Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Fam Cancer ; 2024 May 19.
Article in English | MEDLINE | ID: mdl-38763984

ABSTRACT

De novo germline pathogenic variants (gPV) of the BReast CAncer 1 (BRCA1) gene are very rare. Only a few have been described up to date, usually in patients with a history of ovarian or breast cancer. Here, we report the first case of an incidental de novo BRCA1 germline pathogenic variant which was identified within the framework of the Plan France Médecine Génomique (PFMG) 2025 French national tumor sequencing program. The proband was a 29-year-old man diagnosed with metastatic osteosarcoma. Tumor whole exome sequencing identified a BRCA1 c.3756_3759del p.(Ser1253Argfs*10) pathogenic variant without loss-of-heterozygosity. A low genomic instability score and the absence of single base substitution signatures of homologous recombination deficiency suggested that the BRCA1 variant was not driver in the osteosarcoma tumorigenesis. Germline whole genome sequencing asserted the germline nature of this variant, with a 36% allele frequency, suggesting a mosaicism caused by a post-zygotic mutational event. The proband's family (parents and siblings) were not carriers of this variant confirming the de novo occurrence. Tumor sequencing programs like the French PFMG 2025 have been implemented worldwide and may help identify new gPV, including de novo variants.

2.
J Med Genet ; 60(12): 1198-1205, 2023 Nov 27.
Article in English | MEDLINE | ID: mdl-37402566

ABSTRACT

BACKGROUND: The MSH3 gene is part of the DNA mismatch repair system, but has never been shown to be involved in Lynch syndrome. A first report of four patients from two families, bearing biallelic MSH3 germline variants, with a phenotype of attenuated colorectal adenomatous polyposis raised the question of its involvement in hereditary cancer predisposition. The patients' tumours exhibited elevated microsatellite alterations at selected tetranucleotide repeats (EMAST), a hallmark of MSH3 deficiency. METHODS: We report five new unrelated patients with MSH3-associated polyposis. We describe their personal and familial history and study the EMAST phenotype in various normal and tumour samples, which are relevant findings based on the rarity of this polyposis subtype so far. RESULTS: All patients had attenuated colorectal adenomatous polyposis, with duodenal polyposis in two cases. Both women had breast carcinomas. EMAST phenotype was present at various levels in different samples of the five patients, confirming the MSH3 deficiency, with a gradient of instability in polyps depending on their degree of dysplasia. The negative EMAST phenotype ruled out the diagnosis of germline MSH3 deficiency for two patients: one homozygous for a benign variant and one with a monoallelic large deletion. CONCLUSION: This report lends further credence to biallelic MSH3 germline pathogenic variants being involved in colorectal and duodenal adenomatous polyposis. Large-scale studies may help clarify the tumour spectrum and associated risks. Ascertainment of EMAST may help with the interpretation of variants of unknown significance. We recommend adding MSH3 to dedicated diagnostic gene panels.


Subject(s)
Adenomatous Polyposis Coli , Colorectal Neoplasms, Hereditary Nonpolyposis , Colorectal Neoplasms , Humans , Female , Adenomatous Polyposis Coli/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Microsatellite Repeats/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Genetic Predisposition to Disease , MutS Homolog 3 Protein/genetics , MutS Homolog 3 Protein/metabolism
3.
Eur J Hum Genet ; 31(9): 1078-1082, 2023 09.
Article in English | MEDLINE | ID: mdl-37088804

ABSTRACT

Some patients with Lynch syndrome (LS) have extreme phenotypes, i.e. cancer before the recommended screening age, or cancer for which there are no screening guidelines. We made the hypothesis that additional germline variants in cancer susceptibility genes (CSG) could explain some of these phenotypes. We compared the prevalence of additional CSG variants in LS patients with a cancer diagnosis before age 30 (early-onset, EO group) and after 40 (usual-onset, UO group). While there was no overall difference, we did find an excess of pathogenic variants and variants of unknown significance in EO cases when only gastrointestinal CSG were considered (OR 2.25; 95% CI: 1.01-5.06, p value = 0.04). Four EO cases stood out: two with POLE/POLD1 variants in the key exonuclease domain, one with a BMPR1A duplication and one with an EPCAM deletion. Additional germline variants should be considered in future screening recommendations, as they might influence cancer risk.


Subject(s)
Colorectal Neoplasms, Hereditary Nonpolyposis , Humans , Colorectal Neoplasms, Hereditary Nonpolyposis/diagnosis , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/pathology , Germ-Line Mutation , Risk , Phenotype
5.
Genes Chromosomes Cancer ; 62(4): 210-222, 2023 04.
Article in English | MEDLINE | ID: mdl-36502525

ABSTRACT

Only a few patients with germline AXIN2 variants and colorectal adenomatous polyposis or cancer have been described, raising questions about the actual contribution of this gene to colorectal cancer (CRC) susceptibility. To assess the clinical relevance for AXIN2 testing in patients suspected of genetic predisposition to CRC, we collected clinical and molecular data from the French Oncogenetics laboratories analyzing AXIN2 in this context. Between 2004 and June 2020, 10 different pathogenic/likely pathogenic AXIN2 variants were identified in 11 unrelated individuals. Eight variants were from a consecutive series of 3322 patients, which represents a frequency of 0.24%. However, loss-of-function AXIN2 variants were strongly associated with genetic predisposition to CRC as compared with controls (odds ratio: 11.89, 95% confidence interval: 5.103-28.93). Most of the variants were predicted to produce an AXIN2 protein devoid of the SMAD3-binding and DIX domains, but preserving the ß-catenin-binding domain. Ninety-one percent of the AXIN2 variant carriers who underwent colonoscopy had adenomatous polyposis. Forty percent of the variant carriers developed colorectal or/and other digestive cancer. Multiple tooth agenesis was present in at least 60% of them. Our report provides further evidence for a role of AXIN2 in CRC susceptibility, arguing for AXIN2 testing in patients with colorectal adenomatous polyposis or cancer.


Subject(s)
Adenomatous Polyposis Coli , Colorectal Neoplasms , Humans , Genetic Predisposition to Disease , Adenomatous Polyposis Coli/genetics , Adenomatous Polyposis Coli/pathology , Germ-Line Mutation , beta Catenin/metabolism , Germ Cells/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Axin Protein/genetics
6.
Hum Genet ; 142(1): 1-9, 2023 Jan.
Article in English | MEDLINE | ID: mdl-35941319

ABSTRACT

Neurofibromatosis type 1 (NF1) is an autosomal dominant disease with complete penetrance but highly variable expressivity. In most patients, Next Generation Sequencing (NGS) technologies allow the identification of a loss-of-function pathogenic variant in the NF1 gene, a negative regulator of the RAS-MAPK pathway. We describe the 5-year diagnosis wandering of a patient with a clear NF1 clinical diagnosis, but no molecular diagnosis using standard molecular technologies. The patient presented with a typical NF1 phenotype but NF1 targeted NGS, NF1 transcript analysis, MLPA, and array comparative genomic hybridization failed to reveal a genetic aberration. After 5 years of unsuccessful investigations, trio WGS finally identified a de novo mosaic (VAF ~ 14%) 24.6 kb germline deletion encompassing the promoter and first exon of NF1. This case report illustrates the relevance of WGS to detect structural variants including copy number variants that would be missed by alternative approaches. The identification of the causal pathogenic variant allowed a tailored genetic counseling with a targeted non-invasive prenatal diagnosis by detecting the deletion in plasmatic cell-free DNA from the proband's pregnant partner. This report clearly highlights the need to make WGS a clinically accessible test, offering a tremendous opportunity to identify a molecular diagnosis for otherwise unsolved cases.


Subject(s)
Neurofibromatosis 1 , Humans , Neurofibromatosis 1/diagnosis , Neurofibromatosis 1/genetics , Genes, Neurofibromatosis 1 , Comparative Genomic Hybridization , Exons , Whole Genome Sequencing
8.
Cancer Discov ; 12(6): 1435-1448, 2022 06 02.
Article in English | MEDLINE | ID: mdl-35398880

ABSTRACT

Missense mutations in the polymerase epsilon (POLE) gene have been reported to generate proofreading defects resulting in an ultramutated genome and to sensitize tumors to checkpoint blockade immunotherapy. However, many POLE-mutated tumors do not respond to such treatment. To better understand the link between POLE mutation variants and response to immunotherapy, we prospectively assessed the efficacy of nivolumab in a multicenter clinical trial in patients bearing advanced mismatch repair-proficient POLE-mutated solid tumors. We found that only tumors harboring selective POLE pathogenic mutations in the DNA binding or catalytic site of the exonuclease domain presented high mutational burden with a specific single-base substitution signature, high T-cell infiltrates, and a high response rate to anti-PD-1 monotherapy. This study illustrates how specific DNA repair defects sensitize to immunotherapy. POLE proofreading deficiency represents a novel agnostic biomarker for response to PD-1 checkpoint blockade therapy. SIGNIFICANCE: POLE proofreading deficiency leads to high tumor mutational burden with high tumor-infiltrating lymphocytes and predicts anti-PD-1 efficacy in mismatch repair-proficient tumors. Conversely, tumors harboring POLE mutations not affecting proofreading derived no benefit from PD-1 blockade. POLE proofreading deficiency is a new tissue-agnostic biomarker for cancer immunotherapy. This article is highlighted in the In This Issue feature, p. 1397.


Subject(s)
DNA Polymerase II , Neoplasms , DNA Polymerase II/genetics , Humans , Immunotherapy , Mutation, Missense , Neoplasms/drug therapy , Neoplasms/enzymology , Neoplasms/genetics , Poly-ADP-Ribose Binding Proteins/genetics , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/genetics
9.
Eur J Med Genet ; 65(2): 104409, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34954152

ABSTRACT

The POLD1 gene is involved in DNA proofreading to ensure accurate DNA replication. Some germline alterations in its exonuclease domain are associated with predisposition to cancers and colonic polyps. Only a few pathogenic variants have been clearly identified so far. Here we report a novel variant: c.1458G>T p.(Lys486Asn) that we classified as pathogenic, detected in two putatively unrelated families. The cancer spectrum was very similar to Lynch syndrome, implying an overlapped tissue susceptibility. The common presence of colonic polyps in carriers and the MMR proficient phenotype in tumors were distinctive features suggesting POLD1 implication. Some clinical characteristics observed in the carriers of this variant differed from those reported previously, suggesting a potential genotype/phenotype correlation, and very likely in relation to the functional importance of affected residues. Our findings provide further insight into understanding the role of POLD1 in cancer-related risk.


Subject(s)
Colonic Polyps/genetics , Colorectal Neoplasms, Hereditary Nonpolyposis/genetics , DNA Polymerase III/genetics , Adult , Aged , Aged, 80 and over , Colonic Polyps/diagnosis , Colorectal Neoplasms, Hereditary Nonpolyposis/diagnosis , Diagnosis, Differential , Female , Heterozygote , Humans , Male , Middle Aged , Mutation , Pedigree , Phenotype
10.
Clin Genet ; 99(5): 662-672, 2021 05.
Article in English | MEDLINE | ID: mdl-33454955

ABSTRACT

Biallelic pathogenic variants in the NTHL1 (Nth like DNA glycosylase 1) gene cause a recently identified autosomal recessive hereditary cancer syndrome predisposing to adenomatous polyposis and colorectal cancer. Half of biallelic carriers also display multiple colonic or extra-colonic primary tumors, mainly breast, endometrium, urothelium, and brain tumors. Published data designate NTHL1 as an important contributor to hereditary cancers but also underline the scarcity of available informations. Thanks to the French oncogenetic consortium (Groupe Génétique et Cancer), we collected NTHL1 variants from 7765 patients attending for hereditary colorectal cancer or polyposis (n = 3936) or other hereditary cancers (n = 3829). Here, we describe 10 patients with pathogenic biallelic NTHL1 germline variants, that is, the second largest NTHL1 series. All carriers were from the "colorectal cancer or polyposis" series. All nine biallelic carriers who underwent colonoscopy presented adenomatous polyps. For digestive cancers, average age at diagnosis was 56.2 and we reported colorectal, duodenal, caecal, and pancreatic cancers. Extra-digestive malignancies included sarcoma, basal cell carcinoma, breast cancer, urothelial carcinoma, and melanoma. Although tumor risks remain to be precisely defined, these novel data support NTHL1 inclusion in diagnostic panel testing. Colonic surveillance should be conducted based on MUTYH recommendations while extra-colonic surveillance has to be defined.


Subject(s)
Deoxyribonuclease (Pyrimidine Dimer)/genetics , Neoplastic Syndromes, Hereditary/genetics , Ovarian Neoplasms/genetics , Adenomatous Polyposis Coli/genetics , Adult , Aged , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Colorectal Neoplasms/genetics , Female , Germ-Line Mutation , Heterozygote , Humans , Male , Middle Aged , Pedigree , Reverse Transcriptase Polymerase Chain Reaction
11.
Genet Med ; 22(9): 1533-1541, 2020 09.
Article in English | MEDLINE | ID: mdl-32424176

ABSTRACT

PURPOSE: Polymerase proofreading-associated polyposis is a dominantly inherited colorectal cancer syndrome caused by exonuclease domain missense variants in the DNA polymerases POLE and POLD1. Manifestations may also include malignancies at extracolonic sites. Cancer risks in this syndrome are not yet accurately quantified. METHODS: We sequenced POLE and POLD1 exonuclease domains in 354 individuals with early/familial colorectal cancer (CRC) or adenomatous polyposis. We assessed the pathogenicity of POLE variants with yeast fluctuation assays and structural modeling. We estimated the penetrance function for each cancer site in variant carriers with a previously published nonparametric method based on survival analysis approach, able to manage unknown genotypes. RESULTS: Pathogenic POLE exonuclease domain variants P286L, M294R, P324L, N363K, D368N, L424V, K425R, and P436S were found in ten families. The estimated cumulative risk of CRC at 30, 50, and 70 years was 11.1% (95% confidence interval [CI]: 4.2-17.5), 48.5% (33.2-60.3), and 74% (51.6-86.1). Cumulative risk of glioblastoma was 18.7% (3.2-25.8) at 70 years. Variants interfering with DNA binding (P286L and N363K) had a significantly higher mutagenic effect than variants disrupting ion metal coordination at the exonuclease site. CONCLUSION: The risk estimates derived from this study provide a rational basis on which to provide genetic counseling to POLE variant carriers.


Subject(s)
Adenomatous Polyposis Coli , Colorectal Neoplasms , Adult , Aged , Colorectal Neoplasms/epidemiology , Colorectal Neoplasms/genetics , DNA Polymerase II/genetics , Genetic Predisposition to Disease , Germ-Line Mutation , Humans , Middle Aged , Poly-ADP-Ribose Binding Proteins/genetics
12.
JAMA Oncol ; 5(10): 1440-1447, 2019 Oct 01.
Article in English | MEDLINE | ID: mdl-31294750

ABSTRACT

IMPORTANCE: The risk stratification of adrenocortical carcinoma (ACC) based on tumor proliferation index and stage is limited. Adjuvant therapy after surgery is recommended for most patients. Pan-genomic studies have identified distinct molecular groups closely associated with outcome. OBJECTIVE: To compare the molecular classification for prognostic assessment of ACC with other known prognostic factors. DESIGN, SETTING, AND PARTICIPANTS: In this retrospective biomarker analysis, ACC tumor samples from 368 patients who had undergone surgical tumor removal were collected from March 1, 2005, to September 30, 2015 (144 in the training cohort and 224 in the validation cohort) at 21 referral centers with a median follow-up of 35 months (interquartile range, 18-74 months). Data were analyzed from March 2016 to March 2018. EXPOSURES: Meta-analysis of pan-genomic studies (transcriptome, methylome, chromosome alteration, and mutational profiles) was performed on the training cohort. Targeted biomarker analysis, including targeted gene expression (BUB1B and PINK1), targeted methylation (PAX5, GSTP1, PYCARD, and PAX6), and targeted next-generation sequencing, was performed on the training and validation cohorts. MAIN OUTCOMES AND MEASURES: Disease-free survival. Cox proportional hazards regression and C indexes were used to assess the prognostic value of each model. RESULTS: Of the 368 patients (mean [SD] age, 49 [16] years), 144 were in the training cohort (100 [69.4%] female) and 224 were in the validation cohort (142 [63.4%] female). In the training cohort, pan-genomic measures classified ACC into 3 molecular groups (A1, A2, and A3-B), with 5-year survival of 9% for group A1, 45% for group A2, and 82% for group A3-B (log-rank P < .001). Molecular class was an independent prognostic factor of recurrence in stage I to III ACC after complete surgery (hazard ratio, 55.91; 95% CI, 8.55-365.40; P < .001). The combination of European Network for the Study of Adrenal Tumors (ENSAT) stage, tumor proliferation index, and molecular class provided the most discriminant prognostic model (C index, 0.88). In the validation cohort, the molecular classification, determined by targeted biomarker measures, was confirmed as an independent prognostic factor of recurrence (hazard ratio, 5.96 [95% CI, 1.81-19.58], P = .003 for the targeted classifier combining expression, methylation, and chromosome alterations; and 2.61 [95% CI, 1.31-5.19], P = .006 for the targeted classifier combining methylation, chromosome alterations, and mutational profile). The prognostic value of the molecular markers was limited for patients with stage IV ACC. CONCLUSIONS AND RELEVANCE: The findings suggest that in localized ACC, targeted classifiers may be used as independent markers of recurrence. The determination of molecular class may improve individual prognostic assessment and thus may spare unnecessary adjuvant treatment.

13.
J Clin Endocrinol Metab ; 102(3): 923-932, 2017 Mar 01.
Article in English | MEDLINE | ID: mdl-27967600

ABSTRACT

CONTEXT: Adrenocortical cancer (ACC) is an aggressive tumor with a heterogeneous outcome. Prognostic stratification is difficult even based on tumor stage and Ki67. Recently integrated genomics studies have demonstrated that CpG islands hypermethylation is correlated with poor survival. OBJECTIVE: The goal of this study was to confirm the prognostic value of CpG islands methylation on an independent cohort. DESIGN: Methylation was measured by methylation-specific multiplex ligation-dependent probe amplification (MS-MLPA). SETTING: MS-MLPA was performed in a training cohort of 50 patients with ACC to identify the best set of probes correlating with disease-free survival (DFS) and overall survival (OS). These outcomes were validated in an independent cohort from 21 ENSAT centers. PATIENTS: The validation cohort included 203 patients (64% women, median age 50 years, 80% localized tumors). MAIN OUTCOME MEASURES: DFS and OS. RESULTS: In the training cohort, mean methylation of 4 genes (PAX5, GSTP1, PYCARD, PAX6) was the strongest methylation marker. In the validation cohort, methylation was a significant prognostic factor of DFS (P < 0.0001) and OS (P < 0.0001). Methylation, Ki67, and ENSAT stage were combined in multivariate models. For DFS, methylation (P = 0.0005) and stage (P < 0.0001) but not Ki67 (P = 0.19) remained highly significant. For OS, methylation (P = 0.0006), stage (P < 0.0001), and Ki67 (P = 0.024) were independent prognostic factors. CONCLUSIONS: Tumor DNA methylation emerges as an independent prognostic factor in ACC. MS-MLPA is readily compatible with clinical routine and should enhance our ability for prognostication and precision medicine.


Subject(s)
Adrenal Cortex Neoplasms/genetics , Adrenocortical Carcinoma/genetics , CpG Islands , DNA Methylation , DNA, Neoplasm/genetics , Adrenal Cortex Neoplasms/mortality , Adrenal Cortex Neoplasms/pathology , Adrenocortical Carcinoma/mortality , Adrenocortical Carcinoma/pathology , Adult , Aged , Disease-Free Survival , Female , Humans , Ki-67 Antigen/metabolism , Male , Middle Aged , Multiplex Polymerase Chain Reaction , Multivariate Analysis , Neoplasm Staging , Prognosis , Proportional Hazards Models , Retrospective Studies , Survival Rate , Tumor Burden
14.
J Clin Endocrinol Metab ; 98(1): E174-84, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23093492

ABSTRACT

PURPOSE: DNA methylation is a mechanism for gene expression silencing in cancer. Limited information is available for adrenocortical tumors. Abnormal methylation at the IGF2/H19 locus is common in adrenocortical carcinomas. Our aim was to characterize the methylation in adrenocortical carcinomas at a whole-genome scale and to assess its clinical significance and its impact on gene expression. EXPERIMENTAL DESIGN: Methylation patterns of CpG islands in promoter regions of 51 adrenocortical carcinomas and 84 adenomas were studied by the Infinium HumanMethylation27 Beadchip (Illumina, San Diego, CA). Methylation of 33 genes was studied by methylation-specific multiplex ligation-dependent probe amplification (MRC-Holland, Amsterdam, The Netherlands) in 15 carcinomas. Gene expression data were available for 87 tumors from a previous study (HG-U133Plus2.0 AffymetrixGeneChip; Affymetrix, Santa Clara, CA). Clinical information, including patient features and survival, were available for all tumors. RESULTS: Methylation was higher in carcinomas than in adenomas (t test P = 3.1 × 10(-9)). Unsupervised clustering of DNA methylation profiles identified two groups of carcinomas, one with an elevated methylation level, evoking a CpG island methylator phenotype (CIMP). The subgroup of hypermethylated carcinomas was further divided in two subgroups, with different levels of methylation (CIMP-high and CIMP-low). This classification could be confirmed by methylation-specific multiplex ligation-dependent probe amplification. Hypermethylation was associated with a poor survival (Cox model P = 0.02). The transcriptome/methylation correlation showed 1741 genes (of 12,250) negatively correlated; among the top genes were H19 and other tumor suppressors (PLAGL-1, G0S2, and NDRG2). CONCLUSIONS: This genome-wide methylation analysis reveals the existence of hypermethylated adrenocortical carcinomas, with a poorer prognosis. Hypermethylation in these tumors is important for silencing specific tumor suppressor genes.


Subject(s)
Adrenal Gland Neoplasms/genetics , Adrenocortical Carcinoma/genetics , CpG Islands/genetics , DNA Methylation/genetics , Adolescent , Adrenal Gland Neoplasms/diagnosis , Adrenal Gland Neoplasms/mortality , Adrenocortical Adenoma/diagnosis , Adrenocortical Adenoma/genetics , Adrenocortical Adenoma/mortality , Adrenocortical Carcinoma/diagnosis , Adrenocortical Carcinoma/mortality , Adult , Aged , Aged, 80 and over , Cohort Studies , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Middle Aged , Phenotype , Prognosis , Young Adult
15.
Gastroenterology ; 137(4): 1358-66.e1-3, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19549529

ABSTRACT

BACKGROUND & AIMS: Cyclooxygenase-2 inhibitors reduce colon cancer risk by mechanisms that are not fully understood. We performed microarray analysis of adenomas from Apc(Delta14/+) mice to identify genes that respond to these drugs. METHODS: Apc(Delta14/+) mice were given a single daily injection of parecoxib for up to 9 weeks; intestinal tracts of these and control mice were analyzed by microarray analysis, immunohistochemistry, in situ hybridization, and quantitative real-time polymerase chain reaction. Findings were further assessed using Apc(lox/lox)vil-CreER(T2) mice, the CT26 cancer cell line, and human colon tumor samples. RESULTS: Microarray analysis revealed that osteopontin, a marker of colon cancer progression, was down-regulated in polyps from Apc(Delta14/+) mice given parecoxib compared with controls. Apc(Delta14/+) mice given parecoxib had longer survival times and reduced polyp burdens. Osteopontin was quickly down-regulated by parecoxib in intestinal polyps from Apc(Delta14/+) mice, and 2 components of the osteopontin regulatory network-the orphan nuclear receptor NR4A2 and Wnt/beta-catenin signaling-were sequentially repressed. NR4A2 activated the osteopontin promoter in CT26 cells; this effect was blocked by mutation of the NR4A2 binding response element, cotransfection of a dominant-negative form of NR4A2, and small inhibitory RNA against NR4A2. NR4A2 levels were increased throughout tumor progression in Apc(Delta14/+) mice but, unlike osteopontin, did not correlate with tumor stage. NR4A2 levels were reduced in adenomas from patients treated with rofecoxib. CONCLUSIONS: Down-regulation of osteopontin, probably through blockade of NR4A2 and Wnt signaling, is an important component of the antitumor activity of cyclooxygenase-2 inhibitors. These factors might be developed as therapeutic targets for intestinal cancers.


Subject(s)
Adenomatous Polyposis Coli/drug therapy , Antineoplastic Agents/pharmacology , Colonic Polyps/drug therapy , Colorectal Neoplasms/drug therapy , Cyclooxygenase 2/metabolism , Cyclooxygenase Inhibitors/therapeutic use , DNA-Binding Proteins/metabolism , Osteopontin/metabolism , Transcription Factors/metabolism , Adenomatous Polyposis Coli/enzymology , Adenomatous Polyposis Coli/genetics , Animals , Cell Line, Tumor , Cell Survival , Colonic Polyps/enzymology , Colonic Polyps/genetics , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/genetics , DNA-Binding Proteins/genetics , Down-Regulation , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Genes, APC , Humans , Isoxazoles/therapeutic use , Lactones/therapeutic use , Mice , Mice, Mutant Strains , Nuclear Receptor Subfamily 4, Group A, Member 2 , Oligonucleotide Array Sequence Analysis , Osteopontin/genetics , Promoter Regions, Genetic , RNA Interference , Signal Transduction , Sulfones/therapeutic use , Time Factors , Transcription Factors/genetics , Transfection , Wnt Proteins/metabolism , beta Catenin/metabolism
16.
Dig Dis Sci ; 53(1): 242-7, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17549633

ABSTRACT

The relationship between plasma interleukin-6 (IL-6) concentration and its soluble receptor in Crohn's disease (CD) is not well elucidated. Twenty healthy volunteers and 94 consecutive patients with CD (44 in relapse and 50 in remission) were studied. Plasma IL-6 concentrations in patients with active disease [80 +/- 9 pg/ml; mean +/- standard error of the mean (SEM)] were significantly higher than in patients with inactive disease (50 +/- 4 pg/ml; P < 0.001) or controls (3 +/- 1 pg/ml; P < 0.001). However, concentrations did not vary with the severity of CD attacks. Plasma concentrations of soluble interleukin-6 receptor (sIL-6R) in active-CD patients (77 +/- 5 ng/ml) did not differ significantly from those with inactive disease (82 +/- 5 ng/ml), while both groups had significantly raised concentrations compared with those of controls (58 +/- 6 ng/ml; P < 0.03 and P < 0.01, respectively). Plasma IL-6 concentrations correlated significantly with serum C-reactive protein (CRP) (r = 0.34; P < 0.001), whereas plasma sIL-6R concentrations did not. Taken together, these data suggest that, although IL6 and sIL6-R are both involved in the inflammatory process of CD, they are poor markers of disease activity.


Subject(s)
Crohn Disease/blood , Interleukin-6/blood , Receptors, Interleukin-6/blood , Adolescent , Adult , Aged , Biomarkers/blood , C-Reactive Protein/metabolism , Disease Progression , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Middle Aged
17.
Cell Microbiol ; 6(9): 817-28, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15272863

ABSTRACT

Beta1 integrins are anchored on the basal membrane of enterocytes, but little is known about their localization in M cells, which are the main entry route into the intestinal mucosa for many bacterial pathogens. In particular, it has been suggested that adhesion of enteropathogenic Yersinia to M cells is mediated by interaction of the bacterial protein invasin and apical beta1 integrins. Using a novel in vitro model of M cells, we demonstrate an augmented apical and basolateral targeting of beta1 integrins in M cells associated with increased total alpha chain synthesis. The alpha3 and alpha6 subunits were targeted to the basal pole, but alpha2 subunit was targeted at both poles. No other alpha subunit was found associated with apical beta1 integrins on M cells. Interestingly, Y. enterocolitica still adhered to the apical surface of M cells, despite the fact that alpha2beta1 is not a receptor for invasin. We therefore studied the adhesive properties of invasin-mutant Y. enterocolitica and invasin-expressing Escherichia coli on the apical surface of M cells. We show that it is not invasin, but the product of an as yet unidentified bacterial chromosomal gene, that is involved in the adhesion of Y. enterocolitica to the apical membrane of M cells.


Subject(s)
Bacterial Adhesion , Epithelial Cells/immunology , Integrin beta1/metabolism , Intestinal Mucosa/immunology , Peyer's Patches/microbiology , Yersinia/pathogenicity , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Bacterial Proteins/metabolism , Epithelial Cells/microbiology , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli/pathogenicity , Gene Expression Regulation , Genes, Bacterial , Integrin alpha2/genetics , Integrin alpha2/metabolism , Integrin alpha3/genetics , Integrin alpha3/metabolism , Integrin alpha6/genetics , Integrin alpha6/metabolism , Integrin beta1/genetics , Intestinal Mucosa/microbiology , Mutation , Peyer's Patches/cytology , Virulence Factors/metabolism , Yersinia/genetics , Yersinia/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...