Z. Role of RECK methylation in gastric cancer and its clinical significance. World J Gastroenterol. 2010;16:904-908.  [PubMed]  [DOI]  [Cited in This Article: ]
79.  Zhang L, Zhong K, Dai Y, Zhou H. Genome-wide analysis of histone H3 lysine 27 trimethylation by ChIP-chip in gastric cancer patients. J Gastroenterol. 2009;44:305-312.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 31]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
80.  Kwon OH, Park JL, Kim M, Kim JH, Lee HC, Kim HJ, Noh SM, Song KS, Yoo HS, Paik SG. Aberrant up-regulation of LAMB3 and LAMC2 by promoter demethylation in gastric cancer. Biochem Biophys Res Commun. 2011;406:539-545.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 48]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
81.  Park YS, Jin MY, Kim YJ, Yook JH, Kim BS, Jang SJ. The global histone modification pattern correlates with cancer recurrence and overall survival in gastric adenocarcinoma. Ann Surg Oncol. 2008;15:1968-1976.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 127]  [Cited by in F6Publishing: 137]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
82.  Kondo Y, Shen L, Issa JP. Critical role of histone methylation in tumor suppressor gene silencing in colorectal cancer. Mol Cell Biol. 2003;23:206-215.  [PubMed]  [DOI]  [Cited in This Article: ]
83.  Watanabe Y, Toyota M, Kondo Y, Suzuki H, Imai T, Ohe-Toyota M, Maruyama R, Nojima M, Sasaki Y, Sekido Y. PRDM5 identified as a target of epigenetic silencing in colorectal and gastric cancer. Clin Cancer Res. 2007;13:4786-4794.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 75]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
84.  Li Q, Wang X, Lu Z, Zhang B, Guan Z, Liu Z, Zhong Q, Gu L, Zhou J, Zhu B. Polycomb CBX7 directly controls trimethylation of histone H3 at lysine 9 at the p16 locus. PLoS One. 2010;5:e13732.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 45]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
85.  Angrisano T, Lembo F, Peluso S, Keller S, Chiariotti L, Pero R. Helicobacter pylori regulates iNOS promoter by histone modifications in human gastric epithelial cells. Med Microbiol Immunol. 2012;201:249-257.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 22]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
86.  Egger G, Liang G, Aparicio A, Jones PA. Epigenetics in human disease and prospects for epigenetic therapy. Nature. 2004;429:457-463.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2210]  [Cited by in F6Publishing: 2104]  [Article Influence: 105.2]  [Reference Citation Analysis (0)]
87.  Jones PA, Taylor SM. Cellular differentiation, cytidine analogs and DNA methylation. Cell. 1980;20:85-93.  [PubMed]  [DOI]  [Cited in This Article: ]
88.  Gal-Yam EN, Saito Y, Egger G, Jones PA. Cancer epigenetics: modifications, screening, and therapy. Annu Rev Med. 2008;59:267-280.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 196]  [Cited by in F6Publishing: 207]  [Article Influence: 12.9]  [Reference Citation Analysis (0)]
89.  Dikken JL, van Sandick JW, Maurits Swellengrebel HA, Lind PA, Putter H, Jansen EP, Boot H, van Grieken NC, van de Velde CJ, Verheij M. Neo-adjuvant chemotherapy followed by surgery and chemotherapy or by surgery and chemoradiotherapy for patients with resectable gastric cancer (CRITICS). BMC Cancer. 2011;11:329.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 145]  [Cited by in F6Publishing: 167]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
90.  Zheng Y, Zhang Y, Huang X, Chen L. Analysis of the RUNX3 gene methylation in serum DNA from esophagus squamous cell carcinoma, gastric and colorectal adenocarcinoma patients. Hepatogastroenterology. 2011;58:2007-2011.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 14]  [Reference Citation Analysis (0)]
91.  Jee CD, Kim MA, Jung EJ, Kim J, Kim WH. Identification of genes epigenetically silenced by CpG methylation in human gastric carcinoma. Eur J Cancer. 2009;45:1282-1293.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 74]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
92.  Hibi K, Goto T, Shirahata A, Saito M, Kigawa G, Nemoto H, Sanada Y. Detection of TFPI2 methylation in the serum of gastric cancer patients. Anticancer Res. 2011;31:3835-3838.  [PubMed]  [DOI]  [Cited in This Article: ]
93.  Kanai Y, Ushijima S, Kondo Y, Nakanishi Y, Hirohashi S. DNA methyltransferase expression and DNA methylation of CPG islands and peri-centromeric satellite regions in human colorectal and stomach cancers. Int J Cancer. 2001;91:205-212.  [PubMed]  [DOI]  [Cited in This Article: ]
94.  Fang JY, Cheng ZH, Chen YX, Lu R, Yang L, Zhu HY, Lu LG. Expression of Dnmt1, demethylase, MeCP2 and methylation of tumor-related genes in human gastric cancer. World J Gastroenterol. 2004;10:3394-3398.  [PubMed]  [DOI]  [Cited in This Article: ]
95.  Ding WJ, Fang JY, Chen XY, Peng YS. The expression and clinical significance of DNA methyltransferase proteins in human gastric cancer. Dig Dis Sci. 2008;53:2083-2089.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 67]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
96.  Yang J, Wei X, Wu Q, Xu Z, Gu D, Jin Y, Shen Y, Huang H, Fan H, Chen J. Clinical significance of the expression of DNA methyltransferase proteins in gastric cancer. Mol Med Report. 2011;4:1139-1143.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 29]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
97.  Mutze K, Langer R, Schumacher F, Becker K, Ott K, Novotny A, Hapfelmeier A, Höfler H, Keller G. DNA methyltransferase 1 as a predictive biomarker and potential therapeutic target for chemotherapy in gastric cancer. Eur J Cancer. 2011;47:1817-1825.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 100]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
98.  Fan H, Liu D, Qiu X, Qiao F, Wu Q, Su X, Zhang F, Song Y, Zhao Z, Xie W. A functional polymorphism in the DNA methyltransferase-3A promoter modifies the susceptibility in gastric cancer but not in esophageal carcinoma. BMC Med. 2010;8:12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 68]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
99.  Su X, Lv C, Qiao F, Qiu X, Huang W, Wu Q, Zhao Z, Fan H. Expression pattern and clinical significance of DNA methyltransferase 3B variants in gastric carcinoma. Oncol Rep. 2010;23:819-826.  [PubMed]  [DOI]  [Cited in This Article: ]
100.  Hu J, Fan H, Liu D, Zhang S, Zhang F, Xu H. DNMT3B promoter polymorphism and risk of gastric cancer. Dig Dis Sci. 2010;55:1011-1016.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 35]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
101.  Wada R, Akiyama Y, Hashimoto Y, Fukamachi H, Yuasa Y. miR-212 is downregulated and suppresses methyl-CpG-binding protein MeCP2 in human gastric cancer. Int J Cancer. 2010;127:1106-1114.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 116]  [Article Influence: 7.7]  [Reference Citation Analysis (0)]
102.  Kanai Y, Ushijima S, Nakanishi Y, Hirohashi S. Reduced mRNA expression of the DNA demethylase, MBD2, in human colorectal and stomach cancers. Biochem Biophys Res Commun. 1999;264:962-966.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 36]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
103.  Ogden SR, Wroblewski LE, Weydig C, Romero-Gallo J, O’Brien DP, Israel DA, Krishna US, Fingleton B, Reynolds AB, Wessler S, Peek RM. p120 and Kaiso regulate Helicobacter pylori-induced expression of matrix metalloproteinase-7. Mol Biol Cell. 2008;19:4110-4121.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 57]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
104.  Lee SH, Kim J, Kim WH, Lee YM. Hypoxic silencing of tumor suppressor RUNX3 by histone modification in gastric cancer cells. Oncogene. 2009;28:184-194.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 106]  [Cited by in F6Publishing: 118]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
105.  Oshimo Y, Oue N, Mitani Y, Nakayama H, Kitadai Y, Yoshida K, Chayama K, Yasui W. Frequent epigenetic inactivation of RIZ1 by promoter hypermethylation in human gastric carcinoma. Int J Cancer. 2004;110:212-218.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 46]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
106.  Pan KF, Lu YY, Liu WG, Zhang L, You WC. Detection of frameshift mutations of RIZ in gastric cancers with microsatellite instability. World J Gastroenterol. 2004;10:2719-2722.  [PubMed]  [DOI]  [Cited in This Article: ]
107.  Yoo EJ, Park SY, Cho NY, Kim N, Lee HS, Kang GH. Helicobacter pylori-infection-associated CpG island hypermethylation in the stomach and its possible association with polycomb repressive marks. Virchows Arch. 2008;452:515-524.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 23]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
108.  Liu JH, Song LB, Zhang X, Guo BH, Feng Y, Li XX, Liao WT, Zeng MS, Huang KH. Bmi-1 expression predicts prognosis for patients with gastric carcinoma. J Surg Oncol. 2008;97:267-272.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 70]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
109.  Xiao J, Deng C. Knockdown of Bmi-1 impairs growth and invasiveness of human gastric carcinoma cells. Oncol Res. 2009;17:613-620.  [PubMed]  [DOI]  [Cited in This Article: ]
110.  Lu YW, Li J, Guo WJ. Expression and clinicopathological significance of Mel-18 and Bmi-1 mRNA in gastric carcinoma. J Exp Clin Cancer Res. 2010;29:143.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 19]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
111.  Zhang XW, Sheng YP, Li Q, Qin W, Lu YW, Cheng YF, Liu BY, Zhang FC, Li J, Dimri GP. BMI1 and Mel-18 oppositely regulate carcinogenesis and progression of gastric cancer. Mol Cancer. 2010;9:40.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 65]  [Cited by in F6Publishing: 73]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
112.  Li W, Li Y, Tan Y, Ma K, Cui J. Bmi-1 is critical for the proliferation and invasiveness of gastric carcinoma cells. J Gastroenterol Hepatol. 2010;25:568-575.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 20]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
113.  Takahata M, Inoue Y, Tsuda H, Imoto I, Koinuma D, Hayashi M, Ichikura T, Yamori T, Nagasaki K, Yoshida M. SKI and MEL1 cooperate to inhibit transforming growth factor-beta signal in gastric cancer cells. J Biol Chem. 2009;284:3334-3344.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 68]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
114.  Mattioli E, Vogiatzi P, Sun A, Abbadessa G, Angeloni G, D’Ugo D, Trani D, Gaughan JP, Vecchio FM, Cevenini G. Immunohistochemical analysis of pRb2/p130, VEGF, EZH2, p53, p16(INK4A), p27(KIP1), p21(WAF1), Ki-67 expression patterns in gastric cancer. J Cell Physiol. 2007;210:183-191.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 56]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
115.  Varambally S, Cao Q, Mani RS, Shankar S, Wang X, Ateeq B, Laxman B, Cao X, Jing X, Ramnarayanan K. Genomic loss of microRNA-101 leads to overexpression of histone methyltransferase EZH2 in cancer. Science. 2008;322:1695-1699.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 808]  [Cited by in F6Publishing: 812]  [Article Influence: 50.8]  [Reference Citation Analysis (0)]
116.  Zhou Y, Du WD, Wu Q, Liu Y, Chen G, Ruan J, Xu S, Yang F, Zhou FS, Tang XF. EZH2 genetic variants affect risk of gastric cancer in the Chinese Han population. Mol Carcinog. 2012;Epub ahead of print.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 14]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
117.  Hudlebusch HR, Santoni-Rugiu E, Simon R, Ralfkiær E, Rossing HH, Johansen JV, Jørgensen M, Sauter G, Helin K. The histone methyltransferase and putative oncoprotein MMSET is overexpressed in a large variety of human tumors. Clin Cancer Res. 2011;17:2919-2933.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 104]  [Cited by in F6Publishing: 104]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
118.  Magerl C, Ellinger J, Braunschweig T, Kremmer E, Koch LK, Höller T, Büttner R, Lüscher B, Gütgemann I. H3K4 dimethylation in hepatocellular carcinoma is rare compared with other hepatobiliary and gastrointestinal carcinomas and correlates with expression of the methylase Ash2 and the demethylase LSD1. Hum Pathol. 2010;41:181-189.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 72]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
119.  Li W, Zhao L, Zang W, Liu Z, Chen L, Liu T, Xu D, Jia J. Histone demethylase JMJD2B is required for tumor cell proliferation and survival and is overexpressed in gastric cancer. Biochem Biophys Res Commun. 2011;416:372-378.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 55]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
120.  Katoh M, Katoh M. Comparative integromics on JMJD1C gene encoding histone demethylase: conserved POU5F1 binding site elucidating mechanism of JMJD1C expression in undifferentiated ES cells and diffuse-type gastric cancer. Int J Oncol. 2007;31:219-223.  [PubMed]  [DOI]  [Cited in This Article: ]
121.  Poplawski T, Tomaszewska K, Galicki M, Morawiec Z, Blasiak J. Promoter methylation of cancer-related genes in gastric carcinoma. Exp Oncol. 2008;30:112-116.  [PubMed]  [DOI]  [Cited in This Article: ]
122.  Tahara T, Shibata T, Nakamura M, Yamashita H, Yoshioka D, Okubo M, Yonemura J, Maeda Y, Maruyama N, Kamano T. Increased number of CpG island hypermethylation in tumor suppressor genes of non-neoplastic gastric mucosa correlates with higher risk of gastric cancer. Digestion. 2010;82:27-36.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 24]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
123.  Lee TB, Park JH, Min YD, Kim KJ, Choi CH. Epigenetic mechanisms involved in differential MDR1 mRNA expression between gastric and colon cancer cell lines and rationales for clinical chemotherapy. BMC Gastroenterol. 2008;8:33.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 30]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
124.  Takada H, Imoto I, Tsuda H, Nakanishi Y, Ichikura T, Mochizuki H, Mitsufuji S, Hosoda F, Hirohashi S, Ohki M. ADAM23, a possible tumor suppressor gene, is frequently silenced in gastric cancers by homozygous deletion or aberrant promoter hypermethylation. Oncogene. 2005;24:8051-8060.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 56]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
125.  Watanabe Y, Kim HS, Castoro RJ, Chung W, Estecio MR, Kondo K, Guo Y, Ahmed SS, Toyota M, Itoh F. Sensitive and specific detection of early gastric cancer with DNA methylation analysis of gastric washes. Gastroenterology. 2009;136:2149-2158.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 100]  [Cited by in F6Publishing: 97]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
126.  Kim JH, Jung EJ, Lee HS, Kim MA, Kim WH. Comparative analysis of DNA methylation between primary and metastatic gastric carcinoma. Oncol Rep. 2009;21:1251-1259.  [PubMed]  [DOI]  [Cited in This Article: ]
127.  Balassiano K, Lima S, Jenab M, Overvad K, Tjonneland A, Boutron-Ruault MC, Clavel-Chapelon F, Canzian F, Kaaks R, Boeing H. Aberrant DNA methylation of cancer-associated genes in gastric cancer in the European Prospective Investigation into Cancer and Nutrition (EPIC-EURGAST). Cancer Lett. 2011;311:85-95.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 53]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
128.  Bernal C, Aguayo F, Villarroel C, Vargas M, Díaz I, Ossandon FJ, Santibáñez E, Palma M, Aravena E, Barrientos C. Reprimo as a potential biomarker for early detection in gastric cancer. Clin Cancer Res. 2008;14:6264-6269.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 83]  [Cited by in F6Publishing: 90]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
129.  Geddert H, zur Hausen A, Gabbert HE, Sarbia M. EBV-infection in cardiac and non-cardiac gastric adenocarcinomas is associated with promoter methylation of p16, p14 and APC, but not hMLH1. Cell Oncol (Dordr). 2011;34:209-214.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 26]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
130.  Maekita T, Nakazawa K, Mihara M, Nakajima T, Yanaoka K, Iguchi M, Arii K, Kaneda A, Tsukamoto T, Tatematsu M. High levels of aberrant DNA methylation in Helicobacter pylori-infected gastric mucosae and its possible association with gastric cancer risk. Clin Cancer Res. 2006;12:989-995.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 474]  [Cited by in F6Publishing: 460]  [Article Influence: 25.6]  [Reference Citation Analysis (0)]
131.  Murai M, Toyota M, Suzuki H, Satoh A, Sasaki Y, Akino K, Ueno M, Takahashi F, Kusano M, Mita H. Aberrant methylation and silencing of the BNIP3 gene in colorectal and gastric cancer. Clin Cancer Res. 2005;11:1021-1027.  [PubMed]  [DOI]  [Cited in This Article: ]
132.  Hiraki M, Kitajima Y, Koga Y, Tanaka T, Nakamura J, Hashiguchi K, Noshiro H, Miyazaki K. Aberrant gene methylation is a biomarker for the detection of cancer cells in peritoneal wash samples from advanced gastric cancer patients. Ann Surg Oncol. 2011;18:3013-3019.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 35]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
133.  Sugita H, Iida S, Inokuchi M, Kato K, Ishiguro M, Ishikawa T, Takagi Y, Enjoji M, Yamada H, Uetake H. Methylation of BNIP3 and DAPK indicates lower response to chemotherapy and poor prognosis in gastric cancer. Oncol Rep. 2011;25:513-518.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 52]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
134.  Ryan JL, Jones RJ, Kenney SC, Rivenbark AG, Tang W, Knight ER, Coleman WB, Gulley ML. Epstein-Barr virus-specific methylation of human genes in gastric cancer cells. Infect Agent Cancer. 2010;5:27.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 46]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
135.  Yamashita S, Tsujino Y, Moriguchi K, Tatematsu M, Ushijima T. Chemical genomic screening for methylation-silenced genes in gastric cancer cell lines using 5-aza-2’-deoxycytidine treatment and oligonucleotide microarray. Cancer Sci. 2006;97:64-71.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 191]  [Cited by in F6Publishing: 201]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
136.  Leal M, Lima E, Silva P, Assumpção P, Calcagno D, Payão S, Burbano RR, Smith M. Promoter hypermethylation of CDH1, FHIT, MTAP and PLAGL1 in gastric adenocarcinoma in individuals from Northern Brazil. World J Gastroenterol. 2007;13:2568-2574.  [PubMed]  [DOI]  [Cited in This Article: ]
137.  Al-Moundhri MS, Al-Nabhani M, Tarantini L, Baccarelli A, Rusiecki JA. The prognostic significance of whole blood global and specific DNA methylation levels in gastric adenocarcinoma. PLoS One. 2010;5:e15585.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 24]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
138.  Oki E, Zhao Y, Yoshida R, Masuda T, Ando K, Sugiyama M, Tokunaga E, Morita M, Kakeji Y, Maehara Y. Checkpoint with forkhead-associated and ring finger promoter hypermethylation correlates with microsatellite instability in gastric cancer. World J Gastroenterol. 2009;15:2520-2525.  [PubMed]  [DOI]  [Cited in This Article: ]
139.  Hiraki M, Kitajima Y, Sato S, Mitsuno M, Koga Y, Nakamura J, Hashiguchi K, Noshiro H, Miyazaki K. Aberrant gene methylation in the lymph nodes provides a possible marker for diagnosing micrometastasis in gastric cancer. Ann Surg Oncol. 2010;17:1177-1186.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 28]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
140.  Hu SL, Kong XY, Cheng ZD, Sun YB, Shen G, Xu WP, Wu L, Xu XC, Jiang XD, Huang DB. Promoter methylation of p16, Runx3, DAPK and CHFR genes is frequent in gastric carcinoma. Tumori. 2010;96:726-733.  [PubMed]  [DOI]  [Cited in This Article: ]
141.  Shi J, Zhang G, Yao D, Liu W, Wang N, Ji M, He N, Shi B, Hou P. Prognostic significance of aberrant gene methylation in gastric cancer. Am J Cancer Res. 2012;2:116-129.  [PubMed]  [DOI]  [Cited in This Article: ]
142.  Akiyama Y, Watkins N, Suzuki H, Jair KW, van Engeland M, Esteller M, Sakai H, Ren CY, Yuasa Y, Herman JG. GATA-4 and GATA-5 transcription factor genes and potential downstream antitumor target genes are epigenetically silenced in colorectal and gastric cancer. Mol Cell Biol. 2003;23:8429-8439.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 177]  [Cited by in F6Publishing: 187]  [Article Influence: 8.9]  [Reference Citation Analysis (0)]
143.  Wen XZ, Akiyama Y, Pan KF, Liu ZJ, Lu ZM, Zhou J, Gu LK, Dong CX, Zhu BD, Ji JF. Methylation of GATA-4 and GATA-5 and development of sporadic gastric carcinomas. World J Gastroenterol. 2010;16:1201-1208.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 41]  [Cited by in F6Publishing: 41]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
144.  Fang JY, Zhu SS, Xiao SD, Jiang SJ, Shi Y, Chen XY, Zhou XM, Qian LF. Studies on the hypomethylation of c-myc, c-Ha-ras oncogenes and histopathological changes in human gastric carcinoma. J Gastroenterol Hepatol. 1996;11:1079-1082.  [PubMed]  [DOI]  [Cited in This Article: ]
145.  Luo J, Li YN, Wang F, Zhang WM, Geng X. S-adenosylmethionine inhibits the growth of cancer cells by reversing the hypomethylation status of c-myc and H-ras in human gastric cancer and colon cancer. Int J Biol Sci. 2010;6:784-795.  [PubMed]  [DOI]  [Cited in This Article: ]
146.  Gigek CO, Leal MF, Lisboa LC, Silva PN, Chen ES, Lima EM, Calcagno DQ, Assumpção PP, Burbano RR, Smith Mde A. Insulin-like growth factor binding protein-3 gene methylation and protein expression in gastric adenocarcinoma. Growth Horm IGF Res. 2010;20:234-238.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 13]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
147.  Chen HY, Zhu BH, Zhang CH, Yang DJ, Peng JJ, Chen JH, Liu FK, He YL. High CpG island methylator phenotype is associated with lymph node metastasis and prognosis in gastric cancer. Cancer Sci. 2012;103:73-79.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 43]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
148.  Tamura G, So K, Miyoshi H, Honda T, Nishizuka S, Motoyama T. Quantitative assessment of gene methylation in neoplastic and non-neoplastic gastric epithelia using methylation-specific DNA microarray. Pathol Int. 2009;59:895-899.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 10]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
149.  Hibi K, Sakata M, Yokomizo K, Kitamura YH, Sakuraba K, Shirahata A, Goto T, Mizukami H, Saito M, Ishibashi K. Methylation of the MGMT gene is frequently detected in advanced gastric carcinoma. Anticancer Res. 2009;29:5053-5055.  [PubMed]  [DOI]  [Cited in This Article: ]
150.  Kim HG, Lee S, Kim DY, Ryu SY, Joo JK, Kim JC, Lee KH, Lee JH. Aberrant methylation of DNA mismatch repair genes in elderly patients with sporadic gastric carcinoma: A comparison with younger patients. J Surg Oncol. 2010;101:28-35.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 28]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
151.  Dong CX, Deng DJ, Pan KF, Zhang L, Zhang Y, Zhou J, You WC. Promoter methylation of p16 associated with Helicobacter pylori infection in precancerous gastric lesions: a population-based study. Int J Cancer. 2009;124:434-439.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 45]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
152.  Shu XS, Geng H, Li L, Ying J, Ma C, Wang Y, Poon FF, Wang X, Ying Y, Yeo W. The epigenetic modifier PRDM5 functions as a tumor suppressor through modulating WNT/β-catenin signaling and is frequently silenced in multiple tumors. PLoS One. 2011;6:e27346.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 54]  [Cited by in F6Publishing: 54]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
153.  Guo W, Dong Z, Chen Z, Yang Z, Wen D, Kuang G, Guo Y, Shan B. Aberrant CpG island hypermethylation of RASSF1A in gastric cardia adenocarcinoma. Cancer Invest. 2009;27:459-465.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 32]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
154.  Sakakura C, Hamada T, Miyagawa K, Nishio M, Miyashita A, Nagata H, Ida H, Yazumi S, Otsuji E, Chiba T. Quantitative analysis of tumor-derived methylated RUNX3 sequences in the serum of gastric cancer patients. Anticancer Res. 2009;29:2619-2625.  [PubMed]  [DOI]  [Cited in This Article: ]
155.  Fan XY, Hu XL, Han TM, Wang NN, Zhu YM, Hu W, Ma ZH, Zhang CJ, Xu X, Ye ZY. Association between RUNX3 promoter methylation and gastric cancer: a meta-analysis. BMC Gastroenterol. 2011;11:92.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 39]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
156.  Carvalho R, Kayademir T, Soares P, Canedo P, Sousa S, Oliveira C, Leistenschneider P, Seruca R, Gött P, Blin N. Loss of heterozygosity and promoter methylation, but not mutation, may underlie loss of TFF1 in gastric carcinoma. Lab Invest. 2002;82:1319-1326.  [PubMed]  [DOI]  [Cited in This Article: ]