Skip to main content
Erschienen in: Hereditary Cancer in Clinical Practice 1/2014

Open Access 01.12.2014 | Research

Genetic screening analysis of patients with hereditary diffuse gastric cancer from northern and northeastern Brazil

verfasst von: Caroline Aquino Moreira-Nunes, Mariceli Baia Leão Barros, Bárbara do Nascimento Borges, Raquel Carvalho Montenegro, Leticia Martins Lamarão, Helem Ferreira Ribeiro, Amanda Braga Bona, Paulo Pimentel Assumpção, Juan Antonio Rey, Giovanny Rebouças Pinto, Rommel Rodriguez Burbano

Erschienen in: Hereditary Cancer in Clinical Practice | Ausgabe 1/2014

Abstract

Background

Hereditary diffuse gastric cancer (HDGC) is a hereditary autosomal inherited syndrome associated with CDH1 germline mutations. In Brazil, gastrointestinal tumors are among the most prevalent tumor types and constitute a serious public health problem, especially in the northern and northeastern regions. This study aimed to investigate germline mutations, methylation pattern and genomic rearrangements in the CDH1 gene and quantitative changes in the DNA of HDGC patients in northern and northeastern Brazil.

Methods

Twenty-seven DNA samples from the members of four families affected by HDGC were analyzed using array comparative genomic hybridization (aCGH), DNA sequencing and methylation pattern.

Results

No evidence of gain and loss events or any rearrangements were found in any of the samples tested using aCGH. No promoter region hypermethylation was observed either. Two of the four families presented different types of germline mutations. The 185G > T and 1018A > G germline mutations detected in this study have been described in Asian and European families, respectively. The ancestors of the two families carrying these mutations had originated from those continents.

Conclusion

This is the first study to evaluate CDH1 gene germline mutations in Brazilian families with HDGC. In our study, 50% of the families showed no CDH1 gene alterations, and it is possible that in regions with a high incidence of gastric cancer, such as northern and northeastern Brazil, environmental factors might have induced the different genetic alterations analyzed in this study.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1897-4287-12-18) contains supplementary material, which is available to authorized users.
Caroline Aquino Moreira-Nunes contributed equally to this work.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

Conceived and designed the experiments: CFAMN, MBLB, BNB, LML, RMRB. Performed the experiments: CFAMN, MBLB, BNB, LML, ABB. Analyzed the data: CFAMN MBLB, BNB, HFR, RMRB. Wrote the paper: CFAMN, PPA, JAR, GRP, RMRB. All authors read and approved the final manuscript.

Introduction

In northern and northeastern Brazil, gastric cancer (GC) is the second most common type of cancer and is considered a serious public health problem because it is usually diagnosed at an advanced stage[1]. In addition to its sporadic manifestation, GC is also associated with various syndromes that predispose the carrier to cancer. Among the familial forms of GC, hereditary diffuse gastric cancer (HDGC) is the only form with a well-defined genetic cause[2, 3].
In order for a family to qualify for a diagnosis of HDGC, the following criteria must be met: two or more documented cases of diffuse gastric cancer in first- or second-degree relatives with at least one diagnosed before the age of 50 years or three or more cases of documented diffuse gastric cancer in first- or second-degree relatives independent of the age of onset[4, 5].
The correlation between an E-cadherin gene germline mutation (CDH1 inactivation) and the predisposition to diffuse gastric cancer was first identified in a large family in New Zealand[6]. Based on this finding, HDGC was characterized and other occurrences were described in patients with different ethnic backgrounds[7, 8].
The CDH1 gene, located on the 16q22.1 chromosome, encodes the E-cadherin intercellular adhesion protein; this protein acts as a tumor suppressor and plays an important role in maintenance of the epithelial tissue architecture[9]. Mutations in this gene primarily affect both the intracellular and extracellular domains of the protein and thus affect the integrity of the protein, leading to disturbances in epithelial tissue cell-cell adhesion, increased cell motility and an enhanced infiltrative capacity and tumor metastasis[10, 11]. As HDGC is an infiltrative tumor, endoscopy and biopsy-based diagnostic strategies are inefficient; furthermore, given the high penetrance of CDH1 mutations, prophylactic gastrectomy is recommended for affected patients[11, 12].
Over one hundred germline mutations have been described for the CDH1 gene. Although several of these mutations have been detected in different families, to date, no hotspot has been characterized[3, 13]. Although the CDH1 gene is the predominantly affected gene in HDGC, other inactivation mechanisms should be investigated. Additionally, large deletions might also be responsible for CDH1 inactivation[8, 1420]. For this reason, an array comparative genomic hybridization (aCGH) analysis is essential to the identification of quantitative alterations in the CDH1 gene or other genome regions in families affected by HDGC[21].
The objective of this study was to identify germline mutations in the CDH1 gene and/or quantitative genomic alterations in four families with HDGC in northern and northeastern Brazil.

Methodology

Patients

The samples evaluated in this study were obtained from patients who fulfilled the clinical criteria according to the latest consensus of the International Gastric Cancer Linkage Consortium[5]. The patients had been treated at referral hospitals in northern and northeastern Brazil. The samples were processed in their states of origin and then sent to the Federal University of Pará (Universidade Federal do Pará; UFPA) for genetic analysis.
A retrospective study of the family members was also conducted to identify previous generations with DGC. All genetically analyzed patients (or their guardians) signed consent forms that had been approved by the Research Ethics Committee of University Hospital João de Barros Barreto (Hospital Universitário João de Barros Barreto; HUJBB) under protocol number 274/12. Assurance was provided that the use of biological materials and participation in the study would not cause any harm to nor have any negative influence on patient treatment.
All biological materials except one sample (that was Formaldehyde Fixed-Paraffin Embedded tissue) was blood DNA. And all genetic screening analysis - mutation, deletion and methylation - were all looking for germline changes, not somatic.

DNA extraction

Peripheral blood samples were collected in EDTA-containing tubes and extracted with the QIAamp® DNA Blood Mini Kit (Qiagen N.V., Venlo, The Netherlands) according to the manufacturer’s instructions.
Paraffin block sample was obtained from one patient, and extracted with the QIAamp DNA® FFPE Tissue Kit (Qiagen) according to the manufacturer’s instructions.

Genotypic analysis

The obtained DNA was used to perform molecular screening of the CDH1 gene. Gene fragments containing the analyzed polymorphisms were amplified by polymerase chain reaction (PCR) using gene-specific reaction conditions and primers (Table 1) according to Brooks-Wilson et al.[22].
Table 1
Primer sequences used in the study and their annealing temperatures
Exon
Forward 5′- 3′
Reverse 5′- 3′
Tm (°C)
1
M13F GTGAACCCTCAGCCAATCAG
M13R TGACGACGGGAGAGGAAG
63
2
M13F TGTTGGTTTCGGTGAGCAG
M13R GGTGT3GGGAGTGCAATTTCT
61
3
M13F CGCTCTTTGGAGAAGGAATG
M13R AACGGTACCAAGGCTGAGAA
58
4
M13F GCTGTCTGGCTAGGTTGGAC
M13R TTTTCCCTTTCTCTCCTTGG
58
5
M13F GAAAGGGAAAAGACCCAGTG
M13R GGATCCAGCATGGGTTGAC
58
6
M13F GCCCCTTCTCCCATGTTT
M13R CTTTGGGCTTGGACAACACT
56
7
M13F GGGCAGAATTGGATTAAGCA
M13R TGTCCACGGGATTGAGCTA
57
8
M13F CTGGGTAGGCCAAAGGT
M13R CCATGAGCAGTGGTGACACTT
57
9
M13F AATCCTTTAGCCCCCTGAGA
M13R AGGGGACAAGGGTATGAACA
61
10
M13F CCAAAAGCAACAGTTAAGGA
M13R CAAATGACAAAATGCCATGA
56
11
M13F AGCGCTTAAGCCGTTTTCA
M13R GAGGGGCAAGGAACTGAACT
60
12
M13F AAGGCAATGGGGATTCATTA
M13R ATTGAAAGGTGGGGATCTGG
59
13
M13F CAATTTTATTCTGGAATGAGCTTTT
M13R CAGGAAATAAACCTCCTCCATTT
55
14
M13F GCTGCTTCTGGCCTTCTTA
M13R GCTGTTTCAAATGCCTACCTCT
55
15
M13F TGAACATAGCCCTGTGTGTATG
M13R TTTTGACACAACTCCTCCTG
58
16
M13F AGACTTCTTGCCCCAGATGA
M13R AACCACCAGCAACGTGATTT
63
Forward primers have – 21M13F tail attached for sequencing: TGTAAAACGACGGCCAGT; reverse primers have M13R tail attached for sequencing: CAGGAAACAGCTATGAC.
The result of each reaction was subjected to direct sequencing on an ABI 3130 capillary sequencing platform (Applied Biosystems/Life Technologies, Carlsbad, CA, USA). The sequences were obtained as electropherograms and analyzed with the software package provided with the equipment. The generated sequences were analyzed with the BIQ Analyzer software package[23].

aCGH

High-density probe microarray analyses were performed to determine the copy number variation (CNV). The complete genomes of all patients were evaluated with the goal of identifying genes related to tumor development.
The Affymetrix® CytoScan HD Array (Affymetrix, Inc., Santa Clara, CA, USA) was used; this system features a total of approximately 1.9 million probes for detecting CNV and 750,000 SNP molecular markers. The standard protocol incorporates the following eight procedures before scanning the chip: genomic DNA digestion, NSP adapter ligation, fragment amplification by PCR (Polymerase Chain Reaction), PCR product purification, PCR product fragmentation, end-labeling, hybridization and washing.
The Chromosome Analysis Suite software v1.2.1 (Affymetrix®) was used for the chip analysis.

Methylation status

For methylation analyses, the samples were subjected to DNA modification using sodium bisulfite[24]. A fragment with 22 CpGs of the CDH1 promoter region was amplified using a nested PCR strategy[25]. Fragments obtained were purified using PCR Purification Kit (Invitrogen/Life Technologies, Carlsbad, CA, USA) and sequenced using an ABI3130 automatic sequencer (Applied Biosystems, Foster City, CA, USA). The sequences were aligned with BioEdit v7.0.5[26]. Methylation analyses were run in BiQ Analyzer[23] software.

Immunohistochemistry

An anti-cadherin (Abcam PLC, Cambridge, UK) commercial primary antibody was used to detect the protein product of the CDH1 gene (HDGC). Streptavidin-biotin-peroxidase staining as described by Hsu et al.[27] was adopted as the immunohistochemical method. The normality parameter was defined with samples from normal (non-tumor) formalin-fixed and paraffin-embedded tissues that had been obtained from routine samples. The World Health Organization histopathological classifications for each tumor were used as well as the Lauren classification for gastric cancer[28].

Results

The detailed patient data are shown in Table 2. A total of 27 patient samples were collected for genetic analysis from four unrelated families with histories of DGC in northern and northeastern Brazil. All biological samples collected were peripheral blood, except for the patient AM05, whose sample was Paraffin-embedded tumor. The sample comprised 18 men and nine women with a mean age of 42 years and an age range of 20–75 years (24–56 years for women and 20–75 years for men).
Table 2
Identification of the patients analyzed in this study
Patient
Gender
Age at diagnosis
Type of cancer
(Family A)
   
^AM05
male
36
*DGC
AM06
female
56
none
AM07
male
31
*DGC
AM08
female
26
none
(Family B)
   
P05
male
29
*DGC
P06
male
27
*DGC
P07
male
23
none
P08
male
20
none
(Family C)
   
C05
male
75
Prostate
C14
male
52
*DGC
C15
male
50
*DGC
C16
female
49
*DGC
C17
female
48
none
C18
female
45
none
C19
female
47
none
C20
male
25
none
C21
male
23
none
C22
female
42
none
C23
female
24
none
C24
male
56
none
C25
male
35
none
C26
male
31
none
C27
male
58
none
C28
female
31
none
(Family D)
   
M08
female
67
*DGC
M09
male
62
*DGC
M10
male
64
none
^- Paraffin-embedded tumor sample.
* - Diffuse Gastric Cancer.
Of these 27 patients, nine (33.4%) had been previously diagnosed with diffuse gastric cancer, whereas the remaining patients did not have any type of gastric tumor. Among the patients diagnosed with DGC, the age at diagnosis ranged from 27–67 years.After the retrospective study and using information collected from the analyzed patients, it was possible to identify relatives with a history of DGC and other types of tumors (Figure 1).

Mutations in the CDH1 gene

Among the four families of patients, two families exhibited germline mutations in the CDH1 gene, namely family A from northern Brazil and family B from northeastern Brazil (Table 3). In the other two families, which originated from northeastern Brazil, no mutations were found in the evaluated CDH1 exons.
Table 3
Germline CDH1 mutations identified in hereditary diffuse gastric cancer families
Family ID
Patient ID
Age at onset/sex
Exon
Base changes
Amino acid change
Mutation consequence
A
AM05
36/male
3
185 G > T
Gly ⋄ Val
Missense
 
AM07
31/male
    
B
P05
29/male
8
1018 A > G
Thr ⋄ Ala
Missense
 
P06
27/male
    

Array comparative genomic hybridization (aCGH)

The aCGH sample analysis showed no gains or losses of DNA in the four tested families (27 samples).

Immunoreactivity in HDGC

CDH1 protein detection was performed only for patient AM05, a carrier of the germline mutation 185 G > T and member of family A. This patient died before the start of the study, and a genetic analysis was performed on a paraffin-embedded tumor tissue. The other study participants with this syndrome donated peripheral blood for genetic analysis, and consequently, there was no need to request paraffin blocks of the resected tumor tissues. The tumor cells of patient AM05, revealed negative immunoreactivity to the E-cadherin protein.

Methylation status in HDGC

We analyzed the methylation pattern of a CDH1 gene promoter region fragment in one CpG island containing 22 CpG dinucleotides, to find possible heritable epimutations. No promoter region hypermethylation was observed in all 27 tested individuals, from the four families, and also in 10 healthy controls.

Discussion

In addition to sporadic manifestation, gastric cancer can be associated with various syndromes that predispose the carrier to cancer. Among the familial forms of gastric cancer, HDGC is the only syndrome with a well-defined genetic cause[2, 3].
The relationship between a genetic germline mutation in the E-cadherin-encoding gene (CDH1 inactivation) and the predisposition to HDGC was first identified in a family in New Zealand[6]. Based on this finding, the HDGC syndrome was characterized, and other occurrences were described in patients with different ethnic backgrounds[29].
Mutations in CDH1 gene affect protein integrity, thus causing disturbances in cell-cell adhesion in epithelial tissues, increasing cell motility and enhancing infiltrative behavior of the tumor and metastasis development[10, 11].
This genetic study of four families that carried the HDGC syndrome allowed us to evaluate a previously unaddressed issue within the Brazilian population. CDH1 germline mutations are observed in 30–40% of cases that meet the clinical criteria for HDGC[30, 31]. In our study, 50% of the families exhibited CDH1 gene mutations. This information is useful in clinical evaluations of patients with family histories of HDGC because in addition to prophylactic measures, the information facilitates monitoring aimed at identifying the occurrence of other tumors such as lobular carcinomas of the breast; these types of cancers had also been detected in members of the families analyzed in this study[13, 20, 32].
The cases described in the literature demonstrate that the most common CDH1 gene alterations associated with HDGC are point mutations and small changes in the reading frame; these occur in approximately 93% of families with CDH1 gene mutations[13, 20, 33].
In this study, two individuals in family A, a father and son (Figure 1A), were carriers of a familial CDH1 gene mutation at nucleotide position 185 (185G > T); this mutation causes a change in the protein structure from the amino acid glycine to valine. This missense mutation was first described by Shinmura et al.[34] in a family of Japanese origin. Our results suggest the hypothesis that this 185G > T mutation was introduced into family A by the grandfather of the proband (first generation pedigree), who was of Japanese origin, and suggest that this mutation might result from this ethnic background.
A CDH1 gene germline mutation was also identified in two brothers from family B who had developed an early form of gastric cancer (before 45 years of age)[35]. The mutation was identified at nucleotide position 1018 (1018A > G) and resulted in a change from the amino acid threonine to alanine. The 1018A > G mutation was previously described in a family of European origin and in a Chinese family[36, 37]. Because the proband of this family was of Portuguese descent, it is possible that their ancestors brought this mutation to Brazil upon immigration. On the other hand, another alternative to ancestral mutations is an increased susceptibility of different CDH1 locations to mutations.
A literature review revealed the occurrence of a total of 122 germline mutations in the CDH1 gene in HDGC[38]. Although several types of mutations have been detected in different families, no hotspot has been characterized to date[3, 13]; this led us to sequence all 16 exons of the gene in the 30 members of the four analyzed families.
Of the mutations reported in the literature, approximately 15% are shared by many families worldwide, suggesting that CDH1 mutation-associated HDGC might share a common ancestry, as was suggested for families A and B in this study[33, 39].
In the other two families (C and D), in which the evaluated subjects carried no CDH1 gene mutations, there were considerable numbers of HDGC-affected members in different generations (Figure 1). Pinheiro et al.[20] have proposed that many HDGC families carry some mutations in non-exonic regulatory regions of CDH1 (or in upstream regulators). Based on this, it may be possible in the future to test families C and D, for allele-specific loss on CDH1.
In other populations in which the incidence of gastric cancer is high, there are also reports of HDGC-carrier families with large numbers of affected individuals who do not carry CDH1 gene germline mutations. This finding might be consequent to exposure to environmental risk factors or the susceptibility of individuals to genetic alterations in low-penetrance genes[40, 41].
In a study conducted in southeastern Brazil on 88 patients with early gastric cancer, 16 of whom had familial histories of gastric cancer, it was possible to detect changes in the E-cadherin protein expression in 41 individuals via immunohistochemical analysis. Although CDH1 gene sequencing was not conducted to identify possible mutations, the immunohistochemical analysis revealed the involvement of CDH1 in the early development of gastric cancer in a Brazilian population[42].
Because the CDH1 gene acts as a tumor suppressor, other gene inactivation mechanisms of this gene besides germline mutations in one allele should be investigated to identify changes to the second allele in somatic cells (second event of the Knudson hypothesis[43]), which was not able to be done in the present work. Mechanisms such as CDH1 gene deletion[33] and promoter region hypermethylation[44], were investigated in germline from those analyzed patients, because are also considered potentially responsible for its inactivation.
Cytosine hypermethylation in the CpG dinucleotides present within the CDH1 promoter region induces transcriptional silencing[45] and might thus explain the lack of E-cadherin immunoreactivity. However, analyses to confirm the presence of CDH1 gene promoter region hypermethylation in patients with HDGC have not been conclusive. For example, studies by Li et al.[46] and Concolino et al.[47] demonstrated CDH1 promoter hypermethylation-mediated gene silencing in 53–57% of patients with HDGC. Conversely, Wu et al.[48] reported an absence of this phenomenon in 140 Chinese gastric cancer patients with a familial history of HDGC.
The analyses conducted in the present study did not detect CDH1 gene promoter region hypermethylation in the families with HDGC syndrome. Our results reinforce the theory put forth by Yamada et al.[19] who, after analyzing 22 Japanese patients with early gastric cancer, suggested that germline CDH1 promoter hypermethylation was not a predisposing factor of gastric cancer.
In addition to epigenetic alterations in the promoter region, the CDH1 gene might also suffer deletions even in the absence of point mutations in the germlines of patients with HDGC[49]. Kim et al.[50] used multiplex ligation-dependent probe amplification (MLPA) to analyze 23 patients with HDGC and, similarly to our study, found no deletions or duplications in these genes.
Oliveira et al.[33] genetically analyzed 160 patients with HDGC from different geographic regions and found that CDH1 gene deletions occurred in the peripheral blood in approximately 4% of families affected by this syndrome. The same authors also observed that all families with CDH1 deletions originated from countries with low gastric cancer incidence rates. A large number of patients with HDGC, in northern and northeastern from Brazil, needs to be analyzed to verify if the absence of CDH1 gene deletions and other quantitative genomic alterations, as found in patients of this work, corroborate this observation because these regions have high gastric cancer incidence rates.
It is widely known that the most common forms of cancer develop as a result of interactions between endogenous and environmental factors such as the diet[51]. Environmental factors might be related to the high incidence of these neoplasms in the northern and northeastern regions, or for some reason the HDGC frequency is higher than elsewhere in the world, where sporadic gastric cancer is also endemic, such as Japan and Korea. In particular, Helicobacter pylori (H. pylori) infection during childhood as well as the high consumption of salt-preserved foods, infrequent use of refrigeration and low consumption rates of cereals, fresh fruits and vegetables are considered risk factors for gastrointestinal tumors[5255].

Conclusions

This is the first study to evaluate CDH1 gene germline mutations and quantitative gene alterations in Brazilian families with HDGC. CDH1 gene germline mutations were found in 50% of the families evaluated. The detected mutations appeared to originate from Asia and Europe. This migratory flow does not rule out the possibility that environmental factors might have caused these same mutations in the affected members of the analyzed families. No quantitative changes were observed in the genomes of any of the analyzed families. It is possible that in regions with high gastric cancer incidence rates such as northern and northeastern Brazil, environmental factors or other molecular mechanisms might induce different genetic alterations from those analyzed in this study.

Acknowledgments

This study was supported by Conseho Nacional de Desenvolvimento Científico e Tecnológico (http://​www.​cnpq.​br) grant number 401976/2010-6, 305220/2013-6 to RRB and Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (http://​www.​capes.​gov.​br) grant number PNPD 2810/2011 to CFAMN. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​4.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly credited. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

Conceived and designed the experiments: CFAMN, MBLB, BNB, LML, RMRB. Performed the experiments: CFAMN, MBLB, BNB, LML, ABB. Analyzed the data: CFAMN MBLB, BNB, HFR, RMRB. Wrote the paper: CFAMN, PPA, JAR, GRP, RMRB. All authors read and approved the final manuscript.
Anhänge

Authors’ original submitted files for images

Below are the links to the authors’ original submitted files for images.
Literatur
2.
Zurück zum Zitat Bresciani C, Perez RO, Gama-Rodrigues J: Familial gastric cancer. Arq Gastroenterol 2003, 40: 114–7.CrossRefPubMed Bresciani C, Perez RO, Gama-Rodrigues J: Familial gastric cancer. Arq Gastroenterol 2003, 40: 114–7.CrossRefPubMed
4.
Zurück zum Zitat Caldas C, Carneiro F, Lynch HT, Yokota J, Wiesner GL, Powell SM, Lewis FR, Huntsman DG, Pharoah PDP, Jankowski JA, MacLeod P, Vogelsang H, Keller G, Park KGM, Richards FM, Maher ER, Gayther SA, Oliveira C, Grehan N, Wight D, Seruca R, Roviello F, Ponder BAJ, Jackson CE: Familial gastric cancer: overview and guidelines for management. J Med Genet 1999, 36: 873–80.PubMedPubMedCentral Caldas C, Carneiro F, Lynch HT, Yokota J, Wiesner GL, Powell SM, Lewis FR, Huntsman DG, Pharoah PDP, Jankowski JA, MacLeod P, Vogelsang H, Keller G, Park KGM, Richards FM, Maher ER, Gayther SA, Oliveira C, Grehan N, Wight D, Seruca R, Roviello F, Ponder BAJ, Jackson CE: Familial gastric cancer: overview and guidelines for management. J Med Genet 1999, 36: 873–80.PubMedPubMedCentral
5.
Zurück zum Zitat Fitzgerald RC, Hardwick R, Huntsman D, Carneiro F, Guilford P, Blair V, Chung DC, Norton J, Ragunath K, Krieken JHV, Dwerryhouse S, Caldas C: Hereditary diffuse gastric cancer: updated consensus guidelines for clinical management and directions for future research. J Med Genet 2010, 47: 436–44.CrossRefPubMedPubMedCentral Fitzgerald RC, Hardwick R, Huntsman D, Carneiro F, Guilford P, Blair V, Chung DC, Norton J, Ragunath K, Krieken JHV, Dwerryhouse S, Caldas C: Hereditary diffuse gastric cancer: updated consensus guidelines for clinical management and directions for future research. J Med Genet 2010, 47: 436–44.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Guilford P, Hopkins J, Harraway J, McLeod M, McLeod N, Harawira P, Taite H, Scoular R, Miller A, Reeve AE: E-cadherin germline mutations in familial gastric cancer. Nature 1998, 392: 402–5.CrossRefPubMed Guilford P, Hopkins J, Harraway J, McLeod M, McLeod N, Harawira P, Taite H, Scoular R, Miller A, Reeve AE: E-cadherin germline mutations in familial gastric cancer. Nature 1998, 392: 402–5.CrossRefPubMed
7.
8.
Zurück zum Zitat Moran CJ, Joyce M, McAnena OJ: CDH1 associated gastric cancer: a report of a family and review of the literature. Eur J Surg Oncol 2005, 31: 259–64.CrossRefPubMed Moran CJ, Joyce M, McAnena OJ: CDH1 associated gastric cancer: a report of a family and review of the literature. Eur J Surg Oncol 2005, 31: 259–64.CrossRefPubMed
9.
Zurück zum Zitat van Roy F, Berx G: The cell-cell adhesion molecule E-cadherin. Cell Mol Life Sci 2008, 65: 3756–88.CrossRefPubMed van Roy F, Berx G: The cell-cell adhesion molecule E-cadherin. Cell Mol Life Sci 2008, 65: 3756–88.CrossRefPubMed
10.
Zurück zum Zitat Mateus AR, Simões-Correia J, Figueiredo J, Heindl S, Alves CC, Suriano G, Luber B, Seruca R: E-cadherin mutations and cell motility: a genotype–phenotype correlation. Exp Cell Res 2009, 315: 1393–402.CrossRefPubMed Mateus AR, Simões-Correia J, Figueiredo J, Heindl S, Alves CC, Suriano G, Luber B, Seruca R: E-cadherin mutations and cell motility: a genotype–phenotype correlation. Exp Cell Res 2009, 315: 1393–402.CrossRefPubMed
11.
Zurück zum Zitat Ghaffari SR, Rafati M, Sabokbar T, Dastan J: A novel truncating mutation in the E- cadherin gene in the first Iranian family with hereditary diffuse gastric cancer. Eur J Surg Oncol 2010, 36: 559–62.CrossRefPubMed Ghaffari SR, Rafati M, Sabokbar T, Dastan J: A novel truncating mutation in the E- cadherin gene in the first Iranian family with hereditary diffuse gastric cancer. Eur J Surg Oncol 2010, 36: 559–62.CrossRefPubMed
12.
Zurück zum Zitat Mayrbaeurl B, Kellerf G, Schauerb W, Burgstaller S, Czompo M, Hoebling W, Knoflach P, Duba HC, Hoefler H, Thaler J: Germline mutation of the E- cadherin gene in three sibling cases with advanced gastric cancer: clinical consequences for the other family members. Eur J Gastroenterol Hepatol 2010, 22: 306–10.CrossRefPubMed Mayrbaeurl B, Kellerf G, Schauerb W, Burgstaller S, Czompo M, Hoebling W, Knoflach P, Duba HC, Hoefler H, Thaler J: Germline mutation of the E- cadherin gene in three sibling cases with advanced gastric cancer: clinical consequences for the other family members. Eur J Gastroenterol Hepatol 2010, 22: 306–10.CrossRefPubMed
13.
Zurück zum Zitat Guilford P, Humar B, Blair V: Hereditary diffuse gastric cancer: translation of CDH1 germline mutations into clinical practice. Gastric Cancer 2010, 13: 1–10.CrossRefPubMed Guilford P, Humar B, Blair V: Hereditary diffuse gastric cancer: translation of CDH1 germline mutations into clinical practice. Gastric Cancer 2010, 13: 1–10.CrossRefPubMed
15.
Zurück zum Zitat Oliveira C, Suriano G, Ferreira P, Canedo P, Kaurah P, Mateus R, Ferreira A, Ferreira AC, Oliveira MJ, Figueiredo C, Carneiro F, Keller G, Huntsman D, Machado JC, Seruca R: Genetic screening for familial gastric cancer. Hered Cancer Clin Pract 2004, 2: 51–64.CrossRefPubMedPubMedCentral Oliveira C, Suriano G, Ferreira P, Canedo P, Kaurah P, Mateus R, Ferreira A, Ferreira AC, Oliveira MJ, Figueiredo C, Carneiro F, Keller G, Huntsman D, Machado JC, Seruca R: Genetic screening for familial gastric cancer. Hered Cancer Clin Pract 2004, 2: 51–64.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Corso G, Roviello F, Paredes J, Pedrazzani C, Novais M, Correia J, Marelli D, Cirnes L, Seruca R, Oliveira C, Suriano G: Characterization of the P373L E-cadherin germline missense mutation and implication for clinical management. Eur J Surg Oncol 2007, 33: 1061–7.CrossRefPubMed Corso G, Roviello F, Paredes J, Pedrazzani C, Novais M, Correia J, Marelli D, Cirnes L, Seruca R, Oliveira C, Suriano G: Characterization of the P373L E-cadherin germline missense mutation and implication for clinical management. Eur J Surg Oncol 2007, 33: 1061–7.CrossRefPubMed
17.
Zurück zum Zitat Barber M, Murrell A, Ito Y, Maia AT, Hyland S, Oliveira C, Save V, Carneiro F, Paterson AL, Grehan N, Dwerryhouse S, Lao-Sirieix P, Caldas C, Fitzgerald RC: Mechanisms and sequelae of E-cadherin silencing in hereditary diffuse gastric cancer. J Pathol 2008, 216: 295–306.CrossRefPubMed Barber M, Murrell A, Ito Y, Maia AT, Hyland S, Oliveira C, Save V, Carneiro F, Paterson AL, Grehan N, Dwerryhouse S, Lao-Sirieix P, Caldas C, Fitzgerald RC: Mechanisms and sequelae of E-cadherin silencing in hereditary diffuse gastric cancer. J Pathol 2008, 216: 295–306.CrossRefPubMed
18.
Zurück zum Zitat Oliveira C, Sousa S, Pinheiro H, Karam R, Bordeira-Carriço R, Senz J, Kaurah P, Carvalho J, Pereira R, Gusmão L, Wen X, Cipriano MA, Yokota J, Carneiro F, Huntsman D, Seruca R: Quantification of epigenetic and genetic 2nd hits in CDH1 during hereditary diffuse gastric cancer syndrome progression. Gastroenterology 2009, 136: 2137–48.CrossRefPubMed Oliveira C, Sousa S, Pinheiro H, Karam R, Bordeira-Carriço R, Senz J, Kaurah P, Carvalho J, Pereira R, Gusmão L, Wen X, Cipriano MA, Yokota J, Carneiro F, Huntsman D, Seruca R: Quantification of epigenetic and genetic 2nd hits in CDH1 during hereditary diffuse gastric cancer syndrome progression. Gastroenterology 2009, 136: 2137–48.CrossRefPubMed
19.
Zurück zum Zitat Yamada H, Shinmura K, Goto M, Iwaizumi M, Konno H, Kataoka H, Yamada M, Ozawa T, Tsuneyoshi T, Tanioka F, Sugimura H: Absence of germline mono-allelic promoter hypermethylation of the CDH1 gene in gastric cancer patients. Mol Cancer 2009, 8: 63.CrossRefPubMedPubMedCentral Yamada H, Shinmura K, Goto M, Iwaizumi M, Konno H, Kataoka H, Yamada M, Ozawa T, Tsuneyoshi T, Tanioka F, Sugimura H: Absence of germline mono-allelic promoter hypermethylation of the CDH1 gene in gastric cancer patients. Mol Cancer 2009, 8: 63.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Pinheiro H, Bordeira-Carriço R, Seixas S, Carvalho J, Senz J, Oliveira P, Inácio P, Gusmão L, Rocha J, Huntsman D, Seruca R, Oliveira C: Allele-specific CDH1 downregulation and hereditary diffuse gastric cancer. Hum Mol Genet 2010, 19: 943–52.CrossRefPubMed Pinheiro H, Bordeira-Carriço R, Seixas S, Carvalho J, Senz J, Oliveira P, Inácio P, Gusmão L, Rocha J, Huntsman D, Seruca R, Oliveira C: Allele-specific CDH1 downregulation and hereditary diffuse gastric cancer. Hum Mol Genet 2010, 19: 943–52.CrossRefPubMed
21.
Zurück zum Zitat Fan B, Dachrut S, Coral H, Yuen ST, Chu KM, Law S, Zhang L, Ji J, Leung SY, Chen X: Integration of DNA copy number alterations and transcriptional expression analysis in human gastric cancer. PLoS ONE 2012, 7: e29824.CrossRefPubMedPubMedCentral Fan B, Dachrut S, Coral H, Yuen ST, Chu KM, Law S, Zhang L, Ji J, Leung SY, Chen X: Integration of DNA copy number alterations and transcriptional expression analysis in human gastric cancer. PLoS ONE 2012, 7: e29824.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Brooks-Wilson AR, Kaurah P, Suriano G, Leach S, Senz J, Grehan N, Butterfield YSN, Jeyes J, Schinas J, Bacani J, Kelsey M, Ferreira P, MacGillivray B, MacLeod P, Micek M, Ford J, Foulkes W, Australie K, Greenberg C, LaPointe M, Gilpin C, Nikkel S, Gilchrist D, Hughes R, Jackson CE, Monagham KG, Oliveira MJ, Seruca R, Gallinger S, Caldas C, Huntsman D: Germline E-cadherin mutations in hereditary diffuse gastric cancer: assessment of 42 new families and review of genetic screening criteria. J Med Genet 2004, 41: 508–17.CrossRefPubMedPubMedCentral Brooks-Wilson AR, Kaurah P, Suriano G, Leach S, Senz J, Grehan N, Butterfield YSN, Jeyes J, Schinas J, Bacani J, Kelsey M, Ferreira P, MacGillivray B, MacLeod P, Micek M, Ford J, Foulkes W, Australie K, Greenberg C, LaPointe M, Gilpin C, Nikkel S, Gilchrist D, Hughes R, Jackson CE, Monagham KG, Oliveira MJ, Seruca R, Gallinger S, Caldas C, Huntsman D: Germline E-cadherin mutations in hereditary diffuse gastric cancer: assessment of 42 new families and review of genetic screening criteria. J Med Genet 2004, 41: 508–17.CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Bock C, Reither S, Mikeska T, Paulsen M, Walter J, Lengauer T: BiQ Analyzer: visualization and quality control for DNA methylation data from bisulfite sequencing. Bioinformatics 2005, 21: 4067–8.CrossRefPubMed Bock C, Reither S, Mikeska T, Paulsen M, Walter J, Lengauer T: BiQ Analyzer: visualization and quality control for DNA methylation data from bisulfite sequencing. Bioinformatics 2005, 21: 4067–8.CrossRefPubMed
24.
Zurück zum Zitat Herman JG, Graff JR, Myohanen S, Nelkin BD, Baylin : Methylation-specific PCR: a novel PCR assay for methylation status of CpG islands. Proc Natl Acad Sci USA 1996, 93: 9821–9826.CrossRefPubMedPubMedCentral Herman JG, Graff JR, Myohanen S, Nelkin BD, Baylin : Methylation-specific PCR: a novel PCR assay for methylation status of CpG islands. Proc Natl Acad Sci USA 1996, 93: 9821–9826.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Nojima D, Nakajima K, Li L, Franks J, Ribeiro-Filho L, Ishii N, Dahiya R: CpG methylation of promoter region inactivates E-cadherin gene in renal cell carcinoma. Molecular Carcinogenesis 2001, 32: 10–27.CrossRef Nojima D, Nakajima K, Li L, Franks J, Ribeiro-Filho L, Ishii N, Dahiya R: CpG methylation of promoter region inactivates E-cadherin gene in renal cell carcinoma. Molecular Carcinogenesis 2001, 32: 10–27.CrossRef
26.
Zurück zum Zitat Hall TA: BioEdit: a user-friendly biological sequence aligment editor and analysis program for Windows 95/98/NT. Nucleic Acids Symp Ser 1999, 41: 95–8. Hall TA: BioEdit: a user-friendly biological sequence aligment editor and analysis program for Windows 95/98/NT. Nucleic Acids Symp Ser 1999, 41: 95–8.
27.
Zurück zum Zitat Hsu SM, Raine L: Protein A, avidin, and biotin in immunohistochemistry. J Histochem Cytochem 1981, 29: 1349–53.CrossRefPubMed Hsu SM, Raine L: Protein A, avidin, and biotin in immunohistochemistry. J Histochem Cytochem 1981, 29: 1349–53.CrossRefPubMed
28.
Zurück zum Zitat 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 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
29.
Zurück zum Zitat Oliveira C, Pinheiro H, Figueiredo J, Seuca R, Carneiro F: E-cadherin alterations in hereditary disorders with emphasis on hereditary diffuse gastric cancer. Prog Mol Biol Transl Sci 2013, 116: 337–59.CrossRefPubMed Oliveira C, Pinheiro H, Figueiredo J, Seuca R, Carneiro F: E-cadherin alterations in hereditary disorders with emphasis on hereditary diffuse gastric cancer. Prog Mol Biol Transl Sci 2013, 116: 337–59.CrossRefPubMed
30.
Zurück zum Zitat Fitzgerald RC, Caldas C: E-cadherin mutations and hereditary gastric cancer: prevention by resection? Dig Dis 2002, 20: 23–31.CrossRefPubMed Fitzgerald RC, Caldas C: E-cadherin mutations and hereditary gastric cancer: prevention by resection? Dig Dis 2002, 20: 23–31.CrossRefPubMed
31.
Zurück zum Zitat Corso G, Pedrazzani C, Pinheiro H, Fernandes E, Marelli D, Rinnovati A, Pascale V, Seruca R, Oliveira C, Roviello F: E-cadherin genetic screening and clinico-pathologic characteristics of early onset gastric cancer. Eur J Cancer 2011, 47: 631–9.CrossRefPubMed Corso G, Pedrazzani C, Pinheiro H, Fernandes E, Marelli D, Rinnovati A, Pascale V, Seruca R, Oliveira C, Roviello F: E-cadherin genetic screening and clinico-pathologic characteristics of early onset gastric cancer. Eur J Cancer 2011, 47: 631–9.CrossRefPubMed
32.
Zurück zum Zitat Benusiglio PR, Malka D, Rouleau E, De Paul A, Buecher B, Nogues C, Forume E, Colas C, Coulet F, Warcoin M, Grandjouan S, Sezeur A, Laurent-Puig P, Moliere D, Tlemsani C, Di Maria M, Byrde V, Delaloge S, Blayau M, Caron O: CDH1 germline mutations and the hereditary diffuse gastric and lobular breast cancer syndrome: a multicentre study. J Med Genet 2013, 50: 486–9.CrossRefPubMed Benusiglio PR, Malka D, Rouleau E, De Paul A, Buecher B, Nogues C, Forume E, Colas C, Coulet F, Warcoin M, Grandjouan S, Sezeur A, Laurent-Puig P, Moliere D, Tlemsani C, Di Maria M, Byrde V, Delaloge S, Blayau M, Caron O: CDH1 germline mutations and the hereditary diffuse gastric and lobular breast cancer syndrome: a multicentre study. J Med Genet 2013, 50: 486–9.CrossRefPubMed
33.
Zurück zum Zitat Oliveira C, Senz J, Kaurah P, Pinheiro H, Sanges R, Haegerte A, Corso G, Schouten J, Fistzgerald R, Vogelsang H, Keller G, Dwerryhouse S, Grimmer D, Chin SF, Yang HK, Jackson CE, Seruca R, Roviello F, Stupka E, Caldas C, Huntsman D: Germline CDH1 deletions in hereditary diffuse gastric cancer families. Hum Mol Genet 2009, 18: 1545–55.CrossRefPubMedPubMedCentral Oliveira C, Senz J, Kaurah P, Pinheiro H, Sanges R, Haegerte A, Corso G, Schouten J, Fistzgerald R, Vogelsang H, Keller G, Dwerryhouse S, Grimmer D, Chin SF, Yang HK, Jackson CE, Seruca R, Roviello F, Stupka E, Caldas C, Huntsman D: Germline CDH1 deletions in hereditary diffuse gastric cancer families. Hum Mol Genet 2009, 18: 1545–55.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Shinmura K, Kohno T, Takahashi M, Sasaki A, Ochiai A, Guilford P, Hunter A, Reeve AE, Sugimura H, Yamaguchi N, Yokota J: Familial gastric cancer: clinicopathological characteristics, RER phenotype and germline p53 and Ecadherin mutations. Carcinogenesis 1999, 20: 1127–31.CrossRefPubMed Shinmura K, Kohno T, Takahashi M, Sasaki A, Ochiai A, Guilford P, Hunter A, Reeve AE, Sugimura H, Yamaguchi N, Yokota J: Familial gastric cancer: clinicopathological characteristics, RER phenotype and germline p53 and Ecadherin mutations. Carcinogenesis 1999, 20: 1127–31.CrossRefPubMed
35.
Zurück zum Zitat Milne AN, Sitarz R, Carvalho R, Carneiro F, Offerhaus GJ: Early onset gastric cancer: on the road to unraveling gastric carcinogenesis. Curr Mol Med 2007, 7: 15–28.CrossRefPubMed Milne AN, Sitarz R, Carvalho R, Carneiro F, Offerhaus GJ: Early onset gastric cancer: on the road to unraveling gastric carcinogenesis. Curr Mol Med 2007, 7: 15–28.CrossRefPubMed
36.
Zurück zum Zitat Oliveira C, Bordin MC, Grehan N, Huntsman D, Suriano G, Machado JC, Kiviluoto T, Aaltonen L, Jackson CE, Seruca R, Caldas C: Screening E-cadherin in gastric cancer families reveals germline mutations only in hereditary diffuse gastric cancer kindred. Hum Mutat 2002, 19: 510–7.CrossRefPubMed Oliveira C, Bordin MC, Grehan N, Huntsman D, Suriano G, Machado JC, Kiviluoto T, Aaltonen L, Jackson CE, Seruca R, Caldas C: Screening E-cadherin in gastric cancer families reveals germline mutations only in hereditary diffuse gastric cancer kindred. Hum Mutat 2002, 19: 510–7.CrossRefPubMed
37.
Zurück zum Zitat Zhang Y, Liu X, Fan Y, Ding J, Xu A, Zhou X, Hu X, Zhu M, Zhang X, Li S, Wu J, Cao H, Li J, Wang Y: Germline mutations and polymorphic variants in MMR, E-cadherin and MYH genes associated with familial gastric cancer in Jiangsu of China. Int J Cancer 2006, 119: 2592–6.CrossRefPubMed Zhang Y, Liu X, Fan Y, Ding J, Xu A, Zhou X, Hu X, Zhu M, Zhang X, Li S, Wu J, Cao H, Li J, Wang Y: Germline mutations and polymorphic variants in MMR, E-cadherin and MYH genes associated with familial gastric cancer in Jiangsu of China. Int J Cancer 2006, 119: 2592–6.CrossRefPubMed
38.
Zurück zum Zitat Corso G, Marrelli D, Roviello F: Familial gastric cancer and germline mutations of E-cadherin. Ann Ital Chir 2012, 83: 177–82.PubMed Corso G, Marrelli D, Roviello F: Familial gastric cancer and germline mutations of E-cadherin. Ann Ital Chir 2012, 83: 177–82.PubMed
39.
Zurück zum Zitat Kaurah P, MacMillan A, Boyd N, Senz J, De Luca A, Chun N, Suriano G, Zaor S, Van Manen L, Gilpin C, Nikkel S, Connolly-Wilson M, Weissman S, Rubinstein WS, Sebold C, Greenstein R, Stroop J, Yim D, Panzini B, McKinnon W, Greenblatt M, Wirtzfeld D, Fontaine D, Coit D, Yoon S, Chung D, Lauwers G, Pizzuti A, Vaccaro C, Redal MA, et al.: Founder and recurrent CDH1 mutations in families with hereditary diffuse gastric cancer. JAMA 2007, 297: 2360–72.CrossRefPubMed Kaurah P, MacMillan A, Boyd N, Senz J, De Luca A, Chun N, Suriano G, Zaor S, Van Manen L, Gilpin C, Nikkel S, Connolly-Wilson M, Weissman S, Rubinstein WS, Sebold C, Greenstein R, Stroop J, Yim D, Panzini B, McKinnon W, Greenblatt M, Wirtzfeld D, Fontaine D, Coit D, Yoon S, Chung D, Lauwers G, Pizzuti A, Vaccaro C, Redal MA, et al.: Founder and recurrent CDH1 mutations in families with hereditary diffuse gastric cancer. JAMA 2007, 297: 2360–72.CrossRefPubMed
40.
Zurück zum Zitat Figueiredo C, Machado JC, Pharoah P, Seruca R, Sousa S, Carvalho R, Capelinha AF, Quint W, Caldas C, van Doorn LJ, Carneiro F, Sobrinho-Simões M: Helicobacter pylori and interleukin 1 genotyping: an opportunity to identify high-risk individuals for gastric carcinoma. J Natl Cancer Inst 2002, 94: 1680–7.CrossRefPubMed Figueiredo C, Machado JC, Pharoah P, Seruca R, Sousa S, Carvalho R, Capelinha AF, Quint W, Caldas C, van Doorn LJ, Carneiro F, Sobrinho-Simões M: Helicobacter pylori and interleukin 1 genotyping: an opportunity to identify high-risk individuals for gastric carcinoma. J Natl Cancer Inst 2002, 94: 1680–7.CrossRefPubMed
41.
Zurück zum Zitat Machado JC, Figueiredo C, Canedo P, Pharoah P, Carvalho R, Nabais S, Castro Alves C, Campos ML, Van Doorn LJ, Caldas C, Seruca R, Carneiro F, Sobrinho-Simões M: A proinflammatory genetic profile increases the risk for chronic atrophic gastritis and gastric carcinoma. Gastroenterology 2003, 125: 364–71.CrossRefPubMed Machado JC, Figueiredo C, Canedo P, Pharoah P, Carvalho R, Nabais S, Castro Alves C, Campos ML, Van Doorn LJ, Caldas C, Seruca R, Carneiro F, Sobrinho-Simões M: A proinflammatory genetic profile increases the risk for chronic atrophic gastritis and gastric carcinoma. Gastroenterology 2003, 125: 364–71.CrossRefPubMed
42.
Zurück zum Zitat Silva EM, Fregnani JHTG, Martel G, Costa WL Jr, Coimbra FJF, Achatz MIW, Hainaut P, Soares FA: Molecular analyses of early-onset gastric cancer in Brazilian patients: TP53 mutations, cadherin-catenin and mucins proteins expression. J Cancer Ther 2013, 4: 33–42.CrossRef Silva EM, Fregnani JHTG, Martel G, Costa WL Jr, Coimbra FJF, Achatz MIW, Hainaut P, Soares FA: Molecular analyses of early-onset gastric cancer in Brazilian patients: TP53 mutations, cadherin-catenin and mucins proteins expression. J Cancer Ther 2013, 4: 33–42.CrossRef
44.
Zurück zum Zitat 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: 241–9.CrossRefPubMed 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: 241–9.CrossRefPubMed
45.
Zurück zum Zitat do Nascimento Borges B, Burbano RM, Harada ML: Analysis of the methylation patterns of the p16 INK4A, p15 INK4B, and APC genes in gastric adenocarcinoma patients from a Brazilian population. Tumour Biol 2013, 34: 2127–33.CrossRefPubMed do Nascimento Borges B, Burbano RM, Harada ML: Analysis of the methylation patterns of the p16 INK4A, p15 INK4B, and APC genes in gastric adenocarcinoma patients from a Brazilian population. Tumour Biol 2013, 34: 2127–33.CrossRefPubMed
46.
Zurück zum Zitat Li XJ, Zhao Y, Ren H: E-cadherin expression and CDH1 promoter methylation in sporadic and hereditary gastric cancer. J South Med Univ 2013, 33: 125–7. Li XJ, Zhao Y, Ren H: E-cadherin expression and CDH1 promoter methylation in sporadic and hereditary gastric cancer. J South Med Univ 2013, 33: 125–7.
47.
Zurück zum Zitat Concolino P, Papa V, Mozzetti S, Ferlini C, Pacelli E, Martinelli E, Ricci R, Filippetti F, Scambia G, Doglietto GB: The unsolved enigma of CDH1 down-regulation in hereditary diffuse gastric cancer. J Surg Res 2004, 121: 50–5.CrossRefPubMed Concolino P, Papa V, Mozzetti S, Ferlini C, Pacelli E, Martinelli E, Ricci R, Filippetti F, Scambia G, Doglietto GB: The unsolved enigma of CDH1 down-regulation in hereditary diffuse gastric cancer. J Surg Res 2004, 121: 50–5.CrossRefPubMed
48.
Zurück zum Zitat Wu PY, Zhang Z, Wang JM, Guo WW, Xiao N, He Q, Wang YP, Fan YM: Germline promoter hypermethylation of tumor suppressor genes in gastric cancer. World J Gastroenterol 2012, 18: 70–8.CrossRefPubMedPubMedCentral Wu PY, Zhang Z, Wang JM, Guo WW, Xiao N, He Q, Wang YP, Fan YM: Germline promoter hypermethylation of tumor suppressor genes in gastric cancer. World J Gastroenterol 2012, 18: 70–8.CrossRefPubMedPubMedCentral
49.
Zurück zum Zitat Yamada M, Fukagawa T, Nakajima T, Asada K, Sekine S, Yamashita S, Okochi-Takada E, Taniguchi H, Kushima R, Oda I, Saito Y, Ushijima T, Katai H: Hereditary diffuse gastric cancer in a Japanese family with a large deletion involving CDH1. Gastric Cancer 2013. Sep 15 [Epub ahead of print] Yamada M, Fukagawa T, Nakajima T, Asada K, Sekine S, Yamashita S, Okochi-Takada E, Taniguchi H, Kushima R, Oda I, Saito Y, Ushijima T, Katai H: Hereditary diffuse gastric cancer in a Japanese family with a large deletion involving CDH1. Gastric Cancer 2013. Sep 15 [Epub ahead of print]
50.
Zurück zum Zitat Kim S, Chung JW, Jeong TD, Park YS, Lee JH, Ahn JY, Kim DH, Choi KD, Lee W, Song HJ, Lee GH, Chun S, Jung HY, Min WK, Kin JH: Searching for E-cadherin gene mutations in early onset diffuse gastric cancer and hereditary diffuse gastric cancer in Korean patients. Fam Cancer 2013, 12: 503–7.CrossRefPubMed Kim S, Chung JW, Jeong TD, Park YS, Lee JH, Ahn JY, Kim DH, Choi KD, Lee W, Song HJ, Lee GH, Chun S, Jung HY, Min WK, Kin JH: Searching for E-cadherin gene mutations in early onset diffuse gastric cancer and hereditary diffuse gastric cancer in Korean patients. Fam Cancer 2013, 12: 503–7.CrossRefPubMed
51.
Zurück zum Zitat Haas P, Anton A, De Francisco A: Câncer colo retal no Brasil: consumo de grãos integrais como prevenção [Colorectal cancer in Brazil: whole grain consumption as prevention]. Rev Bras Análises Clínicas 2007, 39: 231–5. Haas P, Anton A, De Francisco A: Câncer colo retal no Brasil: consumo de grãos integrais como prevenção [Colorectal cancer in Brazil: whole grain consumption as prevention]. Rev Bras Análises Clínicas 2007, 39: 231–5.
52.
Zurück zum Zitat Moutinho V, Makino E: Epidemiological features of the gastric cancer in Belém (Brasil). Arq Bras Cir Dig 1988, 3: 69–74. Moutinho V, Makino E: Epidemiological features of the gastric cancer in Belém (Brasil). Arq Bras Cir Dig 1988, 3: 69–74.
53.
Zurück zum Zitat Koifman S, Koifman RJ: Environment and cancer in Brazil: an overview from a public health perspective. Mutat Res 2003, 544: 305–11.CrossRefPubMed Koifman S, Koifman RJ: Environment and cancer in Brazil: an overview from a public health perspective. Mutat Res 2003, 544: 305–11.CrossRefPubMed
54.
Zurück zum Zitat Neves FJ, Koifman RJ, Mattos IE: Mortalidade por câncer de cólon e reto e consumo alimentar em capitais brasileiras selecionada [Colorectal cancer mortality and dietary patterns in selected Brazilian state capitals]. Rev Bras Epidemiol 2006, 9: 112–20.CrossRef Neves FJ, Koifman RJ, Mattos IE: Mortalidade por câncer de cólon e reto e consumo alimentar em capitais brasileiras selecionada [Colorectal cancer mortality and dietary patterns in selected Brazilian state capitals]. Rev Bras Epidemiol 2006, 9: 112–20.CrossRef
55.
Zurück zum Zitat Nakashima JP, Koifman RJ, Koifman S: Cancer incidence in the Western Amazon: population-based estimates in Rio Branco, Acre State, Brazil, 2007–2009. Cad Saude Publica 2012, 28: 2125–32.CrossRefPubMed Nakashima JP, Koifman RJ, Koifman S: Cancer incidence in the Western Amazon: population-based estimates in Rio Branco, Acre State, Brazil, 2007–2009. Cad Saude Publica 2012, 28: 2125–32.CrossRefPubMed
Metadaten
Titel
Genetic screening analysis of patients with hereditary diffuse gastric cancer from northern and northeastern Brazil
verfasst von
Caroline Aquino Moreira-Nunes
Mariceli Baia Leão Barros
Bárbara do Nascimento Borges
Raquel Carvalho Montenegro
Leticia Martins Lamarão
Helem Ferreira Ribeiro
Amanda Braga Bona
Paulo Pimentel Assumpção
Juan Antonio Rey
Giovanny Rebouças Pinto
Rommel Rodriguez Burbano
Publikationsdatum
01.12.2014
Verlag
BioMed Central
Erschienen in
Hereditary Cancer in Clinical Practice / Ausgabe 1/2014
Elektronische ISSN: 1897-4287
DOI
https://doi.org/10.1186/1897-4287-12-18

Weitere Artikel der Ausgabe 1/2014

Hereditary Cancer in Clinical Practice 1/2014 Zur Ausgabe

Nodal-negativ nach neoadjuvanter Chemo: Axilladissektion verzichtbar?

03.05.2024 Mammakarzinom Nachrichten

Wenn bei Mammakarzinomen durch eine neoadjuvante Chemotherapie ein Downstaging von nodal-positiv zu nodal-negativ gelingt, scheint es auch ohne Axilladissektion nur selten zu axillären Rezidiven zu kommen.

Wo hapert es noch bei der Umsetzung der POMGAT-Leitlinie?

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

Bestrahlung nach Prostatektomie: mehr Schaden als Nutzen?

02.05.2024 Prostatakarzinom Nachrichten

Eine adjuvante Radiotherapie nach radikaler Prostata-Op. bringt den Betroffenen wahrscheinlich keinen Vorteil. Im Gegenteil: Durch die Bestrahlung steigt offenbar das Risiko für Harn- und Stuhlinkontinenz.

Endlich: Zi zeigt, mit welchen PVS Praxen zufrieden sind

IT für Ärzte Nachrichten

Darauf haben viele Praxen gewartet: Das Zi hat eine Liste von Praxisverwaltungssystemen veröffentlicht, die von Nutzern positiv bewertet werden. Eine gute Grundlage für wechselwillige Ärzte und Psychotherapeuten.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.