Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters











Publication year range
1.
Eur J Endocrinol ; 179(3): 153-160, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29903750

ABSTRACT

OBJECTIVE: Epigenetic changes contribute to pancreatic neuroendocrine tumor (PanNET) development. Hypermethylation of promoter DNA as a cause of tumor suppressor gene silencing is a well-established oncogenic mechanism that is potentially reversible and therefore an interesting therapeutic target. Multiple endocrine neoplasia type 1 (MEN1) is the most frequent cause of inherited PanNETs. The aim of this study was to determine promoter methylation profiles in MEN1-related PanNETs. DESIGN AND METHODS: Methylation-specific multiplex ligation-dependent probe amplification was used to assess promoter methylation of 56 tumor suppressor genes in MEN1-related (n = 61) and sporadic (n = 34) PanNETs. Differences in cumulative methylation index (CMI), individual methylation percentages and frequency of promoter hypermethylation between subgroups were analyzed. RESULTS: We found promoter methylation of a large number of potential tumor suppressor genes. CMI (median CMI: 912 vs 876, P = 0.207) was the same in MEN1-related and sporadic PanNETs. We found higher methylation percentages of CASP8 in MEN1-related PanNETs (median: 59% vs 16.5%, P = 0.002). In MEN1-related non-functioning PanNETs, the CMI was higher in larger PanNETs (>2 cm) (median: 969.5 vs 838.5; P = 0.021) and in PanNETs with liver metastases (median: 1036 vs 869; P = 0.013). Hypermethylation of MGMT2 was more frequent in non-functioning PanNETs compared to insulinomas (median: 44.7% vs 8.3%; P = 0.022). Hypermethylation of the Von Hippel-Lindau gene promoter was observed in one MEN1-related PanNET and was associated with loss of protein expression. CONCLUSION: Promoter hypermethylation is a frequent event in MEN1-related and sporadic PanNETs. Targeting DNA methylation could be of therapeutic value in MEN1 patients with advanced PanNETs.


Subject(s)
DNA Methylation/genetics , Epigenesis, Genetic/genetics , Multiple Endocrine Neoplasia Type 1/genetics , Neuroendocrine Tumors/genetics , Pancreatic Neoplasms/genetics , Promoter Regions, Genetic/genetics , Adult , Aged , Aged, 80 and over , Female , Genes, Tumor Suppressor , Humans , Male , Middle Aged , Von Hippel-Lindau Tumor Suppressor Protein/genetics
2.
J Pediatr Surg ; 50(8): 1304-9, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25783404

ABSTRACT

BACKGROUND: The murine model of biliary atresia (BA) is used for examining the pathogenesis of BA. The aim of the study was description of the morphological features and illustrating the detailed development of fibrosis using the Biliary Atresia Research Consortium (BARC) system. METHODS: Neonatal mice were injected intraperitoneally with rhesus rotavirus (RRV) strain (N=17). Healthy mice were the control group (N=29). All mice were sacrificed at 7 or 14days after birth. Two pathologists examined the morphological features using the BARC system; CK19, αSMA and collagen type I were assessed by immunohistochemistry. RESULTS: In RRV mice, portal fibrous expansion with focal bile duct proliferation and strong portal cellular infiltrate was found in contrast to healthy mice. In RRV mice, CK19 bile duct staining was significantly less or absent (p<0.01), with stronger portal staining of collagen type I (p=0.02). Expansion of staining for αSMA was more in RRV mice (p<0.01), but αSMA portal staining was stronger in healthy mice (p=0.02). CONCLUSIONS: The morphological features observed in the murine model of BA correspond with the BA characteristics according to the BARC criteria. Fibrosis is an important feature of the model. Therefore, this murine model is useful for investigating the pathogenesis of BA.


Subject(s)
Biliary Atresia/pathology , Liver Cirrhosis/etiology , Liver/pathology , Animals , Biliary Atresia/virology , Disease Models, Animal , Disease Progression , Liver Cirrhosis/pathology , Liver Cirrhosis/virology , Mice , Mice, Inbred BALB C , Rotavirus Infections/complications , Severity of Illness Index
3.
Oncogene ; 33(3): 347-57, 2014 Jan 16.
Article in English | MEDLINE | ID: mdl-23318448

ABSTRACT

The incidence of Barrett's esophagus (BE)-associated esophageal adenocarcinoma (EAC) is increasing. Next-generation sequencing (NGS) provides an unprecedented opportunity to uncover genomic alterations during BE pathogenesis and progression to EAC, but treatment-naive surgical specimens are scarce. The objective of this study was to establish the feasibility of using widely available endoscopic mucosal biopsies for successful NGS, using samples obtained from a BE 'progressor'. Paired-end whole-genome NGS was performed on the Illumina platform using libraries generated from mucosal biopsies of normal squamous epithelium (NSE), BE and EAC obtained from a patient who progressed to adenocarcinoma during endoscopic surveillance. Selective validation studies, including Sanger sequencing, immunohistochemistry and functional assays, were performed to confirm the NGS findings. NGS identified somatic nonsense mutations of AT-rich interactive domain 1A (SWI like) (ARID1A) and PPIE and an additional 37 missense mutations in BE and/or EAC, which were confirmed by Sanger sequencing. ARID1A mutations were detected in 15% (3/20) high-grade dysplasia (HGD)/EAC patients. Immunohistochemistry performed on an independent archival cohort demonstrated ARID1A protein loss in 0% (0/76), 4.9% (2/40), 14.3% (4/28), 16.0% (8/50) and 12.2% (12/98) of NSE, BE, low-grade dysplasia, HGD and EAC tissues, respectively, and was inversely associated with nuclear p53 accumulation (P=0.028). Enhanced cell growth, proliferation and invasion were observed on ARID1A knockdown in EAC cells. In addition, genes downstream of ARID1A that potentially contribute to the ARID1A knockdown phenotype were identified. Our studies establish the feasibility of using mucosal biopsies for NGS, which should enable the comparative analysis of larger 'progressor' versus 'non-progressor' cohorts. Further, we identify ARID1A as a novel tumor-suppressor gene in BE pathogenesis, reiterating the importance of aberrant chromatin in the metaplasia-dysplasia sequence.


Subject(s)
Barrett Esophagus/genetics , High-Throughput Nucleotide Sequencing/methods , Nuclear Proteins/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Aged , Barrett Esophagus/metabolism , Barrett Esophagus/pathology , Biopsy , Blotting, Western , Cell Line, Tumor , Cell Proliferation , DNA-Binding Proteins , Endoscopes , Epithelium/metabolism , Epithelium/pathology , Esophageal Neoplasms/genetics , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Female , Gene Regulatory Networks , Humans , Immunohistochemistry , Male , Middle Aged , Mutation, Missense , Nuclear Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Polymorphism, Single Nucleotide , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/metabolism , Transcriptome , Tumor Suppressor Proteins/metabolism
4.
Dis Esophagus ; 25(7): 630-7, 2012.
Article in English | MEDLINE | ID: mdl-22221671

ABSTRACT

Barrett's esophagus (BE) is a premalignant condition with an increased risk of developing esophageal adenocarcinoma (EAC). Risk factors for EAC overlap with those for esophageal squamous cell carcinoma (ESCC), but ESCC is surprisingly rare in BE. We report two cases of ESCC directly surrounded by BE. Both patients had a previous medical history of cancers, i.e., head and neck squamous cell carcinomas, and were using alcohol and smoking tobacco. Using immunohistochemistry for p63, CK5, CK7, and CDX2, it was confirmed that these carcinomas were pure squamous cell carcinomas, and not EACs or esophageal adenosquamous carcinomas arising from BE. Using TP53 mutation and loss of heterozygosity analysis, we established that the ESCCs in BE were not metastases of the previously diagnosed head and neck squamous cell carcinomas but de novo primary ESCCs. This study shows the strength of molecular analysis as an adjunct to the histopathologic diagnosis for distinguishing between metastases of prior cancers and primary cancers. Furthermore, these cases imply that presence of BE is not protective with regards to developing ESCC in the lower one third of the esophagus. We suggest that their ESCCs arose from islets of squamous epithelium in BE.


Subject(s)
Barrett Esophagus/complications , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/complications , Esophageal Neoplasms/complications , Aged , Barrett Esophagus/pathology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma , Humans , Immunohistochemistry , Male , Neoplasm Metastasis
5.
Cell Oncol ; 32(5-6): 347-59, 2010.
Article in English | MEDLINE | ID: mdl-20448329

ABSTRACT

BACKGROUND: CDC4/FBXW7, encoding a ubiquitin ligase, maps to 4q32 and has been implicated as a tumor suppressor gene and therapeutic target in many tumor types. Mutations in colonic adenomas, and the frequent losses on 4q described in gastric cancer prompt speculation about the role of CDC4/FBXW7 in gastric carcinogenesis. METHODS: We assessed the role of CDC4/FBXW7 in gastric cancer, through loss of heterozygosity (LOH) and multiplex ligation-dependent probe amplification (MLPA) on 47 flow-sorted gastric carcinomas including early-onset gastric cancers (EOGC) and xenografted conventional gastric carcinomas. Ploidy analysis was carried out on 39 EOGCs and immunohistochemistry of CDC4/FBXW7 and its substrates c-myc, c-jun, Notch and cyclin E was performed on 204 gastric carcinomas using tissue microarrays (TMAs). Sequence analysis of CDC4/FBXW7 was carried out on gastric carcinoma cell lines and xenografts. RESULTS: Loss of heterozygosity of CDC4/FBXW7 occurred in 32% of EOGCs, and correlated with loss of expression in 26%. Loss of expression was frequent in both EOGC and conventional gastric cancers. No CDC4/FBXW7 mutations were found and loss of CDC4/FBXW7 did not correlate with ploidy status. There was a significant correlation between loss of CDC4/FBXW7 expression and upregulation of c-myc. CONCLUSION: Loss of CDC4/FBXW7 appears to play a role in both EOGC and conventional gastric carcinogenesis, and c-myc overexpression is likely to be an important oncogenic consequence of CDC4/FBXW7 loss.


Subject(s)
Carcinoma/genetics , Cell Cycle Proteins/metabolism , F-Box Proteins/metabolism , Loss of Heterozygosity , Proto-Oncogene Proteins c-myc/metabolism , Stomach Neoplasms/genetics , Ubiquitin-Protein Ligases/metabolism , Carcinoma/metabolism , Carcinoma/pathology , Cell Cycle Proteins/genetics , Cell Separation , Cell Transformation, Neoplastic , Chromosomes, Human, Pair 4 , DNA Mutational Analysis , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Flow Cytometry , Gene Expression Regulation, Neoplastic , Genetic Association Studies , Humans , Immunohistochemistry , Middle Aged , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins c-myc/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Tumor Cells, Cultured , Ubiquitin-Protein Ligases/genetics
6.
Cell Oncol ; 31(6): 475-85, 2009.
Article in English | MEDLINE | ID: mdl-19940363

ABSTRACT

BACKGROUND: COX-2 and E-cadherin, involved in invasion and metastasis, are molecules critical for gastric carcinogenesis. A relationship between them is documented in non-small cell lung and prostate cancer. We present novel evidence of a relationship between COX-2 and E-cadherin expression in gastric cancer. METHODS: Using qPCR and Western blots analysis on celecoxib and PGE2 treated and untreated gastric cancer cell lines derived from tumours of the intestinal type (MKN45, MKN28, AGS3, MKN7) and immunohistochemistry of 178 gastric cancers on tissue microarrays (TMA), we examined the COX-2/E-cadherin relationship. RESULTS: Down-regulation of COX-2 by celecoxib led to up-regulation of E-cadherin mRNA and protein levels in conventional gastric cancer cell lines, whereas expression was down regulated in the early-onset gastric cancer (EOGC) cell line. Immunohistochemistry on TMAs of 178 gastric cancers showed no correlation between COX-2 and E-cadherin expression in the conventional or early gastric cancer groups. CONCLUSION: The results suggest that COX-2 has an impact on transcriptional regulation of E-cadherin in gastric cancer and our findings further highlight the intriguing nature of EOGCs which appear to have a molecular phenotype distinct from conventional gastric cancer. In addition, our findings also suggest that reduction of COX-2 using nonsteroidal anti-inflammatory drugs in gastric cancer chemoprevention may only be relevant for older patients.


Subject(s)
Cadherins/metabolism , Cyclooxygenase 2/metabolism , Stomach Neoplasms/metabolism , Adult , Age of Onset , Blotting, Western , Cadherins/genetics , Celecoxib , Cell Line, Tumor , Cyclooxygenase 2/genetics , Cyclooxygenase 2 Inhibitors/pharmacology , DNA Mutational Analysis , Dinoprostone/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunohistochemistry , Middle Aged , Mutation , Netherlands/epidemiology , Pyrazoles/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Stomach Neoplasms/epidemiology , Stomach Neoplasms/pathology , Sulfonamides/pharmacology , Tissue Array Analysis
8.
Virchows Arch ; 453(3): 249-55, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18688641

ABSTRACT

It has been reported that interleukin-1beta (IL-1B) genes play a crucial role in the genetic predisposition to gastric cancer although there is no information about their role in different subtypes of gastric cancer. We performed single nucleotide polymorphism analysis of IL-1B in 241 gastric cancers including early onset gastric cancers (EOGC), conventional gastric cancers, and gastric stump cancers (GSCs) as well as 100 control patients, using real-time polymerase chain reaction and sequence analysis. The C allele was present in 60% of EOGCs, 59% of conventional gastric cancers, and 90% of GSCs, compared to 62% in the control group. Interestingly, there was no difference between early onset and conventional gastric cancer with respect to the IL-1B -31T>C polymorphism distribution. A statistically significant difference in the presence of the C allele compared to the control group was found in patients with gastric stump cancer (p = 0.008) with the T allele conferring protection against gastric stump cancer. In summary, we have shown that the IL-1B -31C allele promoter polymorphism is significantly associated with gastric stump cancer compared to the control group. Although several molecular differences have been identified between conventional gastric cancer and early onset gastric cancer, the IL-1B -31 allele distribution is similar between these two groups.


Subject(s)
Gastric Stump , Interleukin-1beta/genetics , Promoter Regions, Genetic , Stomach Neoplasms/genetics , Adult , Alleles , Humans , Middle Aged , Polymorphism, Single Nucleotide , Stomach Neoplasms/complications
9.
Gut ; 57(5): 623-7, 2008 May.
Article in English | MEDLINE | ID: mdl-18178612

ABSTRACT

BACKGROUND/AIMS: Juvenile polyposis syndrome (JPS) is a rare autosomal dominant disorder characterised by multiple gastrointestinal juvenile polyps and an increased risk of colorectal cancer. This syndrome is caused by germline mutation of either SMAD4 or BMPR1A, and possibly ENG. PTEN, originally linked to Cowden syndrome and Bannayan-Riley-Ruvalcaba syndrome, has also been associated with JPS. By direct sequencing, germline mutations are found in only 30-40% of patients with a JPS phenotype. Therefore, alternative ways of inactivation of the known JPS genes, or additional genes predisposing to JPS may be involved. In this study, a comprehensive genetic analysis of SMAD4, BMPR1A, PTEN and ENG is performed through direct sequencing and multiplex ligation-dependent probe amplification (MLPA) in JPS patients. METHODS: Archival material of 29 patients with JPS from 27 families was collected. Direct sequencing and MLPA analysis were performed to search for germline defects in SMAD4, BMPR1A, PTEN and ENG. RESULTS: A germline defect in SMAD4, BMPR1A or PTEN was found in 13 of 27 (48.1%) unrelated JPS patients. Nine mutations (33.3%) were detected by direct sequencing, including six (22.2%) SMAD4 mutations and three (11.1%) BMPR1A mutations. MLPA identified four additional patients (14.8%) with germline hemizygous large genomic deletions, including one deletion of SMAD4, one deletion of exons 10 and 11 of BMPR1A, and two unrelated patients with deletion of both BMPR1A and PTEN. No ENG gene mutations were found. CONCLUSION: Large genomic deletions of SMAD4, BMPR1A and PTEN are a common cause of JPS. Using direct sequencing and MLPA, a germline defect was detected in 48.1% of JPS patients. MLPA identified 14.8% (4/27) of these mutations. Since a substantial percentage of JPS patients carry a germline deletion and MLPA is a reliable and user-friendly technique, it is concluded that MLPA is a valuable adjunct in JPS diagnosis.


Subject(s)
Adenomatous Polyposis Coli/genetics , Base Sequence/genetics , Bone Morphogenetic Protein Receptors, Type I/genetics , PTEN Phosphohydrolase/genetics , Sequence Deletion/genetics , Smad4 Protein/genetics , Base Sequence/physiology , DNA Mutational Analysis/methods , Female , Genome , Genotype , Germ-Line Mutation/genetics , Humans , Male , Pedigree , Phenotype , Sequence Deletion/physiology
11.
Lab Anim ; 18(3): 252-7, 1984 Jul.
Article in English | MEDLINE | ID: mdl-6235399

ABSTRACT

The numbers of IgA-producing cells in intestinal mucosa, mesenteric lymph nodes, lungs and bronchial lymph nodes were scored in rats and mice. A statistically significant difference in the scores existed between germ-free and SPF mice and between gnotobiotic and SPF rats. In a group of SPF rats a statistically significant difference in the scores was demonstrated in relation to several bacterial and viral agents.


Subject(s)
Antibody-Producing Cells/cytology , Immunoglobulin A/biosynthesis , Mice/microbiology , Rats/microbiology , Animals , Animals, Laboratory , Antibody-Producing Cells/immunology , Germ-Free Life , Intestinal Mucosa/immunology , Lung/immunology , Lymph Nodes/immunology , Male , Mice/immunology , Mice, Inbred Strains , Rats/immunology , Rats, Inbred Strains , Specific Pathogen-Free Organisms
SELECTION OF CITATIONS
SEARCH DETAIL