Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Proc Natl Acad Sci U S A ; 111(43): 15544-9, 2014 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-25313082

RESUMO

Previous studies have established that a subset of head and neck tumors contains human papillomavirus (HPV) sequences and that HPV-driven head and neck cancers display distinct biological and clinical features. HPV is known to drive cancer by the actions of the E6 and E7 oncoproteins, but the molecular architecture of HPV infection and its interaction with the host genome in head and neck cancers have not been comprehensively described. We profiled a cohort of 279 head and neck cancers with next generation RNA and DNA sequencing and show that 35 (12.5%) tumors displayed evidence of high-risk HPV types 16, 33, or 35. Twenty-five cases had integration of the viral genome into one or more locations in the human genome with statistical enrichment for genic regions. Integrations had a marked impact on the human genome and were associated with alterations in DNA copy number, mRNA transcript abundance and splicing, and both inter- and intrachromosomal rearrangements. Many of these events involved genes with documented roles in cancer. Cancers with integrated vs. nonintegrated HPV displayed different patterns of DNA methylation and both human and viral gene expressions. Together, these data provide insight into the mechanisms by which HPV interacts with the human genome beyond expression of viral oncoproteins and suggest that specific integration events are an integral component of viral oncogenesis.


Assuntos
Genoma Humano/genética , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/virologia , Interações Hospedeiro-Patógeno/genética , Papillomaviridae/fisiologia , Sequência de Bases , Metilação de DNA/genética , Regulação Neoplásica da Expressão Gênica , Genes Neoplásicos , Humanos , Dados de Sequência Molecular , Integração Viral/genética
2.
Sci Adv ; 8(6): eabm2382, 2022 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-35138907

RESUMO

Fusion genes represent a class of attractive therapeutic targets. Thousands of fusion genes have been identified in patients with cancer, but the functional consequences and therapeutic implications of most of these remain largely unknown. Here, we develop a functional genomic approach that consists of efficient fusion reconstruction and sensitive cell viability and drug response assays. Applying this approach, we characterize ~100 fusion genes detected in patient samples of The Cancer Genome Atlas, revealing a notable fraction of low-frequency fusions with activating effects on tumor growth. Focusing on those in the RTK-RAS pathway, we identify a number of activating fusions that can markedly affect sensitivity to relevant drugs. Last, we propose an integrated, level-of-evidence classification system to prioritize gene fusions systematically. Our study reiterates the urgent clinical need to incorporate similar functional genomic approaches to characterize gene fusions, thereby maximizing the utility of gene fusions for precision oncology.


Assuntos
Neoplasias , Fusão Gênica , Genoma , Genômica , Humanos , Neoplasias/genética , Medicina de Precisão
3.
Genome Biol ; 20(1): 209, 2019 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-31610796

RESUMO

BACKGROUND: Genomic rearrangements exert a heavy influence on the molecular landscape of cancer. New analytical approaches integrating somatic structural variants (SSVs) with altered gene features represent a framework by which we can assign global significance to a core set of genes, analogous to established methods that identify genes non-randomly targeted by somatic mutation or copy number alteration. While recent studies have defined broad patterns of association involving gene transcription and nearby SSV breakpoints, global alterations in DNA methylation in the context of SSVs remain largely unexplored. RESULTS: By data integration of whole genome sequencing, RNA sequencing, and DNA methylation arrays from more than 1400 human cancers, we identify hundreds of genes and associated CpG islands (CGIs) for which the nearby presence of a somatic structural variant (SSV) breakpoint is recurrently associated with altered expression or DNA methylation, respectively, independently of copy number alterations. CGIs with SSV-associated increased methylation are predominantly promoter-associated, while CGIs with SSV-associated decreased methylation are enriched for gene body CGIs. Rearrangement of genomic regions normally having higher or lower methylation is often involved in SSV-associated CGI methylation alterations. Across cancers, the overall structural variation burden is associated with a global decrease in methylation, increased expression in methyltransferase genes and DNA damage response genes, and decreased immune cell infiltration. CONCLUSION: Genomic rearrangement appears to have a major role in shaping the cancer DNA methylome, to be considered alongside commonly accepted mechanisms including histone modifications and disruption of DNA methyltransferases.


Assuntos
Epigenoma , Variação Estrutural do Genoma , Neoplasias/genética , Ilhas de CpG , Humanos
4.
Cell Rep ; 24(2): 515-527, 2018 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-29996110

RESUMO

A systematic cataloging of genes affected by genomic rearrangement, using multiple patient cohorts and cancer types, can provide insight into cancer-relevant alterations outside of exomes. By integrative analysis of whole-genome sequencing (predominantly low pass) and gene expression data from 1,448 cancers involving 18 histopathological types in The Cancer Genome Atlas, we identified hundreds of genes for which the nearby presence (within 100 kb) of a somatic structural variant (SV) breakpoint is associated with altered expression. While genomic rearrangements are associated with widespread copy-number alteration (CNA) patterns, approximately 1,100 genes-including overexpressed cancer driver genes (e.g., TERT, ERBB2, CDK12, CDK4) and underexpressed tumor suppressors (e.g., TP53, RB1, PTEN, STK11)-show SV-associated deregulation independent of CNA. SVs associated with the disruption of topologically associated domains, enhancer hijacking, or fusion transcripts are implicated in gene upregulation. For cancer-relevant pathways, SVs considerably expand our understanding of how genes are affected beyond point mutation or CNA.


Assuntos
Regulação Neoplásica da Expressão Gênica , Rearranjo Gênico/genética , Genes Neoplásicos , Genoma Humano , Neoplasias/genética , Sequência de Bases , Carcinogênese/genética , Carcinogênese/patologia , Variações do Número de Cópias de DNA/genética , Elementos Facilitadores Genéticos/genética , Humanos
5.
Cell Rep ; 19(4): 875-889, 2017 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-28445736

RESUMO

The extent and nature of epigenomic changes associated with melanoma progression is poorly understood. Through systematic epigenomic profiling of 35 epigenetic modifications and transcriptomic analysis, we define chromatin state changes associated with melanomagenesis by using a cell phenotypic model of non-tumorigenic and tumorigenic states. Computation of specific chromatin state transitions showed loss of histone acetylations and H3K4me2/3 on regulatory regions proximal to specific cancer-regulatory genes in important melanoma-driving cell signaling pathways. Importantly, such acetylation changes were also observed between benign nevi and malignant melanoma human tissues. Intriguingly, only a small fraction of chromatin state transitions correlated with expected changes in gene expression patterns. Restoration of acetylation levels on deacetylated loci by histone deacetylase (HDAC) inhibitors selectively blocked excessive proliferation in tumorigenic cells and human melanoma cells, suggesting functional roles of observed chromatin state transitions in driving hyperproliferative phenotype. Through these results, we define functionally relevant chromatin states associated with melanoma progression.


Assuntos
Cromatina/metabolismo , Epigenômica , Histonas/metabolismo , Acetilação , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Imunoprecipitação da Cromatina , Intervalo Livre de Doença , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/química , Histona Desacetilases/metabolismo , Humanos , Ácidos Hidroxâmicos/farmacologia , Estimativa de Kaplan-Meier , Melanoma/metabolismo , Melanoma/mortalidade , Melanoma/patologia , PTEN Fosfo-Hidrolase/antagonistas & inibidores , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Análise de Componente Principal , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Vorinostat
6.
Cancer Cell ; 31(6): 820-832.e3, 2017 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-28528867

RESUMO

Molecular alterations involving the PI3K/AKT/mTOR pathway (including mutation, copy number, protein, or RNA) were examined across 11,219 human cancers representing 32 major types. Within specific mutated genes, frequency, mutation hotspot residues, in silico predictions, and functional assays were all informative in distinguishing the subset of genetic variants more likely to have functional relevance. Multiple oncogenic pathways including PI3K/AKT/mTOR converged on similar sets of downstream transcriptional targets. In addition to mutation, structural variations and partial copy losses involving PTEN and STK11 showed evidence for having functional relevance. A substantial fraction of cancers showed high mTOR pathway activity without an associated canonical genetic or genomic alteration, including cancers harboring IDH1 or VHL mutations, suggesting multiple mechanisms for pathway activation.


Assuntos
Neoplasias/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteogenômica , Proteínas Proto-Oncogênicas c-akt/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Bases de Dados Genéticas , Perfilação da Expressão Gênica , Humanos , Mutação , Neoplasias/metabolismo , Transdução de Sinais , Análise de Sobrevida
7.
Cancer Res ; 75(18): 3865-3878, 2015 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-26139243

RESUMO

The SWI/SNF multisubunit complex modulates chromatin structure through the activity of two mutually exclusive catalytic subunits, SMARCA2 and SMARCA4, which both contain a bromodomain and an ATPase domain. Using RNAi, cancer-specific vulnerabilities have been identified in SWI/SNF-mutant tumors, including SMARCA4-deficient lung cancer; however, the contribution of conserved, druggable protein domains to this anticancer phenotype is unknown. Here, we functionally deconstruct the SMARCA2/4 paralog dependence of cancer cells using bioinformatics, genetic, and pharmacologic tools. We evaluate a selective SMARCA2/4 bromodomain inhibitor (PFI-3) and characterize its activity in chromatin-binding and cell-functional assays focusing on cells with altered SWI/SNF complex (e.g., lung, synovial sarcoma, leukemia, and rhabdoid tumors). We demonstrate that PFI-3 is a potent, cell-permeable probe capable of displacing ectopically expressed, GFP-tagged SMARCA2-bromodomain from chromatin, yet contrary to target knockdown, the inhibitor fails to display an antiproliferative phenotype. Mechanistically, the lack of pharmacologic efficacy is reconciled by the failure of bromodomain inhibition to displace endogenous, full-length SMARCA2 from chromatin as determined by in situ cell extraction, chromatin immunoprecipitation, and target gene expression studies. Furthermore, using inducible RNAi and cDNA complementation (bromodomain- and ATPase-dead constructs), we unequivocally identify the ATPase domain, and not the bromodomain of SMARCA2, as the relevant therapeutic target with the catalytic activity suppressing defined transcriptional programs. Taken together, our complementary genetic and pharmacologic studies exemplify a general strategy for multidomain protein drug-target validation and in case of SMARCA2/4 highlight the potential for drugging the more challenging helicase/ATPase domain to deliver on the promise of synthetic-lethality therapy.


Assuntos
Compostos Azabicíclicos/farmacologia , Montagem e Desmontagem da Cromatina/efeitos dos fármacos , Proteínas Cromossômicas não Histona/deficiência , DNA Helicases/antagonistas & inibidores , Terapia de Alvo Molecular , Proteínas de Neoplasias/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Proteínas Nucleares/antagonistas & inibidores , Piridinas/farmacologia , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/deficiência , Ligação Competitiva , Catálise , Linhagem Celular Tumoral , Cromatina/metabolismo , Proteínas Cromossômicas não Histona/genética , DNA Helicases/química , DNA Helicases/deficiência , DNA Complementar/genética , Técnicas de Inativação de Genes , Teste de Complementação Genética , Humanos , Neoplasias Pulmonares/patologia , Análise em Microsséries , Neoplasias/genética , Proteínas Nucleares/química , Proteínas Nucleares/deficiência , Estrutura Terciária de Proteína , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Tumor Rabdoide/genética , Tumor Rabdoide/patologia , Sarcoma Sinovial/genética , Sarcoma Sinovial/patologia , Fatores de Transcrição/química , Fatores de Transcrição/genética
8.
Science ; 346(6206): 256-9, 2014 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-25301631

RESUMO

Cancers are composed of populations of cells with distinct molecular and phenotypic features, a phenomenon termed intratumor heterogeneity (ITH). ITH in lung cancers has not been well studied. We applied multiregion whole-exome sequencing (WES) on 11 localized lung adenocarcinomas. All tumors showed clear evidence of ITH. On average, 76% of all mutations and 20 out of 21 known cancer gene mutations were identified in all regions of individual tumors, which suggested that single-region sequencing may be adequate to identify the majority of known cancer gene mutations in localized lung adenocarcinomas. With a median follow-up of 21 months after surgery, three patients have relapsed, and all three patients had significantly larger fractions of subclonal mutations in their primary tumors than patients without relapse. These data indicate that a larger subclonal mutation fraction may be associated with increased likelihood of postsurgical relapse in patients with localized lung adenocarcinomas.


Assuntos
Adenocarcinoma/genética , Heterogeneidade Genética , Neoplasias Pulmonares/genética , Recidiva Local de Neoplasia/genética , Adenocarcinoma/patologia , Adenocarcinoma de Pulmão , Análise Mutacional de DNA , Exoma/genética , Genes Neoplásicos , Humanos , Neoplasias Pulmonares/patologia , Mutação , Recidiva Local de Neoplasia/patologia
9.
Nat Genet ; 46(4): 376-379, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24633157

RESUMO

Angiosarcoma is an aggressive malignancy that arises spontaneously or secondarily to ionizing radiation or chronic lymphoedema. Previous work has identified aberrant angiogenesis, including occasional somatic mutations in angiogenesis signaling genes, as a key driver of angiosarcoma. Here we employed whole-genome, whole-exome and targeted sequencing to study the somatic changes underpinning primary and secondary angiosarcoma. We identified recurrent mutations in two genes, PTPRB and PLCG1, which are intimately linked to angiogenesis. The endothelial phosphatase PTPRB, a negative regulator of vascular growth factor tyrosine kinases, harbored predominantly truncating mutations in 10 of 39 tumors (26%). PLCG1, a signal transducer of tyrosine kinases, encoded a recurrent, likely activating p.Arg707Gln missense variant in 3 of 34 cases (9%). Overall, 15 of 39 tumors (38%) harbored at least one driver mutation in angiogenesis signaling genes. Our findings inform and reinforce current therapeutic efforts to target angiogenesis signaling in angiosarcoma.


Assuntos
Hemangiossarcoma/tratamento farmacológico , Hemangiossarcoma/genética , Neovascularização Patológica/tratamento farmacológico , Fosfolipase C gama/genética , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/genética , Análise de Variância , Sequência de Bases , Exoma/genética , Células Endoteliais da Veia Umbilical Humana , Humanos , Dados de Sequência Molecular , Mutação/genética , Neovascularização Patológica/genética , Interferência de RNA , Análise de Sequência de RNA , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA