Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
EMBO J ; 36(19): 2829-2843, 2017 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-28814448

RESUMO

The development of many sporadic cancers is directly initiated by carcinogen exposure. Carcinogens induce malignancies by creating DNA lesions (i.e., adducts) that can result in mutations if left unrepaired. Despite this knowledge, there has been remarkably little investigation into the regulation of susceptibility to acquire DNA lesions. In this study, we present the first quantitative human genome-wide map of DNA lesions induced by ultraviolet (UV) radiation, the ubiquitous carcinogen in sunlight that causes skin cancer. Remarkably, the pattern of carcinogen susceptibility across the genome of primary cells significantly reflects mutation frequency in malignant melanoma. Surprisingly, DNase-accessible euchromatin is protected from UV, while lamina-associated heterochromatin at the nuclear periphery is vulnerable. Many cancer driver genes have an intrinsic increase in carcinogen susceptibility, including the BRAF oncogene that has the highest mutation frequency in melanoma. These findings provide a genome-wide snapshot of DNA injuries at the earliest stage of carcinogenesis. Furthermore, they identify carcinogen susceptibility as an origin of genome instability that is regulated by nuclear architecture and mirrors mutagenesis in cancer.


Assuntos
Carcinógenos/toxicidade , Transformação Celular Neoplásica , Resistência a Medicamentos/genética , Instabilidade Genômica/efeitos dos fármacos , Instabilidade Genômica/genética , Mutagênese , Sequência de Bases/fisiologia , Transformação Celular Neoplásica/efeitos dos fármacos , Transformação Celular Neoplásica/genética , Células Cultivadas , Dano ao DNA , Resistência a Medicamentos/efeitos dos fármacos , Epigênese Genética/efeitos dos fármacos , Humanos , Melanoma/etiologia , Melanoma/genética , Mutagênese/efeitos dos fármacos , Mutagênese/genética , Neoplasias Cutâneas/etiologia , Neoplasias Cutâneas/genética , Raios Ultravioleta , Melanoma Maligno Cutâneo
2.
Life Sci Alliance ; 5(4)2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34983823

RESUMO

Carcinogenic insult, such as UV light exposure, creates DNA lesions that evolve into mutations if left unrepaired. These resulting mutations can contribute to carcinogenesis and drive malignant phenotypes. Susceptibility to carcinogens (i.e., the propensity to form a carcinogen-induced DNA lesion) is regulated by both genetic and epigenetic factors. Importantly, carcinogen susceptibility is a critical contributor to cancer mutagenesis. It is known that mutations can be prevented by tumor suppressor regulation of DNA damage response pathways; however, their roles carcinogen susceptibility have not yet been reported. In this study, we reveal that the retinoblastoma (RB1) tumor suppressor regulates UV susceptibility across broad regions of the genome. In particular, centromere and telomere-proximal regions exhibit significant increases in UV lesion susceptibility when RB1 is deleted. Several cancer-related genes are located within genomic regions of increased susceptibility, including telomerase reverse transcriptase, TERT, thereby accelerating mutagenic potential in cancers with RB1 pathway alterations. These findings reveal novel genome stability mechanisms of a tumor suppressor and uncover new pathways to accumulate mutations during cancer evolution.


Assuntos
Carcinogênese , Carcinógenos/farmacologia , Neoplasias , Proteínas de Ligação a Retinoblastoma/genética , Ubiquitina-Proteína Ligases/genética , Sistemas CRISPR-Cas , Carcinogênese/efeitos dos fármacos , Carcinogênese/genética , Linhagem Celular , Técnicas de Inativação de Genes , Predisposição Genética para Doença/genética , Humanos , Mutação/genética , Neoplasias/genética , Neoplasias/patologia , Oncogenes/genética
3.
Mutat Res ; 823: 111758, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34333390

RESUMO

Exposure to the ultraviolet (UV) radiation in sunlight creates DNA lesions, which if left unrepaired can induce mutations and contribute to skin cancer. The two most common UV-induced DNA lesions are the cis-syn cyclobutane pyrimidine dimers (CPDs) and pyrimidine (6-4) pyrimidone photoproducts (6-4PPs), both of which can initiate mutations. Interestingly, mutation frequency across the genomes of many cancers is heterogenous with significant increases in heterochromatin. Corresponding increases in UV lesion susceptibility and decreases in repair are observed in heterochromatin versus euchromatin. However, the individual contributions of CPDs and 6-4PPs to mutagenesis have not been systematically examined in specific genomic and epigenomic contexts. In this study, we compared genome-wide maps of 6-4PP and CPD lesion abundances in primary cells and conducted comprehensive analyses to determine the genetic and epigenetic features associated with susceptibility. Overall, we found a high degree of similarity between 6-4PP and CPD formation, with an enrichment of both in heterochromatin regions. However, when examining the relative levels of the two UV lesions, we found that bivalent and Polycomb-repressed chromatin states were uniquely more susceptible to 6-4PPs. Interestingly, when comparing UV susceptibility and repair with melanoma mutation frequency in these regions, disparate patterns were observed in that susceptibility was not always inversely associated with repair and mutation frequency. Functional enrichment analysis hint at mechanisms of negative selection for these regions that are essential for cell viability, immune function and induce cell death when mutated. Ultimately, these results reveal both the similarities and differences between UV-induced lesions that contribute to melanoma.


Assuntos
Reparo do DNA , Epigênese Genética/efeitos da radiação , Melanoma/genética , Mutação , Neoplasias Cutâneas/genética , Raios Ultravioleta/efeitos adversos , Dano ao DNA , Bases de Dados Genéticas , Eucromatina/química , Eucromatina/metabolismo , Eucromatina/efeitos da radiação , Fibroblastos/citologia , Fibroblastos/metabolismo , Fibroblastos/efeitos da radiação , Genoma Humano/efeitos da radiação , Heterocromatina/química , Heterocromatina/metabolismo , Heterocromatina/efeitos da radiação , Histonas/genética , Histonas/metabolismo , Humanos , Melanoma/etiologia , Melanoma/metabolismo , Melanoma/patologia , Mutagênese , Proteínas do Grupo Polycomb/genética , Proteínas do Grupo Polycomb/metabolismo , Cultura Primária de Células , Dímeros de Pirimidina/agonistas , Dímeros de Pirimidina/metabolismo , Neoplasias Cutâneas/etiologia , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia
4.
Nat Genet ; 52(11): 1178-1188, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33020667

RESUMO

Somatic mutations in driver genes may ultimately lead to the development of cancer. Understanding how somatic mutations accumulate in cancer genomes and the underlying factors that generate somatic mutations is therefore crucial for developing novel therapeutic strategies. To understand the interplay between spatial genome organization and specific mutational processes, we studied 3,000 tumor-normal-pair whole-genome datasets from 42 different human cancer types. Our analyses reveal that the change in somatic mutational load in cancer genomes is co-localized with topologically-associating-domain boundaries. Domain boundaries constitute a better proxy to track mutational load change than replication timing measurements. We show that different mutational processes lead to distinct somatic mutation distributions where certain processes generate mutations in active domains, and others generate mutations in inactive domains. Overall, the interplay between three-dimensional genome organization and active mutational processes has a substantial influence on the large-scale mutation-rate variations observed in human cancers.


Assuntos
Cromatina/química , Genoma Humano , Mutação , Neoplasias/genética , Linhagem Celular Tumoral , Cromossomos Humanos X/genética , Reparo de Erro de Pareamento de DNA , Análise Mutacional de DNA , DNA de Neoplasias , Conjuntos de Dados como Assunto , Feminino , Humanos , Masculino , Conformação Proteica , Domínios Proteicos , Dobramento de Proteína , Inativação do Cromossomo X
5.
Mol Cell Biol ; 22(3): 856-65, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11784861

RESUMO

The retinoblastoma protein, pRb, controls transcription through recruitment of histone deacetylase to particular E2F-responsive genes. We determined the acetylation level of individual nucleosomes present in the cyclin E promoter of RB(+/+) and RB(-/-) mouse embryo fibroblasts. We also determined the effects of pRb on nucleosomal conformation by examining the thiol reactivity of histone H3 of individual nucleosomes. We found that pRb represses the cyclin E promoter through histone deacetylation of a single nucleosome, to which it and histone deacetylase 1 bind. In addition, the conformation of this nucleosome is modulated by pRb-directed histone deacetylase activity. Thus, the repressive role of pRb in cyclin E transcription and therefore cell cycle progression can be mapped to its control of the acetylation status and conformation of a single nucleosome.


Assuntos
Histonas/metabolismo , Nucleossomos/metabolismo , Proteína do Retinoblastoma/metabolismo , Acetilação , Animais , Células Cultivadas , Mapeamento Cromossômico , Ciclina E/genética , Histona Desacetilases/metabolismo , Histonas/química , Camundongos , Camundongos Knockout , Regiões Promotoras Genéticas , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Proteína do Retinoblastoma/deficiência , Proteína do Retinoblastoma/genética , Transcrição Gênica
6.
Mol Cell Biol ; 23(23): 8626-36, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14612406

RESUMO

The linker histone H1 is involved in maintaining higher-order chromatin structures and displays dynamic nuclear mobility, which may be regulated by posttranslational modifications. To analyze the effect of H1 tail phosphorylation on the modulation of the histone's nuclear dynamics, we generated a mutant histone H1, referred to as M1-5, in which the five cyclin-dependent kinase phosphorylation consensus sites were mutated from serine or threonine residues into alanines. Cyclin E/CDK2 or cyclin A/CDK2 cannot phosphorylate the mutant in vitro. Using the technique of fluorescence recovery after photobleaching, we observed that the mobility of a green fluorescent protein (GFP)-M1-5 fusion protein is decreased compared to that of a GFP-wild-type H1 fusion protein. In addition, recovery of H1 correlated with CDK2 activity, as GFP-H1 mobility was decreased in cells with low CDK2 activity. Blocking the activity of CDK2 by p21 expression decreased the mobility of GFP-H1 but not that of GFP-M1-5. Finally, the level and rate of recovery of cyan fluorescent protein (CFP)-M1-5 were lower than those of CFP-H1 specifically in heterochromatic regions. These data suggest that CDK2 phosphorylates histone H1 in vivo, resulting in a more open chromatin structure by destabilizing H1-chromatin interactions.


Assuntos
Quinases Ciclina-Dependentes/metabolismo , Ciclinas/metabolismo , Histonas/metabolismo , Sítios de Ligação/genética , Quinases relacionadas a CDC2 e CDC28/metabolismo , Linhagem Celular , Ciclina A/metabolismo , Ciclina E/metabolismo , Quinase 2 Dependente de Ciclina , Proteínas de Fluorescência Verde , Células HeLa , Heterocromatina/metabolismo , Histonas/genética , Humanos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Mutagênese Sítio-Dirigida , Fosforilação , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
7.
Mol Endocrinol ; 17(8): 1543-54, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12730327

RESUMO

Repression of the transcriptional activities of the estrogen receptor (ER) is a main goal in the treatment of breast cancer. The antiestrogen tamoxifen is an effective therapy for breast cancer patients because it inhibits estrogen-stimulated gene expression and cell proliferation. Previous studies have implicated a complex containing the nuclear receptor corepressor (N-CoR) in the mechanism by which tamoxifen represses ER-mediated transcriptional activity. In the present study a truncated N-CoR construct was used to inhibit endogenous N-CoR activity in an ER-positive breast cancer cell line. This dominant-negative N-CoR was successful in relieving repression conferred by the unliganded retinoic acid receptor, but it failed to affect the transcriptional activity of the ER in the presence of tamoxifen. Correspondingly, the histone acetylation levels of nucleosomes on endogenous estrogen-responsive genes were unaltered in cells expressing the N-CoR dominant-negative, regardless of ligand. In addition, in vitro cell proliferation and in vivo tumor growth were unchanged in cells that express dominant-negative N-CoR. In conclusion, these results may reveal that N-CoR affects tamoxifen-liganded ER in a manner distinct from its influence on retinoic acid receptor-mediated transcriptional activity or that corepressors other than N-CoR may be involved in the ability of tamoxifen to repress estrogen-responsive transcription and tumor growth.


Assuntos
Proteínas Nucleares/metabolismo , Receptores de Estrogênio/genética , Receptores do Ácido Retinoico/genética , Proteínas Repressoras/metabolismo , Moduladores Seletivos de Receptor Estrogênico/metabolismo , Tamoxifeno/metabolismo , Animais , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Divisão Celular/efeitos dos fármacos , Divisão Celular/genética , Estrogênios/farmacologia , Feminino , Regulação Neoplásica da Expressão Gênica , Genes Dominantes , Humanos , Camundongos , Camundongos Nus , Mutação , Proteínas Nucleares/genética , Correpressor 1 de Receptor Nuclear , Receptores de Estrogênio/agonistas , Receptores de Estrogênio/antagonistas & inibidores , Receptores do Ácido Retinoico/metabolismo , Proteínas Repressoras/genética , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Tamoxifeno/farmacologia , Transcrição Gênica , Células Tumorais Cultivadas
8.
Mol Cell ; 22(5): 693-9, 2006 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-16762841

RESUMO

Two key components of mammalian heterochromatin that play a structural role in higher order chromatin organization are the heterochromatin protein 1alpha (HP1alpha) and the linker histone H1. Here, we show that these proteins interact in vivo and in vitro through their hinge and C-terminal domains, respectively. The phosphorylation of H1 by CDK2, which is required for efficient cell cycle progression, disrupts this interaction. We propose that phosphorylation of H1 provides a signal for the disassembly of higher order chromatin structures during interphase, independent of histone H3-lysine 9 (H3-K9) methylation, by reducing the affinity of HP1alpha for heterochromatin.


Assuntos
Proteínas Cromossômicas não Histona/metabolismo , Quinase 2 Dependente de Ciclina/metabolismo , Histonas/metabolismo , Animais , Sítios de Ligação , Cromatina/genética , Cromatina/metabolismo , Homólogo 5 da Proteína Cromobox , Proteínas Cromossômicas não Histona/genética , Quinase 2 Dependente de Ciclina/genética , Imunofluorescência , Regulação da Expressão Gênica , Humanos , Interfase , Camundongos , Células NIH 3T3 , Fosforilação , Ligação Proteica , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA