Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Cytogenet Genome Res ; 152(3): 117-121, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28854430

RESUMO

A 41-year-old Asian woman with bilateral renal angiomyolipomas (AML) was incidentally identified to have a balanced translocation, 46,XX,t(11;12)(p15.4;q15). She had no other features or family history to suggest a diagnosis of tuberous sclerosis. Her healthy daughter had the same translocation and no renal AML at the age of 3 years. Whole-genome sequencing was performed on genomic maternal DNA isolated from blood. A targeted de novo assembly was then conducted with ABySS for chromosomes 11 and 12. Sanger sequencing was used to validate the translocation breakpoints. As a result, genomic characterization of chromosomes 11 and 12 revealed that the 11p breakpoint disrupted the NUP98 gene in intron 1, causing a separation of the promoter and transcription start site from the rest of the gene. The translocation breakpoint on chromosome 12q was located in a gene desert. NUP98 has not yet been associated with renal AML pathogenesis, but somatic NUP98 alterations are recurrently implicated in hematological malignancies, most often following a gene fusion event. We also found evidence for complex structural events involving chromosome 12, which appear to disrupt the TDG gene. We identified a TDGP1 partially processed pseudogene at 12p12.1, which adds complexity to the de novo assembly. In conclusion, this is the first report of a germline constitutional structural chromosome rearrangement disrupting NUP98 that occurred in a generally healthy woman with bilateral renal AML.


Assuntos
Angiomiolipoma/genética , Cromossomos Humanos Par 11/genética , Cromossomos Humanos Par 12/genética , Neoplasias Renais/genética , Complexo de Proteínas Formadoras de Poros Nucleares/genética , Translocação Genética , Adulto , Amniocentese , Análise Citogenética/métodos , Feminino , Proteínas Ligadas por GPI/genética , Estudo de Associação Genômica Ampla , Genômica/métodos , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Proteínas de Neoplasias/genética , Regiões Promotoras Genéticas , Pseudogenes , Sítio de Iniciação de Transcrição , Esclerose Tuberosa/diagnóstico , Esclerose Tuberosa/genética
2.
Blood ; 119(4): 1029-31, 2012 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-22144180

RESUMO

Women treated at young ages with supradiaphragmatic radiotherapy for Hodgkin lymphoma (HL) have a highly increased risk of breast cancer. For personalized advice and follow-up regimens for patients, information is needed on how the radiotherapy-related risk is affected by other breast cancer risk factors. Genome-wide association studies have identified 14 independently replicated common single nucleotide polymorphisms that influence breast cancer risk. To examine whether these variants contribute to risk of radiation-associated breast cancer in HL, we analyzed 2 independent case-control series, from the United Kingdom and The Netherlands, totaling 693 HL patients, 232 with breast cancer and 461 without. rs1219648, which annotates the FGFR2 gene, was associated with risk in both series (combined per-allele odds ratio = 1.59, 95% confidence interval: 1.26-2.02; P = .000111). These data provide evidence that genetic variation in FGFR2 influences radiation-induced breast cancer risk.


Assuntos
Neoplasias da Mama/genética , Doença de Hodgkin/radioterapia , Glândulas Mamárias Humanas/efeitos da radiação , Polimorfismo de Nucleotídeo Único , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Adulto , Fatores Etários , Neoplasias da Mama/complicações , Neoplasias da Mama/etiologia , Estudos de Casos e Controles , Feminino , Predisposição Genética para Doença , Doença de Hodgkin/complicações , Doença de Hodgkin/genética , Humanos , Pessoa de Meia-Idade , Países Baixos/epidemiologia , Medicina de Precisão , Risco , Reino Unido/epidemiologia
3.
Genes Chromosomes Cancer ; 52(10): 954-60, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23893660

RESUMO

Over 90% of infants (< 1-year-old) diagnosed with leukemia have pro-B acute lymphoblastic leukemia (ALL) containing the MLL-AF4 fusion. When compared with other forms of paediatric ALL affecting later B-cell differentiation, MLL-AF4 pro-B is associated with a dismal prognosis with a typical 5-year disease-free survival of <20%. MLL-AF4 may be sufficient on its own for leukemogenesis or the gene-fusion product may alternatively predispose transformed cells to global genetic instability, enhancing the acquisition of additional key mutations. To gain insight into the genomic landscape of infant MLL-AF4 pro-B ALL we performed whole genome sequencing of diagnostic leukemic blasts and matched germline samples from three MLL-AF4 pro-B ALL infants. Our analysis revealed few somatic changes (copy number abnormalities, loss of heterozygosity, or single nucleotide variants), demonstrating that only a very small number of mutations are necessary to generate infant MLL-leukemia.


Assuntos
Proteína de Leucina Linfoide-Mieloide/genética , Proteínas de Fusão Oncogênica/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Variações do Número de Cópias de DNA , Análise Mutacional de DNA , Genoma Humano , Genômica , Humanos , Mutação INDEL/genética , Lactente , Recém-Nascido , Perda de Heterozigosidade , Polimorfismo de Nucleotídeo Único , Análise de Sequência de DNA , Translocação Genética
4.
Blood ; 118(3): 670-4, 2011 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-21596858

RESUMO

Since an association between the human leukocyte antigen (HLA) region and Hodgkin lymphoma (HL) was first reported in 1967, many studies have reported associations between HL risk and both single nucleotide polymorphism (SNP) and classic HLA allele variation in the major histocompatibility complex. However, population stratification and the extent and complexity of linkage disequilibrium within the major histocompatibility complex have hindered efforts to fine-map causal signals. Using SNP data to impute alleles at classic HLA loci, we have conducted an integrated analysis of HL risk within the HLA region in 582 early-onset HL cases and 4736 controls. We confirm that the strongest signal of association comes from an SNP located in the class II region, rs6903608 (odds ratio [OR] = 1.79, P = 6.63 × 10(-19)), which is unlikely to be driven by association to HLA-DRB, DQA, or DQB alleles. In addition, we identify independent signals at rs2281389 (OR = 1.73, P = 6.31 × 10(-13)), a SNP that maps closely to HLA-DPB1, and the class II HLA allele DQA1*02:01 (OR = 0.56, P = 1.51 × 10(-7)). These data suggest that multiple independent loci within the HLA class II region contribute to the risk of developing early-onset HL.


Assuntos
Cromossomos Humanos Par 6 , Antígenos HLA/genética , Doença de Hodgkin/epidemiologia , Doença de Hodgkin/genética , Idade de Início , Predisposição Genética para Doença/epidemiologia , Predisposição Genética para Doença/genética , Humanos , Modelos Genéticos , Polimorfismo de Nucleotídeo Único , Fatores de Risco
5.
Artigo em Inglês | MEDLINE | ID: mdl-30833417

RESUMO

We report a case of early-onset pancreatic ductal adenocarcinoma in a patient harboring biallelic MUTYH germline mutations, whose tumor featured somatic mutational signatures consistent with defective MUTYH-mediated base excision repair and the associated driver KRAS transversion mutation p.Gly12Cys. Analysis of an additional 730 advanced cancer cases (N = 731) was undertaken to determine whether the mutational signatures were also present in tumors from germline MUTYH heterozygote carriers or if instead the signatures were only seen in those with biallelic loss of function. We identified two patients with breast cancer each carrying a pathogenic germline MUTYH variant with a somatic MUTYH copy loss leading to the germline variant being homozygous in the tumor and demonstrating the same somatic signatures. Our results suggest that monoallelic inactivation of MUTYH is not sufficient for C:G>A:T transversion signatures previously linked to MUTYH deficiency to arise (N = 9), but that biallelic complete loss of MUTYH function can cause such signatures to arise even in tumors not classically seen in MUTYH-associated polyposis (N = 3). Although defective MUTYH is not the only determinant of these signatures, MUTYH germline variants may be present in a subset of patients with tumors demonstrating elevated somatic signatures possibly suggestive of MUTYH deficiency (e.g., COSMIC Signature 18, SigProfiler SBS18/SBS36, SignatureAnalyzer SBS18/SBS36).


Assuntos
Neoplasias da Mama/genética , Carcinoma Ductal Pancreático/genética , DNA Glicosilases/genética , Mutação , Neoplasias Pancreáticas/genética , Idade de Início , DNA Glicosilases/deficiência , Feminino , Mutação em Linhagem Germinativa , Humanos , Perda de Heterozigosidade , Pessoa de Meia-Idade , Proteínas Proto-Oncogênicas p21(ras)/genética
6.
NPJ Precis Oncol ; 2(1): 8, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29872726

RESUMO

Eccrine porocarcinomas (EPs) are rare malignant tumours of the intraepidermic sweat gland duct and most often arise from benign eccrine poromas. Some recurrent somatic genomic events have been identified in these malignancies, but very little is known about the complexity of their molecular pathophysiology. We describe the whole genome and whole transcriptome genomic profiling of a metastatic EP in a 66-year-old male patient with a previous history of localized porocarcinoma of the scalp. Whole genome and whole transcriptome genomic profiling was performed on the metastatic EP. Whole genome sequencing was performed on blood-derived DNA in order to allow a comparison between germline and somatic events. We found somatic copy losses of several tumour suppressor genes including APC, PTEN and CDKN2A, CDKN2B and CDKN1A. We identified a somatic hemizygous CDKN2A pathogenic splice site variant. De novo transcriptome assembly revealed abnormal splicing of CDKN2A p14ARF and p16INK4a. Elevated expression of oncogenes EGFR and NOTCH1 was noted and no somatic mutations were found in these genes. Wnt pathway somatic alterations were also observed. In conclusion, our results suggest that the molecular pathophysiology of malignant EP features high complexity and subtle interactions of multiple key genes. Cell cycle dysregulation and CDKN2A loss of function was found to be a new potential driver in EP tumourigenesis. Moreover, the combination of somatic copy number variants and abnormal gene expression perhaps partly related to epigenetic mechanisms, all likely contribute to the development of this rare malignancy in our patient.

7.
Artigo em Inglês | MEDLINE | ID: mdl-28514723

RESUMO

We describe a woman with the known pathogenic germline variant CHEK2:c.1100delC and synchronous diagnoses of both pelvic genital type leiomyosarcoma (LMS) and metastatic invasive ductal breast carcinoma. CHEK2 (checkpoint kinase 2) is a tumor-suppressor gene encoding a serine/threonine-protein kinase (CHEK2) involved in double-strand DNA break repair and cell cycle arrest. The CHEK2:c.1100delC variant is a moderate penetrance allele resulting in an approximately twofold increase in breast cancer risk. Whole-genome and whole-transcriptome sequencing were performed on the leiomyosarcoma and matched blood-derived DNA. Despite the presence of several genomic hits within the double-strand DNA damage pathway (CHEK2 germline variant and multiple RAD51B somatic structural variants), tumor profiling did not show an obvious DNA repair deficiency signature. However, even though the LMS displayed clear malignant features, its genomic profiling revealed several characteristics classically associated with leiomyomas including a translocation, t(12;14), with one breakpoint disrupting RAD51B and the other breakpoint upstream of HMGA2 with very high expression of HMGA2 and PLAG1 This is the first report of LMS genomic profiling in a patient with the germline CHEK2:c.1100delC variant and an additional diagnosis of metastatic invasive ductal breast carcinoma. We also describe a possible mechanistic relationship between leiomyoma and LMS based on genomic and transcriptome data. Our findings suggest that RAD51B translocation and HMGA2 overexpression may play an important role in LMS oncogenesis.


Assuntos
Quinase do Ponto de Checagem 2/genética , Leiomiossarcoma/genética , Alelos , Neoplasias da Mama/genética , Carcinoma Ductal de Mama/genética , Quinase do Ponto de Checagem 2/metabolismo , Reparo do DNA/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Feminino , Predisposição Genética para Doença/genética , Genômica , Mutação em Linhagem Germinativa , Proteína HMGA2/genética , Proteína HMGA2/metabolismo , Humanos , Leiomiossarcoma/metabolismo , Pessoa de Meia-Idade , Mutação/genética , Metástase Neoplásica
8.
Nat Commun ; 7: 11883, 2016 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-27329137

RESUMO

Colorectal cancer (CRC) displays a complex pattern of inheritance. It is postulated that much of the missing heritability of CRC is enshrined in high-impact rare alleles, which are mechanistically and clinically important. In this study, we assay the impact of rare germline mutations on CRC, analysing high-coverage exome sequencing data on 1,006 early-onset familial CRC cases and 1,609 healthy controls, with additional sequencing and array data on up to 5,552 cases and 6,792 controls. We identify highly penetrant rare mutations in 16% of familial CRC. Although the majority of these reside in known genes, we identify POT1, POLE2 and MRE11 as candidate CRC genes. We did not identify any coding low-frequency alleles (1-5%) with moderate effect. Our study clarifies the genetic architecture of CRC and probably discounts the existence of further major high-penetrance susceptibility genes, which individually account for >1% of the familial risk. Our results inform future study design and provide a resource for contextualizing the impact of new CRC genes.


Assuntos
Neoplasias Colorretais/genética , Predisposição Genética para Doença , Mutação em Linhagem Germinativa , Adulto , Fatores Etários , Idade de Início , Alelos , Estudos de Casos e Controles , Exoma , Feminino , Variação Genética , Humanos , Masculino , Pessoa de Meia-Idade , Linhagem , Fenótipo , Fatores de Risco , Reino Unido
9.
J Clin Oncol ; 33(5): 426-32, 2015 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-25559809

RESUMO

PURPOSE: Knowledge of the contribution of high-penetrance susceptibility to familial colorectal cancer (CRC) is relevant to the counseling, treatment, and surveillance of CRC patients and families. PATIENTS AND METHODS: To quantify the impact of germline mutation to familial CRC, we sequenced the mismatch repair genes (MMR) APC, MUTYH, and SMAD4/BMPR1A in 626 early-onset familial CRC cases ascertained through a population-based United Kingdom national registry. In addition, we evaluated the contribution of mutations in the exonuclease domain (exodom) of POLE and POLD1 genes that have recently been reported to confer CRC risk. RESULTS: Overall mutations (pathogenic, likely pathogenic) in MMR genes make the highest contribution to familial CRC (10.9%). Mutations in the other established CRC genes account for 3.3% of cases. POLE/POLD1 exodom mutations were identified in three patients with family histories consistent with dominant transmission of CRC. Collectively, mutations in the known genes account for 14.2% of familial CRC (89 of 626 cases; 95% CI = 11.5, 17.2). CONCLUSION: A genetic diagnosis is feasible in a high proportion of familial CRC. Mainstreaming such analysis in clinical practice should enable the medical management of patients and their families to be optimized. Findings suggest CRC screening of POLE and POLD1 mutation carriers should be comparable to that afforded to those at risk of HNPCC. Although the risk of CRC associated with unexplained familial CRC is in general moderate, in some families the risk is substantive and likely to be the consequence of unidentified genes, as exemplified by POLE and POLD1. Our findings have utility in the design of genetic analyses to identify such novel CRC risk genes.


Assuntos
Neoplasias Colorretais/diagnóstico , Neoplasias Colorretais/genética , Reparo de Erro de Pareamento de DNA , Exoma , Testes Genéticos/métodos , Mutação em Linhagem Germinativa , Proteína da Polipose Adenomatosa do Colo/genética , Adulto , Idoso , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/genética , Neoplasias Colorretais/epidemiologia , Neoplasias Colorretais Hereditárias sem Polipose/diagnóstico , Neoplasias Colorretais Hereditárias sem Polipose/genética , DNA Glicosilases/genética , Reparo de Erro de Pareamento de DNA/genética , DNA Polimerase II/genética , DNA Polimerase III/genética , Estudos de Viabilidade , Feminino , Predisposição Genética para Doença , Mutação em Linhagem Germinativa/genética , Humanos , Masculino , Pessoa de Meia-Idade , Linhagem , Proteínas de Ligação a Poli-ADP-Ribose , Valor Preditivo dos Testes , Sistema de Registros , Medição de Risco , Fatores de Risco , Análise de Sequência de DNA , Proteína Smad4/genética , Reino Unido/epidemiologia
10.
PLoS One ; 9(7): e102398, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25062255

RESUMO

JAGuaR is an alignment protocol for RNA-seq reads that uses an extended reference to increase alignment sensitivity. It uses BWA to align reads to the genome and reference transcript models (including annotated exon-exon junctions) specifically allowing for the possibility of a single read spanning multiple exons. Reads aligned to the transcript models are then re-mapped on to genomic coordinates, transforming alignments that span multiple exons into large-gapped alignments on the genome. While JAGuaR does not detect novel junctions, we demonstrate how JAGuaR generates fast and accurate transcriptome alignments, which allows for both sensitive and specific SNV calling.


Assuntos
RNA/genética , Alinhamento de Sequência/métodos , Análise de Sequência de RNA/métodos , Software , Algoritmos , Animais , Sequência de Bases , Éxons , Perfilação da Expressão Gênica/métodos , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Splicing de RNA/genética
11.
Nat Genet ; 45(5): 522-525, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23502783

RESUMO

A number of specific chromosomal abnormalities define the subgroups of multiple myeloma. In a meta-analysis of two genome-wide association studies of multiple myeloma including a total of 1,661 affected individuals, we investigated risk for developing a specific tumor karyotype. The t(11;14)(q13;q32) translocation in which CCND1 is placed under the control of the immunoglobulin heavy chain enhancer was strongly associated with the CCND1 c.870G>A polymorphism (P = 7.96 × 10(-11)). These results provide a model in which a constitutive genetic factor is associated with risk of a specific chromosomal translocation.


Assuntos
Cromossomos Humanos Par 11/genética , Cromossomos Humanos Par 14/genética , Ciclina D1/genética , Estudo de Associação Genômica Ampla , Mieloma Múltiplo/etiologia , Polimorfismo de Nucleotídeo Único/genética , Translocação Genética , Estudos de Casos e Controles , Genoma Humano , Genótipo , Humanos , Hibridização in Situ Fluorescente , Cariotipagem , Fenótipo , Fatores de Risco
13.
Nat Genet ; 43(9): 825-7, 2011 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-21804547

RESUMO

To identify susceptibility loci for meningioma, we conducted a genome-wide association study of 859 affected individuals (cases) and 704 controls with validation in two independent sample sets totaling 774 cases and 1,764 controls. We identified a new susceptibility locus for meningioma at 10p12.31 (MLLT10, rs11012732, odds ratio = 1.46, P(combined) = 1.88 × 10(-14)). This finding advances our understanding of the genetic basis of meningioma development.


Assuntos
Cromossomos Humanos Par 10/genética , Predisposição Genética para Doença , Neoplasias Meníngeas/genética , Meningioma/genética , Fatores de Transcrição/genética , Loci Gênicos , Estudo de Associação Genômica Ampla , Humanos , Masculino , Neoplasias Meníngeas/epidemiologia , Meningioma/epidemiologia , Polimorfismo Genético , Polimorfismo de Nucleotídeo Único , Risco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA