Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros

Bases de datos
Tipo de estudio
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
EMBO J ; 30(16): 3383-96, 2011 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-21750527

RESUMEN

Genomic instability due to telomere dysfunction and defective repair of DNA double-strand breaks (DSBs) is an underlying cause of ageing-related diseases. 53BP1 is a key factor in DNA DSBs repair and its deficiency is associated with genomic instability and cancer progression. Here, we uncover a novel pathway regulating the stability of 53BP1. We demonstrate an unprecedented role for the cysteine protease Cathepsin L (CTSL) in the degradation of 53BP1. Overexpression of CTSL in wild-type fibroblasts leads to decreased 53BP1 protein levels and changes in its cellular distribution, resulting in defective repair of DNA DSBs. Importantly, we show that the defects in DNA repair associated with 53BP1 deficiency upon loss of A-type lamins are due to upregulation of CTSL. Furthermore, we demonstrate that treatment with vitamin D stabilizes 53BP1 and promotes DNA DSBs repair via inhibition of CTSL, providing an as yet unsuspected link between vitamin D action and DNA repair. Given that CTSL upregulation is a hallmark of cancer and progeria, regulation of this pathway could be of great therapeutic significance for these diseases.


Asunto(s)
Catepsina L/fisiología , Proteínas Cromosómicas no Histona/metabolismo , Reparación del ADN/fisiología , Proteínas de Unión al ADN/metabolismo , Lamina Tipo A/fisiología , Vitamina D/fisiología , Animales , Calcitriol/farmacología , Catepsina L/antagonistas & inhibidores , Catepsina L/biosíntesis , Catepsina L/genética , Línea Celular , Precursores Enzimáticos/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Lamina Tipo A/deficiencia , Lamina Tipo A/genética , Leupeptinas/farmacología , Ratones , Complejo de la Endopetidasa Proteasomal/metabolismo , Estabilidad Proteica , Proteínas Recombinantes de Fusión/fisiología , Especificidad de la Especie , Transfección , Proteína 1 de Unión al Supresor Tumoral P53
2.
EMBO J ; 28(16): 2414-27, 2009 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-19629036

RESUMEN

A-type lamins are intermediate filament proteins that provide a scaffold for protein complexes regulating nuclear structure and function. Mutations in the LMNA gene are linked to a variety of degenerative disorders termed laminopathies, whereas changes in the expression of lamins are associated with tumourigenesis. The molecular pathways affected by alterations of A-type lamins and how they contribute to disease are poorly understood. Here, we show that A-type lamins have a key role in the maintenance of telomere structure, length and function, and in the stabilization of 53BP1, a component of the DNA damage response (DDR) pathway. Loss of A-type lamins alters the nuclear distribution of telomeres and results in telomere shortening, defects in telomeric heterochromatin, and increased genomic instability. In addition, A-type lamins are necessary for the processing of dysfunctional telomeres by non-homologous end joining, putatively through stabilization of 53BP1. This study shows new functions for A-type lamins in the maintenance of genomic integrity, and suggests that alterations of telomere biology and defects in DDR contribute to the pathogenesis of lamin-related diseases.


Asunto(s)
Reparación del ADN , Lamina Tipo A/genética , Lamina Tipo A/metabolismo , Telómero/metabolismo , Animales , Línea Celular , Núcleo Celular/química , Núcleo Celular/metabolismo , Proteínas Cromosómicas no Histona , Proteínas de Unión al ADN , Fibroblastos/citología , Fibroblastos/metabolismo , Eliminación de Gen , Inestabilidad Genómica , Péptidos y Proteínas de Señalización Intracelular/análisis , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Telómero/química , Proteína 1 de Unión al Supresor Tumoral P53
3.
Cell Cycle ; 12(23): 3629-39, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24091731

RESUMEN

BRCA1 and 53BP1 play decisive roles in the choice of DNA double-strand break repair mechanisms. BRCA1 promotes DNA end resection and homologous recombination (HR) during S/G 2 phases of the cell cycle, while 53BP1 inhibits end resection and facilitates non-homologous end-joining (NHEJ), primarily during G 1. This competitive relationship is critical for genome integrity during cell division. However, their relationship in the many cells in our body that are not cycling is unknown. We discovered profound differences in 53BP1 and BRCA1 regulation between cycling and non-cycling cells. Cellular growth arrest results in transcriptional downregulation of BRCA1 and activation of cathepsin-L (CTSL)-mediated degradation of 53BP1. Accordingly, growth-arrested cells do not form BRCA1 or 53BP1 ionizing radiation-induced foci (IRIF). Interestingly, cell cycle re-entry reverts this scenario, with upregulation of BRCA1, downregulation of CTSL, stabilization of 53BP1, and 53BP1 IRIF formation throughout the cycle, indicating that BRCA1 and 53BP1 are important in replicating cells and dispensable in non-cycling cells. We show that CTSL-mediated degradation of 53BP1, previously associated with aggressive breast cancers, is an endogenous mechanism of non-cycling cells to balance NHEJ (53BP1) and HR (BRCA1). Breast cancer cells exploit this mechanism to ensure genome stability and viability, providing an opportunity for targeted therapy.


Asunto(s)
Proteína BRCA1/metabolismo , Regulación de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteína BRCA1/genética , Catepsina L/antagonistas & inhibidores , Catepsina L/genética , Catepsina L/metabolismo , Puntos de Control del Ciclo Celular/efectos de la radiación , Línea Celular , Roturas del ADN de Doble Cadena/efectos de la radiación , Reparación del ADN por Unión de Extremidades , Replicación del ADN/efectos de los fármacos , Células HeLa , Humanos , Hidroxiurea/farmacología , Péptidos y Proteínas de Señalización Intracelular/genética , Leucina/análogos & derivados , Leucina/farmacología , Células MCF-7 , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Radiación Ionizante , Proteína 1 de Unión al Supresor Tumoral P53
4.
Nucleus ; 4(5): 410-9, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24153156

RESUMEN

Over 300 mutations in the LMNA gene, encoding A-type lamins, are associated with 15 human degenerative disorders and premature aging syndromes. Although genomic instability seems to contribute to the pathophysiology of some laminopathies, there is limited information about what mutations cause genomic instability and by which molecular mechanisms. Mouse embryonic fibroblasts depleted of A-type lamins or expressing mutants lacking exons 8-11 (Lmna(Δ8-11/Δ8-11)) exhibit alterations in telomere biology and DNA repair caused by cathepsin L-mediated degradation of 53BP1 and reduced expression of BRCA1 and RAD51. Thus, a region encompassing exons 8-11 seems essential for genome integrity. Given that deletion of lamin A exon 9 in the mouse (Lmna(Δ9/Δ9)) results in a progeria phenotype, we tested if this domain is important for genome integrity. Lmna(Δ9/Δ9) MEFs exhibit telomere shortening and heterochromatin alterations but do not activate cathepsin L-mediated degradation of 53BP1 and maintain expression of BRCA1 and RAD51. Accordingly, Lmna(Δ9/Δ9) MEFs do not present genomic instability, and expression of mutant lamin A Δexon9 in lamin-depleted cells restores DNA repair factors levels and partially rescues nuclear abnormalities. These data reveal that the domain encoded by exon 9 is important to maintain telomere homeostasis and heterochromatin structure but does not play a role in DNA repair, thus pointing to other exons in the lamin A tail as responsible for the genomic instability phenotype in Lmna(Δ8-11/Δ8-11) mice. Our study also suggests that the levels of DNA repair factors 53BP1, BRCA1 and RAD51 could potentially serve as biomarkers to identify laminopathies that present with genomic instability.


Asunto(s)
Cromatina/genética , Exones/genética , Inestabilidad Genómica/genética , Lamina Tipo A/genética , Eliminación de Secuencia/genética , Telómero/genética , Animales , Proteína BRCA1/metabolismo , Línea Celular , Cromatina/química , Cromatina/patología , Proteínas Cromosómicas no Histona/metabolismo , Reparación del ADN/genética , Proteínas de Unión al ADN/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Heterocromatina/química , Heterocromatina/genética , Heterocromatina/metabolismo , Humanos , Ratones , Recombinasa Rad51/metabolismo , Telómero/patología , Acortamiento del Telómero/genética , Proteína 1 de Unión al Supresor Tumoral P53
5.
J Cell Biol ; 200(2): 187-202, 2013 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-23337117

RESUMEN

Loss of 53BP1 rescues BRCA1 deficiency and is associated with BRCA1-deficient and triple-negative breast cancers (TNBC) and with resistance to genotoxic drugs. The mechanisms responsible for decreased 53BP1 transcript and protein levels in tumors remain unknown. Here, we demonstrate that BRCA1 loss activates cathepsin L (CTSL)-mediated degradation of 53BP1. Activation of this pathway rescued homologous recombination repair and allowed BRCA1-deficient cells to bypass growth arrest. Importantly, depletion or inhibition of CTSL with vitamin D or specific inhibitors stabilized 53BP1 and increased genomic instability in response to radiation and poly(adenosine diphosphate-ribose) polymerase inhibitors, compromising proliferation. Analysis of human breast tumors identified nuclear CTSL as a positive biomarker for TNBC, which correlated inversely with 53BP1. Importantly, nuclear levels of CTSL, vitamin D receptor, and 53BP1 emerged as a novel triple biomarker signature for stratification of patients with BRCA1-mutated tumors and TNBC, with potential predictive value for drug response. We identify here a novel pathway with prospective relevance for diagnosis and customization of breast cancer therapy.


Asunto(s)
Proteína BRCA1/genética , Neoplasias de la Mama/genética , Catepsina L/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Catepsina L/genética , Línea Celular Tumoral , Reparación del ADN/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Inestabilidad Genómica , Mutación de Línea Germinal , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteína 1 de Unión al Supresor Tumoral P53
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA