Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
NAR Cancer ; 5(3): zcad043, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37609662

RESUMEN

Poly(ADP-ribosylation) (PARylation) by poly(ADP-ribose) polymerases (PARPs) is a highly regulated process that consists of the covalent addition of polymers of ADP-ribose (PAR) through post-translational modifications of substrate proteins or non-covalent interactions with PAR via PAR binding domains and motifs, thereby reprogramming their functions. This modification is particularly known for its central role in the maintenance of genomic stability. However, how genomic integrity is controlled by an intricate interplay of covalent PARylation and non-covalent PAR binding remains largely unknown. Of importance, PARylation has caught recent attention for providing a mechanistic basis of synthetic lethality involving PARP inhibitors (PARPi), most notably in homologous recombination (HR)-deficient breast and ovarian tumors. The molecular mechanisms responsible for the anti-cancer effect of PARPi are thought to implicate both catalytic inhibition and trapping of PARP enzymes on DNA. However, the relative contribution of each on tumor-specific cytotoxicity is still unclear. It is paramount to understand these PAR-dependent mechanisms, given that resistance to PARPi is a challenge in the clinic. Deciphering the complex interplay between covalent PARylation and non-covalent PAR binding and defining how PARP trapping and non-trapping events contribute to PARPi anti-tumour activity is essential for developing improved therapeutic strategies. With this perspective, we review the current understanding of PARylation biology in the context of the DNA damage response (DDR) and the mechanisms underlying PARPi activity and resistance.

2.
Nat Commun ; 14(1): 381, 2023 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-36693839

RESUMEN

Fanconi Anemia (FA) is a rare, genome instability-associated disease characterized by a deficiency in repairing DNA crosslinks, which are known to perturb several cellular processes, including DNA transcription, replication, and repair. Formaldehyde, a by-product of metabolism, is thought to drive FA by generating DNA interstrand crosslinks (ICLs) and DNA-protein crosslinks (DPCs). However, the impact of formaldehyde on global cellular pathways has not been investigated thoroughly. Herein, using a pangenomic CRISPR-Cas9 screen, we identify EXO1 as a critical regulator of formaldehyde-induced DNA lesions. We show that EXO1 knockout cell lines exhibit formaldehyde sensitivity leading to the accumulation of replicative stress, DNA double-strand breaks, and quadriradial chromosomes, a typical feature of FA. After formaldehyde exposure, EXO1 is recruited to chromatin, protects DNA replication forks from degradation, and functions in parallel with the FA pathway to promote cell survival. In vitro, EXO1-mediated exonuclease activity is proficient in removing DPCs. Collectively, we show that EXO1 limits replication stress and DNA damage to counteract formaldehyde-induced genome instability.


Asunto(s)
Sistemas CRISPR-Cas , Tolerancia a Medicamentos , Exodesoxirribonucleasas , Anemia de Fanconi , Formaldehído , Humanos , ADN , Daño del ADN/efectos de los fármacos , Daño del ADN/genética , Reparación del ADN/efectos de los fármacos , Reparación del ADN/genética , Enzimas Reparadoras del ADN/genética , Enzimas Reparadoras del ADN/metabolismo , Replicación del ADN/efectos de los fármacos , Replicación del ADN/genética , Exodesoxirribonucleasas/genética , Exodesoxirribonucleasas/metabolismo , Anemia de Fanconi/inducido químicamente , Anemia de Fanconi/genética , Formaldehído/toxicidad , Inestabilidad Genómica/efectos de los fármacos , Inestabilidad Genómica/genética , Tolerancia a Medicamentos/genética
3.
Elife ; 112022 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-36269050

RESUMEN

The tumour suppressor PALB2 stimulates RAD51-mediated homologous recombination (HR) repair of DNA damage, whilst its steady-state association with active genes protects these loci from replication stress. Here, we report that the lysine acetyltransferases 2A and 2B (KAT2A/2B, also called GCN5/PCAF), two well-known transcriptional regulators, acetylate a cluster of seven lysine residues (7K-patch) within the PALB2 chromatin association motif (ChAM) and, in this way, regulate context-dependent PALB2 binding to chromatin. In unperturbed cells, the 7K-patch is targeted for KAT2A/2B-mediated acetylation, which in turn enhances the direct association of PALB2 with nucleosomes. Importantly, DNA damage triggers a rapid deacetylation of ChAM and increases the overall mobility of PALB2. Distinct missense mutations of the 7K-patch render the mode of PALB2 chromatin binding, making it either unstably chromatin-bound (7Q) or randomly bound with a reduced capacity for mobilisation (7R). Significantly, both of these mutations confer a deficiency in RAD51 foci formation and increase DNA damage in S phase, leading to the reduction of overall cell survival. Thus, our study reveals that acetylation of the ChAM 7K-patch acts as a molecular switch to enable dynamic PALB2 shuttling for HR repair while protecting active genes during DNA replication.


Asunto(s)
Cromatina , Proteínas Supresoras de Tumor , Acetilación , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Reparación del ADN , Daño del ADN , Nucleosomas
4.
JCO Precis Oncol ; 6: e2100450, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35442721

RESUMEN

PURPOSE: The BRCA2 p.K3326* variant is considered a low-penetrance variant for breast cancer. Aldehydes that accumulate in cells under insufficient aldehyde oxidation were most recently shown to trigger carcinogenesis by promoting depletion of BRCA2 protein. Allele T of the common variant rs10744777 in the ALDH2 gene was associated with reduced expression of aldehyde dehydrogenase, the main enzyme in aldehyde oxidation. We hypothesized that this allele could modify breast cancer risk in women with the BRCA2 p.K3326* low-penetrance variant through reduced function of ALDH2, increased accumulation of cellular aldehydes, and depletion of BRCA2 protein. MATERIALS AND METHODS: We genotyped 11,873 Polish women diagnosed with breast cancer and 7,615 ethnically matched controls for these two variants. Next, we extended our analysis of rs10744777 to 231 carriers of pathogenic BRCA2 mutations. RESULTS: BRCA2 p.K3326* variant was associated with significant increase in breast cancer risk only in those who were homozygous for the T allele of the ALDH2 rs10744777 variant (odds ratio = 1.72; 95% CI, 1.19 to 2.48; P = .003). The BRCA2 p.K3326* variant did not increase the risk of breast cancer among those who were heterozygous or homozygous for the C allele of the ALDH2 rs10744777 variant (odds ratio = 1.05; 95% CI, 0.73 to 1.51; P = .81). In the carriers of high-risk BRCA2 mutations, the TT genotype of rs10744777 conferred a modest (18%) and not significant increase in breast cancer risk. CONCLUSION: Our results suggest that BRCA2 p.K3326* variant, which is low-penetrance by itself, confers increased breast cancer risk on the background of the TT genotype of the ALDH2 rs10744777 variant in the Polish population.


Asunto(s)
Proteína BRCA2 , Neoplasias de la Mama , Aldehído Deshidrogenasa Mitocondrial/genética , Aldehídos , Proteína BRCA2/genética , Neoplasias de la Mama/epidemiología , Femenino , Predisposición Genética a la Enfermedad/genética , Humanos , Polimorfismo de Nucleótido Simple/genética
5.
Trends Cancer ; 7(3): 188-197, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33139182

RESUMEN

Since its discovery, partner and localizer of breast cancer 2 (BRCA2) (PALB2) has emerged as a major tumor suppressor gene linked to breast cancer (BC), pancreatic cancer (PC), and ovarian cancer (OC) susceptibility. Its protein product plays a pivotal role in the maintenance of genome integrity. Here we discuss the first functional evaluation of a large set of PALB2 missense variants of uncertain significance (VUSs). Assessment of 136 VUSs interrogating a range of PALB2 biological functions resulted in the identification of 15 variants with consistent loss of function across different assays. All loss-of-function variants are located at the PALB2 coiled coil (CC) or at the WD40 domain, highlighting the importance of modular domains mechanistically involved in the DNA damage response (DDR) and pinpointing their roles in tumor suppression.


Asunto(s)
Proteína del Grupo de Complementación N de la Anemia de Fanconi/genética , Genes Supresores de Tumor , Predisposición Genética a la Enfermedad , Neoplasias/genética , Humanos , Mutación con Pérdida de Función , Mutación Missense , Dominios Proteicos/genética , Reparación del ADN por Recombinación
6.
Fam Cancer ; 19(2): 123-131, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32048105

RESUMEN

The PALB2 protein is essential to RAD51-mediated homologous recombination (HR) repair. Germline monoallelic PALB2 pathogenic variants confer significant risks for breast cancer. However, the majority of PALB2 variants remain classified as variants of unknown significance (VUS). We aim to functionally and mechanistically evaluate three novel PALB2 VUS. Patient-derived lymphoblastoid cell lines containing the VUS were analyzed for nuclear localization and foci formation of RAD51 as a measure of HR efficiency. To understand the mechanism underlying the HR deficiency, PALB2 nuclear localization was assessed using immunofluorescence studies. Among these VUS, c.3251C>T (p.Ser1084Leu) occurred in a patient with metastatic breast cancer while c.1054G>C (p.Glu352Gln) and c.1057A>G (p.Lys353Glu) were seen in patients with squamous cell carcinoma of skin and renal cell carcinoma respectively. Variant c.3251C>T was located within the WD40 domain which normally masked the nuclear export signal sequence responsible for nuclear delocalization of PALB2. Correspondingly, c.3251C>T displayed aberrant cytoplasmic localization of PALB2 which led to an impaired RAD51 nuclear localization and foci formation. On the other hand, both c.1054G>C and c.1057A>G showed intact HR functions and nuclear localization of PALB2, consistent with their locations within domains of no known function. Additionally, the prevalence of c.1054G>C was similar among healthy controls and patients with breast cancer (as seen in other studies), suggestive of its non-pathogenicity. In conclusion, our studies provided the functional evidence showing the deleterious effect of c.3251C>T, and non-deleterious effects of c.1054G>C and c.1057A>G. Using the ClinGen Pathogenicity calculator, c.3251C>T remains a VUS while c.1054G>C and c.1057A>G may be classified as likely benign variants.


Asunto(s)
Núcleo Celular/metabolismo , Proteína del Grupo de Complementación N de la Anemia de Fanconi/genética , Mutación de Línea Germinal , Mutación Missense , Neoplasias/genética , Recombinasa Rad51/metabolismo , Adulto , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Carcinoma Ductal de Mama/genética , Carcinoma Ductal de Mama/metabolismo , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/metabolismo , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Citoplasma/metabolismo , Proteína del Grupo de Complementación N de la Anemia de Fanconi/metabolismo , Femenino , Variación Genética , Humanos , Neoplasias Renales/genética , Neoplasias Renales/metabolismo , Masculino , Persona de Mediana Edad , Neoplasias/metabolismo , Linaje , Reparación del ADN por Recombinación , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo
7.
Genet Med ; 22(3): 622-632, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31636395

RESUMEN

PURPOSE: Inherited pathogenic variants in PALB2 are associated with increased risk of breast and pancreatic cancer. However, the functional and clinical relevance of many missense variants of uncertain significance (VUS) identified through clinical genetic testing is unclear. The ability of patient-derived germline missense VUS to disrupt PALB2 function was assessed to identify variants with potential clinical relevance. METHODS: The influence of 84 VUS on PALB2 function was evaluated using a cellular homology directed DNA repair (HDR) assay and VUS impacting activity were further characterized using secondary functional assays. RESULTS: Four (~5%) variants (p.L24S,c.71T>C; p.L35P,c.104T>C; pI944N,c.2831T>A; and p.L1070P,c.3209T>C) disrupted PALB2-mediated HDR activity. These variants conferred sensitivity to cisplatin and a poly(ADP-ribose) polymerase (PARP) inhibitor and reduced RAD51 foci formation in response to DNA damage. The p.L24S and p.L35P variants disrupted BRCA1-PALB2 protein complexes, p.I944N was associated with protein instability, and both p.I944N and p.L1070P mislocalized PALB2 to the cytoplasm. CONCLUSION: These findings show that the HDR assay is an effective method for screening the influence of inherited variants on PALB2 function, that four missense variants impact PALB2 function and may influence cancer risk and response to therapy, and suggest that few inherited PALB2 missense variants disrupt PALB2 function in DNA repair.


Asunto(s)
Proteína BRCA1/genética , Neoplasias de la Mama/genética , Proteína del Grupo de Complementación N de la Anemia de Fanconi/genética , Recombinasa Rad51/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Daño del ADN/genética , Reparación del ADN/efectos de los fármacos , Femenino , Factor de Transcripción GATA3/genética , Predisposición Genética a la Enfermedad , Humanos , Mutación Missense/genética , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Reparación del ADN por Recombinación/genética
8.
Nat Commun ; 10(1): 5296, 2019 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-31757951

RESUMEN

Heterozygous carriers of germ-line loss-of-function variants in the DNA repair gene PALB2 are at a highly increased lifetime risk for developing breast cancer. While truncating variants in PALB2 are known to increase cancer risk, the interpretation of missense variants of uncertain significance (VUS) is in its infancy. Here we describe the development of a relatively fast and easy cDNA-based system for the semi high-throughput functional analysis of 48 VUS in human PALB2. By assessing the ability of PALB2 VUS to rescue the DNA repair and checkpoint defects in Palb2 knockout mouse embryonic stem (mES) cells, we identify various VUS in PALB2 that impair its function. Three VUS in the coiled-coil domain of PALB2 abrogate the interaction with BRCA1, whereas several VUS in the WD40 domain dramatically reduce protein stability. Thus, our functional assays identify damaging VUS in PALB2 that may increase cancer risk.


Asunto(s)
Neoplasias de la Mama/genética , Proteína del Grupo de Complementación N de la Anemia de Fanconi/genética , Técnicas Genéticas , Células Madre Embrionarias de Ratones/metabolismo , Proteínas Mutantes/metabolismo , Mutación Missense , Animales , ADN Complementario , Proteína del Grupo de Complementación N de la Anemia de Fanconi/metabolismo , Citometría de Flujo , Predisposición Genética a la Enfermedad , Inestabilidad Genómica , Humanos , Ratones , Ratones Noqueados
9.
Nucleic Acids Res ; 47(20): 10662-10677, 2019 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-31586400

RESUMEN

While biallelic mutations in the PALB2 tumor suppressor cause Fanconi anemia subtype FA-N, monoallelic mutations predispose to breast and familial pancreatic cancer. Although hundreds of missense variants in PALB2 have been identified in patients to date, only a few have clear functional and clinical relevance. Herein, we investigate the effects of 44 PALB2 variants of uncertain significance found in breast cancer patients and provide detailed analysis by systematic functional assays. Our comprehensive functional analysis reveals two hotspots for potentially deleterious variations within PALB2, one at each terminus. PALB2 N-terminus variants p.P8L [c.23C>T], p.Y28C [c.83A>G], and p.R37H [c.110G>A] compromised PALB2-mediated homologous recombination. At the C-terminus, PALB2 variants p.L947F [c.2841G>T], p.L947S [c.2840T>C], and most strikingly p.T1030I [c.3089C>T] and p.W1140G [c.3418T>C], stood out with pronounced PARP inhibitor sensitivity and cytoplasmic accumulation in addition to marked defects in recruitment to DNA damage sites, interaction with BRCA2 and homologous recombination. Altogether, our findings show that a combination of functional assays is necessary to assess the impact of germline missense variants on PALB2 function, in order to guide proper classification of their deleteriousness.


Asunto(s)
Neoplasias de la Mama/genética , Proteína del Grupo de Complementación N de la Anemia de Fanconi/genética , Mutación Missense/genética , Línea Celular Tumoral , Simulación por Computador , Daño del ADN , Femenino , Sitios Genéticos , Recombinación Homóloga/genética , Humanos , Cinética , Recombinasa Rad51/metabolismo , Reproducibilidad de los Resultados
10.
Nat Commun ; 10(1): 2954, 2019 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-31273204

RESUMEN

PARP-1 is rapidly recruited and activated by DNA double-strand breaks (DSBs). Upon activation, PARP-1 synthesizes a structurally complex polymer composed of ADP-ribose units that facilitates local chromatin relaxation and the recruitment of DNA repair factors. Here, we identify a function for PARP-1 in DNA DSB resection. Remarkably, inhibition of PARP-1 leads to hyperresected DNA DSBs. We show that loss of PARP-1 and hyperresection are associated with loss of Ku, 53BP1 and RIF1 resection inhibitors from the break site. DNA curtains analysis show that EXO1-mediated resection is blocked by PARP-1. Furthermore, PARP-1 abrogation leads to increased DNA resection tracks and an increase of homologous recombination in cellulo. Our results, therefore, place PARP-1 activation as a critical early event for DNA DSB repair activation and regulation of resection. Hence, our work has direct implications for the clinical use and effectiveness of PARP inhibition, which is prescribed for the treatment of various malignancies.


Asunto(s)
Roturas del ADN de Doble Cadena , ADN/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Animales , Cromatina/metabolismo , Técnicas de Silenciamiento del Gen , Células HeLa , Recombinación Homóloga/genética , Humanos , Ratones , Modelos Biológicos , Proteínas Nucleares/metabolismo , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Proteínas de Unión a Telómeros/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo
11.
Trends Biochem Sci ; 44(3): 226-240, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30638972

RESUMEN

Partner and Localizer of BRCA2 (PALB2) has emerged as an important and versatile player in genome integrity maintenance. Biallelic mutations in PALB2 cause Fanconi anemia (FA) subtype FA-N, whereas monoallelic mutations predispose to breast, and pancreatic familial cancers. Herein, we review recent developments in our understanding of the mechanisms of regulation of the tumor suppressor PALB2 and its functional domains. Regulation of PALB2 functions in DNA damage response and repair occurs on multiple levels, including homodimerization, phosphorylation, and ubiquitylation. With a molecular emphasis, we present PALB2-associated cancer mutations and their detailed analysis by functional assays.


Asunto(s)
Proteína BRCA2/metabolismo , Anemia de Fanconi/metabolismo , Animales , Proteína BRCA2/genética , Daño del ADN/genética , Anemia de Fanconi/genética , Femenino , Humanos , Mutación/genética , Ubiquitinación/genética , Ubiquitinación/fisiología
12.
Nucleic Acids Res ; 45(5): 2644-2657, 2017 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-28158555

RESUMEN

One typical mechanism to promote genomic instability, a hallmark of cancer, is to inactivate tumor suppressors, such as PALB2. It has recently been reported that mutations in PALB2 increase the risk of breast cancer by 8-9-fold by age 40 and the life time risk is ∼3-4-fold. To date, predicting the functional consequences of PALB2 mutations has been challenging as they lead to different cancer risks. Here, we performed a structure-function analysis of PALB2, using PALB2 truncated mutants (R170fs, L531fs, Q775X and W1038X), and uncovered a new mechanism by which cancer cells could drive genomic instability. Remarkably, the PALB2 W1038X mutant, harboring a mutation in its C-terminal domain, is still proficient in stimulating RAD51-mediated recombination in vitro, although it is unusually localized to the cytoplasm. After further investigation, we identified a hidden NES within the WD40 domain of PALB2 and found that the W1038X truncation leads to the exposure of this NES to CRM1, an export protein. This concept was also confirmed with another WD40-containing protein, RBBP4. Consequently, our studies reveal an unreported mechanism linking the nucleocytoplasmic translocation of PALB2 mutants to cancer formation.


Asunto(s)
Mutación , Neoplasias/genética , Proteínas Nucleares/genética , Proteínas Supresoras de Tumor/genética , Citoplasma/metabolismo , ADN/metabolismo , Proteína del Grupo de Complementación N de la Anemia de Fanconi , Células HEK293 , Humanos , Carioferinas/metabolismo , Señales de Exportación Nuclear , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Recombinasa Rad51/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Eliminación de Secuencia , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/metabolismo , Repeticiones WD40 , Proteína Exportina 1
13.
Mol Cell ; 65(2): 336-346, 2017 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-28089683

RESUMEN

ATR is a key regulator of cell-cycle checkpoints and homologous recombination (HR). Paradoxically, ATR inhibits CDKs during checkpoint responses, but CDK activity is required for efficient HR. Here, we show that ATR promotes HR after CDK-driven DNA end resection. ATR stimulates the BRCA1-PALB2 interaction after DNA damage and promotes PALB2 localization to DNA damage sites. ATR enhances BRCA1-PALB2 binding at least in part by inhibiting CDKs. The optimal interaction of BRCA1 and PALB2 requires phosphorylation of PALB2 at S59, an ATR site, and hypo-phosphorylation of S64, a CDK site. The PALB2-S59A/S64E mutant is defective for localization to DNA damage sites and HR, whereas the PALB2-S59E/S64A mutant partially bypasses ATR for its localization. Thus, HR is a biphasic process requiring both high-CDK and low-CDK periods. As exemplified by the regulation of PALB2 by ATR, ATR promotes HR by orchestrating a "CDK-to-ATR switch" post-resection, directly coupling the checkpoint to HR.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN por Recombinación , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Quinasas Ciclina-Dependientes/genética , Quinasas Ciclina-Dependientes/metabolismo , Proteína del Grupo de Complementación N de la Anemia de Fanconi , Células HeLa , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosforilación , Unión Proteica , Transducción de Señal , Factores de Tiempo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
14.
Nucleic Acids Res ; 44(22): 10879-10897, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27924011

RESUMEN

APRIN (PDS5 cohesin associated factor B) interacts with both the cohesin complex and the BRCA2 tumor suppressor. How APRIN influences cohesion and DNA repair processes is not well understood. Here, we show that APRIN is recruited to DNA damage sites. We find that APRIN interacts directly with RAD51, PALB2 and BRCA2. APRIN stimulates RAD51-mediated DNA strand invasion. APRIN also binds DNA with an affinity for D-loop structures and single-strand (ss) DNA. APRIN is a new homologous recombination (HR) mediator as it counteracts the RPA inhibitory effect on RAD51 loading to ssDNA. We show that APRIN strongly improves the annealing of complementary-strand DNA and that it can stimulate this process in synergy with BRCA2. Unlike cohesin constituents, its depletion has no impact on class switch recombination, supporting a specific role for this protein in HR. Furthermore, we show that low APRIN expression levels correlate with a better survival in ovarian cancer patients and that APRIN depletion sensitizes cells to the PARP inhibitor Olaparib in xenografted zebrafish. Our findings establish APRIN as an important and specific actor of HR, with cohesin-independent functions.


Asunto(s)
Biomarcadores de Tumor/fisiología , Proteínas de Unión al ADN/fisiología , Neoplasias Ováricas/metabolismo , Lesiones Intraepiteliales Escamosas de Cuello Uterino/metabolismo , Factores de Transcripción/fisiología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Antineoplásicos/farmacología , Proteína BRCA2/metabolismo , Bencimidazoles/farmacología , Biomarcadores de Tumor/química , Línea Celular Tumoral , Daño del ADN , Proteínas de Unión al ADN/química , Resistencia a Antineoplásicos , Proteína del Grupo de Complementación N de la Anemia de Fanconi , Femenino , Humanos , Estimación de Kaplan-Meier , Persona de Mediana Edad , Proteínas Nucleares/metabolismo , Neoplasias Ováricas/diagnóstico , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/mortalidad , Ftalazinas/farmacología , Piperazinas/farmacología , Unión Proteica , Transporte de Proteínas , Curva ROC , Recombinasa Rad51/metabolismo , Reparación del ADN por Recombinación , Lesiones Intraepiteliales Escamosas de Cuello Uterino/diagnóstico , Lesiones Intraepiteliales Escamosas de Cuello Uterino/tratamiento farmacológico , Lesiones Intraepiteliales Escamosas de Cuello Uterino/mortalidad , Factores de Transcripción/química , Proteínas Supresoras de Tumor/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Pez Cebra
15.
Sci Rep ; 6: 23302, 2016 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-27001483

RESUMEN

2-Ethylphenyl 4-(3-ethylureido)benzenesulfonate (SFOM-0046) is a novel anticancer agent that arrests cell cycle in S-phase and causes DNA replication stress leading to the phosphorylation of H2AX into γ-H2AX. First, using the M21, HT29, HT-1080 and HeLa cell lines, we confirmed that S-phase cell cycle arrest and γ-H2AX foci induction by SFOM-0046 is a general mechanism occurring in diverse cancer cell lines. In addition to γ-H2AX, SFOM-0046 activates preferentially ATR-Chk1 in M21 and HT29 cells while both ATR-Chk1 and ATM-Chk2 pathways are activated in HCT116 cells. Co-localization of SFOM-0046-induced 53BP1 foci with γ-H2AX foci validates that the DNA damage generated corresponds to double-strand-breaks (DSBs). Consistent with an S-phase arrest, SFOM-0046 treatment induces RAD51 foci formation but not DNA-PKcs foci, confirming that homologous recombination is the major DSB repair pathway targeted by the drug. Furthermore, using isogenic HCT116 p53+/+ and HCT116 p53-/- cells, we showed that p53 plays a key role in the survival mechanism to SFOM-0046. Finally, SFOM-0046 exhibits a dose-dependent antitumor activity on human fibrosarcoma HT-1080 tumours grafted onto chick chorioallantoic membranes without showing embryo toxicity even at high doses. Altogether, our results highlight SFOM-0046 as a very promising drug that induces a replication stress response.


Asunto(s)
Bencenosulfonatos/farmacología , Daño del ADN , Reparación del ADN , Compuestos de Fenilurea/farmacología , Antineoplásicos/farmacología , Línea Celular Tumoral , Replicación del ADN , Humanos
16.
Biomolecules ; 5(4): 3204-59, 2015 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-26610585

RESUMEN

For decades, radiotherapy and chemotherapy were the two only approaches exploiting DNA repair processes to fight against cancer. Nowadays, cancer therapeutics can be a major challenge when it comes to seeking personalized targeted medicine that is both effective and selective to the malignancy. Over the last decade, the discovery of new targeted therapies against DNA damage signalling and repair has offered the possibility of therapeutic improvements in oncology. In this review, we summarize the current knowledge of DNA damage signalling and repair inhibitors, their molecular and cellular effects, and future therapeutic use.


Asunto(s)
Antineoplásicos/farmacología , Daño del ADN , Enzimas Reparadoras del ADN/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Neoplasias/genética , Animales , Antineoplásicos/uso terapéutico , Enzimas Reparadoras del ADN/genética , Enzimas Reparadoras del ADN/metabolismo , Inhibidores Enzimáticos/uso terapéutico , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo
17.
Biochem J ; 460(3): 331-42, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24870022

RESUMEN

PALB2 [partner and localizer of BRCA2 (breast cancer early-onset 2)] [corrected] has emerged as a key player in the maintenance of genome integrity. Biallelic mutations in PALB2 cause FA (Fanconi's anaemia) subtype FA-N, a devastating inherited disorder marked by developmental abnormalities, bone marrow failure and childhood cancer susceptibility, whereas monoallelic mutations predispose to breast, ovarian and pancreatic cancer. The tumour suppressor role of PALB2 has been intimately linked to its ability to promote HR (homologous recombination)-mediated repair of DNA double-strand breaks. Because PALB2 lies at the crossroads between FA, HR and cancer susceptibility, understanding its function has become the primary focus of several studies. The present review discusses a current synthesis of the contribution of PALB2 to these pathways. We also provide a molecular description of FA- or cancer-associated PALB2 mutations.


Asunto(s)
Reparación del ADN , Recombinación Homóloga , Neoplasias/fisiopatología , Proteínas Nucleares/fisiología , Proteínas Supresoras de Tumor/fisiología , Animales , Proteína BRCA2/fisiología , Neoplasias de la Mama/genética , Neoplasias de la Mama Masculina/genética , Anemia de Fanconi/genética , Proteína del Grupo de Complementación N de la Anemia de Fanconi , Femenino , Humanos , Masculino , Ratones , Neoplasias/genética , Neoplasias Ováricas/genética , Neoplasias Pancreáticas/genética , Factores de Transcripción/fisiología
18.
Microbiol Mol Biol Rev ; 78(1): 40-73, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24600040

RESUMEN

All living organisms are continuously faced with endogenous or exogenous stress conditions affecting genome stability. DNA repair pathways act as a defense mechanism, which is essential to maintain DNA integrity. There is much to learn about the regulation and functions of these mechanisms, not only in human cells but also equally in divergent organisms. In trypanosomatids, DNA repair pathways protect the genome against mutations but also act as an adaptive mechanism to promote drug resistance. In this review, we scrutinize the molecular mechanisms and DNA repair pathways which are conserved in trypanosomatids. The recent advances made by the genome consortiums reveal the complete genomic sequences of several pathogens. Therefore, using bioinformatics and genomic sequences, we analyze the conservation of DNA repair proteins and their key protein motifs in trypanosomatids. We thus present a comprehensive view of DNA repair processes in trypanosomatids at the crossroads of DNA repair and drug resistance.


Asunto(s)
Reparación del ADN/fisiología , Resistencia a Medicamentos/genética , Trypanosomatina/efectos de los fármacos , Trypanosomatina/genética , ADN , Humanos , Leishmaniasis/tratamiento farmacológico , Leishmaniasis/parasitología , Tripanosomiasis/tratamiento farmacológico , Tripanosomiasis/parasitología
19.
Proc Natl Acad Sci U S A ; 111(1): 285-90, 2014 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-24347639

RESUMEN

The cellular response to highly genotoxic DNA double-strand breaks (DSBs) involves the exquisite coordination of multiple signaling and repair factors. Here, we conducted a functional RNAi screen and identified BAP1 as a deubiquitinase required for efficient assembly of the homologous recombination (HR) factors BRCA1 and RAD51 at ionizing radiation (IR) -induced foci. BAP1 is a chromatin-associated protein frequently inactivated in cancers of various tissues. To further investigate the role of BAP1 in DSB repair, we used a gene targeting approach to knockout (KO) this deubiquitinase in chicken DT40 cells. We show that BAP1-deficient cells are (i) sensitive to IR and other agents that induce DSBs, (ii) defective in HR-mediated immunoglobulin gene conversion, and (iii) exhibit an increased frequency of chromosomal breaks after IR treatment. We also show that BAP1 is recruited to chromatin in the proximity of a single site-specific I-SceI-induced DSB. Finally, we identified six IR-induced phosphorylation sites in BAP1 and showed that mutation of these residues inhibits BAP1 recruitment to DSB sites. We also found that both BAP1 catalytic activity and its phosphorylation are critical for promoting DNA repair and cellular recovery from DNA damage. Our data reveal an important role for BAP1 in DSB repair by HR, thereby providing a possible molecular basis for its tumor suppressor function.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , Regulación Neoplásica de la Expresión Génica , Recombinación Homóloga , Neoplasias/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Animales , Proteína BRCA1/metabolismo , Línea Celular , Línea Celular Tumoral , Pollos , Daño del ADN , Células HEK293 , Células HeLa , Humanos , Inmunoglobulinas/genética , Células MCF-7 , Microscopía Fluorescente , Mutación , Neoplasias/genética , Fenotipo , Fosforilación , Recombinasa Rad51 , Radiación Ionizante
20.
J Cell Sci ; 126(Pt 1): 348-59, 2013 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23108668

RESUMEN

The interplay between homologous DNA recombination and mitotic progression is poorly understood. The five RAD51 paralogs (RAD51B, RAD51C, RAD51D, XRCC2 and XRCC3) are key enzymes for DNA double-strand break repair. In our search for specific functions of the various RAD51 paralogs, we found that inhibition of XRCC3 elicits checkpoint defects, while inhibition of RAD51B or RAD51C induces G2/M cell cycle arrest in HeLa cells. Using live-cell microscopy we show that in XRCC3-knockdown cells the spindle assembly checkpoint persists and there is a higher frequency of chromosome misalignments, anaphase bridges, and aneuploidy. We observed centrosome defects in the absence of XRCC3. While RAD51B and RAD51C act early in homologous recombination, XRCC3 functions jointly with GEN1 later in the pathway at the stage of Holliday junction resolution. Our data demonstrate that Holliday junction resolution has critical functions for preventing aberrant mitosis and aneuploidy in mitotic cells.


Asunto(s)
Aneuploidia , Proteínas de Unión al ADN/metabolismo , Western Blotting , ADN Cruciforme/genética , Proteínas de Unión al ADN/genética , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Células HeLa , Resolvasas de Unión Holliday/genética , Resolvasas de Unión Holliday/metabolismo , Humanos , Mitosis/genética , Interferencia de ARN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...