Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 655
Filtrar
Más filtros

País/Región como asunto
Intervalo de año de publicación
1.
Cell ; 184(24): 5970-5984.e18, 2021 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-34793701

RESUMEN

Numerous DNA double-strand breaks (DSBs) arise during meiosis to initiate homologous recombination. These DSBs are usually repaired faithfully, but here, we uncover a distinct type of mutational event in which deletions form via joining of ends from two closely spaced DSBs (double cuts) within a single hotspot or at adjacent hotspots on the same or different chromatids. Deletions occur in normal meiosis but are much more frequent when DSB formation is dysregulated in the absence of the ATM kinase. Events between chromosome homologs point to multi-chromatid damage and aborted gap repair. Some deletions contain DNA from other hotspots, indicating that double cutting at distant sites creates substrates for insertional mutagenesis. End joining at double cuts can also yield tandem duplications or extrachromosomal circles. Our findings highlight the importance of DSB regulation and reveal a previously hidden potential for meiotic mutagenesis that is likely to affect human health and genome evolution.


Asunto(s)
Eliminación de Gen , Duplicación de Gen , Células Germinativas/metabolismo , Recombinación Genética/genética , Animales , Proteínas de la Ataxia Telangiectasia Mutada/deficiencia , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Secuencia de Bases , Cromátides/metabolismo , Cromosomas de los Mamíferos/genética , Cruzamientos Genéticos , Roturas del ADN de Doble Cadena , ADN Circular/genética , Femenino , Genoma , Haplotipos/genética , Recombinación Homóloga/genética , Masculino , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Mutagénesis Insercional/genética , Mutación/genética
2.
Annu Rev Biochem ; 87: 239-261, 2018 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-29494238

RESUMEN

The number of DNA polymerases identified in each organism has mushroomed in the past two decades. Most newly found DNA polymerases specialize in translesion synthesis and DNA repair instead of replication. Although intrinsic error rates are higher for translesion and repair polymerases than for replicative polymerases, the specialized polymerases increase genome stability and reduce tumorigenesis. Reflecting the numerous types of DNA lesions and variations of broken DNA ends, translesion and repair polymerases differ in structure, mechanism, and function. Here, we review the unique and general features of polymerases specialized in lesion bypass, as well as in gap-filling and end-joining synthesis.


Asunto(s)
Daño del ADN , Enzimas Reparadoras del ADN/química , Enzimas Reparadoras del ADN/metabolismo , Reparación del ADN , ADN Polimerasa Dirigida por ADN/química , ADN Polimerasa Dirigida por ADN/metabolismo , Enzimas Reparadoras del ADN/clasificación , ADN Polimerasa Dirigida por ADN/clasificación , Humanos , Modelos Biológicos , Modelos Moleculares
3.
Cell ; 173(4): 972-988.e23, 2018 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-29656893

RESUMEN

Repair of damaged DNA is essential for maintaining genome integrity and for preventing genome-instability-associated diseases, such as cancer. By combining proximity labeling with quantitative mass spectrometry, we generated high-resolution interaction neighborhood maps of the endogenously expressed DNA repair factors 53BP1, BRCA1, and MDC1. Our spatially resolved interaction maps reveal rich network intricacies, identify shared and bait-specific interaction modules, and implicate previously concealed regulators in this process. We identified a novel vertebrate-specific protein complex, shieldin, comprising REV7 plus three previously uncharacterized proteins, RINN1 (CTC-534A2.2), RINN2 (FAM35A), and RINN3 (C20ORF196). Recruitment of shieldin to DSBs, via the ATM-RNF8-RNF168-53BP1-RIF1 axis, promotes NHEJ-dependent repair of intrachromosomal breaks, immunoglobulin class-switch recombination (CSR), and fusion of unprotected telomeres. Shieldin functions as a downstream effector of 53BP1-RIF1 in restraining DNA end resection and in sensitizing BRCA1-deficient cells to PARP inhibitors. These findings have implications for understanding cancer-associated PARPi resistance and the evolution of antibody CSR in higher vertebrates.


Asunto(s)
Reparación del ADN por Unión de Extremidades/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Proteínas Adaptadoras Transductoras de Señales , Proteína BRCA1/antagonistas & inhibidores , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Proteínas de Ciclo Celular , Línea Celular Tumoral , Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Humanos , Cambio de Clase de Inmunoglobulina/efectos de los fármacos , Proteínas Mad2/antagonistas & inhibidores , Proteínas Mad2/genética , Proteínas Mad2/metabolismo , Mutagénesis Sitio-Dirigida , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Proteínas de Unión a Telómeros/antagonistas & inhibidores , Proteínas de Unión a Telómeros/genética , Proteínas de Unión a Telómeros/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53/antagonistas & inhibidores , Proteína 1 de Unión al Supresor Tumoral P53/genética , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
4.
Mol Cell ; 83(5): 698-714.e4, 2023 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-36724784

RESUMEN

Non-homologous end joining is the major double-strand break repair (DSBR) pathway in mammals. DNA-PK is the hub and organizer of multiple steps in non-homologous end joining (NHEJ). Recent high-resolution structures show how two distinct NHEJ complexes "synapse" two DNA ends. One complex includes a DNA-PK dimer mediated by XLF, whereas a distinct DNA-PK dimer forms via a domain-swap mechanism where the C terminus of Ku80 from one DNA-PK protomer interacts with another DNA-PK protomer in trans. Remarkably, the distance between the two synapsed DNA ends in both dimers is the same (∼115 Å), which matches the distance observed in the initial description of an NHEJ long-range synaptic complex. Here, a mutational strategy is used to demonstrate distinct cellular function(s) of the two dimers: one promoting fill-in end processing, while the other promotes DNA end resection. Thus, the specific DNA-PK dimer formed (which may be impacted by DNA end structure) dictates the mechanism by which ends will be made ligatable.


Asunto(s)
Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN , Animales , Proteínas de Unión al ADN/genética , Subunidades de Proteína/metabolismo , Reparación del ADN por Unión de Extremidades , Reparación del ADN , ADN/genética , Proteína Quinasa Activada por ADN/genética , Autoantígeno Ku/genética , Mamíferos/metabolismo
5.
Mol Cell ; 82(20): 3932-3942.e6, 2022 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-36130596

RESUMEN

The DNA-PKcs kinase mediates the repair of DNA double-strand breaks via classical non-homologous end joining (NHEJ). DNA-PKcs is also recruited to active replication forks, although a role for DNA-PKcs in the control of fork dynamics is unclear. Here, we identify a crucial role for DNA-PKcs in promoting fork reversal, a process that stabilizes stressed replication forks and protects genome integrity. DNA-PKcs promotes fork reversal and slowing in response to several replication stress-inducing agents in a manner independent of its role in NHEJ. Cells lacking DNA-PKcs activity show increased DNA damage during S-phase and cellular sensitivity to replication stress. Notably, prevention of fork slowing and reversal via DNA-PKcs inhibition efficiently restores chemotherapy sensitivity in BRCA2-deficient mammary tumors with acquired PARPi resistance. Together, our data uncover a new key regulator of fork reversal and show how DNA-PKcs signaling can be manipulated to alter fork dynamics and drug resistance in cancer.


Asunto(s)
Roturas del ADN de Doble Cadena , Resistencia a Antineoplásicos , Resistencia a Antineoplásicos/genética , Daño del ADN , Reparación del ADN por Unión de Extremidades , ADN/genética , Replicación del ADN , Reparación del ADN
6.
Mol Cell ; 82(1): 177-189.e4, 2022 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-34936881

RESUMEN

The DNA-dependent protein kinase (DNA-PK) initially protects broken DNA ends but then promotes their processing during non-homologous end joining (NHEJ). Before ligation by NHEJ, DNA hairpin ends generated during V(D)J recombination must be opened by the Artemis nuclease, together with autophosphorylated DNA-PK. Structures of DNA-PK bound to DNA before and after phosphorylation, and in complex with Artemis and a DNA hairpin, reveal an essential functional switch. When bound to open DNA ends in its protection mode, DNA-PK is inhibited for cis-autophosphorylation of the so-called ABCDE cluster but activated for phosphorylation of other targets. In contrast, DNA hairpin ends promote cis-autophosphorylation. Phosphorylation of four Thr residues in ABCDE leads to gross structural rearrangement of DNA-PK, widening the DNA binding groove for Artemis recruitment and hairpin cleavage. Meanwhile, Artemis locks DNA-PK into the kinase-inactive state. Kinase activity and autophosphorylation of DNA-PK are regulated by different DNA ends, feeding forward to coordinate NHEJ events.


Asunto(s)
Daño del ADN , Reparación del ADN por Unión de Extremidades , ADN de Neoplasias/metabolismo , Proteína Quinasa Activada por ADN/metabolismo , Neoplasias del Cuello Uterino/enzimología , ADN de Neoplasias/genética , Proteína Quinasa Activada por ADN/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Endonucleasas/genética , Endonucleasas/metabolismo , Activación Enzimática , Femenino , Células HEK293 , Células HeLa , Humanos , Autoantígeno Ku/genética , Autoantígeno Ku/metabolismo , Conformación de Ácido Nucleico , Fosforilación , Unión Proteica , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/patología
7.
Mol Cell ; 82(7): 1359-1371.e9, 2022 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-35216668

RESUMEN

The chromatin-binding protein 53BP1 promotes DNA repair by orchestrating the recruitment of downstream effectors including PTIP, RIF1, and shieldin to DNA double-strand break sites. While we know how PTIP recognizes 53BP1, the molecular details of RIF1 recruitment to DNA-damage sites remains undefined. Here, we report that RIF1 is a phosphopeptide-binding protein that directly interacts with three phosphorylated 53BP1 epitopes. The RIF1-binding sites on 53BP1 share an essential LxL motif followed by two closely apposed phosphorylated residues. Simultaneous mutation of these sites on 53BP1 abrogates RIF1 accumulation into ionizing-radiation-induced foci, but surprisingly, only fully compromises 53BP1-dependent DNA repair when an alternative mode of shieldin recruitment to DNA-damage sites is also disabled. Intriguingly, this alternative mode of recruitment still depends on RIF1 but does not require its interaction with 53BP1. RIF1 therefore employs phosphopeptide recognition to promote DNA repair but also modifies shieldin action independently of 53BP1 binding.


Asunto(s)
Fosfopéptidos , Proteínas de Unión a Telómeros , Proteína BRCA1/genética , Proteínas Portadoras/metabolismo , ADN/metabolismo , Reparación del ADN por Unión de Extremidades , Reparación del ADN , Fosfopéptidos/genética , Fosfopéptidos/metabolismo , 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
8.
Mol Cell ; 82(10): 1940-1955.e7, 2022 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-35447080

RESUMEN

Previously, we showed that CDYL1 is recruited to DNA double-strand breaks (DSBs) to promote homologous recombination (HR) repair and foster transcriptional silencing. However, how CDYL1 elicits DSB-induced silencing is not fully understood. Here, we identify a CDYL1-dependent local decrease in the transcriptionally active marks histone lysine crotonylation (Kcr) and crotonylated lysine 9 of H3 (H3K9cr) at AsiSI-induced DSBs, which correlates with transcriptional silencing. Mechanistically, we reveal that CDYL1 crotonyl-CoA hydratase activity counteracts Kcr and H3K9cr at DSB sites, which triggers the eviction of the transcription elongation factor ENL and fosters transcriptional silencing. Furthermore, genetic inhibition of CDYL1 hydratase activity blocks the reduction in H3K9cr and alleviates DSB-induced silencing, whereas HR efficiency unexpectedly remains intact. Therefore, our results functionally uncouple the repair and silencing activity of CDYL1 at DSBs. In a broader context, we address a long-standing question concerning the functional relationship between HR repair and DSB-induced silencing, suggesting that they may occur independently.


Asunto(s)
Roturas del ADN de Doble Cadena , Lisina , ADN , Reparación del ADN por Unión de Extremidades , Reparación del ADN , Histonas/genética , Histonas/metabolismo , Lisina/genética
9.
Annu Rev Biochem ; 83: 409-39, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24606144

RESUMEN

Current technology enables the production of highly specific genome modifications with excellent efficiency and specificity. Key to this capability are targetable DNA cleavage reagents and cellular DNA repair pathways. The break made by these reagents can produce localized sequence changes through inaccurate nonhomologous end joining (NHEJ), often leading to gene inactivation. Alternatively, user-provided DNA can be used as a template for repair by homologous recombination (HR), leading to the introduction of desired sequence changes. This review describes three classes of targetable cleavage reagents: zinc-finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and CRISPR/Cas RNA-guided nucleases (RGNs). As a group, these reagents have been successfully used to modify genomic sequences in a wide variety of cells and organisms, including humans. This review discusses the properties, advantages, and limitations of each system, as well as the specific considerations required for their use in different biological systems.


Asunto(s)
Endonucleasas/genética , Ingeniería Genética/métodos , Genoma , Animales , Arabidopsis , ADN/química , Daño del ADN , Reparación del ADN por Unión de Extremidades , Reparación del ADN , Drosophila , Drosophila melanogaster , Eliminación de Gen , Genómica , Humanos , Ratones , Ingeniería de Proteínas/métodos , Estructura Terciaria de Proteína , Ratas , Recombinación Genética , Pez Cebra , Dedos de Zinc
10.
Mol Cell ; 81(16): 3400-3409.e3, 2021 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-34352203

RESUMEN

Non-homologous end joining (NHEJ) is one of two critical mechanisms utilized in humans to repair DNA double-strand breaks (DSBs). Unrepaired or incorrect repair of DSBs can lead to apoptosis or cancer. NHEJ involves several proteins, including the Ku70/80 heterodimer, DNA-dependent protein kinase catalytic subunit (DNA-PKcs), X-ray cross-complementing protein 4 (XRCC4), XRCC4-like factor (XLF), and ligase IV. These core proteins bind DSBs and ligate the damaged DNA ends. However, details of the structural assembly of these proteins remain unclear. Here, we present cryo-EM structures of NHEJ supercomplexes that are composed of these core proteins and DNA, revealing the detailed structural architecture of this assembly. We describe monomeric and dimeric forms of this supercomplex and also propose the existence of alternate dimeric forms of long-range synaptic complexes. Finally, we show that mutational disruption of several structural features within these NHEJ complexes negatively affects DNA repair.


Asunto(s)
ADN Ligasa (ATP)/ultraestructura , Enzimas Reparadoras del ADN/ultraestructura , Proteína Quinasa Activada por ADN/ultraestructura , Proteínas de Unión al ADN/ultraestructura , Complejos Multiproteicos/ultraestructura , Apoptosis/genética , Microscopía por Crioelectrón , Roturas del ADN de Doble Cadena , Daño del ADN/genética , Reparación del ADN por Unión de Extremidades/genética , ADN Ligasa (ATP)/genética , Reparación del ADN/genética , Enzimas Reparadoras del ADN/genética , Proteína Quinasa Activada por ADN/genética , Proteínas de Unión al ADN/genética , Humanos , Autoantígeno Ku/genética , Autoantígeno Ku/ultraestructura , Complejos Multiproteicos/genética , Fosforilación/genética
11.
Mol Cell ; 81(10): 2216-2230.e10, 2021 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-33848455

RESUMEN

DNA double-strand break (DSB) repair is mediated by multiple pathways. It is thought that the local chromatin context affects the pathway choice, but the underlying principles are poorly understood. Using a multiplexed reporter assay in combination with Cas9 cutting, we systematically measure the relative activities of three DSB repair pathways as a function of chromatin context in >1,000 genomic locations. This reveals that non-homologous end-joining (NHEJ) is broadly biased toward euchromatin, while the contribution of microhomology-mediated end-joining (MMEJ) is higher in specific heterochromatin contexts. In H3K27me3-marked heterochromatin, inhibition of the H3K27 methyltransferase EZH2 reverts the balance toward NHEJ. Single-stranded template repair (SSTR), often used for precise CRISPR editing, competes with MMEJ and is moderately linked to chromatin context. These results provide insight into the impact of chromatin on DSB repair pathway balance and guidance for the design of Cas9-mediated genome editing experiments.


Asunto(s)
Proteína 9 Asociada a CRISPR/metabolismo , Cromatina/metabolismo , Roturas del ADN de Doble Cadena , Reparación del ADN , Secuencia de Bases , Reparación del ADN por Unión de Extremidades , Eucromatina/metabolismo , Reordenamiento Génico , Genoma Humano , Heterocromatina/metabolismo , Humanos , Mutación INDEL/genética , Células K562 , Cinética , Unión Proteica , Reproducibilidad de los Resultados
12.
Mol Cell ; 81(5): 1084-1099.e6, 2021 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-33450211

RESUMEN

Cells have evolved an elaborate DNA repair network to ensure complete and accurate DNA replication. Defects in these repair machineries can fuel genome instability and drive carcinogenesis while creating vulnerabilities that may be exploited in therapy. Here, we use nascent chromatin capture (NCC) proteomics to characterize the repair of replication-associated DNA double-strand breaks (DSBs) triggered by topoisomerase 1 (TOP1) inhibitors. We reveal profound changes in the fork proteome, including the chromatin environment and nuclear membrane interactions, and identify three classes of repair factors according to their enrichment at broken and/or stalled forks. ATM inhibition dramatically rewired the broken fork proteome, revealing that ataxia telangiectasia mutated (ATM) signalling stimulates DNA end resection, recruits PLK1, and concomitantly suppresses the canonical DSB ubiquitination response by preventing accumulation of RNF168 and BRCA1-A. This work and collection of replication fork proteomes provide a new framework to understand how cells orchestrate homologous recombination repair of replication-associated DSBs.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de Ciclo Celular/genética , Replicación del ADN , ADN-Topoisomerasas de Tipo I/genética , ADN/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Reparación del ADN por Recombinación , Proteínas de la Ataxia Telangiectasia Mutada/antagonistas & inhibidores , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Camptotecina/farmacología , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Cromatina/química , Cromatina/metabolismo , ADN/metabolismo , Roturas del ADN de Doble Cadena , ADN-Topoisomerasas de Tipo I/metabolismo , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Regulación de la Expresión Génica , Células HeLa , Humanos , Unión Proteica , Proteínas Serina-Treonina Quinasas/metabolismo , Proteómica/métodos , Proteínas Proto-Oncogénicas/metabolismo , Piridinas/farmacología , Quinolinas/farmacología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Inhibidores de Topoisomerasa I/farmacología , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación/efectos de los fármacos , Quinasa Tipo Polo 1
13.
Genes Dev ; 35(1-2): 1-21, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33361389

RESUMEN

In this perspective, we introduce shelterin and the mechanisms of ATM activation and NHEJ at telomeres, before discussing the following questions: How are t-loops proposed to protect chromosome ends and what is the evidence for this model? Can other models explain how TRF2 mediates end protection? Could t-loops be pathological structures? How is end protection achieved in pluripotent cells? What do the insights into telomere end protection in pluripotent cells mean for the t-loop model of end protection? Why might different cell states have evolved different mechanisms of end protection? Finally, we offer support for an updated t-loop model of end protection, suggesting that the data is supportive of a critical role for t-loops in protecting chromosome ends from NHEJ and ATM activation, but that other mechanisms are involved. Finally, we propose that t-loops are likely dynamic, rather than static, structures.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades/fisiología , Telómero/metabolismo , Telómero/patología , Animales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Inestabilidad Cromosómica , Reparación del ADN , Células Madre Embrionarias , Humanos , Modelos Biológicos , Células Madre Pluripotentes , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo
14.
Trends Biochem Sci ; 49(5): 391-400, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38490833

RESUMEN

One of the two chromosomal breakage events in recurring translocations in B cell neoplasms is often due to the recombination-activating gene complex (RAG complex) releasing DNA ends before end joining. The other break occurs in a fragile zone of 20-600 bp in a non-antigen receptor gene locus, with a more complex and intriguing set of mechanistic factors underlying such narrow fragile zones. These factors include activation-induced deaminase (AID), which acts only at regions of single-stranded DNA (ssDNA). Recent work leads to a model involving the tethering of AID to the nascent RNA as it emerges from the RNA polymerase. This mechanism may have relevance in class switch recombination (CSR) and somatic hypermutation (SHM), as well as broader relevance for other DNA enzymes.


Asunto(s)
ARN , Translocación Genética , Humanos , ARN/metabolismo , ARN/genética , Citidina Desaminasa/metabolismo , Citidina Desaminasa/genética , Sitios Frágiles del Cromosoma
15.
Mol Cell ; 77(5): 1080-1091.e8, 2020 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-31862156

RESUMEN

Enzymatic processing of DNA underlies all DNA repair, yet inappropriate DNA processing must be avoided. In vertebrates, double-strand breaks are repaired predominantly by non-homologous end joining (NHEJ), which directly ligates DNA ends. NHEJ has the potential to be highly mutagenic because it uses DNA polymerases, nucleases, and other enzymes that modify incompatible DNA ends to allow their ligation. Using frog egg extracts that recapitulate NHEJ, we show that end processing requires the formation of a "short-range synaptic complex" in which DNA ends are closely aligned in a ligation-competent state. Furthermore, single-molecule imaging directly demonstrates that processing occurs within the short-range complex. This confinement of end processing to a ligation-competent complex ensures that DNA ends undergo ligation as soon as they become compatible, thereby minimizing mutagenesis. Our results illustrate how the coordination of enzymatic catalysis with higher-order structural organization of substrate maximizes the fidelity of DNA repair.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades , Inestabilidad Genómica , Animales , ADN Ligasas/genética , ADN Ligasas/metabolismo , Enzimas Reparadoras del ADN/genética , Enzimas Reparadoras del ADN/metabolismo , Proteína Quinasa Activada por ADN/genética , Proteína Quinasa Activada por ADN/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Autoantígeno Ku/genética , Autoantígeno Ku/metabolismo , Modelos Genéticos , Complejos Multiproteicos , Hidrolasas Diéster Fosfóricas/genética , Hidrolasas Diéster Fosfóricas/metabolismo , Imagen Individual de Molécula , Factores de Tiempo , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Xenopus laevis
16.
Mol Cell ; 73(5): 885-899.e6, 2019 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-30686591

RESUMEN

BRCA1 or BRCA2 inactivation drives breast and ovarian cancer but also creates vulnerability to poly(ADP-ribose) polymerase (PARP) inhibitors. To search for additional targets whose inhibition is synthetically lethal in BRCA2-deficient backgrounds, we screened two pairs of BRCA2 isogenic cell lines with DNA-repair-focused small hairpin RNA (shRNA) and CRISPR (clustered regularly interspaced short palindromic repeats)-based libraries. We found that BRCA2-deficient cells are selectively dependent on multiple pathways including base excision repair, ATR signaling, and splicing. We identified APEX2 and FEN1 as synthetic lethal genes with both BRCA1 and BRCA2 loss of function. BRCA2-deficient cells require the apurinic endonuclease activity and the PCNA-binding domain of Ape2 (APEX2), but not Ape1 (APEX1). Furthermore, BRCA2-deficient cells require the 5' flap endonuclease but not the 5'-3' exonuclease activity of Fen1, and chemically inhibiting Fen1 selectively targets BRCA-deficient cells. Finally, we developed a microhomology-mediated end-joining (MMEJ) reporter and showed that Fen1 participates in MMEJ, underscoring the importance of MMEJ as a collateral repair pathway in the context of homologous recombination (HR) deficiency.


Asunto(s)
Proteína BRCA2/genética , Sistemas CRISPR-Cas , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Endonucleasas de ADN Solapado/genética , Genes Letales , Neoplasias/genética , Interferencia de ARN , Mutaciones Letales Sintéticas , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Proteína BRCA2/metabolismo , Muerte Celular , Línea Celular Tumoral , Daño del ADN , Reparación del ADN por Unión de Extremidades , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Endonucleasas , Endonucleasas de ADN Solapado/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Enzimas Multifuncionales , Neoplasias/tratamiento farmacológico , Neoplasias/enzimología , Neoplasias/patología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Antígeno Nuclear de Célula en Proliferación/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , ARN Interferente Pequeño/genética
17.
Mol Cell ; 74(3): 466-480.e4, 2019 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-30930055

RESUMEN

The mTOR pathway integrates both extracellular and intracellular signals and serves as a central regulator of cell metabolism, growth, survival, and stress responses. Neurotropic viruses, such as herpes simplex virus-1 (HSV-1), also rely on cellular AKT-mTORC1 signaling to achieve viral latency. Here, we define a novel genotoxic response whereby spatially separated signals initiated by extracellular neurotrophic factors and nuclear DNA damage are integrated by the AKT-mTORC1 pathway. We demonstrate that endogenous DNA double-strand breaks (DSBs) mediated by Topoisomerase 2ß-DNA cleavage complex (TOP2ßcc) intermediates are required to achieve AKT-mTORC1 signaling and maintain HSV-1 latency in neurons. Suppression of host DNA-repair pathways that remove TOP2ßcc trigger HSV-1 reactivation. Moreover, perturbation of AKT phosphorylation dynamics by downregulating the PHLPP1 phosphatase led to AKT mis-localization and disruption of DSB-induced HSV-1 reactivation. Thus, the cellular genome integrity and environmental inputs are consolidated and co-opted by a latent virus to balance lifelong infection with transmission.


Asunto(s)
ADN-Topoisomerasas de Tipo II/genética , Herpesvirus Humano 1/genética , Proteínas Nucleares/genética , Proteínas Proto-Oncogénicas c-akt/genética , Latencia del Virus/genética , Animales , Roturas del ADN de Doble Cadena , Daño del ADN/genética , Reparación del ADN por Unión de Extremidades/genética , Reparación del ADN/genética , Enzimas Reparadoras del ADN/genética , Proteínas de Unión al ADN/genética , Herpesvirus Humano 1/patogenicidad , Humanos , Proteína Homóloga de MRE11/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Neuronas/metabolismo , Neuronas/virología , Fosforilación , Ratas , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/genética
18.
Trends Genet ; 39(12): 924-940, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37806853

RESUMEN

Genome integrity and maintenance are essential for the viability of all organisms. A wide variety of DNA damage types have been described, but double-strand breaks (DSBs) stand out as one of the most toxic DNA lesions. Two major pathways account for the repair of DSBs: homologous recombination (HR) and non-homologous end joining (NHEJ). Both pathways involve complex DNA transactions catalyzed by proteins that sequentially or cooperatively work to repair the damage. Single-molecule methods allow visualization of these complex transactions and characterization of the protein:DNA intermediates of DNA repair, ultimately allowing a comprehensive breakdown of the mechanisms underlying each pathway. We review current understanding of the HR and NHEJ responses to DSBs in eukaryotic cells, with a particular emphasis on recent advances through the use of single-molecule techniques.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , Reparación del ADN/genética , ADN/genética , Daño del ADN , Reparación del ADN por Unión de Extremidades/genética
19.
EMBO J ; 41(1): e108813, 2022 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-34817085

RESUMEN

Heterochromatin is a conserved feature of eukaryotic chromosomes, with central roles in gene expression regulation and maintenance of genome stability. How heterochromatin proteins regulate DNA repair remains poorly described. In the yeast Saccharomyces cerevisiae, the silent information regulator (SIR) complex assembles heterochromatin-like chromatin at sub-telomeric chromosomal regions. SIR-mediated repressive chromatin limits DNA double-strand break (DSB) resection, thus protecting damaged chromosome ends during homologous recombination (HR). As resection initiation represents the crossroads between repair by non-homologous end joining (NHEJ) or HR, we asked whether SIR-mediated heterochromatin regulates NHEJ. We show that SIRs promote NHEJ through two pathways, one depending on repressive chromatin assembly, and the other relying on Sir3 in a manner that is independent of its heterochromatin-promoting function. Via physical interaction with the Sae2 protein, Sir3 impairs Sae2-dependent functions of the MRX (Mre11-Rad50-Xrs2) complex, thereby limiting Mre11-mediated resection, delaying MRX removal from DSB ends, and promoting NHEJ.


Asunto(s)
Reparación del ADN por Unión de Extremidades , Endonucleasas/metabolismo , Heterocromatina/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas Reguladoras de Información Silente de Saccharomyces cerevisiae/metabolismo , Secuencia de Aminoácidos , Endonucleasas/química , Mutación Puntual/genética , Unión Proteica , Dominios Proteicos , Proteínas de Saccharomyces cerevisiae/química , Proteínas Reguladoras de Información Silente de Saccharomyces cerevisiae/química , Proteínas Reguladoras de Información Silente de Saccharomyces cerevisiae/genética , Telómero/metabolismo
20.
Development ; 150(10)2023 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-37129004

RESUMEN

Fluorescent protein (FP) tagging is a key method for observing protein distribution, dynamics and interaction with other proteins in living cells. However, the typical approach using overexpression of tagged proteins can perturb cell behavior and introduce localization artifacts. To preserve native expression, fluorescent proteins can be inserted directly into endogenous genes. This approach has been widely used in yeast for decades, and more recently in invertebrate model organisms with the advent of CRISPR/Cas9. However, endogenous FP tagging has not been widely used in mammalian cells due to inefficient homology-directed repair. Recently, the CRISPaint system used non-homologous end joining for efficient integration of FP tags into native loci, but it only allows C-terminal knock-ins. Here, we have enhanced the CRISPaint system by introducing new universal donors for N-terminal insertion and for multi-color tagging with orthogonal selection markers. We adapted the procedure for mouse embryonic stem cells, which can be differentiated into diverse cell types. Our protocol is rapid and efficient, enabling live imaging in less than 2 weeks post-transfection. These improvements increase the versatility and applicability of FP knock-in in mammalian cells.


Asunto(s)
Sistemas CRISPR-Cas , Células Madre Embrionarias de Ratones , Animales , Ratones , Sistemas CRISPR-Cas/genética , Proteínas/genética , Técnicas de Sustitución del Gen , Edición Génica/métodos , Mamíferos/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA