Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Genes Chromosomes Cancer ; 63(1): e23208, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37795928

ABSTRACT

Polyketide synthase (pks) island harboring Escherichia coli are, under the right circumstances, able to produce the genotoxin colibactin. Colibactin is a risk factor for the development of colorectal cancer and associated with mutational signatures SBS88 and ID18. This study explores colibactin-associated mutational signatures in biallelic NTHL1 and MUTYH patients. Targeted Next Generation Sequencing (NGS) was performed on colorectal adenomas and carcinomas of one biallelic NTHL and 12 biallelic MUTYH patients. Additional fecal metagenomics and genome sequencing followed by mutational signature analysis was conducted for the NTHL1 patient. Targeted NGS of the NTHL1 patient showed somatic APC variants fitting SBS88 which was confirmed using WGS. Furthermore, fecal metagenomics revealed pks genes. Also, in 1 out of 11 MUTYH patient a somatic variant was detected fitting SBS88. This report shows that colibactin may influence development of colorectal neoplasms in predisposed patients.


Subject(s)
Adenomatous Polyposis Coli , Colorectal Neoplasms , Humans , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/pathology , Mutation , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Deoxyribonuclease (Pyrimidine Dimer)/genetics
2.
J Pathol ; 262(2): 137-146, 2024 02.
Article in English | MEDLINE | ID: mdl-37850614

ABSTRACT

The identification of causal BRCA1/2 pathogenic variants (PVs) in epithelial ovarian carcinoma (EOC) aids the selection of patients for genetic counselling and treatment decision-making. Current recommendations therefore stress sequencing of all EOCs, regardless of histotype. Although it is recognised that BRCA1/2 PVs cluster in high-grade serous ovarian carcinomas (HGSOC), this view is largely unsubstantiated by detailed analysis. Here, we aimed to analyse the results of BRCA1/2 tumour sequencing in a centrally revised, consecutive, prospective series including all EOC histotypes. Sequencing of n = 946 EOCs revealed BRCA1/2 PVs in 125 samples (13%), only eight of which were found in non-HGSOC histotypes. Specifically, BRCA1/2 PVs were identified in high-grade endometrioid (3/20; 15%), low-grade endometrioid (1/40; 2.5%), low-grade serous (3/67; 4.5%), and clear cell (1/64; 1.6%) EOCs. No PVs were identified in any mucinous ovarian carcinomas tested. By re-evaluation and using loss of heterozygosity and homologous recombination deficiency analyses, we then assessed: (1) whether the eight 'anomalous' cases were potentially histologically misclassified and (2) whether the identified variants were likely causal in carcinogenesis. The first 'anomalous' non-HGSOC with a BRCA1/2 PV proved to be a misdiagnosed HGSOC. Next, germline BRCA2 variants, found in two p53-abnormal high-grade endometrioid tumours, showed substantial evidence supporting causality. One additional, likely causal variant, found in a p53-wildtype low-grade serous ovarian carcinoma, was of somatic origin. The remaining cases showed retention of the BRCA1/2 wildtype allele, suggestive of non-causal secondary passenger variants. We conclude that likely causal BRCA1/2 variants are present in high-grade endometrioid tumours but are absent from the other EOC histotypes tested. Although the findings require validation, these results seem to justify a transition from universal to histotype-directed sequencing. Furthermore, in-depth functional analysis of tumours harbouring BRCA1/2 variants combined with detailed revision of cancer histotypes can serve as a model in other BRCA1/2-related cancers. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
BRCA1 Protein , Ovarian Neoplasms , Female , Humans , BRCA1 Protein/genetics , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Tumor Suppressor Protein p53 , Carcinoma, Ovarian Epithelial/genetics
3.
Eur J Surg Oncol ; 46(10 Pt A): 1925-1931, 2020 10.
Article in English | MEDLINE | ID: mdl-32354538

ABSTRACT

INTRODUCTION: Mitomycin C (MMC) is commonly used in patients with colorectal peritoneal metastases (CPM) treated with cytoreductive surgery plus hyperthermic intraperitoneal chemotherapy (CRS + HIPEC). MMC requires metabolic activation prior to exert its cytotoxic effect of which the main activating enzymes are NQO1 and POR. However, not all patients are able to activate MMC for example due to polymorphisms in the genes encoding these enzymes. The aim of this study was to investigate the association of NQO1∗2, NQO1∗3, and POR∗28 with the efficacy of CRS + HIPEC with MMC in patients with CPM. METHOD: A retrospective follow-up design was used to study genetic association in patients with histologically proven CPM treated with CRS + HIPEC with MMC with respect to peritoneal recurrence rate after 3 months (primary endpoint), after 6 months, disease-free survival and overall survival. Genetic polymorphisms NQO1∗2, NQO1∗3, and POR∗28 were tested for association. RESULTS: A total of 253 patients were included. In NQO1∗3 carriers the peritoneal recurrence rate 3 and 6 months after HIPEC was significantly higher than in wild type patients, respectively 30.0% vs 3.8% (p = 0.009) and 40.0% vs 12.1% (p = 0.031). In line with these results, NQO1∗3 was associated with a shorter disease-free survival (HR 2.04, 95% CI [1.03-4.03]). There was no significant association with overall survival (HR 1.42, 95% CI [0.66-3.07]). CONCLUSION: Carriership of the NQO1∗3 allele is associated with worse peritoneal recurrence rate and disease-free survival. These results suggest that individualization of patients treated with CRS + HIPEC based upon pharmacogenetics may be beneficial.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Carcinoma/therapy , Colorectal Neoplasms/therapy , Cytochrome P-450 Enzyme System/genetics , Mitomycin/therapeutic use , NAD(P)H Dehydrogenase (Quinone)/genetics , Peritoneal Neoplasms/therapy , Pharmacogenomic Variants/genetics , Aged , Alleles , Antibiotics, Antineoplastic/metabolism , Carcinoma/secondary , Colorectal Neoplasms/pathology , Cytochrome P-450 Enzyme System/metabolism , Cytoreduction Surgical Procedures , Disease-Free Survival , Female , Humans , Hyperthermic Intraperitoneal Chemotherapy , Male , Middle Aged , Mitomycin/metabolism , NAD(P)H Dehydrogenase (Quinone)/metabolism , Peritoneal Neoplasms/secondary , Prognosis , Retrospective Studies
4.
Fam Cancer ; 19(1): 15-21, 2020 01.
Article in English | MEDLINE | ID: mdl-31598872

ABSTRACT

We report a case of a 22-year-old female patient who was diagnosed with a cribriform-morular variant of papillary thyroid carcinoma (CMV-PTC). While at early ages this thyroid cancer variant is highly suggestive for familial adenomatous polyposis (FAP), there was no family history of FAP. In the tumor biallelic, inactivating APC variants were identified. The patient tested negative for germline variants based on analysis of genomic DNA from peripheral blood leukocytes. Somatic mosaicism was excluded by subsequent deep sequencing of leukocyte and normal thyroid DNA using next generation sequencing (NGS). This report presents a rare sporadic case of CMV-PTC, and to the best of our knowledge the first featuring two somatic APC mutations underlying the disease, with an overview of CMV-PTC cases with detected APC and CTNNB1 pathogenic variants from the literature.


Subject(s)
Genes, APC , Mutation , Thyroid Cancer, Papillary/genetics , Thyroid Neoplasms/genetics , Adenomatous Polyposis Coli/genetics , Female , Humans , Rare Diseases/genetics , Young Adult , beta Catenin/genetics
5.
Eur J Endocrinol ; 180(4): 235-241, 2019 Apr 01.
Article in English | MEDLINE | ID: mdl-30668525

ABSTRACT

Objective Gene alterations leading to activation of the MAPK pathway are of interest for targeted therapy in patients with advanced radioactive iodine refractory (RAI-R) thyroid carcinoma. Due to technical reasons gene fusion analysis in RNA isolated from formalin-fixed tumor tissues has till now been limited. The objective of the present study was to identify targetable gene rearrangements in RNA isolated from formalin-fixed RAI-R thyroid carcinomas. Design Retrospective study in 132 patients with RAI-R thyroid carcinoma (59 papillary-, 24 follicular-, 35 Hürthle cell- and 14 anaplastic thyroid carcinoma). Methods Total nucleic acid (undivided DNA and RNA) was isolated from formalin-fixed tissue. Extensive gene fusion analysis was performed in all samples that tested negative for pathogenic BRAF, NRAS, HRAS and KRAS variants. Results Seven targetable gene fusions were identified in the remaining 60 samples without known DNA variants. This includes frequently reported gene fusions such as CCDC6/RET (PTC1), PRKAR1A/RET (PTC2) and ETV6/NTRK3 , and gene fusions that are less common in thyroid cancer (TPM3/NTRK1, EML4/ALK and EML4/NTRK3). Of note, most gene fusions were detected in papillary thyroid carcinoma and MAPK-associated alterations in Hürthle cell carcinomas are rare (2/35). Conclusion Targetable gene fusions were found in 12% of RAI-R thyroid carcinoma without DNA variants and can be effectively identified in formalin-fixed tissue. These gene fusions might provide a preclinical rationale to include specific kinase inhibitors in the treatment regimen for these patients. The latter intends to restore iodine transport and/or take advantage of the direct effect on tumor cell vitality once progressive disease is seen.


Subject(s)
Gene Fusion/genetics , Gene Targeting/methods , Iodine , Thyroid Neoplasms/genetics , Adolescent , Aged , Aged, 80 and over , Female , Humans , Iodine/administration & dosage , Male , Middle Aged , Retrospective Studies , Thyroid Neoplasms/diagnosis , Thyroid Neoplasms/drug therapy
6.
Eur J Cancer ; 72: 215-225, 2017 02.
Article in English | MEDLINE | ID: mdl-28049106

ABSTRACT

BACKGROUND: Uterine serous carcinoma (USC) shows greater morphological, clinical and molecular similarities to high-grade ovarian tubal serous carcinoma than to other types of endometrial cancer. As high-grade ovarian tubal serous carcinoma is known to be associated with BRCA1/2 pathogenic germline mutations (PMs), we aimed to explore whether USC is also a constituent of hereditary breast and ovarian cancer syndrome. METHODS: Pubmed, EMBASE and Web of Science were searched in July 2016 for articles assessing the association between USC and germline BRCA1/2-PMs. Pooled analysis and comparisons were performed using a random effects logistic model, stratifying for ethnicity (Ashkenazi versus non-Ashkenazi). In addition, tumour tissue from an USC case with a hereditary BRCA1-PM was analysed for loss of heterozygosity at the BRCA1 locus and was functionally analysed for homologous recombination proficiency. RESULTS: The search yielded 1893 citations, 10 studies were included describing 345 USC patients. For Ashkenazi Jews, the pooled odds ratio of having a germline BRCA1/2-PM was increased in USC patients compared with the general Ashkenazi population: odds ratio 5.4 (95%confidence interval: 2.2-13.1). In the patient with USC, we identified the known germline BRCA1-PM in the tumour DNA. Furthermore, we showed both loss of heterozygosity of the wild-type allele and a deficiency of homologous recombination. CONCLUSION: This study suggests that USC may be an overlooked component of BRCA1/2-associated hereditary breast and ovarian cancer syndrome. Screening for germline BRCA1/2-PMs should be considered in patients diagnosed with USC, especially in cases with a positive first-degree family history for breast and/or ovarian cancer.


Subject(s)
Carcinoma/genetics , Genes, BRCA1 , Genes, BRCA2 , Germ-Line Mutation , Uterine Neoplasms/genetics , Female , Humans , Middle Aged
7.
Ann Oncol ; 28(1): 96-102, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27742654

ABSTRACT

Background: Mismatch repair (MMR)-deficiency analysis is increasingly recommended for all endometrial cancers, as it identifies Lynch syndrome patients, and is emerging as a prognostic classifier to guide adjuvant treatment. The aim of this study was to define the optimal approach for MMR-deficiency testing and to clarify discrepancies between microsatellite instability (MSI) analysis and immunohistochemical (IHC) analysis of MMR protein expression. Patients and methods: Six hundred ninety- six endometrial cancers were analyzed for MSI (pentaplex panel) and MMR protein expression (IHC). Agreement between methodologies was calculated using Cohen's Kappa. MLH1 promoter hypermethylation, dinucleotide microsatellite markers and somatic MMR and POLE exonuclease domain (EDM) gene variants (using next-generation/Sanger sequencing) were analyzed in discordant cases. Results: MSI was found in 180 patients. Complete loss of expression of one or more MMR proteins was observed in 196 cases. A PMS2- and MSH6-antibody panel detected all cases with loss of MMR protein expression. The results of MSI and MMR protein expression were concordant in 655/696 cases (kappa = 0.854, P < 0.001). Ambiguous cases (n = 41, 6%) included: subclonal loss of MMR protein expression (n = 18), microsatellite stable or MSI-low cases with loss of MMR protein expression (n = 20), and MSI-low or MSI-high cases with retained MMR protein expression (n = 3). Most of these cases could be explained by MLH1 promoter hypermethylation. Five of seven cases with solitary loss of PMS2 or MSH6 protein expression carried somatic gene variants. Two MSI-high cases with retained MMR protein expression carried a POLE-EDM variant. Conclusion: MSI and IHC analysis are highly concordant in endometrial cancer. This holds true for cases with subclonal loss of MMR protein expression. Discordant MMR-proficient/MSI-high cases (<1%), may be explained by POLE-EDM variants.


Subject(s)
Brain Neoplasms/diagnosis , Colorectal Neoplasms/diagnosis , DNA Mismatch Repair/genetics , Endometrial Neoplasms/genetics , Microsatellite Instability , Neoplastic Syndromes, Hereditary/diagnosis , Brain Neoplasms/complications , Colorectal Neoplasms/complications , Female , Humans , Immunohistochemistry , Neoplastic Syndromes, Hereditary/complications , Polymerase Chain Reaction
8.
Fam Cancer ; 15(4): 563-70, 2016 10.
Article in English | MEDLINE | ID: mdl-26880076

ABSTRACT

Familial adenomatous polyposis (FAP) is a dominantly inherited syndrome caused by germline mutations in the APC gene and characterized by the development of multiple colorectal adenomas and a high risk of developing colorectal cancer (CRC). The severity of polyposis is correlated with the site of the APC mutation. However, there is also phenotypic variability within families with the same underlying APC mutation, suggesting that additional factors influence the severity of polyposis. Genome-wide association studies identified several single nucleotide polymorphisms (SNPs) that are associated with CRC. We assessed whether these SNPs are associated with polyp multiplicity in proven APC mutation carriers. Sixteen CRC-associated SNPs were analysed in a cohort of 419 APC germline mutation carriers from 182 families. Clinical data were retrieved from the Dutch Polyposis Registry. Allele frequencies of the SNPs were compared for patients with <100 colorectal adenomas versus patients with ≥100 adenomas, using generalized estimating equations with the APC genotype as a covariate. We found a trend of association of two of the tested SNPs with the ≥100 adenoma phenotype: the C alleles of rs16892766 at 8q23.3 (OR 1.71, 95 % CI 1.05-2.76, p = 0.03, dominant model) and rs3802842 at 11q23.1 (OR 1.51, 95 % CI 1.03-2.22, p = 0.04, dominant model). We identified two risk variants that are associated with a more severe phenotype in APC mutation carriers. These risk variants may partly explain the phenotypic variability in families with the same APC gene defect. Further studies with a larger sample size are recommended to evaluate and confirm the phenotypic effect of these SNPs in FAP.


Subject(s)
Adenomatous Polyposis Coli Protein/genetics , Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 8 , Colorectal Neoplasms/genetics , Adenoma/genetics , Adenomatous Polyposis Coli/genetics , Adult , Gene Frequency , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Male , Mutation , Polymorphism, Single Nucleotide
9.
Invest New Drugs ; 33(6): 1242-7, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26386973

ABSTRACT

INTRODUCTION: Cetuximab is registered for use in colorectal cancer (CRC) patients with RAS wild-type tumours only. Simvastatin blocks the mevalonate pathway and thereby interferes with the post-translational modification (prenylation) of KRAS. We hypothesize that the activated KRAS pathway in KRAS mutant tumors can be inhibited by simvastatin rendering these tumors sensitive to the EGFR inhibitor cetuximab. METHODS: A Simon two-stage, single-arm, phase II study was performed to test the efficacy and safety of the addition of simvastatin to cetuximab in patients with a KRAS mutation in their CRC tumour who were previously treated with fluoropyrimidine, oxaliplatin and irinotecan based regimens. The primary endpoint was to test the percentage of patients alive and free from progression 12.5 weeks after the first administration of cetuximab. Our hypothesis was that at least 40% was free from progression, comparable to, though slightly lower than in KRAS wild-type patients. RESULTS: Four of 18 included patients (22.2%) were free from progression at the primary endpoint time. The time to progression in these 4 patients ranged from 20.3 to 47 weeks. CONCLUSION: Based on the current study we conclude that the theoretical concept of KRAS modulation with simvastatin was not applicable in the clinic, as we were not able to restore sensitivity to cetuximab in CRC patients harbouring a somatic KRAS mutation.


Subject(s)
Cetuximab/administration & dosage , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Mutation/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Simvastatin/administration & dosage , Aged , Antineoplastic Combined Chemotherapy Protocols , Cetuximab/adverse effects , Colorectal Neoplasms/diagnosis , Exanthema/chemically induced , Fatigue/chemically induced , Female , Humans , Male , Middle Aged , Simvastatin/adverse effects , Treatment Outcome
10.
Int J Endocrinol ; 2015: 348124, 2015.
Article in English | MEDLINE | ID: mdl-26294908

ABSTRACT

Objective. Until recently, advanced medullary thyroid cancer (MTC) had few treatment options except surgery. The mTOR inhibitor everolimus has shown encouraging results in neuroendocrine tumors. As part of a prospective phase II study, we analyzed the safety and efficacy of everolimus in advanced MTC. Methods. Seven patients with per RECIST 1.1 documented advanced MTC were included and received everolimus 10 mg daily. The primary objective was determining treatment efficacy. Secondary endpoints included progression-free survival (PFS), overall survival (OS), toxicity, and pharmacokinetics (PK). Results. Median follow-up duration was 28 weeks (17-147). Five patients (71%) showed SD, of which 4 (57%) showed SD >24 weeks. Median PFS and OS were 33 (95%CI: 8-56) and 30 (95%CI: 15-45) weeks, respectively. Toxicity was predominantly grade 1/2 and included mucositis (43%), fatigue (43%), and hypertriglyceridemia (43%). Four MTCs harbored the somatic RET mutation c.2753T>C, p.Met918Thr. The best clinical response was seen in a MEN2A patient. PK characteristics were consistent with phase I data. One patient exhibited extensive toxicity accompanying elevated everolimus plasma concentrations. Conclusions. This study suggests that everolimus exerts clinically relevant antitumor activity in patients with advanced MTC. Given the high level of clinical benefit and the relatively low toxicity profile, further investigation of everolimus in these patients is warranted.

11.
Int J Cancer ; 130(4): 837-46, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-21445971

ABSTRACT

Many hereditary nonpolyposis colorectal cancers (CRCs) cannot be explained by Lynch syndrome. Other high penetrance genetic risk factors are likely to play a role in these mismatch repair (MMR)-proficient CRC families. Because genomic profiles of CRC tend to vary with CRC susceptibility syndromes, our aim is to analyze the genomic profile of MMR-proficient familial CRC to obtain insight into the biological basis of MMR-proficient familial CRC. We studied 30 MMR-proficient familial colorectal carcinomas, from 15 families, for genomic aberrations, including gains, physical losses, and copy-neutral loss of heterozygosity LOH (cnLOH) using SNP array comparative genomic hybridization. In addition, we performed somatic mutation analysis for KRAS, BRAF, PIK3CA and GNAS. The frequency of 20q gain (77%) is remarkably increased when compared with sporadic CRC, suggesting that 20q gain is involved in tumor progression of familial CRC. There is also a significant increase in the frequency of cnLOH and, as a consequence, a reduced frequency of physical loss compared with sporadic CRC. The most frequent aberrations observed included gains of 7p, 7q, 8q, 13q, 20p and 20q as well as physical losses of 17p, 18p and 18q. Most of these changes are also observed in sporadic CRC. Mutations in KRAS were identified in 37% of the MMR-proficient CRCs, and mutations in BRAF were identified in 16%. No mutations were identified in PIK3CA or chromosome 20 candidate gene GNAS. We show that the patterns of chromosomal instability of MMR-proficient familial CRC are clearly distinct from those from sporadic CRC. Both the increased gain on chromosome 20 and the increased levels of cnLOH suggest the presence of yet undiscovered germline defects that can, in part, underlie the cancer risk in these families.


Subject(s)
Chromosome Aberrations , Colorectal Neoplasms/genetics , DNA Mismatch Repair , Loss of Heterozygosity , Adult , Aged , Chromosomes, Human, Pair 20 , Heterozygote , Humans , Middle Aged , Mutation , Polymorphism, Single Nucleotide
12.
Br J Cancer ; 102(2): 447-54, 2010 Jan 19.
Article in English | MEDLINE | ID: mdl-19920828

ABSTRACT

It is now recognised that a part of the inherited risk of colorectal cancer (CRC) can be explained by the co-inheritance of low-penetrance genetic variants. The accumulated experience to date in identifying these variants has served to highlight difficulties in conducting statistically and methodologically rigorous studies and follow-up analyses. The COGENT (COlorectal cancer GENeTics) consortium includes 20 research groups in Europe, Australia, the Americas, China and Japan. The overarching goal of COGENT is to identify and characterise low-penetrance susceptibility variants for CRC through association-based analyses. In this study, we review the rationale for identifying low-penetrance variants for CRC and our proposed strategy for establishing COGENT.


Subject(s)
Colorectal Neoplasms/genetics , Polymorphism, Genetic , Genetic Predisposition to Disease , Humans , Penetrance , Prognosis , Risk , Risk Factors
13.
J Pathol ; 216(1): 25-31, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18506705

ABSTRACT

Genetic instability is known to drive colorectal carcinogenesis. Generally, a distinction is made between two types of genetic instability: chromosomal instability (CIN) and microsatellite instability (MIN or MSI). Most CIN tumours are aneuploid, whereas MSI tumours are considered near-diploid. However, for MUTYH-associated polyposis (MAP) the genetic instability involved in the carcinogenesis remains unclear, as near-diploid adenomas, aneuploid adenomas and near-diploid carcinomas have been reported. Remarkably, our analysis of 26 MAP carcinomas, using SNP arrays and flow sorting, showed that these tumours are often near-diploid (52%) and mainly contain chromosomal regions of copy-neutral loss of heterozygosity (LOH) (71%). This is in contrast to sporadic colon cancer, where physical loss is the main characteristic. The percentage of chromosomal gains (24%) is comparable to sporadic colorectal cancers with CIN. Furthermore, we verified our scoring of copy-neutral LOH versus physical loss in MAP carcinomas by two methods: fluorescence in situ hybridization, and LOH analysis using polymorphic markers on carcinoma fractions purified by flow sorting. The results presented in this study suggest that copy-neutral LOH is an important mechanism in the tumorigenesis of MAP.


Subject(s)
Adenomatous Polyposis Coli/genetics , Chromosomal Instability/genetics , DNA Glycosylases/genetics , Loss of Heterozygosity/genetics , Adenomatous Polyposis Coli/pathology , Adult , Aged , Biomarkers, Tumor/genetics , DNA Mutational Analysis/methods , Humans , Microsatellite Instability , Middle Aged
14.
Clin Endocrinol (Oxf) ; 67(3): 370-6, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17555500

ABSTRACT

OBJECTIVE: Parathyroid carcinoma remains difficult to diagnose. Recently, it has been shown that mutations in the HRPT2 gene (encoding parafibromin) are associated with the development of parathyroid carcinoma. Although MEN1 is not typically thought to be involved in carcinoma formation, parathyroid carcinoma may be an extremely rare feature of the multiple endocrine neoplasia type 1 (MEN1) syndrome. We recently concluded that loss of heterozygosity (LOH) of the MEN1 gene is present in a relatively large number of parathyroid carcinomas, often in combination with LOH at the HRPT2 locus. The aim of this study was to evaluate the role of MEN1 and HRPT2 mutations in sporadic parathyroid tumours fulfilling histological criteria for malignancy. PATIENTS AND DESIGN: Formalin-fixed, paraffin-embedded (FFPE) parathyroid carcinoma tissue from 28 cases identified in the period 1985-2000 in the Netherlands was studied. HRPT2 (27/28 cases) and MEN1 (23/28 cases) were analysed by direct sequencing. RESULTS: Somatic MEN1 mutations were found in three of 23 (13%) sporadic parathyroid carcinoma cases; these consisted of one missense and two frameshift mutations. One of the latter two cases displayed lymph-node and lung metastases during follow-up. Six HRPT2 mutations were found in 4/27 cases (15%): five were truncating mutations and one was a missense mutation. Consistent with previously published reports, we found double mutations (2x) and germline mutations (2x) in apparently sporadic parathyroid carcinomas. CONCLUSIONS: These results suggest that not only HRPT2 but also MEN1 mutations may play a role in sporadic parathyroid cancer formation.


Subject(s)
Multiple Endocrine Neoplasia Type 1/genetics , Parathyroid Neoplasms/genetics , Proto-Oncogene Proteins/genetics , Tumor Suppressor Proteins/genetics , Adult , Aged , Aged, 80 and over , Biological Specimen Banks , Cohort Studies , DNA Mutational Analysis , Female , Frameshift Mutation , Humans , Loss of Heterozygosity/genetics , Male , Middle Aged , Multiple Endocrine Neoplasia Type 1/pathology , Mutation, Missense , Netherlands , Paraffin Embedding , Parathyroid Neoplasms/pathology
15.
Br J Cancer ; 94(5): 661-71, 2006 Mar 13.
Article in English | MEDLINE | ID: mdl-16495925

ABSTRACT

Using genome-wide expression profiling of a panel of 27 human mammary cell lines with different mechanisms of E-cadherin inactivation, we evaluated the relationship between E-cadherin status and gene expression levels. Expression profiles of cell lines with E-cadherin (CDH1) promoter methylation were significantly different from those with CDH1 expression or, surprisingly, those with CDH1 truncating mutations. Furthermore, we found no significant differentially expressed genes between cell lines with wild-type and mutated CDH1. The expression profile complied with the fibroblastic morphology of the cell lines with promoter methylation, suggestive of epithelial-mesenchymal transition (EMT). All other lines, also the cases with CDH1 mutations, had epithelial features. Three non-tumorigenic mammary cell lines derived from normal breast epithelium also showed CDH1 promoter methylation, a fibroblastic phenotype and expression profile. We suggest that CDH1 promoter methylation, but not mutational inactivation, is part of an entire programme, resulting in EMT and increased invasiveness in breast cancer. The molecular events that are part of this programme can be inferred from the differentially expressed genes and include genes from the TGFbeta pathway, transcription factors involved in CDH1 regulation (i.e. ZFHX1B, SNAI2, but not SNAI1, TWIST), annexins, AP1/2 transcription factors and members of the actin and intermediate filament cytoskeleton organisation.


Subject(s)
Breast Neoplasms/pathology , Cadherins/biosynthesis , DNA Methylation , Gene Expression Profiling , Cell Line, Tumor , Cell Transformation, Neoplastic , DNA Mutational Analysis , Down-Regulation , Epithelial Cells , Female , Gene Expression Regulation, Neoplastic , Humans , Mesoderm/cytology , Neoplasm Invasiveness , Polymerase Chain Reaction , Promoter Regions, Genetic , Transcription, Genetic , Transforming Growth Factor beta/physiology
16.
Cancer Res ; 59(17): 4216-8, 1999 Sep 01.
Article in English | MEDLINE | ID: mdl-10485458

ABSTRACT

Germ-line mutations in APC and mismatch repair genes explain only a small percentage of all colorectal cancer cases. We have used the recombinant congenic strain mouse model to find new loci that are involved in the control of susceptibility to colon cancer. Five different colon cancer susceptibility genes, Scc1-Scc5, have been described previously using the recombinant congenic strains. Two of these loci, Scc4 and Scc5, show a reciprocal, genetic interaction. Here we report the mapping of four new colon tumor susceptibility genes: (a) Scc6 on chromosome 11; (b) Scc7 on chromosome 3; (c) Scc8 on chromosome 8; and (d) Scc9 on chromosome 10. Scc7 and Scc8 show a genetic interaction; Scc7 is only detected by virtue of its interaction with Scc8.


Subject(s)
Chromosome Mapping , Colonic Neoplasms/genetics , Genetic Predisposition to Disease , Alleles , Animals , Genetic Linkage , Mice , Mice, Inbred BALB C
17.
Mamm Genome ; 9(5): 377-80, 1998 May.
Article in English | MEDLINE | ID: mdl-9545495

ABSTRACT

Apoptosis, a mechanism for removal of genetically damaged cells and for maintenance of desired size of cell populations, has been implicated in tumor development. Previously, we defined polymorphic loci for susceptibility to apoptosis of thymocytes Rapop1, Rapop2, and Rapop3 on mouse Chromosomes 16, 9, and 3, respectively, using recombinant congenic CcS/Dem strains, each of which contains a random set of 12.5% STS/A genome in the genetic background of BALB/cHeA. The STS/A alleles at these loci confer lower susceptibility to radiation-induced apoptosis of thymocytes than the BALB/cHeA. In the present study, we tested susceptibility of colon crypt cells to radiation-induced apoptosis. In contrast to apoptosis in thymus, the STS/A mice were more susceptible to apoptosis in colon than the BALB/cHeA. Among the CcS/Dem strains, CcS-4, CcS-7, and CcS-16 were more susceptible to apoptosis in colon than the BALB/cHeA; in thymus, the CcS-7 mice are less susceptible, and the CcS-4 and CcS-16 are not different from the BALB/cHeA. Thus, individual CcS/Dem strains showed different apoptosis susceptibility in the two organs. Analysis of (CcS-7 x BALB/cHeA)F2 hybrids revealed linkage of susceptibility to radiation-induced apoptosis of colon crypt cells to two loci on Chrs 9 and 16, to which Rapop2 and Rapop1 are mapped. The STS/A allele at the locus on chromosome 9 results in high susceptibility to apoptosis of colon crypt cells in mice homozygous for the BALB/cHeA allele at the locus on Chr 16. Although these two loci may be identical to Rapop1 and Rapop2, they affect apoptosis in colon in a way different from that in thymus.


Subject(s)
Apoptosis/genetics , Apoptosis/radiation effects , Chromosome Mapping , Colon/radiation effects , Radiation Tolerance/genetics , Animals , Colon/cytology , Crosses, Genetic , Female , Gamma Rays , Genetic Linkage , Humans , Infant, Newborn , Intestinal Mucosa/cytology , Intestinal Mucosa/radiation effects , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Species Specificity , Thymus Gland/cytology , Thymus Gland/radiation effects
18.
Nat Genet ; 14(4): 468-70, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8944029

ABSTRACT

To dissect the multigenic control of colon tumour susceptibility in the mouse we used the set of 20 CcS/Dem (CcS) recombinant congenic (RC) strains. Each CcS strain carries a unique, random subset of approximately 12.5% of the genome of strain STS/A (STS) on the genetic background of BALB/cHeA (BALB/c). Previously, applying a protocol of 26 injections of 1,2-dimethylhydrazine (DMH), we detected two susceptibility loci, Scc1 and Scc2, on chromosome 2 (refs 4, 5). Using a shorter tumour-induction procedure, combining DMH and N-ethyl-N-nitrosourea (ENU) treatment, we demonstrate that BALB/c, STS and most CcS strains are relatively resistant. The strain CcS-19, however, is susceptible, probably due to a combination of BALB/c and STS alleles at several loci. Analysis of 192 (BALB/c x CcS-19) F2 mice revealed, in addition to the Scc1/Scc2 region, three new susceptibility loci: Scc3 on chromosome 1, Scc4 on chromosome 17 and Scc5 on chromosome 18. Scc4 and Scc5 have no apparent individual effect, but show a strong reciprocal interaction. Their BALB/c and STS alleles are not a priori susceptible or resistant but the genotype at one locus determines the effect of the allele at the second locus and vice versa. These findings and the accompanying paper on lung tumour susceptibility show that interlocus interactions are likely to be an important component of tumour susceptibility.


Subject(s)
Colonic Neoplasms/genetics , Animals , Genetic Predisposition to Disease , Mice , Mice, Inbred BALB C , Mice, Inbred Strains
19.
J Bacteriol ; 178(7): 1955-61, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8606170

ABSTRACT

Previously we have shown that flagella and the O-specific polysaccharide of lipopolysaccharide play a role in colonization of the potato root by plant growth-promoting Pseudomonas strains WCS374 and WCS358. In this paper, we describe a novel cell surface-exposed structure in Pseudomonas putida WCS358 examined with a specific monoclonal antibody. This cell surface structure appeared to be a polysaccharide, which was accessible to the monoclonal antibody at the outer cell surface. Further study revealed that it does not contain 2-keto-3-deoxyoctonate, heptose, or lipid A, indicating that it is not a second type of lipopolysaccharide. Instead, the polysaccharide shared some characteristics with K antigen described for Escherichia coli. From a series of 49 different soil bacteria tested, only one other potato plant growth-promoting Pseudomonas strain reacted positively with the monoclonal antibody. Mutant cells lacking the novel antigen were efficiently isolated by an enrichment method involving magnetic antibodies. Mutant strains defective in the novel antigen contained normal lipopolysaccharide. One of these mutants was affected in neither its ability to adhere to sterile potato root pieces nor its ability to colonize potato roots. We conclude that the bacterial cell surface of P. putida WCS358 contains at least two different polysaccharide structures. These are the O-specific polysaccharide of lipopolysaccharide, which is relevant for potato root colonization, and the novel polysaccharide, which is not involved in adhesion to or colonization of the potato root.


Subject(s)
Antigens, Bacterial , Antigens, Surface/chemistry , Polysaccharides, Bacterial/isolation & purification , Pseudomonas putida/chemistry , Animals , Antibodies, Monoclonal/immunology , Antibody Specificity/immunology , Antigens, Surface/immunology , Bacterial Adhesion/physiology , Cell Membrane/chemistry , Escherichia coli/immunology , Mice , Mice, Inbred BALB C , Mutation , Plant Roots/microbiology , Polysaccharides, Bacterial/chemistry , Polysaccharides, Bacterial/immunology , Pseudomonas putida/genetics , Pseudomonas putida/immunology , Pseudomonas putida/isolation & purification , Rabbits , Solanum tuberosum/microbiology , Tumor Cells, Cultured
20.
Infect Immun ; 56(2): 432-8, 1988 Feb.
Article in English | MEDLINE | ID: mdl-3123391

ABSTRACT

We incorporated the major outer membrane protein (PI) of Neisseria gonorrhoeae into immunostimulating complexes (iscoms) and examined some analytical, physicochemical, and immunological properties of these structures. The immunogenicity was compared with that of three other PI-containing structures, i.e., liposomes, outer membrane complexes produced by the bacterium, and protein-detergent-adjuvant complexes. AIPO4 and dioctadecyldimethylammonium bromide were used as adjuvants. Our results show that iscoms are much more immunogenic than liposomes and protein-detergent complexes but are also much more toxic. The localization of PI in iscoms was investigated. Therefore, the chymotrypsin susceptibility of PI in iscoms was tested, and the incorporation of fragments of PI was determined. Amphiphilic fragments of PI were incorporated in iscoms, but hydrophilic and hydrophobic fragments were not. Chymotrypsin degradation of PI in iscoms indicated that the protein is exposed to the environment in a similar manner as PI in outer membrane complexes, i.e., with both termini anchored in the iscom.


Subject(s)
Aluminum Compounds , Antigens, Bacterial/administration & dosage , Bacterial Outer Membrane Proteins/immunology , Neisseria gonorrhoeae/immunology , Adjuvants, Immunologic , Aluminum , Animals , Bacterial Outer Membrane Proteins/administration & dosage , Chemical Phenomena , Chemistry, Physical , Freeze Fracturing , Lipids , Liposomes , Mice , Peptide Fragments/analysis , Phosphates , Saponins
SELECTION OF CITATIONS
SEARCH DETAIL
...