SciELO - Scientific Electronic Library Online

 
vol.41 número3Daucus carota as a novel model to evaluate the effect of light on carotenogenic gene expressionEthanol Intake during Lactation Alters Milk Nutrient Composition and Growth and Mineral Status of Rat Pups índice de autoresíndice de materiabúsqueda de artículos
Home Pagelista alfabética de revistas  

Servicios Personalizados

Revista

Articulo

Indicadores

Links relacionados

Compartir


Biological Research

versión impresa ISSN 0716-9760

Biol. Res. v.41 n.3 Santiago  2008

http://dx.doi.org/10.4067/S0716-97602008000300007 

 

Biol Res41: 303-315, 2008

ARTICLES

 

DNA methylation profile in diffuse type gastric cancer: evidence for hypermethylation of the BRCA1 promoter region in early-onset gastric carcinogenesis

 

CAROLINA BERNAL1, MACARENA VARGAS1, FRANCISCO OSSANDÓN1, EUDOCIA SANTIBÁÑEZ1, JULIO URRUTIA1, VÍCTOR LUENGO1, LUIS F. ZAVALA1, CLAUDIA BACKHOUSE3, MARIANA PALMA3, JORGE ARGANDOÑA3, FRANCISCO AGUAYO2, and ALEJANDRO CORVALÁN1,2

1 Departmento Anatomía Patológica y 2Laboratorio Patología Molecular y Epidemiología, Centro Investigaciones Médicas, Pontificia Universidad Católica de Chile e 3Instituto Chileno Japonés de Enfermedades Digestivas, Hospital Clínico San Borja Arriarán, Santiago Chile.

Dirección para correspondencia

JOURNAL CATEGORY: Tumor genetics

NOVELTY AND IMPACT: We identified that hypermethylation of BRCA1 is associated with young age, suggesting a role in early-onset gastric carcinoma. The low frequency of hypermethylation of the p73 gene in normal adjacent mucosa suggests that it might play a role in the early stages of diffuse type gastric carcinoma.


ABSTRACT

Diffuse type gastric carcinoma is the most aggressive type of gastric cáncer. This type of tumor is not preceded by precancerous changes and is associated with early-onset and hereditary syndromes. To test the hypothesis that DNA methylation profile would be useful for molecular classification of the diffuse type gastric carcinoma, DNA methylation patterns of the CpG Island of 17 genes were studied in 104 cases and 47 normal adjacent gastric mucosa by Methylation-specific PCR, Immunohistochemistry and Hierarchical clustering analysis. The most frequent methylated genes were FHIT, E-cadherin, BRCA1 and APC (>50%), followed by pl4, pl6, pl5, p73, MGMT and SEMA3B (20-49%). Hierarchical clustering analysis reveáis four groups with different clinical features. The first was characterized by hypermethylation of BRCA1 and younger age (<45 years oíd), and the second by hypermethylation of pl4 and pl6 genes, male predominance and Epstein-Barr virus infection. The third group was characterized by hypermethylation of FHIT and antrum located tumors and the fourth was not associated with any clinical variables. In normal adjacent mucosa only the p73 gene was significantly less methylated in comparison to tumor mucosa. DNA methylation identified subgroups of diffuse type gastric cáncer. Hypermethylation of BRCA1 associated with young age suggests a role in early-onset gastric carcinoma.

Key terms: gastric cáncer, diffuse type, early-onset, BRCA-1


 

Introduction 

Gastric cáncer is the fourth most common cáncer and the second leading cause of cáncer-related death worldwide (Pisani et al., 2002). Although its incidence and mortality have fallen dramatically, gastric cáncer remains a worldwide public health problem (Pisani et al., 2002; Rastogi et al., 2004). According to Lauren's classification, there are two major histological types of gastric cáncer, intestinal and diffuse (Lauren 1965). The diffuse type is the most aggressive form of gastric cáncer and the mortality rate is increasing in spite of the decline of the intestinal type (Henson et al., 2004; Crew and Neugut 2006). In addition, the diffuse type is not preceded by sequential stages of precancerous changes, and tends to arise "de novo" (Vauhkonen et al., 2006). Furthermore, early-onset gastric cáncer and hereditary diffuse gastric cáncer (HDGC) are both associated with diffuse type histology (Dunbier and Guilford 2001; Milne et al., 2007).

Particular alterations at the genetic and epigenetic levéis in oncogenes and tumor-suppressor genes have been associated with the multistage process of gastric cáncer (Tahara 2004; Yasui et al., 2005). In the diffuse type, the best characterized genetic alteration is the loss of heterozygosity (LOH) of chromosome 17p and mutations of p53 and E-cadherin genes (Tamura 2006). In addition, amplification of K-sam and c-met, LOH at lp and reduced p27 and nm23 expression have been associated with advanced stage disease and low survival rates (Yasui et al., 2005; Vauhkonen et al., 2006). In spite of these findings, no consistent gene alterations have been detected in diffuse type gastric cáncer.

The identification and characterization of genes selectively hypermethylated in cáncer may improve our understanding of gastric carcinoma (Esteller 2003). Several reports have shown frequent hypermethylation of tumor suppressor genes in the intestinal type of gastric cáncer (Tamura 2006; Vauhkonen et al., 2006). However, besides hypermetylation of the promoter región of E-cadherin gene (Graziano et al., 2004), and more recently, PGP9.5 (Yamashita et al, 2006), no consistent information of the role of epigenetics in the diffuse type is currently available. In this study we used a candidate gene approach of 17 genes, covering all cellular pathways, to test the hypothesis that a hypermethylation profile would be useful for molecular classification of diffuse type gastric cáncer. We also assessed the role of specific genes as molecular markers for the "de novo" precancerous changes in normal adjacent mucosa of paired tumor samples.

MATERIALS AND METHODS

Clinical Samples

We studied 104 formalin-fixed paraffin-embedded archival specimens of diffuse type gastric cáncer. All cases were selected using the histological criteria according to the Lauren's classification (Lauren 1965). In 47 of these cases, normal gastric mucosa adjacent to the tumor was also available. Clinical characteristics of these cases are shown in Table 1. In this series, 66 (63.5%) cases were males with an age average of 58 years oíd and 17 (16.3%) patients were under 45 years of age. Forty-three (41.3%) tumors were located in cardia and 27 (26%) in the antrum. In 3 cases (2.9%) the location was not consigned. Fourteen (13.5%) cases were at early stage. Seventy-one (68.3%) of these cases were lymph node positive and 37 (35.6%) had signet-ring cell features. In this series, 30 (28.8%) cases were positive for EBV infection and had been reported previously (Corvalan et al., 2001). The date of the last follow-up and status (alive or dead) was available in 100 cases. The Institutional Review Boards of the Pontificia Universidad Católica de Chile and Hospital Clínico San Borja Arriarán, Santiago Chile approved this study.

DNA extraction

Five 15 μm paraffin sections of representative áreas of diffuse type gastric carcinoma (>70% tumor cells) were cut and placed into a 0.5 mL tube for DNA extraction. DNA extraction was performed in a 100 μL extraction solution (1 M Tris pH 8.0, 50 mM EDTA, 0.5% and Tween 20) with 1 mg/mL Proteinase K (Sigma) for 12 hrs at 55°C. Proteinase K was inactivated by boiling at 100°C for 10 minutes and DNA was purified by phenol-chloroform extraction and ethanol precipitation according to standard protocols. DNA concentration was determined by spectroscopy using 1 OD260 for 50 μg/ml.

DNA Methylation Assays

DNA was treated with sodium bisulphite as described previously (Riquelme et al., 2007). Briefly, 1 μg of genomic DNA was denatured by incubation with 0.2 M NaOH for 10 min at 37°C. Aliquots of lOmM hydroquinone (30 ul; Sigma Chemical Co., St. Louis, MO) and 3 M sodium bisulphite (pH 5.0; 520 ul; Sigma Chemical Co.) were added, and the solution was incubated at 50 °C for 16 h. Treated DNA was purified by use of the Wizard DNA Purification System (Promega Corp., Madison, WI), desulfonated with 0.3 M NaOH, precipitated with ethanol, and resuspended in water. Modified DNA was stored at -80° C until used. The methylation status of 17 genes (APC, BRCA1, DAPK, ER, E-cadherin, FHIT, GSTP, hMLHl, MGMT, p14, p15, p16, p73, RARb, SEMA3B, SOCS, TIMP3) was determined by methylation-specific polymerase chain reaction (MS-PCR) (Hermán and Baylin 2003) and details on primer sequences and PCR conditions are available upon request. These genes were chosen because they are known to be tumor suppressor genes, methylation at these CpG sites is associated with gene silencing, they cover essential alterations in cell physiology that collectively dictate malignant growth and they had previously been described as undergoing hypermethylation in other tumor types (Hanahan and Weinberg 2000; Virmani et al., 2000; Chan et al., 2002; Li et al., 2002; Oka et al., 2002; Sato et al., 2002; Kuroki et al., 2003; Miyamoto et al., 2003; Wild et al., 2003; Sarbia et al., 2004; Takahashi et al., 2004; Xu et al., 2004; Kim et al., 2005; Kim et al., 2005; Liu et al., 2005; Takahashi et al., 2005; Kawaguchi et al., 2006; Morí et al., 2006; Tamura 2006; Riquelme et al., 2007). Gene ñames, gene location and function for each gene selected in this study is summarized in Table 2. Only cases with positive unmethylated bands were considered informative for methylation status in this study. All reactions were done in triplícate. In vitro methylated Sssl methyltransferase (New England Biolabs) and bisulfite-modified DNA from the MKN-45 cell line were used as a positive control. S ampies without DNA témplate (water only) were included as negative control for each set of PCR reaction.

Protein expression assays

To establish the association between CpG island hypermethylation and gene silencing we determined protein expression by immunohistochemistry assay on tissue microarray (TMA) slides. Tissue microarrays were performed using a Manual Tissue Arrayer II instrument (Beecher Instruments, Silver Spring, Maryland, USA). Archival tumor tissue blocks from 104 tumors were selected, cut and stained with hematoxylin and eosin for the best tumor área identification. After whole-section glass slide evaluation, tumor área was selected for placement into the TMA by a circling on the glass slide and identified in the corresponding paraffin block. Six hundred [im stylets in inner diameter were used to take three cylindrical core biopsies from each tumor tissue block (donor block), with subsequent arraying into a new recipient paraffin block. In this way, all 104 cases were held in the 3 recipient blocks. An adequate case was defined as a tumor occupying more than 10% of the core área. Immunohistochemistry was performed on 4-μm-thick section TMA blocks. Sections were dewaxed in xylene, rehydrated through graded alcohol, and placed in an endogenous peroxide block for 15 minutes. Antigen retrieval was performed in a citrate buffer (10% citrate buffer stock in distilled water, pH 6.0) and microwaved for 10 minutes. Non-reactive staining was blocked by 1% horse serum in Tris-buffered saline, pH6.0 for 3 minutes. After primary incubation, antibody binding was detected using two-stage visualization systems based on an enzyme-conjugated polymer backbone carrying secondary antibody molecules (EnVision™ Systems, Dako Cytomation). Protein expression of E-cadherin, pl6 and p73 on TMA was determined in which cell membrane (E-cadherin) and nuclear (pl6 and p73) staining from 10% or more tumor cells was considered positive for expression (Fig. 2).

Data Analysis

In order to identify clinically relevant groups based on to the DNA methylation pattern, we performed hierarchical clustering analysis in a similar fashion to cDNA expression microarrays in breast tumors and lymphomas (Alizadeh et al., 2000; van 't Veer et al., 2002) or more recently, DNA methylation signature in neuroblastoma (10 genes) or hepatocellular carcinoma (18 genes) (Alaminos et al., 2004; Nishida et al., 2007). TIGR MultiExperiment Viewer was applied to the DNA methylation dataset using unsupervised hierarchical clustering analysis with Pearson correlation and complete linkage to cluster the tumors. In addition, we analyzed this data using the Methylation index, defined as the number of methylated genes, divided by the total number of genes analyzed, as a way to compare the methylation status of each cluster. Clinical variables and follow-up data of cases from each particular cluster were compared using c2 tests. Survival analysis was performed using the Kaplan-Meier method and differences among groups were compared with the Log-rank testing and Cox regression models (Stata 8.0, Stata Corporation; College Station, TX). For all tests, probability valúes of p<0.05 were regarded as statistically significant.

RESULTS

Frequency of the DNA methylation in diffuse type gastric cáncer

As shown in Fig. 1A, the most frequent methylated genes (>50% cases) were FHIT, E-cadherin, BRCA1, APC. Genes with intermedíate frequency of methylation (20-49%) were pl4, pl6, pl5, p73, MGMT and SEMA3B. Genes with a low level of methylation (<20%) were RARbeta, hMLHl, DAPK, ER, TIMP3, GSTP and SOCS. Representative examples are shown in Fig. 1B.


Correlations between DNA methylation and loss of protein expression

To establish the association between DNA methylation and gene silencing, we determined protein expression of three genes that were methylated in a relatively large number of cases (E-cadherin, pl6 and p73). This analysis was performed in all 104 tested cases. For all three genes, the presence of CpG island methylation was associated with loss of protein expression. Conversely, in those cases harbouring an unmethylated CpG island, each gene was expressed. Representative examples are shown in Fig .2.

DNA methylation patterns and clinical variables

To explore the relationship among DNA methylation patterns and clinical variables, we performed unsupervised hierarchical clustering analysis. As shown in Fig. 3, clustering analysis revealed two different clusters, both further subdivided into two major branches. The most upper branch was characterized by hypermethylation of BRCA1 gene (p=0.0006). The second branch was characterized by hypermethylation of pl4 and pl6 (p<0.0001). The upper branch of the second major cluster was characterized by hypermethylation of FHIT (p<0.0001) and the lower branch was not associated with hypermethylation of any specific gene. Accordingly, methylation index analysis revealed that this cluster was significantly less methylated in comparison to all other clusters (p<0.0001)(data not shown). Subsequently, clinical variables of cases from each cluster were compared and are shown in Table 1. The most upper branch (BRCA1 cluster) was associated with patients of younger age (age <45 y.o.) (p=0.009). The second branch (pl4/pl6 cluster) was associated with male predominance (p=0.002) and Epstein-Barr virus infection (p=0.008). The upper branch of the second major cluster (FHIT cluster) was associated with antrum as the predominant location (p=0.02). Finally, the lower branch of the second major group (low methylation index cluster) was not related to any specific clinical variables.

Survival Analysis

The impact in prognosis of clusters and individual genes was examined by survival analysis using Kaplan-Meier method, Log-rank testing, and Cox regression models. The average follow-up period was 64 months (Standard deviation = 48, range 1-146). Univariate analysis, using all four branches or individual genes, demonstrated that no clusters or single gene was significantly correlated with worse prognosis. Multivariate Cox regression analysis showed that only the lymph node metástasis was prognostic determinant (HR=3.06, 95% CI= 1.07-8.7; p=0.03)(data not shown).

DNA methylation patterns in normal adjacent mucosa from diffuse type gastric cáncer

The frequency of DNA methylation patterns of the CpG Island of 8 genes was studied in a subgroup of 47 paired tumor and normal adjacent mucosa. The most representative genes of each cluster (BRCA1, p14/p16 and FHIT) and 4 non-cluster related genes (APC, MGMT, pl5 and p73 genes) were included in this analysis. Case-informative incidence ranged from 8 to 39 samples. As shown in Fig 4A, only the p73 gene was significant less methylated in normal adjacent mucosa in comparison to tumor mucosa (p=0.006). Representative examples are shown in Fig. 4B.

DISCUSSION

In order to define the DNA methylation signature in diffuse type gastric carcinoma, the most aggressive and increasing form of gastric cáncer, we used the candidate gene approach by searching the hypermethylation profile of CpG Island of 17 genes. To identify associations between genes and clinical variables, we performed hierarchical clustering analysis in a similar fashion to cDNA expression microarrays in breast tumors and lymphomas (Alizadeh et al., 2000; van 't Veer et al., 2002) or DNA methylation signature in Neuroblastoma (Alaminos et al., 2004) or Hepatocellular carcinoma (Nishida et al., 2007). Here, we found that DNA methylation is a frequent event in diffuse type gastric cáncer and clustering analysis reveáis different branches associated with hypermethylation of specific genes. Interestingly, these branches were associated with distinct clinical variables. For example, BRCA1 was frequently more methylated in a group of tumors associated with young age (<45 years oíd). Tumors at this age or less have been considered early-onset gastric cáncer (Milne et al., 2007) and although they represent less than 10% of gastric carcinoma, they have unique clinico-pathological features including diffuse type histology (Milne et al., 2007). Although early onset also has unique molecular features (lack of microsatellite instability, infrequent LOH, low COX2 expression, infrequent loss of TFF1 expression, no loss of RUNX3, gains at chromosomes 17q, 19q and 20q) (Milne et al., 2007), DNA methylation has not been extensively explored in this type of gastric cáncer. Only Kim et al (Kim et al., 2005) assayed the hypermethylation status in genes associated with the APC-beta-catenin axis and the mismatch repair system (hMLHl, TIMP3, THBS1, DAPK, GSTP1, APC, and MINT2) and found that hypermethylation is a frequent phenomenon in early-onset gastric carcinoma. However, no specific genes were hypermethylated. Thus, to our knowledge this is the first report that has identified hypermethylation of CpG island of the BRCA1 gene in association with early-onset gastric carcinoma. Interestingly, Varis et al (Varis et al., 2003) identified increases of DNA copy number at chromosome 17q, the location of the BRCA1 gene, in 52% of 22 cases of early-onset gastric cáncer and Semba et al (Semba et al., 1998) described LOH on chromosome 17ql2-21 with several neighbouring markers in this región, while no mutation was found in the BRCA1 gene. In addition, although associated with hereditary and not early-onset gastric cáncer, studies exploring additional tumors on relatives of BRCA1 carriers identified gastric cáncer as one of the most common sites for malignancies (Gallardo et al., 2006). Taken together, these findings suggest that alterations of the BRCA1 gene should be included as one of the molecular features of early-onset gastric carcinoma.

An association of hypermethylation of pl4 and pl6 and the presence of Epstein-Barr virus infection were characteristic of the lower cluster of the upper branch in the clustering analysis. This association has been described previously (Koriyama et al., 2004). However, our male gender association is contrary to previous studies for pl6 methylation (Vauhkonen et al., 2006). Hypermethylation of FHIT has been associated with antrum as a predominant location. Interestingly, recent data showed that FHIT knock-out mice develop tumors in the forestomach and small intestines (Fujishita et al., 2004). These findings suggest that FHIT plays an important role in the integrity of gastrointestinal mucosal structures. Chang et al (Chang et al., 2002) described frequent LOH at the FHIT locus and loss of Fhit protein expression in a series of 7 signet-ring cell gastric cáncer. Although these authors have proposed that alteration of the FHIT gene might be the hallmark of signet-ring cell gastric cáncer, we did not confirm this.

The finding that seven out of 8 tested genes (with the only exception being the p73 gene) were hypermethylated in a similar frequency in normal adjacent mucosa in comparison to tumor mucosa, suggests the presence of an epigenetic field for cancerization (Ushijima 2007). Epigenetic field for cancerization has been demonstrated for Barrett's esophagus, liver, lung and urothelial cancers (Eads et al., 2000; Wistuba et al., 2002). Since changes in DNA methylation status are specific for each tumor, it is likely that specific genes are methylated according to unique carcinogenic factors (Ushijima 2007). The p73 gene was the only one not hypermethylated in normal adjacent mucosa in comparison to tumor mucosa. Diffuse type gastric cáncer does not have sequential stages of precancerous changes, as does intestinal-type gastric cáncer, and consequently is considered to arise "de novo" (Vauhkonen et al., 2006). Thus, this finding suggests that hypermethylation of p73 might play an important role in the early stages of diffuse type gastric carcinoma. Extremely low levéis of p73 expression has been observed in gastric cáncer cell lines, although reports have shown that mutations of p73 are rare in primary human cancers (Pilozzi et al., 2003). These findings suggest that p73 could be a target of epigenetic regulation in gastric carcinogenesis.

In summary, we found that in DNA, methylation is a frequent event in diffuse type gastric cáncer. Clustering analysis reveáis specific association between genes and clinical variables, in particular BRCA1 to early-onset gastric carcinoma. The finding that the p73 gene was significantly less methylated in normal adjacent mucosa suggests that it may play a role in the early stages of diffuse type gastric carcinoma.

ACKNOWLEDGMENTS

Supported by FONDECYT Grant 1030130 and FONIS Grant SA06I20019 from Chilean government.

REFERENCES

ALAMINOS, M, V DAVALOS, N-K V CHEUNG, W L GERALD and M ESTELLER (2004). "Clustering of Gene Hypermethylation Associated With Clinical Risk Groups in Neuroblastoma." J Nati Cáncer Inst 96: 1208-1219

ALIZADEH, A A, M B EISEN, R E DAVIS, C MA, I S LOSSOS, A ROSENWALD, J C BOLDRICK, H SABET, T TRAN, X YU, J I POWELL, L YANG, G E MARTÍ, T MOORE, J HUDSON, JR., L LU, D B LEWIS, R TIBSHIRANI, G SHERLOCK, W C CHAN, T C GREINER, D D WEISENBURGER, J O ARMITAGE, R WARNKE, R LEVY, W WILSON, M R GREVER, J C BYRD, D BOTSTEIN, P O BROWN and L M STAUDT (2000). "Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling". Nature 403: 503-11

CHAN, K Y, H OZCELIK, A N CHEUNG, H Y NGAN and U S KHOO (2002). "Epigenetic factors controlling the BRCA1 and BRCA2 genes in sporadic ovarían cáncer." Cáncer Res 62: 4151-6

CHANG, Y T, M S WU, C J CHANG, P H HUANG, S M HSU and J T LIN (2002). "Preferential loss of Fhit expression in signet-ring cell and Krukenberg subtypes of gastric cáncer." Lab Invest 82: 1201-8

CORVALÁN, A, C KORIYAMA, S AKIBA, Y EIZURU, C BACKHOUSE, M PALMA, J ARGANDOÑA and M TOKUNAGA (2001). "Epstein-Barr virus in gastric carcinoma is associated with location in the cardia and with a diffuse histology: a study in one área of Chile." Int J Cáncer 94: 527-30

CREW, K D and A I NEUGUT (2006). "Epidemiology of gastric cáncer." World J Gastroenterol 12: 354-62

DUNBIER, A and P GUILFORD (2001). "Hereditary diffuse gastric cáncer." Adv Cáncer Res 83: 55-65

EADS, C A, R V LORD, S K KURUMBOOR, K WICKRAMASINGHE, M L SKINNER, T I LONG, J H PETERS, T R DEMEESTER, K D DANENBERG, P V DANENBERG, P W LAIRD and K A SKINNER (2000). "Fields of aberrant CpG island hypermethylation in Barrett's esophagus and associated adenocarcinoma." Cáncer Res 60: 5021-6

ESTELLER, M (2003). "Relevance of DNA methylation in the management of cáncer." Lancet Oncol 4: 351-8

FUJISHITA, T, Y DOI, M SONOSHITA, H HIAI, M OSHIMA, K HUEBNER, C M CROCE and M M TAKETO (2004). "Development of spontaneous tumours and intestinal lesions in Fhit gene knockout mice." BrJ Cáncer 91: 1571-4

GALLARDO, M, A SILVA, L RUBIO, C ÁLVAREZ, C TORREALBA, M SALINAS, T TAPIA, P FAÚNDEZ, L PALMA, M E RICCIO, H PAREDES, M RODRÍGUEZ, A CRUZ, C ROUSSEAU, M C KING, M CAMUS, M ÁLVAREZ and P CARVALLO (2006). "Incidence of BRCA1 and BRCA2 mutations in 54 Chilean families with breast/ovarian cáncer, genotype-phenotype correlations." Breast Cáncer Res Treat 95: 81-7

GRAZIANO, F, F ARDUINI, A RUZZO, I BEARZI, B HUMAR, H MORE, R SILVA, P MURETTO, P GUILFORD, E TESTA, D MARI, M MAGNANI and S CASCINU (2004). "Prognostic Analysis of E-Cadherin Gene Promoter Hypermethylation in Patients with Surgically Resected, Node-Positive, Diffuse Gastric Cáncer 10.1158/1078-0432.CCR-03-0320." Clin Cáncer Res 10: 2784-2789

HANAHAN, D and R A WEINBERG (2000). "The hallmarks of cáncer." Cell 100: 57-70

HENSON, D E, C DITTUS, M YOUNES, H NGUYEN and J ALBORES-SAAVEDRA (2004). "Differential trends in the intestinal and diffuse types of gastric carcinoma in the United States, 1973-2000: increase in the signet ring cell type." Arch Pathol Lab Med 128: 765-70

HERMÁN, J G and S B BAYLIN (2003). "Gene silencing in cáncer in association with promoter hypermethylation." N Engl J Med 349: 2042-54

KAWAGUCHI, K, Y ODA, T SAITO, H YAMAMOTO, T TAKAHIRA, C KOBAYASHI, S TAMIYA, N TATEISHI, Y IWAMOTO and M TSUNEYOSHI (2006). "DNA hypermethylation status of múltiple genes in soft tissue sarcomas." Mod Pathol 19: 106-14

KIM, H C, J C KIM, S A ROH, C S YU, J H YOOK, S T OH, B S KIM, K C PARK and R CHANO (2005). "Aberrant CpG island methylation in early-onset sporadic gastric carcinoma." J Cáncer Res Clin Oncol 131: 733-40

KIM, H C, S A ROH, I H GA, J S KIM, C S YU and J C KIM (2005). "CpG island methylation as an early event during adenoma progression in carcinogenesis of sporadic colorectal cáncer." J Gastroenterol Hepatol 20: 1920-6

KIM, J, H S LEE, S I BAE, Y M LEE and W H KIM (2005). "Silencing and CpG island methylation of GSTP1 is rare in ordinary gastric carcinomas but common in Epstein-Barr virus-associated gastric carcinomas." Anticancer Res 25: 4013-9

KORIYAMA, C, S KITAJIMA, M NOMOTO, K SUEYOSHI, T ITOH, Y EIZURU, S YONEZAWA and S AKIBA (2004). "Loss of pl6/CDKN2A protein in Epstein-Barr virus-associated gastric carcinoma." Oncology 67: 81-7

KUROKI, T, F TRAPASSO, S YENDAMURI, A MATSUYAMA, H ALDER, N N WILLIAMS, L R KAISER and C M CROCE (2003). "Allelic loss on chromosome 3p21.3 and promoter hypermethylation of semaphorin 3B in non-small cell lung cáncer." Cáncer Res 63: 3352-5

LAUREN, P (1965). "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 64: 31-49

LI, Q L, K ITO, C SAKAKURA, H FUKAMACHI, K INOUE, X Z CHI, K Y LEE, S NOMURA, C W LEE, S B HAN, H M KIM, W J KIM, H YAMAMOTO, N YAMASHITA, T YANO, T IKEDA, S ITOHARA, J INAZAWA, T ABE, A HAGIWARA, H YAMAGISHI, A OOE, A KANEDA, T SUGIMURA, T USHIJIMA, S C BAE and Y ITO (2002). "Causal relationship between the loss of RUNX3 expression and gastric cáncer." Cell 109: 113-24

LIU, Z, L E WANG, L WANG, K H LU, G B MILLS, M L BONDY and Q WEI (2005). "Methylation and messenger RNA expression of pl5INK4b but not pl6INK4a are independent risk factors for ovarian cáncer." Clin Cáncer Res 11: 4968-76

MILNE, A N, R SITARZ, R CARVALHO, F CARNEIRO and G J OFFERHAUS (2007). "Early onset gastric cáncer: on the road to unraveling gastric carcinogenesis." Curr Mol Med 7: 15-28

MIYAMOTO, K, K ASADA, T FUKUTOMI, E OKOCHI, Y YAGI, T HASEGAWA, T ASAHARA, T SUGIMURA and T USHIJIMA (2003). "Methylation-associated silencing of heparan sulfate D-glucosaminyl 3-0-sulfotransferase-2 (3-OST-2) in human breast, colon, lung and pancreatic cancers." Oncogene 22: 274-80

MORÍ, T, S R MARTÍNEZ, S J O'DAY, D L MORTON, N UMETANI, M KITAGO, A TANEMURA, S L NGUYEN, A N TRAN, H J WANG and D S HOON (2006). "Estrogen receptor-alpha methylation predicts melanoma progression." Cáncer Res 66: 6692-8

NISHIDA, N, T NISHIMURA, T NAGASAKA, I IKAI, G AJAY and C R BOLAND (2007). "Extensive Methylation Is Associated with {beta}-Catenin Mutations in Hepatocellular Carcinoma: Evidence for Two Distinct Pathways of Human Hepatocarcinogenesis." Cáncer Res 67: 4586-4594

OKA, T, M OUCHIDA, M KOYAMA, Y OGAMA, S TARADA, Y NAKATANI, T TANAKA, T YOSHINO, K HAYASHI, N OHARA, E KONDO, K TAKAHASHI, J TSUCHIYAMA, M TANIMOTO, K SHIMIZU and T AKAGI (2002). "Gene silencing of the tyrosine phosphatase SHP1 gene by aberrant methylation in leukemias/lymphomas." Cáncer Res 62: 6390-4

PILOZZI, E, C TALERICO, A PLATT, C FIDLER and L RUCO (2003). "p73 gene mutations in gastric adenocarcinomas." Mol Pathol 56: 60-2

PISANI, P, F BRAY and D M PARKIN (2002). "Estimates of the world-wide prevalence of cáncer for 25 sites in the adult population." Int J Cáncer 97: 72-81

RASTOGI, T, A HILDESHEIM and R SINHA (2004). "Opportunities for cáncer epidemiology in developing countries." Nat Rev Cáncer 4: 909-17

RIQUELME, E, M TANG, S BÁEZ, A DÍAZ, M PRUYAS, WISTUBA, II and A CORVALÁN (2007). "Frequent epigenetic inactivation of chromosome 3p candidate tumor suppressor genes in gallbladder carcinoma." Cáncer Lett 250: 100-6

SARBIA, M, H GEDDERT, B KLUMP, S KIEL, E ISKENDER and H E GABBERT (2004). "Hypermethylation of tumor suppressor genes (pl6INK4A, pl4ARF and APC) in adenocarcinomas of the upper gastrointestinal tract." Int J Cáncer 111: 224-8

SATO, K, G TAMURA, T TSUCHIYA, Y ENDOH, K SAKATA, T MOTOYAMA, O USUBA, W KIMURA, M TERASHIMA, S NISHIZUKA, T ZOU and S J MELTZER (2002). "Analysis of genetic and epigenetic alterations of the PTEN gene in gastric cáncer." Virchows Arch 440: 160-5

SEMBA, S, H YOKOZAKI, W YASUI and E TAHARA (1998). "Frequent microsatellite instability and loss of heterozygosity in the región including BRCA1 (17q21) in young patients with gastric cáncer." Int J Oncol 12: 1245-51

TAHARA, E (2004). "Genetic pathways of two types of gastric cáncer." IARC Sci Publ: 327-49

TAKAHASHI, T, N SHIVAPURKAR, E RIQUELME, H SHIGEMATSU, J REDDY, M SUZUKI, K MIYAJIMA, X ZHOU, B N BEKELE, A F GAZDAR and WISTUBA, II (2004). "Aberrant promoter hypermethylation of múltiple genes in gallbladder carcinoma and chronic cholecystitis." Clin Cáncer Res 10: 6126-33

TAKAHASHI, T, M SUZUKI, H SHIGEMATSU, N SHIVAPURKAR, C ECHEBIRI, M NOMURA, V STASTNY, M AUGUSTUS, C W WU, WISTUBA, II, S J MELTZER and A F GAZDAR (2005). "Aberrant methylation of Reprimo in human malignancies." Int J Cáncer 115: 503-10

TAMURA, G (2006). "Alterations of tumor suppressor and tumor-related genes in the development and progression of gastric cáncer." World J Gastroenterol 12: 192-8

USHIJIMA, T (2007). "Epigenetic field for cancerization." J Biochem Mol Biol 40: 142-50

VAN 'T VEER, L J, H DAI, M J VAN DE VIJVER, Y D HE, A A HART, M MAO, H L PETERSE, K VAN DER KOOY, M J MARTON, A T WITTEVEEN, G J SCHREIBER, R M KERKHOVEN, C ROBERTS, P S LINSLEY, R BERNARDS and S H FRIEND (2002). "Gene expression profiling predicts clinical outcome of breast cáncer." Nature 415: 530-6

VARIS, A, B VAN REES, M WETERMAN, A RISTIMAKI, J OFFERHAUS AND S KNUUTILA (2003). "DNA copy number changes in young gastric cáncer patients with special reference to chromosome 19." BrJ Cáncer 88: 1914-9

VAUHKONEN, M, H VAUHKONEN and P SIPPONEN (2006). "Pathology and molecular biology of gastric cáncer." Best Practice & Research Clinical Gastroenterology 20: 651-674

VIRMANI, A K, A RATHI, S ZOCHBAUER-MULLER, N SACCHI, Y FUKUYAMA, D BRYANT, A MAITRA, S HEDA, K M FONG, F THUNNISSEN, J D MINNA and A F GAZDAR (2000). "Promoter methylation and silencing of the retinoic acid receptor-beta gene in lung carcinomas." J Nati Cáncer Inst 92: 1303-7

WILD, A, A RAMASWAMY, P LANGER, I CELIK, V FENDRICH, B CHALOUPKA, B SIMÓN and D K BARTSCH (2003). "Frequent methylation-associated silencing of the tissue inhibitor of metalloproteinase-3 gene in pancreatic endocrine tumors." J Clin Endocrinol Metab 88: 1367-73

WISTUBA, I, L MAO and A F GAZDAR (2002). "Smoking molecular damage in bronchial epithelium." Oncogene21: 7298-306

XU, X L, J YU, H Y ZHANG, M H SUN, J GU, X DU, D R SHI, P WANG, Z H YANG and J D ZHU (2004). "Methylation profile of the promoter CpG islands of 31 genes that may contribute to colorectal carcinogenesis." World J Gastroenterol 10: 3441-54

YAMASHITA, K, H L PARK, M S KIM, M OSADA, Y TOKUMARU, H INOUE, M MORÍ and D SIDRANSKY (2006). "PGP9.5 Methylation in Diffuse type Gastric Cáncer." Cáncer Res 66: 3921-3927

YASUI, W, N OUE, P P AUNG, S MATSUMURA, M SHUTOH and H NAKAYAMA (2005). "Molecular-pathological prognostic factors of gastric cáncer: a review." Gastric Cáncer 8: 86-94

Corresponding Author: Dr. Alejandro Corvalan, Laboratorio Patología Molecular y Epidemiología, Centro Investigaciones Médicas, Pontificia Universidad Católica de Chile, Marcoleta 391 Santiago 8330074 Chile, Phone: 56(2) 3548289, E-mail: corvalan@med.puc.cl

Received: August 27, 2008. In Revised form: May 26, 2008. Accepted: September 12, 2008

 

Creative Commons License Todo el contenido de esta revista, excepto dónde está identificado, está bajo una Licencia Creative Commons