Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
EMBO J ; 36(19): 2829-2843, 2017 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-28814448

RESUMEN

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.


Asunto(s)
Carcinógenos/toxicidad , Transformación Celular Neoplásica , Resistencia a Medicamentos/genética , Inestabilidad Genómica/efectos de los fármacos , Inestabilidad Genómica/genética , Mutagénesis , Secuencia de Bases/fisiología , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/genética , Células Cultivadas , Daño del ADN , Resistencia a Medicamentos/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Humanos , Melanoma/etiología , Melanoma/genética , Mutagénesis/efectos de los fármacos , Mutagénesis/genética , Neoplasias Cutáneas/etiología , Neoplasias Cutáneas/genética , Rayos Ultravioleta , Melanoma Cutáneo Maligno
2.
Life Sci Alliance ; 5(4)2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34983823

RESUMEN

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.


Asunto(s)
Carcinogénesis , Carcinógenos/farmacología , Neoplasias , Proteínas de Unión a Retinoblastoma/genética , Ubiquitina-Proteína Ligasas/genética , Sistemas CRISPR-Cas , Carcinogénesis/efectos de los fármacos , Carcinogénesis/genética , Línea Celular , Técnicas de Inactivación de Genes , Predisposición Genética a la Enfermedad/genética , Humanos , Mutación/genética , Neoplasias/genética , Neoplasias/patología , Oncogenes/genética
3.
Mutat Res ; 823: 111758, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34333390

RESUMEN

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.


Asunto(s)
Reparación del ADN , Epigénesis Genética/efectos de la radiación , Melanoma/genética , Mutación , Neoplasias Cutáneas/genética , Rayos Ultravioleta/efectos adversos , Daño del ADN , Bases de Datos Genéticas , Eucromatina/química , Eucromatina/metabolismo , Eucromatina/efectos de la radiación , Fibroblastos/citología , Fibroblastos/metabolismo , Fibroblastos/efectos de la radiación , Genoma Humano/efectos de la radiación , Heterocromatina/química , Heterocromatina/metabolismo , Heterocromatina/efectos de la radiación , Histonas/genética , Histonas/metabolismo , Humanos , Melanoma/etiología , Melanoma/metabolismo , Melanoma/patología , Mutagénesis , Proteínas del Grupo Polycomb/genética , Proteínas del Grupo Polycomb/metabolismo , Cultivo Primario de Células , Dímeros de Pirimidina/agonistas , Dímeros de Pirimidina/metabolismo , Neoplasias Cutáneas/etiología , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología
4.
Nat Genet ; 52(11): 1178-1188, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33020667

RESUMEN

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.


Asunto(s)
Cromatina/química , Genoma Humano , Mutación , Neoplasias/genética , Línea Celular Tumoral , Cromosomas Humanos X/genética , Reparación de la Incompatibilidad de ADN , Análisis Mutacional de ADN , ADN de Neoplasias , Conjuntos de Datos como Asunto , Femenino , Humanos , Masculino , Conformación Proteica , Dominios Proteicos , Pliegue de Proteína , Inactivación del Cromosoma X
5.
Mol Cell Biol ; 22(3): 856-65, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11784861

RESUMEN

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.


Asunto(s)
Histonas/metabolismo , Nucleosomas/metabolismo , Proteína de Retinoblastoma/metabolismo , Acetilación , Animales , Células Cultivadas , Mapeo Cromosómico , Ciclina E/genética , Histona Desacetilasas/metabolismo , Histonas/química , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Proteína de Retinoblastoma/deficiencia , Proteína de Retinoblastoma/genética , Transcripción Genética
6.
Mol Cell Biol ; 23(23): 8626-36, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14612406

RESUMEN

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.


Asunto(s)
Quinasas Ciclina-Dependientes/metabolismo , Ciclinas/metabolismo , Histonas/metabolismo , Sitios de Unión/genética , Quinasas CDC2-CDC28/metabolismo , Línea Celular , Ciclina A/metabolismo , Ciclina E/metabolismo , Quinasa 2 Dependiente de la Ciclina , Proteínas Fluorescentes Verdes , Células HeLa , Heterocromatina/metabolismo , Histonas/genética , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Mutagénesis Sitio-Dirigida , Fosforilación , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo
7.
Mol Endocrinol ; 17(8): 1543-54, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12730327

RESUMEN

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.


Asunto(s)
Proteínas Nucleares/metabolismo , Receptores de Estrógenos/genética , Receptores de Ácido Retinoico/genética , Proteínas Represoras/metabolismo , Moduladores Selectivos de los Receptores de Estrógeno/metabolismo , Tamoxifeno/metabolismo , Animales , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , División Celular/efectos de los fármacos , División Celular/genética , Estrógenos/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica , Genes Dominantes , Humanos , Ratones , Ratones Desnudos , Mutación , Proteínas Nucleares/genética , Co-Represor 1 de Receptor Nuclear , Receptores de Estrógenos/agonistas , Receptores de Estrógenos/antagonistas & inhibidores , Receptores de Ácido Retinoico/metabolismo , Proteínas Represoras/genética , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Tamoxifeno/farmacología , Transcripción Genética , Células Tumorales Cultivadas
8.
Mol Cell ; 22(5): 693-9, 2006 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-16762841

RESUMEN

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.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Quinasa 2 Dependiente de la Ciclina/metabolismo , Histonas/metabolismo , Animales , Sitios de Unión , Cromatina/genética , Cromatina/metabolismo , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/genética , Quinasa 2 Dependiente de la Ciclina/genética , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica , Humanos , Interfase , Ratones , Células 3T3 NIH , Fosforilación , Unión Proteica , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA