Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
EMBO J ; 43(12): 2397-2423, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38760575

RESUMEN

The nucleoside analogue decitabine (or 5-aza-dC) is used to treat several haematological cancers. Upon its triphosphorylation and incorporation into DNA, 5-aza-dC induces covalent DNA methyltransferase 1 DNA-protein crosslinks (DNMT1-DPCs), leading to DNA hypomethylation. However, 5-aza-dC's clinical outcomes vary, and relapse is common. Using genome-scale CRISPR/Cas9 screens, we map factors determining 5-aza-dC sensitivity. Unexpectedly, we find that loss of the dCMP deaminase DCTD causes 5-aza-dC resistance, suggesting that 5-aza-dUMP generation is cytotoxic. Combining results from a subsequent genetic screen in DCTD-deficient cells with the identification of the DNMT1-DPC-proximal proteome, we uncover the ubiquitin and SUMO1 E3 ligase, TOPORS, as a new DPC repair factor. TOPORS is recruited to SUMOylated DNMT1-DPCs and promotes their degradation. Our study suggests that 5-aza-dC-induced DPCs cause cytotoxicity when DPC repair is compromised, while cytotoxicity in wild-type cells arises from perturbed nucleotide metabolism, potentially laying the foundations for future identification of predictive biomarkers for decitabine treatment.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasa 1 , Decitabina , Ubiquitina-Proteína Ligasas , Decitabina/farmacología , Humanos , ADN (Citosina-5-)-Metiltransferasa 1/metabolismo , ADN (Citosina-5-)-Metiltransferasa 1/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina-Proteína Ligasas/genética , Metilación de ADN/efectos de los fármacos , Antimetabolitos Antineoplásicos/farmacología , Animales , Sumoilación/efectos de los fármacos
2.
Nat Cell Biol ; 26(5): 797-810, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38600235

RESUMEN

Covalent DNA-protein cross-links (DPCs) are toxic DNA lesions that block replication and require repair by multiple pathways. Whether transcription blockage contributes to the toxicity of DPCs and how cells respond when RNA polymerases stall at DPCs is unknown. Here we find that DPC formation arrests transcription and induces ubiquitylation and degradation of RNA polymerase II. Using genetic screens and a method for the genome-wide mapping of DNA-protein adducts, DPC sequencing, we discover that Cockayne syndrome (CS) proteins CSB and CSA provide resistance to DPC-inducing agents by promoting DPC repair in actively transcribed genes. Consequently, CSB- or CSA-deficient cells fail to efficiently restart transcription after induction of DPCs. In contrast, nucleotide excision repair factors that act downstream of CSB and CSA at ultraviolet light-induced DNA lesions are dispensable. Our study describes a transcription-coupled DPC repair pathway and suggests that defects in this pathway may contribute to the unique neurological features of CS.


Asunto(s)
Síndrome de Cockayne , ADN Helicasas , Enzimas Reparadoras del ADN , Reparación del ADN , Proteínas de Unión a Poli-ADP-Ribosa , ARN Polimerasa II , Transcripción Genética , Ubiquitinación , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/genética , Enzimas Reparadoras del ADN/metabolismo , Enzimas Reparadoras del ADN/genética , Humanos , ADN Helicasas/metabolismo , ADN Helicasas/genética , ARN Polimerasa II/metabolismo , ARN Polimerasa II/genética , Síndrome de Cockayne/genética , Síndrome de Cockayne/metabolismo , Síndrome de Cockayne/patología , Daño del ADN , Rayos Ultravioleta , ADN/metabolismo , ADN/genética , Aductos de ADN/metabolismo , Aductos de ADN/genética , Reparación por Escisión , Factores de Transcripción , Receptores de Interleucina-17
3.
Elife ; 92020 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-32441252

RESUMEN

CRISPR-Cas9 genome engineering has revolutionised high-throughput functional genomic screens. However, recent work has raised concerns regarding the performance of CRISPR-Cas9 screens using TP53 wild-type human cells due to a p53-mediated DNA damage response (DDR) limiting the efficiency of generating viable edited cells. To directly assess the impact of cellular p53 status on CRISPR-Cas9 screen performance, we carried out parallel CRISPR-Cas9 screens in wild-type and TP53 knockout human retinal pigment epithelial cells using a focused dual guide RNA library targeting 852 DDR-associated genes. Our work demonstrates that although functional p53 status negatively affects identification of significantly depleted genes, optimal screen design can nevertheless enable robust screen performance. Through analysis of our own and published screen data, we highlight key factors for successful screens in both wild-type and p53-deficient cells.


The invention of CRISPR-Cas9 genome editing has unlocked a greater understanding of the human genome. Researchers can use this system to make targeted cuts in any gene in the genome, forcing the cell to perform a rapid repair at the cut site. These repairs often introduce mutations into the damaged area, adding or removing DNA letters and disrupting the gene. This allows researchers to study what happens to cells when specific genes are missing, which can help to uncover what each gene is for. One of the most comprehensive ways to use this technique is to perform a CRISPR-Cas9 screen, which disrupts each gene in the genome one by one. For a CRISPR-Cas9 screen to work well, a cell needs to survive the cuts to its genome. But there is a crucial gene that can stop this happening. Often described as the 'guardian of the genome', this gene codes for a protein called p53, a tumour suppressor that helps to stop a cell turning cancerous when its DNA becomes damaged. This protein activates when the cell senses a cut in its genetic material and can kill the cell if it fails to make a successful repair. Recent work has shown that the presence of a working copy of the gene for the p53 protein might limit the ability of CRISPR-Cas9 to edit genes. But the evidence was inconclusive. So, Bowden, Morales-Juarez et al. performed two parallel CRISPR-Cas9 screens in human cells with and without p53 to find out more. This revealed that CRISPR-Cas9 can inactivate genes in both normal cells and cells lacking the p53 protein, but that it works better in cells without p53. This was because, when p53 was active, the cells initiated a protective response against the CRISPR-Cas9 cuts. This changed the patterns of genes successfully inactivated by the screen, but it did not make the results unusable. Careful experimental design and thorough data analysis made it possible to get useful results even in cells with functional p53 protein. The gene for p53 has mutations in around half of human cancers. So, understanding how it affects CRISPR-Cas9 screens could influence the design of future experiments. It is possible that the effects of the p53 protein could vary from cell type to cell type, and with different p53 mutations. Comparisons like the one performed here could help to further unpick how the cell's DNA repair systems might interfere with future CRISPR experiments.


Asunto(s)
Sistemas CRISPR-Cas , Proteína p53 Supresora de Tumor/metabolismo , Apoptosis , Línea Celular , Daño del ADN , Células Epiteliales , Eliminación de Gen , Edición Génica , Regulación de la Expresión Génica , Humanos , Proteína p53 Supresora de Tumor/genética
4.
Nat Commun ; 10(1): 5191, 2019 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-31729360

RESUMEN

Histone H2AX and MDC1 are key DNA repair and DNA-damage signalling proteins. When DNA double-strand breaks (DSBs) occur, H2AX is phosphorylated and then recruits MDC1, which in turn serves as a docking platform to promote the localization of other factors, including 53BP1, to DSB sites. Here, by using CRISPR-Cas9 engineered human cell lines, we identify a hitherto unknown, H2AX-independent, function of MDC1 mediated by its PST-repeat region. We show that the PST-repeat region directly interacts with chromatin via the nucleosome acidic patch and mediates DNA damage-independent association of MDC1 with chromatin. We find that this region is largely functionally dispensable when the canonical γH2AX-MDC1 pathway is operative but becomes critical for 53BP1 recruitment to DNA-damage sites and cell survival following DSB induction when H2AX is not available. Consequently, our results suggest a role for MDC1 in activating the DDR in areas of the genome lacking or depleted of H2AX.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Cromatina/metabolismo , Daño del ADN , Histonas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Secuencias de Aminoácidos , Proteínas de Ciclo Celular/genética , Línea Celular , Cromatina/genética , Roturas del ADN de Doble Cadena , Reparación del ADN , Histonas/genética , Humanos , Proteína 1 de Unión al Supresor Tumoral P53/genética , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo
5.
Nat Commun ; 10(1): 87, 2019 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-30622252

RESUMEN

Mutations in the ATM tumor suppressor gene confer hypersensitivity to DNA-damaging chemotherapeutic agents. To explore genetic resistance mechanisms, we performed genome-wide CRISPR-Cas9 screens in cells treated with the DNA topoisomerase I inhibitor topotecan. Thus, we here establish that inactivating terminal components of the non-homologous end-joining (NHEJ) machinery or of the BRCA1-A complex specifically confer topotecan resistance to ATM-deficient cells. We show that hypersensitivity of ATM-mutant cells to topotecan or the poly-(ADP-ribose) polymerase (PARP) inhibitor olaparib reflects delayed engagement of homologous recombination at DNA-replication-fork associated single-ended double-strand breaks (DSBs), allowing some to be subject to toxic NHEJ. Preventing DSB ligation by NHEJ, or enhancing homologous recombination by BRCA1-A complex disruption, suppresses this toxicity, highlighting a crucial role for ATM in preventing toxic LIG4-mediated chromosome fusions. Notably, suppressor mutations in ATM-mutant backgrounds are different to those in BRCA1-mutant scenarios, suggesting new opportunities for patient stratification and additional therapeutic vulnerabilities for clinical exploitation.


Asunto(s)
Antineoplásicos/farmacología , Proteínas de la Ataxia Telangiectasia Mutada/genética , Reparación del ADN por Unión de Extremidades/genética , Resistencia a Antineoplásicos/genética , Animales , Antineoplásicos/uso terapéutico , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteína BRCA1/metabolismo , Sistemas CRISPR-Cas/genética , Línea Celular Tumoral , Supervivencia Celular/genética , Roturas del ADN de Doble Cadena/efectos de los fármacos , ADN Ligasa (ATP)/metabolismo , Replicación del ADN/efectos de los fármacos , Replicación del ADN/genética , Femenino , Humanos , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Células Madre Embrionarias de Ratones , Mutación , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Ftalazinas/farmacología , Ftalazinas/uso terapéutico , Piperazinas/farmacología , Piperazinas/uso terapéutico , Topotecan/farmacología , Topotecan/uso terapéutico
6.
Nat Cell Biol ; 20(8): 954-965, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30022119

RESUMEN

BRCA1 deficiencies cause breast, ovarian, prostate and other cancers, and render tumours hypersensitive to poly(ADP-ribose) polymerase (PARP) inhibitors. To understand the resistance mechanisms, we conducted whole-genome CRISPR-Cas9 synthetic-viability/resistance screens in BRCA1-deficient breast cancer cells treated with PARP inhibitors. We identified two previously uncharacterized proteins, C20orf196 and FAM35A, whose inactivation confers strong PARP-inhibitor resistance. Mechanistically, we show that C20orf196 and FAM35A form a complex, 'Shieldin' (SHLD1/2), with FAM35A interacting with single-stranded DNA through its C-terminal oligonucleotide/oligosaccharide-binding fold region. We establish that Shieldin acts as the downstream effector of 53BP1/RIF1/MAD2L2 to promote DNA double-strand break (DSB) end-joining by restricting DSB resection and to counteract homologous recombination by antagonizing BRCA2/RAD51 loading in BRCA1-deficient cells. Notably, Shieldin inactivation further sensitizes BRCA1-deficient cells to cisplatin, suggesting how defining the SHLD1/2 status of BRCA1-deficient tumours might aid patient stratification and yield new treatment opportunities. Highlighting this potential, we document reduced SHLD1/2 expression in human breast cancers displaying intrinsic or acquired PARP-inhibitor resistance.


Asunto(s)
Proteína BRCA1/genética , Neoplasias Óseas/tratamiento farmacológico , Neoplasias de la Mama/tratamiento farmacológico , Reparación del ADN por Unión de Extremidades , Resistencia a Antineoplásicos , Osteosarcoma/tratamiento farmacológico , Neoplasias Ováricas/tratamiento farmacológico , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Proteínas/metabolismo , Reparación del ADN por Recombinación , Animales , Proteína BRCA1/deficiencia , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proteínas de Ciclo Celular , Línea Celular Tumoral , Cisplatino/farmacología , Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos/genética , Femenino , Células HEK293 , Humanos , Proteínas Mad2/genética , Proteínas Mad2/metabolismo , Ratones , Complejos Multiproteicos , Osteosarcoma/genética , Osteosarcoma/metabolismo , Osteosarcoma/patología , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Proteínas/genética , Proteínas de Unión a Telómeros/genética , Proteínas de Unión a Telómeros/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53/genética , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Nat Cell Biol ; 17(11): 1458-1470, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26502057

RESUMEN

Ubiquitylation is crucial for proper cellular responses to DNA double-strand breaks (DSBs). If unrepaired, these highly cytotoxic lesions cause genome instability, tumorigenesis, neurodegeneration or premature ageing. Here, we conduct a comprehensive, multilayered screen to systematically profile all human ubiquitin E2 enzymes for impacts on cellular DSB responses. With a widely applicable approach, we use an exemplary E2 family, UBE2Ds, to identify ubiquitylation-cascade components downstream of E2s. Thus, we uncover the nuclear E3 ligase RNF138 as a key homologous recombination (HR)-promoting factor that functions with UBE2Ds in cells. Mechanistically, UBE2Ds and RNF138 accumulate at DNA-damage sites and act at early resection stages by promoting CtIP ubiquitylation and accrual. This work supplies insights into regulation of DSB repair by HR. Moreover, it provides a rich information resource on E2s that can be exploited by follow-on studies.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas Nucleares/metabolismo , Reparación del ADN por Recombinación , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Portadoras/genética , Ciclo Celular/genética , Línea Celular Tumoral , Supervivencia Celular/genética , Roturas del ADN de Doble Cadena/efectos de los fármacos , Roturas del ADN de Doble Cadena/efectos de la radiación , Endodesoxirribonucleasas , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , Humanos , Immunoblotting , Microscopía Confocal , Proteínas Nucleares/genética , Interferencia de ARN , Enzimas Ubiquitina-Conjugadoras/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
8.
Cell Rep ; 11(5): 704-14, 2015 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-25921528

RESUMEN

The activities of many DNA-repair proteins are controlled through reversible covalent modification by ubiquitin and ubiquitin-like molecules. Nonhomologous end-joining (NHEJ) is the predominant DNA double-strand break (DSB) repair pathway in mammalian cells and is initiated by DSB ends being recognized by the Ku70/Ku80 (Ku) heterodimer. By using MLN4924, an anti-cancer drug in clinical trials that specifically inhibits conjugation of the ubiquitin-like protein, NEDD8, to target proteins, we demonstrate that NEDD8 accumulation at DNA-damage sites is a highly dynamic process. In addition, we show that depleting cells of the NEDD8 E2-conjugating enzyme, UBE2M, yields ionizing radiation hypersensitivity and reduced cell survival following NHEJ. Finally, we demonstrate that neddylation promotes Ku ubiquitylation after DNA damage and release of Ku and Ku-associated proteins from damage sites following repair. These studies provide insights into how the NHEJ core complex dissociates from repair sites and highlight its importance for cell survival following DSB induction.


Asunto(s)
Antígenos Nucleares/metabolismo , Daño del ADN , Proteínas de Unión al ADN/metabolismo , Ubiquitinas/metabolismo , Antígenos Nucleares/química , Línea Celular , Supervivencia Celular/efectos de los fármacos , Ciclopentanos/toxicidad , Daño del ADN/efectos de los fármacos , Daño del ADN/efectos de la radiación , Reparación del ADN por Unión de Extremidades , Proteínas de Unión al ADN/química , Histonas/metabolismo , Humanos , Autoantígeno Ku , Proteína NEDD8 , Unión Proteica , Estructura Terciaria de Proteína , Proteómica , Pirimidinas/toxicidad , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Radiación Ionizante , Enzimas Ubiquitina-Conjugadoras/antagonistas & inhibidores , Enzimas Ubiquitina-Conjugadoras/genética , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitinación/efectos de los fármacos , Ubiquitinas/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA