Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Br J Cancer ; 117(6): 1215-1223, 2017 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-28742792

RESUMEN

BACKGROUND: A substantial fraction of familial colorectal cancer (CRC) and polyposis heritability remains unexplained. This study aimed to identify predisposing loci in patients with these disorders. METHODS: Homozygosity mapping was performed using 222 563 SNPs in 302 index patients with various colorectal neoplasms and 3367 controls. Linkage analysis, exome and whole-genome sequencing were performed in a family affected by microsatellite stable CRCs. Candidate variants were genotyped in 10 554 cases and 21 480 controls. Gene expression was assessed at the mRNA and protein level. RESULTS: Homozygosity mapping revealed a disease-associated region at 1q32.3 which was part of the linkage region 1q32.2-42.2 identified in the CRC family. This includes a region previously associated with risk of CRC. Sequencing identified the p.Asp1432Glu variant in the MIA3 gene (known as TANGO1 or TANGO) and 472 additional rare, shared variants within the linkage region. In both cases and controls the population frequency was 0.02% for this MIA3 variant. The MIA3 mutant allele showed predominant mRNA expression in normal, cancer and precancerous tissues. Furthermore, immunohistochemistry revealed increased expression of MIA3 in adenomatous tissues. CONCLUSIONS: Taken together, our two independent strategies associate genetic variations in chromosome 1q loci and predisposition to familial CRC and polyps, which warrants further investigation.


Asunto(s)
Poliposis Adenomatosa del Colon/genética , Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Cromosomas Humanos Par 1/genética , Neoplasias Colorrectales/genética , Predisposición Genética a la Enfermedad , Proteínas de Neoplasias/genética , Translocador Nuclear del Receptor de Aril Hidrocarburo/metabolismo , Mapeo Cromosómico , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Ligamiento Genético , Genotipo , Homocigoto , Humanos , Repeticiones de Microsatélite , Proteínas de Neoplasias/metabolismo , Polimorfismo de Nucleótido Simple , Lesiones Precancerosas/genética , Lesiones Precancerosas/metabolismo , ARN Mensajero/metabolismo
4.
Fam Cancer ; 21(1): 79-83, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-33683519

RESUMEN

In addition to classic germline APC gene variants, APC mosaicism and deep intronic germline APC variants have also been reported to be causes of adenomatous polyposis. In this study, we investigated 80 unexplained colorectal polyposis patients without germline pathogenic variants in known polyposis predisposing genes to detect mosaic and deep intronic APC variants. All patients developed more than 50 colorectal polyps, with adenomas being predominantly observed. To detect APC mosaicism, we performed next-generation sequencing (NGS) in leukocyte DNA. Furthermore, using Sanger sequencing, the cohort was screened for the following previously reported deep intronic pathogenic germline APC variants: c.1408 + 731C > T, p.(Gly471Serfs*55), c.1408 + 735A > T, p.(Gly471Serfs*55), c.1408 + 729A > G, p.(Gly471Serfs*55) and c.532-941G > A, p.(Phe178Argfs*22). We did not detect mosaic or intronic APC variants in the screened unexplained colorectal polyposis patients. The results of this study indicate that the deep intronic APC variants investigated in this study are not a cause of colorectal polyposis in this Dutch population. In addition, NGS did not detect any further mosaic variants in our cohort.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/genética , Poliposis Adenomatosa del Colon , Neoplasias Colorrectales , Poliposis Adenomatosa del Colon/genética , Poliposis Adenomatosa del Colon/patología , Neoplasias Colorrectales/genética , Genes APC , Mutación de Línea Germinal , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Mutación
5.
Hum Pathol ; 125: 11-17, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35417733

RESUMEN

Small intestine-neuroendocrine tumors (SI-NETs) are one of the most common tumors of the small bowel. Despite an increasing incidence, the exact mechanisms driving underlying pathology remain to be determined. Interestingly, recent studies linked the development of (SI-)NETs to both Lynch syndrome (LS) and MUTYH variants. If confirmed, these associations would have important consequences for treatment. In this study we therefore investigated the prevalence of mismatch repair (MMR) deficiency and MUTYH variants in 64 primary resected SI-NETs. Immunohistochemistry was used to assess the expression of the MMR genes, and competitive allele-specific PCR (KASPar) targeting two hotspot MUTYH variants [p.(Tyr179Cys), p.(Gly396Asp)] was performed to determine their prevalence in SI-NETs. Strikingly, all 64 SI-NETs stained positive for MSH6 and PMS2, indicating MMR proficiency. In addition, no MUTYH hotspot variant was found in any of the 64 SI-NETs. As such, these results do not support an association between SI-NET development and LS or MUTYH variants. In order to gain insight into SI-NET pathogenesis and optimally manage patients, future research should therefore focus on other candidate genes.


Asunto(s)
Neoplasias Colorrectales Hereditarias sin Poliposis , Neoplasias Colorrectales , ADN Glicosilasas , Tumores Neuroendocrinos , Neoplasias Encefálicas , Neoplasias Colorrectales/genética , Neoplasias Colorrectales Hereditarias sin Poliposis/genética , ADN Glicosilasas/genética , Reparación de la Incompatibilidad de ADN/genética , Mutación de Línea Germinal , Humanos , Intestino Delgado , Endonucleasa PMS2 de Reparación del Emparejamiento Incorrecto/genética , Homólogo 1 de la Proteína MutL/genética , Síndromes Neoplásicos Hereditarios , Tumores Neuroendocrinos/metabolismo
6.
Mol Genet Genomic Med ; 7(4): e00603, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30827058

RESUMEN

BACKGROUND: Germline mutations affecting the exonuclease domains of POLE and POLD1 predispose to colorectal adenomas and carcinoma. Here, we aimed to screen the exonuclease domains to find the genetic causes of multiple colorectal polyps in unexplained cases. METHODS: Using a custom next-generation sequencing panel, we sequenced the exonuclease domains of POLE and POLD1 in 332 index patients diagnosed with multiple colorectal polyps without germline alteration in colorectal polyposis predisposing genes. RESULTS: We identified two variants of unknown significance. One germline POLD1 c.961G>A, p.(Gly321Ser) variant was found in two cases. The first patient was diagnosed with multiple polyps at age 35 and colorectal cancer (CRC) at age 37, with no known family history of CRC. The second patient was diagnosed with CRC at age 44 and cumulatively developed multiple polyps; this patient had two sisters with endometrial cancer who did not carry the variant. Furthermore, we identified a novel POLD1 c.955 T>G, p.(Cys319Gly) variant in a patient diagnosed with multiple colorectal adenomas at age 40. Co-segregation analysis showed that one sister who cumulatively developed multiple adenomas from age 34, and another sister who developed CRC at age 38 did not carry the variant. We did not identify pathogenic variants in POLE and POLD1. CONCLUSION: This study confirms the low frequency of causal variants in these genes in the predisposition for multiple colorectal polyps, and also establishes that these genes are a rare cause of the disease.


Asunto(s)
Pólipos del Colon/genética , Neoplasias Colorrectales/genética , ADN Polimerasa III/genética , ADN Polimerasa II/genética , Tasa de Mutación , Proteínas de Unión a Poli-ADP-Ribosa/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Dominio Catalítico , ADN Polimerasa II/química , ADN Polimerasa III/química , Femenino , Mutación de Línea Germinal , Humanos , Masculino , Persona de Mediana Edad , Mutación Missense , Proteínas de Unión a Poli-ADP-Ribosa/química
7.
Cancer Cell ; 35(2): 256-266.e5, 2019 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-30753826

RESUMEN

Biallelic germline mutations affecting NTHL1 predispose carriers to adenomatous polyposis and colorectal cancer, but the complete phenotype is unknown. We describe 29 individuals carrying biallelic germline NTHL1 mutations from 17 families, of which 26 developed one (n = 10) or multiple (n = 16) malignancies in 14 different tissues. An unexpected high breast cancer incidence was observed in female carriers (60%). Mutational signature analysis of 14 tumors from 7 organs revealed that NTHL1 deficiency underlies the main mutational process in all but one of the tumors (93%). These results reveal NTHL1 as a multi-tumor predisposition gene with a high lifetime risk for extracolonic cancers and a typical mutational signature observed across tumor types, which can assist in the recognition of this syndrome.


Asunto(s)
Biomarcadores de Tumor/genética , Análisis Mutacional de ADN , Desoxirribonucleasa (Dímero de Pirimidina)/genética , Perfilación de la Expresión Génica , Mutación de Línea Germinal , Síndromes Neoplásicos Hereditarios/genética , Transcriptoma , Adulto , Anciano , Biomarcadores de Tumor/deficiencia , Reparación del ADN/genética , Desoxirribonucleasa (Dímero de Pirimidina)/deficiencia , Europa (Continente) , Femenino , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Herencia , Humanos , Masculino , Persona de Mediana Edad , Síndromes Neoplásicos Hereditarios/enzimología , Síndromes Neoplásicos Hereditarios/patología , Linaje , Fenotipo , Medición de Riesgo , Factores de Riesgo , Adulto Joven
8.
Fam Cancer ; 17(4): 507-515, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29147930

RESUMEN

Lynch syndrome (LS) patients are at high risk of developing colorectal cancer (CRC). Phenotypic variability might in part be explained by common susceptibility loci identified in Genome Wide Association Studies (GWAS). Previous studies focused mostly on MLH1, MSH2 and MSH6 carriers, with conflicting results. We aimed to determine the role of GWAS SNPs in PMS2 mutation carriers. A cohort study was performed in 507 PMS2 carriers (124 CRC cases), genotyped for 24 GWAS SNPs, including SNPs at 11q23.1 and 8q23.3. Hazard ratios (HRs) were calculated using a weighted Cox regression analysis to correct for ascertainment bias. Discrimination was assessed with a concordance statistic in a bootstrap cross-validation procedure. Individual SNPs only had non-significant associations with CRC occurrence with HRs lower than 2, although male carriers of allele A at rs1321311 (6p21.31) may have increased risk of CRC (HR = 2.1, 95% CI 1.2-3.0). A polygenic risk score (PRS) based on 24 HRs had an HR of 2.6 (95% CI 1.5-4.6) for the highest compared to the lowest quartile, but had no discriminative ability (c statistic 0.52). Previously suggested SNPs do not modify CRC risk in PMS2 carriers. Future large studies are needed for improved risk stratification among Lynch syndrome patients.


Asunto(s)
Neoplasias Colorrectales Hereditarias sin Poliposis/genética , Neoplasias Colorrectales/genética , Endonucleasa PMS2 de Reparación del Emparejamiento Incorrecto/genética , Polimorfismo de Nucleótido Simple , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Cromosomas Humanos Par 11 , Cromosomas Humanos Par 8 , Neoplasias Colorrectales/mortalidad , Neoplasias Colorrectales Hereditarias sin Poliposis/mortalidad , Femenino , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Heterocigoto , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad
9.
Eur J Hum Genet ; 23(8): 1080-4, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25370038

RESUMEN

Germline variants affecting the exonuclease domains of POLE and POLD1 predispose to multiple colorectal adenomas and/or colorectal cancer (CRC). The aim of this study was to estimate the prevalence of previously described heterozygous germline variants POLE c.1270C>G, p.(Leu424Val) and POLD1 c.1433G>A, p.(Ser478Asn) in a Dutch series of unexplained familial, early onset CRC and polyposis index cases. We examined 1188 familial CRC and polyposis index patients for POLE p.(Leu424Val) and POLD1 p.(Ser478Asn) variants using competitive allele-specific PCR. In addition, protein expression of the POLE and DNA mismatch repair genes was studied by immunohistochemistry in tumours from POLE carriers. Somatic mutations were screened using semiconductor sequencing. We detected three index patients (0.25%) with a POLE p.(Leu424Val) variant. In one patient, the variant was found to be de-novo. Tumours from three patients from two families were microsatellite instable, and immunohistochemistry showed MSH6/MSH2 deficiency suggestive of Lynch syndrome. Somatic mutations but no germline MSH6 and MSH2 variants were subsequently found, and one tumour displayed a hypermutator phenotype. None of the 1188 patients carried the POLD1 p.(Ser478Asn) variant. POLE germline variant carriers are also associated with a microsatellite instable CRC. POLE DNA analysis now seems warranted in microsatellite instable CRC, especially in the absence of a causative DNA mismatch repair gene germline variant.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Colorrectales/genética , ADN Polimerasa III/genética , ADN Polimerasa II/genética , Síndromes Neoplásicos Hereditarios/genética , Adolescente , Adulto , Neoplasias Encefálicas/complicaciones , Neoplasias Encefálicas/patología , Neoplasias Colorrectales/complicaciones , Neoplasias Colorrectales/patología , Reparación de la Incompatibilidad de ADN , Proteínas de Unión al ADN/genética , Femenino , Mutación de Línea Germinal , Humanos , Masculino , Persona de Mediana Edad , Proteína 2 Homóloga a MutS/genética , Síndromes Neoplásicos Hereditarios/complicaciones , Síndromes Neoplásicos Hereditarios/patología , Proteínas de Unión a Poli-ADP-Ribosa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA