Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Elife ; 102021 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-34477552

RESUMEN

DNA double-strand break (DSB) repair by homologous recombination (HR) is thought to be restricted to the S- and G2- phases of the cell cycle in part due to 53BP1 antagonizing DNA end resection in G1-phase and non-cycling quiescent (G0) cells. Here, we show that LIN37, a component of the DREAM transcriptional repressor, functions in a 53BP1-independent manner to prevent DNA end resection and HR in G0 cells. Loss of LIN37 leads to the expression of HR proteins, including BRCA1, BRCA2, PALB2, and RAD51, and promotes DNA end resection in G0 cells even in the presence of 53BP1. In contrast to 53BP1-deficiency, DNA end resection in LIN37-deficient G0 cells depends on BRCA1 and leads to RAD51 filament formation and HR. LIN37 is not required to protect DNA ends in cycling cells at G1-phase. Thus, LIN37 regulates a novel 53BP1-independent cell phase-specific DNA end protection pathway that functions uniquely in quiescent cells.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades , Recombinasa Rad51/metabolismo , Transactivadores/metabolismo , Proteína BRCA1/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Replicación del ADN , Fase G1 , Fase G2 , Recombinación Homóloga , Humanos , Fase S , Transactivadores/genética , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo
2.
Nature ; 586(7828): 292-298, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32999459

RESUMEN

The RecQ DNA helicase WRN is a synthetic lethal target for cancer cells with microsatellite instability (MSI), a form of genetic hypermutability that arises from impaired mismatch repair1-4. Depletion of WRN induces widespread DNA double-strand breaks in MSI cells, leading to cell cycle arrest and/or apoptosis. However, the mechanism by which WRN protects MSI-associated cancers from double-strand breaks remains unclear. Here we show that TA-dinucleotide repeats are highly unstable in MSI cells and undergo large-scale expansions, distinct from previously described insertion or deletion mutations of a few nucleotides5. Expanded TA repeats form non-B DNA secondary structures that stall replication forks, activate the ATR checkpoint kinase, and require unwinding by the WRN helicase. In the absence of WRN, the expanded TA-dinucleotide repeats are susceptible to cleavage by the MUS81 nuclease, leading to massive chromosome shattering. These findings identify a distinct biomarker that underlies the synthetic lethal dependence on WRN, and support the development of therapeutic agents that target WRN for MSI-associated cancers.


Asunto(s)
Roturas del ADN de Doble Cadena , Expansión de las Repeticiones de ADN/genética , Repeticiones de Dinucleótido/genética , Neoplasias/genética , Helicasa del Síndrome de Werner/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Línea Celular Tumoral , Cromosomas Humanos/genética , Cromosomas Humanos/metabolismo , Cromotripsis , División del ADN , Replicación del ADN , Proteínas de Unión al ADN/metabolismo , Endodesoxirribonucleasas/metabolismo , Endonucleasas/metabolismo , Inestabilidad Genómica , Humanos , Recombinasas/metabolismo
3.
Proc Natl Acad Sci U S A ; 117(33): 20100-20108, 2020 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-32727902

RESUMEN

Mutation of HELLS (Helicase, Lymphoid-Specific)/Lsh in human DNA causes a severe immunodeficiency syndrome, but the nature of the defect remains unknown. We assessed here the role of Lsh in hematopoiesis using conditional Lsh knockout mice with expression of Mx1 or Vav Cre-recombinase. Bone marrow transplantation studies revealed that Lsh depletion in hematopoietic stem cells severely reduced B cell numbers and impaired B cell development in a hematopoietic cell-autonomous manner. Lsh-deficient mice without bone marrow transplantation exhibited lower Ig levels in vivo compared to controls despite normal peripheral B cell numbers. Purified B lymphocytes proliferated normally but produced less immunoglobulins in response to in vitro stimulation, indicating a reduced capacity to undergo class switch recombination (CSR). Analysis of germline transcripts, examination of double-stranded breaks using biotin-labeling DNA break assay, and End-seq analysis indicated that the initiation of the recombination process was unscathed. In contrast, digestion-circularization PCR analysis and high-throughput sequencing analyses of CSR junctions and a chromosomal break repair assay indicated an impaired ability of the canonical end-joining pathway in Lsh-deficient B cells. Our data suggest a hematopoietic cell-intrinsic role of Lsh in B cell development and in CSR providing a potential target for immunodeficiency therapy.


Asunto(s)
Linfocitos B/fisiología , ADN Helicasas/metabolismo , Inmunoglobulinas/metabolismo , Animales , Línea Celular , ADN Helicasas/genética , Silenciador del Gen , Humanos , Inmunoglobulinas/genética , Ratones , Ratones Noqueados , Mutación
4.
EMBO J ; 39(14): e104036, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32484965

RESUMEN

Mechanistic understanding of how ionizing radiation induces type I interferon signaling and how to amplify this signaling module should help to maximize the efficacy of radiotherapy. In the current study, we report that inhibitors of the DNA damage response kinase ATR can significantly potentiate ionizing radiation-induced innate immune responses. Using a series of mammalian knockout cell lines, we demonstrate that, surprisingly, both the cGAS/STING-dependent DNA-sensing pathway and the MAVS-dependent RNA-sensing pathway are responsible for type I interferon signaling induced by ionizing radiation in the presence or absence of ATR inhibitors. The relative contributions of these two pathways in type I interferon signaling depend on cell type and/or genetic background. We propose that DNA damage-elicited double-strand DNA breaks releases DNA fragments, which may either activate the cGAS/STING-dependent pathway or-especially in the case of AT-rich DNA sequences-be transcribed and initiate MAVS-dependent RNA sensing and signaling. Together, our results suggest the involvement of two distinct pathways in type I interferon signaling upon DNA damage. Moreover, radiation plus ATR inhibition may be a promising new combination therapy against cancer.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/inmunología , Roturas del ADN de Doble Cadena/efectos de la radiación , Interferón Tipo I/inmunología , Radiación Ionizante , Transducción de Señal/efectos de la radiación , Proteínas de la Ataxia Telangiectasia Mutada/genética , Línea Celular Tumoral , Humanos , Interferón Tipo I/genética , Transducción de Señal/genética , Transducción de Señal/inmunología
5.
Clin Cancer Res ; 26(13): 3468-3480, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32253233

RESUMEN

PURPOSE: The standard treatment of patients with locally advanced rectal cancer consists of preoperative chemoradiotherapy (CRT) followed by surgery. However, the response of individual tumors to CRT is extremely diverse, presenting a clinical dilemma. This broad variability in treatment response is likely attributable to intratumor heterogeneity (ITH). EXPERIMENTAL DESIGN: We addressed the impact of ITH on response to CRT by establishing single-cell-derived cell lines (SCDCL) from a treatment-naïve rectal cancer biopsy after xenografting. RESULTS: Individual SCDCLs derived from the same tumor responded profoundly different to CRT in vitro. Clonal reconstruction of the tumor and derived cell lines based on whole-exome sequencing revealed nine separate clusters with distinct proportions in the SCDCLs. Missense mutations in SV2A and ZWINT were clonal in the resistant SCDCL, but not detected in the sensitive SCDCL. Single-cell genetic analysis by multiplex FISH revealed the expansion of a clone with a loss of PIK3CA in the resistant SCDCL. Gene expression profiling by tRNA-sequencing identified the activation of the Wnt, Akt, and Hedgehog signaling pathways in the resistant SCDCLs. Wnt pathway activation in the resistant SCDCLs was confirmed using a reporter assay. CONCLUSIONS: Our model system of patient-derived SCDCLs provides evidence for the critical role of ITH for treatment response in patients with rectal cancer and shows that distinct genetic aberration profiles are associated with treatment response. We identified specific pathways as the molecular basis of treatment response of individual clones, which could be targeted in resistant subclones of a heterogenous tumor.


Asunto(s)
Heterogeneidad Genética , Neoplasias del Recto/etiología , Neoplasias del Recto/patología , Análisis de la Célula Individual , Animales , Biomarcadores de Tumor , Línea Celular Tumoral , Terapia Combinada , Hibridación Genómica Comparativa , Modelos Animales de Enfermedad , Humanos , Inmunohistoquímica , Hibridación Fluorescente in Situ , Ratones , Neoplasias del Recto/metabolismo , Neoplasias del Recto/terapia , Transducción de Señal , Resultado del Tratamiento , Secuenciación del Exoma , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Cell ; 174(5): 1127-1142.e19, 2018 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-30078706

RESUMEN

Replication origins, fragile sites, and rDNA have been implicated as sources of chromosomal instability. However, the defining genomic features of replication origins and fragile sites are among the least understood elements of eukaryote genomes. Here, we map sites of replication initiation and breakage in primary cells at high resolution. We find that replication initiates between transcribed genes within nucleosome-depleted structures established by long asymmetrical poly(dA:dT) tracts flanking the initiation site. Paradoxically, long (>20 bp) (dA:dT) tracts are also preferential sites of polar replication fork stalling and collapse within early-replicating fragile sites (ERFSs) and late-replicating common fragile sites (CFSs) and at the rDNA replication fork barrier. Poly(dA:dT) sequences are fragile because long single-strand poly(dA) stretches at the replication fork are unprotected by the replication protein A (RPA). We propose that the evolutionary expansion of poly(dA:dT) tracts in eukaryotic genomes promotes replication initiation, but at the cost of chromosome fragility.


Asunto(s)
Replicación del ADN , ADN Ribosómico/química , Nucleosomas/metabolismo , Poli dA-dT/química , Origen de Réplica , Secuencias de Aminoácidos , Animales , Línea Celular , Inmunoprecipitación de Cromatina , Inestabilidad Cromosómica , Sitios Frágiles del Cromosoma , Fragilidad Cromosómica , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Saccharomyces cerevisiae , Schizosaccharomyces , Sitio de Iniciación de la Transcripción , Transcripción Genética
8.
Elife ; 62017 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-28362262

RESUMEN

Macrophages produce genotoxic agents, such as reactive oxygen and nitrogen species, that kill invading pathogens. Here we show that these agents activate the DNA damage response (DDR) kinases ATM and DNA-PKcs through the generation of double stranded breaks (DSBs) in murine macrophage genomic DNA. In contrast to other cell types, initiation of this DDR depends on signaling from the type I interferon receptor. Once activated, ATM and DNA-PKcs regulate a genetic program with diverse immune functions and promote inflammasome activation and the production of IL-1ß and IL-18. Indeed, following infection with Listeria monocytogenes, DNA-PKcs-deficient murine macrophages produce reduced levels of IL-18 and are unable to optimally stimulate IFN-γ production by NK cells. Thus, genomic DNA DSBs act as signaling intermediates in murine macrophages, regulating innate immune responses through the initiation of a type I IFN-dependent DDR.


Asunto(s)
Regulación de la Expresión Génica , Inmunidad Innata , Inflamasomas/metabolismo , Interferón Tipo I/metabolismo , Listeria monocytogenes/inmunología , Macrófagos/inmunología , Animales , Roturas del ADN de Doble Cadena , Daño del ADN , Ratones , Proteínas Quinasas/metabolismo
9.
Cell ; 168(4): 644-656, 2017 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-28187286

RESUMEN

Genome instability, defined as higher than normal rates of mutation, is a double-edged sword. As a source of genetic diversity and natural selection, mutations are beneficial for evolution. On the other hand, genomic instability can have catastrophic consequences for age-related diseases such as cancer. Mutations arise either from inactivation of DNA repair pathways or in a repair-competent background due to genotoxic stress from celluar processes such as transcription and replication that overwhelm high-fidelity DNA repair. Here, we review recent studies that shed light on endogenous sources of mutation and epigenomic features that promote genomic instability during cancer evolution.


Asunto(s)
Daño del ADN , Inestabilidad Genómica , Neoplasias/genética , Cromatina/química , Reparación del ADN , Replicación del ADN , Humanos , Mutación , Activación Transcripcional
10.
Cell ; 167(6): 1571-1585.e18, 2016 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-27839864

RESUMEN

Cell migration in confined 3D tissue microenvironments is critical for both normal physiological functions and dissemination of tumor cells. We discovered a cytoskeletal structure that prevents damage to the nucleus during migration in confined microenvironments. The formin-family actin filament nucleator FMN2 associates with and generates a perinuclear actin/focal adhesion (FA) system that is distinct from previously characterized actin/FA structures. This system controls nuclear shape and positioning in cells migrating on 2D surfaces. In confined 3D microenvironments, FMN2 promotes cell survival by limiting nuclear envelope damage and DNA double-strand breaks. We found that FMN2 is upregulated in human melanomas and showed that disruption of FMN2 in mouse melanoma cells inhibits their extravasation and metastasis to the lung. Our results indicate a critical role for FMN2 in generating a perinuclear actin/FA system that protects the nucleus and DNA from damage to promote cell survival during confined migration and thus promote cancer metastasis.


Asunto(s)
Núcleo Celular/metabolismo , Adhesiones Focales , Neoplasias Pulmonares/secundario , Melanoma/patología , Proteínas de Microfilamentos/metabolismo , Metástasis de la Neoplasia , Proteínas Nucleares/metabolismo , Actinas/metabolismo , Animales , Roturas del ADN de Doble Cadena , Embrión de Mamíferos/citología , Matriz Extracelular/metabolismo , Femenino , Forminas , Humanos , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso
11.
Sci Signal ; 9(445): ra91, 2016 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-27625305

RESUMEN

Among the various subtypes of acute myeloid leukemia (AML), those with chromosomal rearrangements of the MLL oncogene (AML-MLL) have a poor prognosis. AML-MLL tumor cells are resistant to current genotoxic therapies because of an attenuated response by p53, a protein that induces cell cycle arrest and apoptosis in response to DNA damage. In addition to chemicals that damage DNA, efforts have focused on targeting DNA repair enzymes as a general chemotherapeutic approach to cancer treatment. Here, we found that inhibition of the kinase ATR, which is the primary sensor of DNA replication stress, induced chromosomal breakage and death of mouse AML(MLL) cells (with an MLL-ENL fusion and a constitutively active N-RAS independently of p53. Moreover, ATR inhibition as a single agent exhibited antitumoral activity, both reducing tumor burden after establishment and preventing tumors from growing, in an immunocompetent allograft mouse model of AML(MLL) and in xenografts of a human AML-MLL cell line. We also found that inhibition of ATM, a kinase that senses DNA double-strand breaks, also promoted the survival of the AML(MLL) mice. Collectively, these data indicated that ATR or ATM inhibition represent potential therapeutic strategies for the treatment of AML, especially MLL-driven leukemias.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada , Reordenamiento Génico , N-Metiltransferasa de Histona-Lisina , Leucemia Mieloide Aguda , Proteína de la Leucemia Mieloide-Linfoide , Neoplasias Experimentales , Animales , Proteínas de la Ataxia Telangiectasia Mutada/antagonistas & inhibidores , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/enzimología , Leucemia Mieloide Aguda/genética , Ratones , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/enzimología , Neoplasias Experimentales/genética , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Mol Cell ; 63(5): 898-911, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27477910

RESUMEN

DNA double-strand breaks (DSBs) arise during physiological transcription, DNA replication, and antigen receptor diversification. Mistargeting or misprocessing of DSBs can result in pathological structural variation and mutation. Here we describe a sensitive method (END-seq) to monitor DNA end resection and DSBs genome-wide at base-pair resolution in vivo. We utilized END-seq to determine the frequency and spectrum of restriction-enzyme-, zinc-finger-nuclease-, and RAG-induced DSBs. Beyond sequence preference, chromatin features dictate the repertoire of these genome-modifying enzymes. END-seq can detect at least one DSB per cell among 10,000 cells not harboring DSBs, and we estimate that up to one out of 60 cells contains off-target RAG cleavage. In addition to site-specific cleavage, we detect DSBs distributed over extended regions during immunoglobulin class-switch recombination. Thus, END-seq provides a snapshot of DNA ends genome-wide, which can be utilized for understanding genome-editing specificities and the influence of chromatin on DSB pathway choice.


Asunto(s)
Cromatina/química , Roturas del ADN de Doble Cadena , ADN/genética , Genoma , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Animales , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/inmunología , Linfocitos B/citología , Linfocitos B/inmunología , Cromatina/inmunología , ADN/inmunología , Replicación del ADN , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Regulación de la Expresión Génica , Histonas/genética , Histonas/inmunología , Cambio de Clase de Inmunoglobulina/genética , Ratones , Células Precursoras de Linfocitos B/citología , Células Precursoras de Linfocitos B/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Recombinación Genética , Timocitos/citología , Timocitos/inmunología
13.
Nat Commun ; 7: 12425, 2016 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-27498558

RESUMEN

Poly (ADP-ribose) polymerase (PARP) inhibitor (PARPi) olaparib has been approved for treatment of advanced ovarian cancer associated with BRCA1 and BRCA2 mutations. BRCA1- and BRCA2-mutated cells, which are homologous recombination (HR) deficient, are hypersensitive to PARPi through the mechanism of synthetic lethality. Here we examine the effect of PARPi on HR-proficient cells. Olaparib pretreatment, PARP1 knockdown or Parp1 heterozygosity of Brca2(cko/ko) mouse embryonic stem cells (mESCs), carrying a null (ko) and a conditional (cko) allele of Brca2, results in viable Brca2(ko/ko) cells. PARP1 deficiency does not restore HR in Brca2(ko/ko) cells, but protects stalled replication forks from MRE11-mediated degradation through its impaired recruitment. The functional consequence of Parp1 heterozygosity on BRCA2 loss is demonstrated by a significant increase in tumorigenesis in Brca2(cko/cko) mice. Thus, while olaparib efficiently kills BRCA2-deficient cells, we demonstrate that it can also contribute to the synthetic viability if PARP is inhibited before BRCA2 loss.


Asunto(s)
Proteína BRCA2/deficiencia , Poli(ADP-Ribosa) Polimerasa-1/deficiencia , Animales , Proteína BRCA2/metabolismo , Supervivencia Celular/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Recombinación Homóloga/efectos de los fármacos , Humanos , Integrasas/metabolismo , Proteína Homóloga de MRE11/metabolismo , Ratones , Modelos Biológicos , Células Madre Embrionarias de Ratones/citología , Células Madre Embrionarias de Ratones/efectos de los fármacos , Células Madre Embrionarias de Ratones/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/antagonistas & inhibidores , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología
14.
Mol Cell ; 56(6): 808-18, 2014 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-25435138

RESUMEN

The structure of broken DNA ends is a critical determinant of the pathway used for DNA double-strand break (DSB) repair. Here, we develop an approach involving the hairpin capture of DNA end structures (HCoDES), which elucidates chromosomal DNA end structures at single-nucleotide resolution. HCoDES defines structures of physiologic DSBs generated by the RAG endonuclease, as well as those generated by nucleases widely used for genome editing. Analysis of G1 phase cells deficient in H2AX or 53BP1 reveals DNA ends that are frequently resected to form long single-stranded overhangs that can be repaired by mutagenic pathways. In addition to 3' overhangs, many of these DNA ends unexpectedly form long 5' single-stranded overhangs. The divergence in DNA end structures resolved by HCoDES suggests that H2AX and 53BP1 may have distinct activities in end protection. Thus, the high-resolution end structures obtained by HCoDES identify features of DNA end processing during DSB repair.


Asunto(s)
Cromosomas Humanos/genética , Análisis de Secuencia de ADN/métodos , Secuencia de Bases , Células Cultivadas , Roturas del ADN de Doble Cadena , Humanos
16.
Mol Cell Biol ; 34(15): 2811-21, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24842905

RESUMEN

The resection of broken DNA ends is required for DNA double-strand break (DSB) repair by homologous recombination (HR) but can inhibit normal repair by nonhomologous end joining (NHEJ), the main DSB repair pathway in G1-phase cells. Antigen receptor gene assembly proceeds through DNA DSB intermediates generated in G1-phase lymphocytes by the RAG endonuclease. These DSBs activate ATM, which phosphorylates H2AX, forming γ-H2AX in flanking chromatin. γ-H2AX prevents CtIP from initiating resection of RAG DSBs. Whether there are additional proteins required to promote resection of these DNA ends is not known. KRAB-associated protein 1 (KAP-1) (TRIM28) is a transcriptional repressor that modulates chromatin structure and has been implicated in the repair of DNA DSBs in heterochromatin. Here, we show that in murine G1-phase lymphocytes, KAP-1 promotes resection of DSBs that are not protected by H2AX and its downstream effector 53BP1. In these murine cells, KAP-1 activity in DNA end resection is attenuated by a single-amino-acid change that reflects a KAP-1 polymorphism between primates and other mammalian species. These findings establish KAP-1 as a component of the machinery that can resect DNA ends in G1-phase cells and suggest that there may be species-specific features to this activity.


Asunto(s)
Reparación del ADN por Unión de Extremidades/genética , Proteínas de Unión al ADN/metabolismo , Fase G1/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Linfocitos/metabolismo , Animales , Células Cultivadas , ADN/genética , Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN/genética , Heterocromatina/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Ratones Endogámicos C57BL , Fosforilación/genética
17.
Mol Cell ; 54(6): 1022-1033, 2014 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-24837676

RESUMEN

The carboxy-terminal binding protein (CtBP)-interacting protein (CtIP) is known to function in 5' strand resection during homologous recombination, similar to the budding yeast Sae2 protein, but its role in this process is unclear. Here, we characterize recombinant human CtIP and find that it exhibits 5' flap endonuclease activity on branched DNA structures, independent of the MRN complex. Phosphorylation of CtIP at known damage-dependent sites and other sites is essential for its catalytic activity, although the S327 and T847 phosphorylation sites are dispensable. A catalytic mutant of CtIP that is deficient in endonuclease activity exhibits wild-type levels of homologous recombination at restriction enzyme-generated breaks but is deficient in processing topoisomerase adducts and radiation-induced breaks in human cells, suggesting that the nuclease activity of CtIP is specifically required for the removal of DNA adducts at sites of DNA breaks.


Asunto(s)
Proteínas Portadoras/metabolismo , Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades/genética , Endonucleasas/metabolismo , Proteínas Nucleares/metabolismo , Reparación del ADN por Recombinación/genética , Sitios de Unión/genética , Proteínas Portadoras/genética , Catálisis , Línea Celular , Supervivencia Celular/genética , ADN/genética , Proteínas de Unión al ADN/genética , Endodesoxirribonucleasas , Endonucleasas/genética , Humanos , Proteínas Nucleares/genética , Fosforilación/genética , Procesamiento Proteico-Postraduccional/genética , Radiación Ionizante , Recombinación Genética
18.
Cell ; 153(6): 1266-80, 2013 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-23727112

RESUMEN

The DNA damage response (DDR) protein 53BP1 protects DNA ends from excessive resection in G1, and thereby favors repair by nonhomologous end-joining (NHEJ) as opposed to homologous recombination (HR). During S phase, BRCA1 antagonizes 53BP1 to promote HR. The pro-NHEJ and antirecombinase functions of 53BP1 are mediated in part by RIF1, the only known factor that requires 53BP1 phosphorylation for its recruitment to double-strand breaks (DSBs). Here, we show that a 53BP1 phosphomutant, 53BP18A, comprising alanine substitutions of the eight most N-terminal S/TQ phosphorylation sites, mimics 53BP1 deficiency by restoring genome stability in BRCA1-deficient cells yet behaves like wild-type 53BP1 with respect to immunoglobulin class switch recombination (CSR). 53BP18A recruits RIF1 but fails to recruit the DDR protein PTIP to DSBs, and disruption of PTIP phenocopies 53BP18A. We conclude that 53BP1 promotes productive CSR and suppresses mutagenic DNA repair through distinct phosphodependent interactions with RIF1 and PTIP.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Reparación del ADN por Unión de Extremidades , Proteínas de Unión al ADN/metabolismo , Cambio de Clase de Inmunoglobulina , Proteínas Nucleares/metabolismo , Proteínas de Unión a Telómeros/metabolismo , Animales , Linfocitos B/metabolismo , Proteína BRCA1/metabolismo , Proteínas Cromosómicas no Histona/genética , Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN/genética , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Fibroblastos/metabolismo , Inestabilidad Genómica , Ratones , Mutación , Proteína 1 de Unión al Supresor Tumoral P53
19.
J Exp Med ; 210(2): 233-9, 2013 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-23382544

RESUMEN

Allelic exclusion is enforced through the ability of antigen receptor chains expressed from one allele to signal feedback inhibition of V-to-(D)J recombination on the other allele. To achieve allelic exclusion by such means, only one allele can initiate V-to-(D)J recombination within the time required to signal feedback inhibition. DNA double-strand breaks (DSBs) induced by the RAG endonuclease during V(D)J recombination activate the Ataxia Telangiectasia mutated (ATM) and DNA-dependent protein kinase (DNA-PK) kinases. We demonstrate that ATM enforces Igκ allelic exclusion, and that RAG DSBs induced during Igκ recombination in primary pre-B cells signal through ATM, but not DNA-PK, to suppress initiation of additional Igκ rearrangements. ATM promotes high-density histone H2AX phosphorylation to create binding sites for MDC1, which functions with H2AX to amplify a subset of ATM-dependent signals. However, neither H2AX nor MDC1 is required for ATM to enforce Igκ allelic exclusion and suppress Igκ rearrangements. Upon activation in response to RAG Igκ cleavage, ATM signals down-regulation of Gadd45α with concomitant repression of the Gadd45α targets Rag1 and Rag2. Our data indicate that ATM kinases activated by RAG DSBs during Igκ recombination transduce transient H2AX/MDC1-independent signals that suppress initiation of further Igκ rearrangements to control Igκ allelic exclusion.


Asunto(s)
Proteínas de Ciclo Celular/inmunología , Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/inmunología , Proteínas de Unión al ADN/metabolismo , Reordenamiento Génico de Cadena Ligera de Linfocito B , Proteínas Serina-Treonina Quinasas/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Supresoras de Tumor/inmunología , Proteínas Supresoras de Tumor/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Alelos , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Linfocitos B/inmunología , Linfocitos B/metabolismo , Secuencia de Bases , Proteínas de Ciclo Celular/genética , Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN/genética , Histonas/deficiencia , Histonas/genética , Histonas/metabolismo , Proteínas de Homeodominio/metabolismo , Cadenas kappa de Inmunoglobulina/genética , Cadenas kappa de Inmunoglobulina/metabolismo , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Ratones de la Cepa 129 , Ratones Noqueados , Modelos Biológicos , Proteínas Serina-Treonina Quinasas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Proteínas Supresoras de Tumor/genética
20.
Nature ; 469(7329): 245-9, 2011 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-21160476

RESUMEN

DNA double-strand breaks (DSBs) are generated by the recombination activating gene (RAG) endonuclease in all developing lymphocytes as they assemble antigen receptor genes. DNA cleavage by RAG occurs only at the G1 phase of the cell cycle and generates two hairpin-sealed DNA (coding) ends that require nucleolytic opening before their repair by classical non-homologous end-joining (NHEJ). Although there are several cellular nucleases that could perform this function, only the Artemis nuclease is able to do so efficiently. Here, in vivo, we show that in murine cells the histone protein H2AX prevents nucleases other than Artemis from processing hairpin-sealed coding ends; in the absence of H2AX, CtIP can efficiently promote the hairpin opening and resection of DNA ends generated by RAG cleavage. This CtIP-mediated resection is inhibited by γ-H2AX and by MDC-1 (mediator of DNA damage checkpoint 1), which binds to γ-H2AX in chromatin flanking DNA DSBs. Moreover, the ataxia telangiectasia mutated (ATM) kinase activates antagonistic pathways that modulate this resection. CtIP DNA end resection activity is normally limited to cells at post-replicative stages of the cell cycle, in which it is essential for homology-mediated repair. In G1-phase lymphocytes, DNA ends that are processed by CtIP are not efficiently joined by classical NHEJ and the joints that do form frequently use micro-homologies and show significant chromosomal deletions. Thus, H2AX preserves the structural integrity of broken DNA ends in G1-phase lymphocytes, thereby preventing these DNA ends from accessing repair pathways that promote genomic instability.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/metabolismo , Roturas del ADN de Doble Cadena , Reparación del ADN , Fase G1 , Reordenamiento Génico de Linfocito B , Histonas/metabolismo , Linfocitos/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Línea Celular Transformada , Cromatina/metabolismo , Proteínas de Unión al ADN/metabolismo , Endonucleasas , Reordenamiento Génico de Linfocito B/genética , Inestabilidad Genómica , Histonas/deficiencia , Histonas/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Linfocitos/citología , Ratones , Proteínas Nucleares , Células Precursoras de Linfocitos B/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Recombinación Genética/genética , Especificidad por Sustrato , Proteínas Supresoras de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...