Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Nature ; 528(7582): 422-6, 2015 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-26649820

RESUMEN

DNA repair by homologous recombination is highly suppressed in G1 cells to ensure that mitotic recombination occurs solely between sister chromatids. Although many homologous recombination factors are cell-cycle regulated, the identity of the events that are both necessary and sufficient to suppress recombination in G1 cells is unknown. Here we report that the cell cycle controls the interaction of BRCA1 with PALB2-BRCA2 to constrain BRCA2 function to the S/G2 phases in human cells. We found that the BRCA1-interaction site on PALB2 is targeted by an E3 ubiquitin ligase composed of KEAP1, a PALB2-interacting protein, in complex with cullin-3 (CUL3)-RBX1 (ref. 6). PALB2 ubiquitylation suppresses its interaction with BRCA1 and is counteracted by the deubiquitylase USP11, which is itself under cell cycle control. Restoration of the BRCA1-PALB2 interaction combined with the activation of DNA-end resection is sufficient to induce homologous recombination in G1, as measured by RAD51 recruitment, unscheduled DNA synthesis and a CRISPR-Cas9-based gene-targeting assay. We conclude that the mechanism prohibiting homologous recombination in G1 minimally consists of the suppression of DNA-end resection coupled with a multi-step block of the recruitment of BRCA2 to DNA damage sites that involves the inhibition of BRCA1-PALB2-BRCA2 complex assembly. We speculate that the ability to induce homologous recombination in G1 cells with defined factors could spur the development of gene-targeting applications in non-dividing cells.


Asunto(s)
Fase G1 , Recombinación Homóloga , Secuencia de Aminoácidos , Proteína BRCA1/metabolismo , Proteína BRCA2/metabolismo , Sistemas CRISPR-Cas/genética , Proteínas Portadoras/metabolismo , Línea Celular , Proteínas Cullin/metabolismo , ADN/metabolismo , Daño del ADN , Reparación del ADN , Proteína del Grupo de Complementación N de la Anemia de Fanconi , Fase G2 , Marcación de Gen , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteína 1 Asociada A ECH Tipo Kelch , Datos de Secuencia Molecular , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Unión Proteica , Recombinasa Rad51/metabolismo , Fase S , Tioléster Hidrolasas/metabolismo , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación
2.
Haematologica ; 105(10): 2457-2470, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-33054086

RESUMEN

GFI1B-related thrombocytopenia (GFI1B-RT) is a rare bleeding disorder mainly caused by the presence of truncated GFI1B proteins with dominant-negative properties. The disease is characterized by low platelet counts, the presence of abnormal platelets, a megakaryocytic expansion and mild erythroid defects. However, no animal models faithfully reproducing the GFI1B-RT phenotype observed in patients exist. We had previously generated mice with floxed Gfi1b alleles that can be eliminated by Cre recombinase, but those animals developed a much more severe phenotype than GFI1B-RT patients and were of limited interest in assessing the disease. Using CRISPR/Cas9 technology, we have now established three independent mouse lines that carry mutated Gfi1b alleles producing proteins lacking DNA binding zinc fingers and thereby acting in a dominant negative (DN) manner. Mice heterozygous for these Gfi1b-DN alleles show reduced platelet counts and an expansion of megakaryocytes similar to features of human GFI1B-RT but lacking the distinctively large agranular platelets. In addition, Gfi1b-DN mice exhibit an expansion of erythroid precursors indicative of a mildly abnormal erythropoiesis but without noticeable red blood cell defects. When associated with megakaryocyte-specific ablation of the remaining allele, the Gfi1b-DN alleles triggered erythroid-specific deleterious defects. Gfi1b-DN mice also showed a delayed recovery from platelet depletion, indicating a defect in stress thrombopoiesis. However, injecting Gfi1b-DN mice with romiplostim, a thrombopoietin receptor super agonist, increased platelet numbers even beyond normal levels. Thus, our data support a causal link between DN mutations in GFI1B and thrombocytopenia and suggest that patients with GFI1B-RT could be treated successfully with thrombopoietin agonists.


Asunto(s)
Trombocitopenia , Trombopoyesis , Animales , Plaquetas , Eritropoyesis , Humanos , Megacariocitos , Ratones , Mutación , Proteínas Proto-Oncogénicas/genética , Proteínas Represoras/genética , Trombocitopenia/genética , Trombopoyesis/genética
3.
Nucleic Acids Res ; 45(18): 10518-10533, 2017 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-29048592

RESUMEN

The yeast 2-µm plasmid is a remarkable genetic parasite, managing efficient maintenance at high-copy number with minimal impact on the host. Equal partitioning of the plasmid upon host cell division requires plasmid proteins Rep1 and Rep2 and the plasmid STB locus. The Rep proteins and the plasmid-encoded Raf protein also regulate plasmid gene transcription. In this study, protein interaction assays, sequence analyses and mutational approaches were used to identify domains and residues in Rep2 and Raf required for association with Rep1 and Rep2 and to delineate the Rep2 DNA-binding domain. Rep2 and Raf displayed similarities in interactions with Rep1 and Rep2, in having Rep1 promote their STB association in vivo, and in stabilizing Rep protein levels. Rep2 mutants impaired for self-association were competent for transcriptional repression while those deficient for Rep1 association were not. Surprisingly, Rep2 mutants impaired for either Rep1 interaction or self-association were able to maintain efficient plasmid inheritance provided Raf was present and competent for Rep protein interaction. Our findings provide insight into the Rep protein complexes required for partitioning and transcriptional repression, and suggest that in addition to its transcriptional function, Raf stabilization of Rep partitioning proteins contributes to the remarkable persistence of the 2-µm plasmid.


Asunto(s)
Plásmidos/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Transactivadores/metabolismo , Quinasas raf/metabolismo , Quinasas raf/fisiología , División Celular , Patrón de Herencia , Organismos Modificados Genéticamente , Unión Proteica , Estabilidad Proteica
4.
Nucleic Acids Res ; 43(19): 9379-92, 2015 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-26429972

RESUMEN

CRISPR is a genome-editing platform that makes use of the bacterially-derived endonuclease Cas9 to introduce DNA double-strand breaks at precise locations in the genome using complementary guide RNAs. We developed a nuclear domain knock-in screen, whereby the insertion of a gene encoding the green fluorescent protein variant Clover is inserted by Cas9-mediated homology directed repair (HDR) within the first exon of genes that are required for the structural integrity of subnuclear domains such as the nuclear lamina and promyelocytic leukemia nuclear bodies (PML NBs). Using this approach, we compared strategies for enhancing CRISPR-mediated HDR, focusing on known genes and small molecules that impact non-homologous end joining (NHEJ) and homologous recombination (HR). Ultimately, we identified the small molecule RS-1 as a potent enhancer of CRISPR-based genome editing, enhancing HDR 3- to 6-fold depending on the locus and transfection method. We also characterized U2OS human osteosarcoma cells expressing Clover-tagged PML and demonstrate that this strategy generates cell lines with PML NBs that are structurally and functionally similar to bodies in the parental cell line. Thus, the nuclear domain knock-in screen that we describe provides a simple means of rapidly evaluating methods and small molecules that have the potential to enhance Cas9-mediated HDR.


Asunto(s)
Sistemas CRISPR-Cas , Endodesoxirribonucleasas/metabolismo , Técnicas de Sustitución del Gen/métodos , Reparación del ADN por Recombinación , Proteína BRCA1/metabolismo , Benzamidas/farmacología , Línea Celular Tumoral , Roturas del ADN de Doble Cadena , Citometría de Flujo , Genoma , Proteínas Fluorescentes Verdes/genética , Células HEK293 , Humanos , Proteínas Nucleares/análisis , Proteínas Nucleares/genética , Oligonucleótidos , Pirimidinas/farmacología , ARN/metabolismo , Recombinasa Rad51/metabolismo , Bases de Schiff/farmacología , Sulfonamidas/farmacología
5.
Haematologica ; 100(1): 70-6, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25281505

RESUMEN

Cancer therapeutics is evolving to precision medicine, with the goal of matching targeted compounds with molecular aberrations underlying a patient's cancer. While murine models offer a pre-clinical tool, associated costs and time are not compatible with actionable patient-directed interventions. Using the paradigm of T-cell acute lymphoblastic leukemia, a high-risk disease with defined molecular underpinnings, we developed a zebrafish human cancer xenotransplantation model to inform therapeutic decisions. Using a focused chemical genomic approach, we demonstrate that xenografted cell lines harboring mutations in the NOTCH1 and PI3K/AKT pathways respond concordantly to their targeted therapies, patient-derived T-cell acute lymphoblastic leukemia can be successfully engrafted in zebrafish and specific drug responses can be quantitatively determined. Using this approach, we identified a mutation sensitive to γ-secretase inhibition in a xenograft from a child with T-cell acute lymphoblastic leukemia, confirmed by Sanger sequencing and validated as a gain-of-function NOTCH1 mutation. The zebrafish xenotransplantation platform provides a novel cost-effective means of tailoring leukemia therapy in real time.


Asunto(s)
Antineoplásicos/farmacología , Embrión no Mamífero/efectos de los fármacos , Genómica/métodos , Mutación/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Pez Cebra/genética , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Animales , Células Cultivadas , Niño , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Inhibidores Enzimáticos/farmacología , Técnica del Anticuerpo Fluorescente , Células HeLa , Humanos , Fosfohidrolasa PTEN/genética , Fosfatidilinositol 3-Quinasas/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Proteínas Proto-Oncogénicas c-akt/genética , Receptor Notch1/genética , Transducción de Señal , Trasplante Heterólogo , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo
6.
Mol Cell Proteomics ; 12(11): 3253-70, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23938469

RESUMEN

Stress-inducible phosphoprotein 1 (STI1), a cochaperone for Hsp90, has been shown to regulate multiple pathways in astrocytes, but its contributions to cellular stress responses are not fully understood. We show that in response to irradiation-mediated DNA damage stress STI1 accumulates in the nucleus of astrocytes. Also, STI1 haploinsufficiency decreases astrocyte survival after irradiation. Using yeast two-hybrid screenings we identified several nuclear proteins as STI1 interactors. Overexpression of one of these interactors, PIAS1, seems to be specifically involved in STI1 nuclear retention and in directing STI1 and Hsp90 to specific sub-nuclear regions. PIAS1 and STI1 co-immunoprecipitate and PIAS1 can function as an E3 SUMO ligase for STI. Using mass spectrometry we identified five SUMOylation sites in STI1. A STI1 mutant lacking these five sites is not SUMOylated, but still accumulates in the nucleus in response to increased expression of PIAS1, suggesting the possibility that a direct interaction with PIAS1 could be responsible for STI1 nuclear retention. To test this possibility, we mapped the interaction sites between PIAS1 and STI1 using yeast-two hybrid assays and surface plasmon resonance and found that a large domain in the N-terminal region of STI1 interacts with high affinity with amino acids 450-480 of PIAS1. Knockdown of PIAS1 in astrocytes impairs the accumulation of nuclear STI1 in response to irradiation. Moreover, a PIAS1 mutant lacking the STI1 binding site is unable to increase STI1 nuclear retention. Interestingly, in human glioblastoma multiforme PIAS1 expression is increased and we found a significant correlation between increased PIAS1 expression and STI1 nuclear localization. These experiments provide evidence that direct interaction between STI1 and PIAS1 is involved in the accumulation of nuclear STI1. This retention mechanism could facilitate nuclear chaperone activity.


Asunto(s)
Astrocitos/metabolismo , Proteínas de Choque Térmico/metabolismo , Proteínas Inhibidoras de STAT Activados/metabolismo , Animales , Astrocitos/citología , Astrocitos/efectos de la radiación , Muerte Celular/genética , Muerte Celular/fisiología , Muerte Celular/efectos de la radiación , Núcleo Celular/metabolismo , Células Cultivadas , Daño del ADN , Rayos gamma , Técnicas de Silenciamiento del Gen , Células HEK293 , Haploinsuficiencia , Proteínas de Choque Térmico/deficiencia , Proteínas de Choque Térmico/genética , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Inhibidoras de STAT Activados/antagonistas & inhibidores , Proteínas Inhibidoras de STAT Activados/genética , Mapas de Interacción de Proteínas , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Estrés Fisiológico , Sumoilación , Técnicas del Sistema de Dos Híbridos
7.
Exp Cell Res ; 319(17): 2554-65, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24036361

RESUMEN

The promyelocytic leukemia protein (PML) is a tumor suppressor protein that regulates a variety of important cellular processes, including gene expression, DNA repair and cell fate decisions. Integral to its function is the ability of PML to form nuclear bodies (NBs) that serve as hubs for the interaction and modification of over 90 cellular proteins. There are seven canonical isoforms of PML, which encode diverse C-termini generated by alternative pre-mRNA splicing. Recruitment of specific cellular proteins to PML NBs is mediated by protein-protein interactions with individual PML isoforms. Using a yeast two-hybrid screen employing peptide sequences unique to PML isoform I (PML-I), we identified an interaction with the eukaryotic initiation factor 3 subunit K (eIF3K), and in the process identified a novel eIF3K isoform, which we term eIF3K-2. We further demonstrate that eIF3K and PML interact both in vitro via pull-down assays, as well as in vivo within human cells by co-immunoprecipitation and co-immunofluorescence. In addition, eIF3K isoform 2 (eIF3K-2) colocalizes to PML bodies, particularly those enriched in PML-I, while eIF3K isoform 1 associates poorly with PML NBs. Thus, we report eIF3K as the first known subunit of the eIF3 translation pre-initiation complex to interact directly with the PML protein, and provide data implicating alternative splicing of both PML and eIF3K as a possible regulatory mechanism for eIF3K localization at PML NBs.


Asunto(s)
Estructuras del Núcleo Celular/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Empalme Alternativo , Células HEK293 , Células HeLa , Humanos , Proteínas Asociadas a Microtúbulos/genética , Proteínas Nucleares/genética , Proteína de la Leucemia Promielocítica , Unión Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Factores de Transcripción/genética , Proteínas Supresoras de Tumor/genética , Técnicas del Sistema de Dos Híbridos
8.
Mol Cell Biol ; 25(10): 4299-310, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15870298

RESUMEN

The 2 microm circle plasmid confers no phenotype in wild-type Saccharomyces cerevisiae but in a nib1 mutant, an elevated plasmid copy number is associated with cell death. Complementation was used to identify nib1 as a mutant allele of the ULP1 gene that encodes a protease required for removal of a ubiquitin-like protein, Smt3/SUMO, from protein substrates. The nib1 mutation replaces conserved tryptophan 490 with leucine in the protease domain of Ulp1. Complete deletion of ULP1 is lethal, even in a strain that lacks the 2 microm circle. Partial deletion of ULP1, like the nib1 mutation, results in clonal variations in plasmid copy number. In addition, a subset of these mutant cells produces lineages in which all cells have reduced proliferative capacity, and this phenotype is dependent upon the presence of the 2 microm circle. Segregation of the 2 microm circle requires two plasmid-encoded proteins, Rep1 and Rep2, which were found to colocalize with Ulp1 protein in the nucleus and interact with Smt3 in a two-hybrid assay. These associations and the observation of missegregation of a fluorescently tagged 2 microm circle reporter plasmid in a subset of ulp1 mutant cells suggest that Smt3 modification plays a role in both plasmid copy number control and segregation.


Asunto(s)
Cisteína Endopeptidasas/genética , Mutación/genética , Plásmidos/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/genética , Alelos , División Celular , Núcleo Celular/metabolismo , Proliferación Celular , Segregación Cromosómica , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Fase G2 , Dosificación de Gen , Genes Reporteros/genética , Fenotipo , Plásmidos/genética , Unión Proteica , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Saccharomyces cerevisiae/enzimología , Proteínas de Saccharomyces cerevisiae/metabolismo , Eliminación de Secuencia/genética , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina , Técnicas del Sistema de Dos Híbridos
9.
Nat Commun ; 9(1): 1418, 2018 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-29651020

RESUMEN

GFI1 is a transcriptional regulator expressed in lymphoid cells, and an "oncorequisite" factor required for development and maintenance of T-lymphoid leukemia. GFI1 deletion causes hypersensitivity to ionizing radiation, for which the molecular mechanism remains unknown. Here, we demonstrate that GFI1 is required in T cells for the regulation of key DNA damage signaling and repair proteins. Specifically, GFI1 interacts with the arginine methyltransferase PRMT1 and its substrates MRE11 and 53BP1. We demonstrate that GFI1 enables PRMT1 to bind and methylate MRE11 and 53BP1, which is necessary for their function in the DNA damage response. Thus, our results provide evidence that GFI1 can adopt non-transcriptional roles, mediating the post-translational modification of proteins involved in DNA repair. These findings have direct implications for treatment responses in tumors overexpressing GFI1 and suggest that GFI1's activity may be a therapeutic target in these malignancies.


Asunto(s)
Linfocitos T CD4-Positivos/efectos de la radiación , Reparación del ADN , Proteínas de Unión al ADN/metabolismo , Proteína Homóloga de MRE11/metabolismo , Procesamiento Proteico-Postraduccional , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/metabolismo , Línea Celular , Daño del ADN , Proteínas de Unión al ADN/genética , Rayos gamma , Humanos , Células Jurkat , Proteína Homóloga de MRE11/genética , Metilación , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína-Arginina N-Metiltransferasas/genética , Proteínas Represoras/genética , Transducción de Señal , Factores de Transcripción/genética , Transcripción Genética , Proteína 1 de Unión al Supresor Tumoral P53/genética
10.
Oncotarget ; 8(12): 19021-19038, 2017 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-28147323

RESUMEN

Ionizing radiation generates a broad spectrum of oxidative DNA lesions, including oxidized base products, abasic sites, single-strand breaks and double-strand breaks. The CUX1 protein was recently shown to function as an auxiliary factor that stimulates enzymatic activities of OGG1 through its CUT domains. In the present study, we investigated the requirement for CUX1 and OGG1 in the resistance to radiation. Cancer cell survival following ionizing radiation is reduced by CUX1 knockdown and increased by higher CUX1 expression. However, CUX1 knockdown is sufficient by itself to reduce viability in many cancer cell lines that exhibit high levels of reactive oxygen species (ROS). Consequently, clonogenic results expressed relative to that of non-irradiated cells indicate that CUX1 knockdown confers no or modest radiosensitivity to cancer cells with high ROS. A recombinant protein containing only two CUT domains is sufficient for rapid recruitment to DNA damage, acceleration of DNA repair and increased survival following radiation. In agreement with these findings, OGG1 knockdown and treatment of cells with OGG1 inhibitors sensitize cancer cells to radiation. Together, these results validate CUX1 and more specifically the CUT domains as therapeutic targets.


Asunto(s)
Reparación del ADN/genética , Proteínas de Homeodominio/metabolismo , Proteínas Nucleares/metabolismo , Tolerancia a Radiación/genética , Proteínas Represoras/metabolismo , Línea Celular Tumoral , ADN Glicosilasas/metabolismo , Técnicas de Silenciamiento del Gen , Proteínas de Homeodominio/genética , Humanos , Immunoblotting , Microscopía Confocal , Proteínas Nucleares/genética , Reacción en Cadena de la Polimerasa , Especies Reactivas de Oxígeno/metabolismo , Proteínas Represoras/genética , Factores de Transcripción
11.
Cancer Discov ; 7(11): 1336-1353, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28974511

RESUMEN

T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive malignancy of thymocytes. Using a transgenic screen in zebrafish, thymocyte selection-associated high mobility group box protein (TOX) was uncovered as a collaborating oncogenic driver that accelerated T-ALL onset by expanding the initiating pool of transformed clones and elevating genomic instability. TOX is highly expressed in a majority of human T-ALL and is required for proliferation and continued xenograft growth in mice. Using a wide array of functional analyses, we uncovered that TOX binds directly to KU70/80 and suppresses recruitment of this complex to DNA breaks to inhibit nonhomologous end joining (NHEJ) repair. Impaired NHEJ is well known to cause genomic instability, including development of T-cell malignancies in KU70- and KU80-deficient mice. Collectively, our work has uncovered important roles for TOX in regulating NHEJ by elevating genomic instability during leukemia initiation and sustaining leukemic cell proliferation following transformation.Significance: TOX is an HMG box-containing protein that has important roles in T-ALL initiation and maintenance. TOX inhibits the recruitment of KU70/KU80 to DNA breaks, thereby inhibiting NHEJ repair. Thus, TOX is likely a dominant oncogenic driver in a large fraction of human T-ALL and enhances genomic instability. Cancer Discov; 7(11); 1336-53. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 1201.


Asunto(s)
Reparación del ADN por Unión de Extremidades/genética , Inestabilidad Genómica/genética , Proteínas HMGB/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Factores de Transcripción/genética , Animales , Animales Modificados Genéticamente , Proliferación Celular/genética , Humanos , Autoantígeno Ku/genética , Ratones , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Linfocitos T/patología , Ensayos Antitumor por Modelo de Xenoinjerto , Pez Cebra/genética
12.
Cell Rep ; 11(1): 149-63, 2015 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-25818296

RESUMEN

Cells experience damage from exogenous and endogenous sources that endanger genome stability. Several cellular pathways have evolved to detect DNA damage and mediate its repair. Although many proteins have been implicated in these processes, only recent studies have revealed how they operate in the context of high-ordered chromatin structure. Here, we identify the nuclear oncogene SET (I2PP2A) as a modulator of DNA damage response (DDR) and repair in chromatin surrounding double-strand breaks (DSBs). We demonstrate that depletion of SET increases DDR and survival in the presence of radiomimetic drugs, while overexpression of SET impairs DDR and homologous recombination (HR)-mediated DNA repair. SET interacts with the Kruppel-associated box (KRAB)-associated co-repressor KAP1, and its overexpression results in the sustained retention of KAP1 and Heterochromatin protein 1 (HP1) on chromatin. Our results are consistent with a model in which SET-mediated chromatin compaction triggers an inhibition of DNA end resection and HR.


Asunto(s)
Cromatina/genética , Proteínas Cromosómicas no Histona/genética , Chaperonas de Histonas/genética , Reparación del ADN por Recombinación/genética , Proteínas Represoras/genética , Factores de Transcripción/genética , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/biosíntesis , Roturas del ADN de Doble Cadena/efectos de los fármacos , Daño del ADN/genética , Proteínas de Unión al ADN/genética , Heterocromatina/genética , Chaperonas de Histonas/antagonistas & inhibidores , Chaperonas de Histonas/metabolismo , Humanos , Proteínas Represoras/biosíntesis , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/metabolismo , Proteína 28 que Contiene Motivos Tripartito
13.
J Cell Biol ; 207(6): 717-33, 2014 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-25533843

RESUMEN

Heterochromatin is a barrier to DNA repair that correlates strongly with elevated somatic mutation in cancer. CHD class II nucleosome remodeling activity (specifically CHD3.1) retained by KAP-1 increases heterochromatin compaction and impedes DNA double-strand break (DSB) repair requiring Artemis. This obstruction is alleviated by chromatin relaxation via ATM-dependent KAP-1S824 phosphorylation (pKAP-1) and CHD3.1 dispersal from heterochromatic DSBs; however, how heterochromatin compaction is actually adjusted after CHD3.1 dispersal is unknown. In this paper, we demonstrate that Artemis-dependent DSB repair in heterochromatin requires ISWI (imitation switch)-class ACF1-SNF2H nucleosome remodeling. Compacted chromatin generated by CHD3.1 after DNA replication necessitates ACF1-SNF2H-mediated relaxation for DSB repair. ACF1-SNF2H requires RNF20 to bind heterochromatic DSBs, underlies RNF20-mediated chromatin relaxation, and functions downstream of pKAP-1-mediated CHD3.1 dispersal to enable DSB repair. CHD3.1 and ACF1-SNF2H display counteractive activities but similar histone affinities (via the plant homeodomains of CHD3.1 and ACF1), which we suggest necessitates a two-step dispersal and recruitment system regulating these opposing chromatin remodeling activities during DSB repair.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Reparación del ADN por Unión de Extremidades , ADN Helicasas/metabolismo , Heterocromatina/genética , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Factores de Transcripción/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Línea Celular Tumoral , Ensamble y Desensamble de Cromatina , Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN , Endonucleasas , Heterocromatina/metabolismo , Histonas/metabolismo , Ratones , Datos de Secuencia Molecular , Células 3T3 NIH , Proteínas Nucleares/metabolismo , Nucleosomas/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Ubiquitina-Proteína Ligasas/metabolismo
14.
PLoS One ; 8(3): e60384, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23555963

RESUMEN

The 2-micron plasmid of the budding yeast Saccharomyces cerevisiae encodes copy-number amplification and partitioning systems that enable the plasmid to persist despite conferring no advantage to its host. Plasmid partitioning requires interaction of the plasmid Rep1 and Rep2 proteins with each other and with the plasmid-partitioning locus STB. Here we demonstrate that Rep1 stability is reduced in the absence of Rep2, and that both Rep proteins are sumoylated. Lysine-to-arginine substitutions in Rep1 and Rep2 that inhibited their sumoylation perturbed plasmid inheritance without affecting Rep protein stability or two-hybrid interaction between Rep1 and Rep2. One-hybrid and chromatin immunoprecipitation assays revealed that Rep1 was required for efficient retention of Rep2 at STB and that sumoylation-deficient mutants of Rep1 and Rep2 were impaired for association with STB. The normal co-localization of both Rep proteins with the punctate nuclear plasmid foci was also lost when Rep1 was sumoylation-deficient. The correlation of Rep protein sumoylation status with plasmid-partitioning locus association suggests a theme common to eukaryotic chromosome segregation proteins, sumoylated forms of which are found enriched at centromeres, and between the yeast 2-micron plasmid and viral episomes that depend on sumoylation of their maintenance proteins for persistence in their hosts.


Asunto(s)
Plásmidos/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Sumoilación , Transactivadores/metabolismo , Sustitución de Aminoácidos , Sitios Genéticos , Plásmidos/genética , Mapas de Interacción de Proteínas , Proteína SUMO-1/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Transactivadores/genética
15.
Front Genet ; 4: 45, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23554604

RESUMEN

Genomic instability is both a hallmark of cancer and a major contributing factor to tumor development. Central to the maintenance of genome stability is the repair of DNA damage, and the most toxic form of DNA damage is the DNA double-strand break. As a consequence the eukaryotic cell harbors an impressive array of protein machinery to detect and repair DNA breaks through the initiation of a multi-branched, highly coordinated signaling cascade. This signaling cascade, known as the DNA damage response (DDR), functions to integrate DNA repair with a host of cellular processes including cell cycle checkpoint activation, transcriptional regulation, and programmed cell death. In eukaryotes, DNA is packaged in chromatin, which provides a mechanism to regulate DNA transactions including DNA repair through an equally impressive array of post-translational modifications to proteins within chromatin, and the DDR machinery itself. Histones, as the major protein component of chromatin, are subject to a host of post-translational modifications including phosphorylation, methylation, and acetylation. More recently, modification of both the histones and DDR machinery by ubiquitin and other ubiquitin-like proteins, such as the small ubiquitin-like modifiers, has been shown to play a central role in coordinating the DDR. In this review, we explore how ubiquitination and sumoylation contribute to the "writing" of key post-translational modifications within chromatin that are in turn "read" by the DDR machinery and chromatin-remodeling factors, which act together to facilitate the efficient detection and repair of DNA damage.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA