Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell Mol Life Sci ; 79(12): 603, 2022 Nov 24.
Artículo en Inglés | MEDLINE | ID: mdl-36434396

RESUMEN

Ataxia telangiectasia mutated (ATM) is a serine-threonine protein kinase and important regulator of the DNA damage response (DDR). One critical ATM target is the structural subunit A (PR65-S401) of protein phosphatase 2A (PP2A), known to regulate diverse cellular processes such as mitosis and cell growth as well as dephosphorylating many proteins during the recovery from the DDR. We generated mouse embryonic fibroblasts expressing PR65-WT, -S401A (cannot be phosphorylated), and -S401D (phospho-mimetic) transgenes. Significantly, S401 mutants exhibited extensive chromosomal aberrations, impaired DNA double-strand break (DSB) repair and underwent increased mitotic catastrophe after radiation. Both S401A and the S401D cells showed impaired DSB repair (nonhomologous end joining and homologous recombination repair) and exhibited delayed DNA damage recovery, which was reflected in reduced radiation survival. Furthermore, S401D cells displayed increased ERK and AKT signaling resulting in enhanced growth rate further underscoring the multiple roles ATM-PP2A signaling plays in regulating prosurvival responses. Time-lapse video and cellular localization experiments showed that PR65 was exported to the cytoplasm after radiation by CRM1, a nuclear export protein, in line with the very rapid pleiotropic effects observed. A putative nuclear export sequence (NES) close to S401 was identified and when mutated resulted in aberrant PR65 shuttling. Our study demonstrates that the phosphorylation of a single, critical PR65 amino acid (S401) by ATM fundamentally controls the DDR, and balances DSB repair quality, cell survival and growth by spatiotemporal PR65 nuclear-cytoplasmic shuttling mediated by the nuclear export receptor CRM1.


Asunto(s)
Ataxia Telangiectasia , Animales , Ratones , Ataxia Telangiectasia/genética , Proteína Fosfatasa 2/genética , Proteína Fosfatasa 2/metabolismo , Transporte Activo de Núcleo Celular , Proteínas de Unión al ADN/metabolismo , Fibroblastos/metabolismo , Proteínas Nucleares/metabolismo , Daño del ADN
2.
Nucleic Acids Res ; 46(2): 520-537, 2018 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-29216365

RESUMEN

Tyrosyl-DNA Phosphodiesterases 1 (TDP1) and 2 (TDP2) are eukaryotic enzymes that clean-up after aberrant topoisomerase activity. While TDP1 hydrolyzes phosphotyrosyl peptides emanating from trapped topoisomerase I (Top I) from the 3' DNA ends, topoisomerase 2 (Top II)-induced 5'-phosphotyrosyl residues are processed by TDP2. Even though the canonical functions of TDP1 and TDP2 are complementary, they exhibit little structural or sequence similarity. Homozygous mutations in genes encoding these enzymes lead to the development of severe neurodegenerative conditions due to the accumulation of transcription-dependent topoisomerase cleavage complexes underscoring the biological significance of these enzymes in the repair of topoisomerase-DNA lesions in the nervous system. TDP1 can promiscuously process several blocked 3' ends generated by DNA damaging agents and nucleoside analogs in addition to hydrolyzing 3'-phosphotyrosyl residues. In addition, deficiency of these enzymes causes hypersensitivity to anti-tumor topoisomerase poisons. Thus, TDP1 and TDP2 are promising therapeutic targets and their inhibitors are expected to significantly synergize the effects of current anti-tumor therapies including topoisomerase poisons and other DNA damaging agents. This review covers the structural aspects, biology and regulation of these enzymes, along with ongoing developments in the process of discovering safe and effective TDP inhibitors.


Asunto(s)
Daño del ADN , Reparación del ADN , Proteínas Nucleares/metabolismo , Hidrolasas Diéster Fosfóricas/metabolismo , Factores de Transcripción/metabolismo , ADN/química , ADN/genética , ADN/metabolismo , ADN-Topoisomerasas de Tipo I/genética , ADN-Topoisomerasas de Tipo I/metabolismo , ADN-Topoisomerasas de Tipo II/genética , ADN-Topoisomerasas de Tipo II/metabolismo , Proteínas de Unión al ADN , Humanos , Neoplasias/enzimología , Neoplasias/genética , Neoplasias/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/genética , Hidrolasas Diéster Fosfóricas/química , Hidrolasas Diéster Fosfóricas/genética , Unión Proteica , Factores de Transcripción/química , Factores de Transcripción/genética
3.
Nucleic Acids Res ; 46(17): 8926-8939, 2018 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-30113698

RESUMEN

The Artemis nuclease and tyrosyl-DNA phosphodiesterase (TDP1) are each capable of resolving protruding 3'-phosphoglycolate (PG) termini of DNA double-strand breaks (DSBs). Consequently, both a knockout of Artemis and a knockout/knockdown of TDP1 rendered cells sensitive to the radiomimetic agent neocarzinostatin (NCS), which induces 3'-PG-terminated DSBs. Unexpectedly, however, a knockdown or knockout of TDP1 in Artemis-null cells did not confer any greater sensitivity than either deficiency alone, indicating a strict epistasis between TDP1 and Artemis. Moreover, a deficiency in Artemis, but not TDP1, resulted in a fraction of unrepaired DSBs, which were assessed as 53BP1 foci. Conversely, a deficiency in TDP1, but not Artemis, resulted in a dramatic increase in dicentric chromosomes following NCS treatment. An inhibitor of DNA-dependent protein kinase, a key regulator of the classical nonhomologous end joining (C-NHEJ) pathway sensitized cells to NCS, but eliminated the sensitizing effects of both TDP1 and Artemis deficiencies. These results suggest that TDP1 and Artemis perform different functions in the repair of terminally blocked DSBs by the C-NHEJ pathway, and that whereas an Artemis deficiency prevents end joining of some DSBs, a TDP1 deficiency tends to promote DSB mis-joining.


Asunto(s)
Reparación del ADN por Unión de Extremidades , ADN/genética , Endonucleasas/genética , Epistasis Genética , Proteínas Nucleares/genética , Hidrolasas Diéster Fosfóricas/genética , Supervivencia Celular/efectos de los fármacos , Citotoxinas/farmacología , ADN/química , ADN/metabolismo , Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN , Endonucleasas/antagonistas & inhibidores , Endonucleasas/deficiencia , Células HCT116 , Células HEK293 , Humanos , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/deficiencia , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Hidrolasas Diéster Fosfóricas/deficiencia , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53/genética , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo , Cinostatina/farmacología
4.
Blood ; 127(18): 2219-30, 2016 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-26851293

RESUMEN

Two classes of novel agents, NEDD8-activating enzyme (NAE) and histone deacetylase (HDAC) inhibitors, have shown single-agent activity in acute myelogenous leukemia (AML)/myelodysplastic syndrome (MDS). Here we examined mechanisms underlying interactions between the NAE inhibitor pevonedistat (MLN4924) and the approved HDAC inhibitor belinostat in AML/MDS cells. MLN4924/belinostat coadministration synergistically induced AML cell apoptosis with or without p53 deficiency or FLT3-internal tandem duplication (ITD), whereas p53 short hairpin RNA (shRNA) knockdown or enforced FLT3-ITD expression significantly sensitized cells to the regimen. MLN4924 blocked belinostat-induced antiapoptotic gene expression through nuclear factor-κB inactivation. Each agent upregulated Bim, and Bim knockdown significantly attenuated apoptosis. Microarrays revealed distinct DNA damage response (DDR) genetic profiles between individual vs combined MLN4924/belinostat exposure. Whereas belinostat abrogated the MLN4924-activated intra-S checkpoint through Chk1 and Wee1 inhibition/downregulation, cotreatment downregulated multiple homologous recombination and nonhomologous end-joining repair proteins, triggering robust double-stranded breaks, chromatin pulverization, and apoptosis. Consistently, Chk1 or Wee1 shRNA knockdown significantly sensitized AML cells to MLN4924. MLN4924/belinostat displayed activity against primary AML or MDS cells, including those carrying next-generation sequencing-defined poor-prognostic cancer hotspot mutations, and CD34(+)/CD38(-)/CD123(+) populations, but not normal CD34(+) progenitors. Finally, combined treatment markedly reduced tumor burden and significantly prolonged animal survival (P < .0001) in AML xenograft models with negligible toxicity, accompanied by pharmacodynamic effects observed in vitro. Collectively, these findings argue that MLN4924 and belinostat interact synergistically by reciprocally disabling the DDR in AML/MDS cells. This strategy warrants further consideration in AML/MDS, particularly in disease with unfavorable genetic aberrations.


Asunto(s)
Ciclopentanos/uso terapéutico , Daño del ADN , Reparación del ADN/efectos de los fármacos , Inhibidores de Histona Desacetilasas/uso terapéutico , Ácidos Hidroxámicos/uso terapéutico , Leucemia Mieloide Aguda/tratamiento farmacológico , Terapia Molecular Dirigida , Síndromes Mielodisplásicos/tratamiento farmacológico , Pirimidinas/uso terapéutico , Sulfonamidas/uso terapéutico , Enzimas Activadoras de Ubiquitina/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Proteína 11 Similar a Bcl2/antagonistas & inhibidores , Proteína 11 Similar a Bcl2/genética , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/genética , Células Cultivadas , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/antagonistas & inhibidores , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/genética , Ciclopentanos/farmacología , Sinergismo Farmacológico , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Ácidos Hidroxámicos/farmacología , Estimación de Kaplan-Meier , Leucemia Mieloide Aguda/patología , Ratones , Síndromes Mielodisplásicos/patología , FN-kappa B/metabolismo , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiología , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/genética , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/genética , Pirimidinas/farmacología , Interferencia de ARN , ARN Interferente Pequeño/genética , Puntos de Control de la Fase S del Ciclo Celular/efectos de los fármacos , Sulfonamidas/farmacología , Células U937 , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Nucleic Acids Res ; 42(5): 3125-37, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24371269

RESUMEN

To track the processing of damaged DNA double-strand break (DSB) ends in vivo, a method was devised for quantitative measurement of 3'-phosphoglycolate (PG) termini on DSBs induced by the non-protein chromophore of neocarzinostatin (NCS-C) in the human Alu repeat. Following exposure of cells to NCS-C, DNA was isolated, and labile lesions were chemically stabilized. All 3'-phosphate and 3'-hydroxyl ends were enzymatically capped with dideoxy termini, whereas 3'-PG ends were rendered ligatable, linked to an anchor, and quantified by real-time Taqman polymerase chain reaction. Using this assay and variations thereof, 3'-PG and 3'-phosphate termini on 1-base 3' overhangs of NCS-C-induced DSBs were readily detected in DNA from the treated lymphoblastoid cells, and both were largely eliminated from cellular DNA within 1 h. However, the 3'-PG termini were processed more slowly than 3'-phosphate termini, and were more persistent in tyrosyl-DNA phosphodiesterase 1-mutant SCAN1 than in normal cells, suggesting a significant role for tyrosyl-DNA phosphodiesterase 1 in removing 3'-PG blocking groups for DSB repair. DSBs with 3'-hydroxyl termini, which are not directly induced by NCS-C, were formed rapidly in cells, and largely eliminated by further processing within 1 h, both in Alu repeats and in heterochromatic α-satellite DNA. Moreover, absence of DNA-PK in M059J cells appeared to accelerate resolution of 3'-PG ends.


Asunto(s)
Roturas del ADN de Doble Cadena , Glicolatos/análisis , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Línea Celular Transformada , ADN/química , Proteína Quinasa Activada por ADN/deficiencia , Humanos , Hidrolasas Diéster Fosfóricas/deficiencia , Ataxias Espinocerebelosas/genética , Cinostatina/toxicidad
6.
Mol Pharm ; 12(1): 287-97, 2015 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-25407898

RESUMEN

TriplatinNC is a highly positively charged, substitution-inert derivative of the phase II clinical anticancer drug, BBR3464. Such substitution-inert complexes form a distinct subset of polynuclear platinum complexes (PPCs) interacting with DNA and other biomolecules through noncovalent interactions. Rapid cellular entry is facilitated via interaction with cell surface glycosoaminoglycans and is a mechanism unique to PPCs. Nanoscale secondary ion mass spectrometry (nanoSIMS) showed rapid distribution within cytoplasmic and nucleolar compartments, but not the nucleus. In this article, the downstream effects of nucleolar localization are described. In human colon carcinoma cells, HCT116, the production rate of 47S rRNA precursor transcripts was dramatically reduced as an early event after drug treatment. Transcriptional inhibition of rRNA was followed by a robust G1 arrest, and activation of apoptotic proteins caspase-8, -9, and -3 and PARP-1 in a p53-independent manner. Using cell synchronization and flow cytometry, it was determined that cells treated while in G1 arrest immediately, but cells treated in S or G2 successfully complete mitosis. Twenty-four hours after treatment, the majority of cells finally arrest in G1, but nearly one-third contained highly compacted DNA; a distinct biological feature that cannot be associated with mitosis, senescence, or apoptosis. This unique effect mirrored the efficient condensation of tRNA and DNA in cell-free systems. The combination of DNA compaction and apoptosis by TriplatinNC treatment conferred striking activity in platinum-resistant and/or p53 mutant or null cell lines. Taken together, our results support that the biological activity of TriplatinNC reflects reduced metabolic deactivation (substitution-inert compound not reactive to sulfur nucleophiles), high cellular accumulation, and novel consequences of high-affinity noncovalent DNA binding, producing a new profile and a further shift in the structure-activity paradigms for antitumor complexes.


Asunto(s)
Antineoplásicos/química , Nucléolo Celular/efectos de los fármacos , ADN/química , Compuestos Organoplatinos/química , Platino (Metal)/uso terapéutico , ARN Ribosómico/química , Proteína p53 Supresora de Tumor/genética , Animales , Apoptosis , Caspasas/metabolismo , Ciclo Celular , Línea Celular Tumoral , Sistema Libre de Células , Citometría de Flujo , Células HCT116 , Humanos , Concentración 50 Inhibidora , Ratones , Microscopía Confocal , Mitosis , Mutación , Péptidos/química , Fosfatos/química , ARN de Transferencia/química , Proteína p53 Supresora de Tumor/metabolismo , beta-Galactosidasa/metabolismo
7.
Subcell Biochem ; 85: 321-36, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25201202

RESUMEN

p53 is a tumor suppressor protein that prevents oncogenic transformation and maintains genomic stability by blocking proliferation of cells harboring unrepaired or misrepaired DNA. A wide range of genotoxic stresses such as DNA damaging anti-cancer drugs and ionizing radiation promote nuclear accumulation of p53 and trigger its ability to activate or repress a number of downstream target genes involved in various signaling pathways. This cascade leads to the activation of multiple cell cycle checkpoints and subsequent cell cycle arrest, allowing the cells to either repair the DNA or undergo apoptosis, depending on the intensity of DNA damage. In addition, p53 has many transcription-independent functions, including modulatory roles in DNA repair and recombination. This chapter will focus on the role of p53 in regulating or influencing the repair of DNA double-strand breaks that mainly includes homologous recombination repair (HRR) and non-homologous end joining (NHEJ). Through this discussion, we will try to establish that p53 acts as an important linchpin between upstream DNA damage signaling cues and downstream cellular events that include repair, recombination, and apoptosis.


Asunto(s)
Daño del ADN , Reparación del ADN por Unión de Extremidades , Genes p53 , Reparación del ADN por Recombinación , Humanos , Mutación
8.
Nucleic Acids Res ; 39(15): 6500-10, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21531702

RESUMEN

Deficiency in Artemis is associated with lack of V(D)J recombination, sensitivity to radiation and radiomimetic drugs, and failure to repair a subset of DNA double-strand breaks (DSBs). Artemis harbors an endonuclease activity that trims both 5'- and 3'-ends of DSBs. To examine whether endonucleolytic trimming of terminally blocked DSBs by Artemis is a biologically relevant function, Artemis-deficient fibroblasts were stably complemented with either wild-type Artemis or an endonuclease-deficient D165N mutant. Wild-type Artemis completely restored resistance to γ-rays, bleomycin and neocarzinostatin, and also restored DSB-repair proficiency in G0/G1 phase as measured by pulsed-field gel electrophoresis and repair focus resolution. In contrast, cells expressing the D165N mutant, even at very high levels, remained as chemo/radiosensitive and repair deficient as the parental cells, as evidenced by persistent γ-H2AX, 53BP1 and Mre11 foci that slowly increased in size and ultimately became juxtaposed with promyelocytic leukemia protein nuclear bodies. In normal fibroblasts, overexpression of wild-type Artemis increased radioresistance, while D165N overexpression conferred partial repair deficiency following high-dose radiation. Restoration of chemo/radioresistance by wild-type, but not D165N Artemis suggests that the lack of endonucleolytic trimming of DNA ends is the principal cause of sensitivity to double-strand cleaving agents in Artemis-deficient cells.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , Endodesoxirribonucleasas/genética , Proteínas Nucleares/genética , Tolerancia a Radiación , Bleomicina/toxicidad , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Proteínas de Unión al ADN , Endodesoxirribonucleasas/deficiencia , Endodesoxirribonucleasas/metabolismo , Endonucleasas , Fase G1 , Humanos , Mutación , Proteínas Nucleares/análisis , Proteínas Nucleares/deficiencia , Proteínas Nucleares/metabolismo , Proteína de la Leucemia Promielocítica , Factores de Transcripción/análisis , Proteínas Supresoras de Tumor/análisis , Cinostatina/toxicidad
9.
Nucleic Acids Res ; 38(16): 5419-31, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20435673

RESUMEN

Differences in the substrate specificity of mammalian family X DNA polymerases are proposed to partly depend on a loop (loop 1) upstream of the polymerase active site. To examine if this is the case in DNA polymerase λ (pol λ), here we characterize a variant of the human polymerase in which nine residues of loop 1 are replaced with four residues from the equivalent position in pol ß. Crystal structures of the mutant enzyme bound to gapped DNA with and without a correct dNTP reveal that the change in loop 1 does not affect the overall structure of the protein. Consistent with these structural data, the mutant enzyme has relatively normal catalytic efficiency for correct incorporation, and it efficiently participates in non-homologous end joining of double-strand DNA breaks. However, DNA junctions recovered from end-joining reactions are more diverse than normal, and the mutant enzyme is substantially less accurate than wild-type pol λ in three different biochemical assays. Comparisons of the binary and ternary complex crystal structures of mutant and wild-type pol λ suggest that loop 1 modulates pol λ's fidelity by controlling dNTP-induced movements of the template strand and the primer-terminal 3'-OH as the enzyme transitions from an inactive to an active conformation.


Asunto(s)
ADN Polimerasa beta/química , Secuencia de Aminoácidos , Biocatálisis , Secuencia Conservada , Cristalografía por Rayos X , ADN/biosíntesis , ADN Polimerasa beta/metabolismo , Desoxirribonucleótidos/metabolismo , Humanos , Cinética , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica
10.
Nucleic Acids Res ; 38(2): 441-54, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19892829

RESUMEN

The repair of DNA double-strand breaks (DSBs) is essential to maintain genomic integrity. In higher eukaryotes, DNA DSBs are predominantly repaired by non-homologous end joining (NHEJ), but DNA ends can also be joined by an alternative error-prone mechanism termed microhomology-mediated end joining (MMEJ). In MMEJ, the repair of DNA breaks is mediated by annealing at regions of microhomology and is always associated with deletions at the break site. In budding yeast, the Mre11/Rad5/Xrs2 complex has been demonstrated to play a role in both classical NHEJ and MMEJ, but the involvement of the analogous MRE11/RAD50/NBS1 (MRN) complex in end joining in higher eukaryotes is less certain. Here we demonstrate that in Xenopus laevis egg extracts, the MRN complex is not required for classical DNA-PK-dependent NHEJ. However, the XMRN complex is necessary for resection-based end joining of mismatched DNA ends. This XMRN-dependent end joining process is independent of the core NHEJ components Ku70 and DNA-PK, occurs with delayed kinetics relative to classical NHEJ and brings about repair at sites of microhomology. These data indicate a role for the X. laevis MRN complex in MMEJ.


Asunto(s)
Proteínas Portadoras/fisiología , Roturas del ADN de Doble Cadena , Reparación del ADN , Proteínas de Unión al ADN/fisiología , Proteínas Supresoras de Tumor/fisiología , Proteínas de Xenopus/fisiología , Animales , Antígenos Nucleares/metabolismo , Proteínas Portadoras/análisis , Proteínas Portadoras/genética , Enzimas Reparadoras del ADN , Proteína Quinasa Activada por ADN/metabolismo , Proteínas de Unión al ADN/análisis , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Autoantígeno Ku , Proteína Homóloga de MRE11 , Óvulo/metabolismo , Recombinación Genética , Proteínas Supresoras de Tumor/análisis , Proteínas Supresoras de Tumor/genética , Proteínas de Xenopus/análisis , Proteínas de Xenopus/genética , Xenopus laevis
11.
Nucleic Acids Res ; 37(12): 4055-62, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19420065

RESUMEN

XLF/Cernunnos is a core protein of the nonhomologous end-joining pathway of DNA double-strand break repair. To better define the role of Cernunnos in end joining, whole-cell extracts were prepared from Cernunnos-deficient human cells. These extracts effected little joining of DNA ends with cohesive 5' or 3' overhangs, and no joining at all of partially complementary 3' overhangs that required gap filling prior to ligation. Assays in which gap-filled but unligated intermediates were trapped using dideoxynucleotides revealed that there was no gap filling on aligned DSB ends in the Cernunnos-deficient extracts. Recombinant Cernunnos protein restored gap filling and end joining of partially complementary overhangs, and stimulated joining of cohesive ends more than twentyfold. XLF-dependent gap filling was nearly eliminated by immunodepletion of DNA polymerase lambda, but was restored by addition of either polymerase lambda or polymerase mu. Thus, Cernunnos is essential for gap filling by either polymerase during nonhomologous end joining, suggesting that it plays a major role in aligning the two DNA ends in the repair complex.


Asunto(s)
ADN Polimerasa beta/metabolismo , Enzimas Reparadoras del ADN/fisiología , Reparación del ADN , Proteínas de Unión al ADN/fisiología , ADN Polimerasa Dirigida por ADN/metabolismo , Extractos Celulares , ADN/química , ADN/metabolismo , Roturas del ADN de Doble Cadena , Enzimas Reparadoras del ADN/química , Enzimas Reparadoras del ADN/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Humanos , Fosforilación , Serina/metabolismo
12.
DNA Repair (Amst) ; 8(5): 654-63, 2009 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-19211312

RESUMEN

A homozygous H493R mutation in the active site of tyrosyl-DNA phosphodiesterase (TDP1) has been implicated in hereditary spinocerebellar ataxia with axonal neuropathy (SCAN1), an autosomal recessive neurodegenerative disease. However, it is uncertain how the H493R mutation elicits the specific pathologies of SCAN1. To address this question, and to further elucidate the role of TDP1 in repair of DNA end modifications and general physiology, we generated a Tdp1 knockout mouse and carried out detailed behavioral analyses as well as characterization of repair deficiencies in extracts of embryo fibroblasts from these animals. While Tdp1(-/-) mice appear phenotypically normal, extracts from Tdp1(-/-) fibroblasts exhibited deficiencies in processing 3'-phosphotyrosyl single-strand breaks and 3'-phosphoglycolate double-strand breaks (DSBs), but not 3'-phosphoglycolate single-strand breaks. Supplementing Tdp1(-/-) extracts with H493R TDP1 partially restored processing of 3'-phosphotyrosyl single-strand breaks, but with evidence of persistent covalent adducts between TDP1 and DNA, consistent with a proposed intermediate-stabilization effect of the SCAN1 mutation. However, H493R TDP1 supplementation had no effect on phosphoglycolate (PG) termini on 3' overhangs of double-strand breaks; these remained completely unprocessed. Altogether, these results suggest that for 3'-phosphoglycolate overhang lesions, the SCAN1 mutation confers loss of function, while for 3'-phosphotyrosyl lesions, the mutation uniquely stabilizes a reaction intermediate.


Asunto(s)
Aductos de ADN/química , Glicolatos/metabolismo , Mutación/genética , Hidrolasas Diéster Fosfóricas/fisiología , Fosfotirosina/metabolismo , Ataxias Espinocerebelosas/genética , Animales , Southern Blotting , Western Blotting , Catálisis , Aductos de ADN/genética , Aductos de ADN/metabolismo , Roturas del ADN de Doble Cadena , Roturas del ADN de Cadena Simple , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Prueba de Complementación Genética , Masculino , Ratones , Ratones Noqueados , Mutagénesis Sitio-Dirigida , Reacción en Cadena de la Polimerasa , Ataxias Espinocerebelosas/metabolismo
13.
Nucleic Acids Res ; 36(9): 2895-905, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18385158

RESUMEN

Ionizing radiation induces various clustered DNA lesions, including double-strand breaks (DSBs) accompanied by nearby oxidative base damage. Previous work showed that, in HeLa nuclear extracts, DSBs with partially complementary 3' overhangs and a one-base gap in each strand are accurately rejoined, with the gaps being filled by DNA polymerase lambda. To determine the possible effect of oxidative base damage on this process, plasmid substrates were constructed containing overhangs with 8-oxoguanine or thymine glycol in base-pairing positions of 3-base (-ACG or -GTA) 3' overhangs. In this context, 8-oxoguanine was well tolerated by the end-joining machinery when present at one end of the break, but not when present at both ends. Thymine glycol was less well tolerated than 8-oxoguanine, reducing gap filling and accurate rejoining by at least 10-fold. The results suggest that complex DSBs can be accurately rejoined despite the presence of accompanying base damage, but that nonplanar bases constitute a major barrier to this process and promote error-prone joining. A chimeric DNA polymerase, in which the catalytic domain of polymerase lambda was replaced with that of polymerase beta, could not substitute for polymerase lambda in these assays, suggesting that this domain is specifically adapted for gap filling on aligned DSB ends.


Asunto(s)
Roturas del ADN de Doble Cadena , ADN Polimerasa beta/metabolismo , Reparación del ADN , Guanina/análogos & derivados , Timina/análogos & derivados , Disparidad de Par Base , Extractos Celulares , Núcleo Celular/metabolismo , ADN Polimerasa beta/genética , Guanina/química , Células HeLa , Humanos , Proteínas Recombinantes de Fusión/metabolismo , Timina/química
14.
Nucleic Acids Res ; 36(10): 3354-65, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18440975

RESUMEN

Previous work showed that, in the presence of DNA-dependent protein kinase (DNA-PK), Artemis slowly trims 3'-phosphoglycolate-terminated blunt ends. To examine the trimming reaction in more detail, long internally labeled DNA substrates were treated with Artemis. In the absence of DNA-PK, Artemis catalyzed extensive 5'-->3' exonucleolytic resection of double-stranded DNA. This resection required a 5'-phosphate, but did not require ATP, and was accompanied by endonucleolytic cleavage of the resulting 3' overhang. In the presence of DNA-PK, Artemis-mediated trimming was more limited, was ATP-dependent and did not require a 5'-phosphate. For a blunt end with either a 3'-phosphoglycolate or 3'-hydroxyl terminus, endonucleolytic trimming of 2-4 nucleotides from the 3'-terminal strand was accompanied by trimming of 6 nt from the 5'-terminal strand. The results suggest that autophosphorylated DNA-PK suppresses the exonuclease activity of Artemis toward blunt-ended DNA, and promotes slow and limited endonucleolytic trimming of the 5'-terminal strand, resulting in short 3' overhangs that are trimmed endonucleolytically. Thus, Artemis and DNA-PK can convert terminally blocked DNA ends of diverse geometry and chemical structure to a form suitable for polymerase-mediated patching and ligation, with minimal loss of terminal sequence. Such processing could account for the very small deletions often found at DNA double-strand break repair sites.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , Proteína Quinasa Activada por ADN/metabolismo , Endodesoxirribonucleasas/metabolismo , ADN/química , ADN/metabolismo , Endodesoxirribonucleasas/antagonistas & inhibidores , Endodesoxirribonucleasas/genética , Exodesoxirribonucleasas/metabolismo , Sueros Inmunes/farmacología , Mutación , Fosfatos/química
15.
Nucleic Acids Res ; 35(12): 3869-78, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17526517

RESUMEN

Previous work suggested that phosphorylation of DNA-PKcs at several serine/threonine (S/T) residues at positions 2609-2647 promotes DNA-PK-dependent end joining. In an attempt to clarify the role of such phosphorylation, end joining was examined in extracts of DNA-PKcs-deficient M059J cells. Joining of ends requiring gap filling prior to ligation was completely dependent on complementation of these extracts with exogenous DNA-PKcs. DNA-PKcs with either S/T --> A or S/T --> D substitutions at all six sites in the 2609-2647 cluster also supported end joining, but with markedly lower efficiency than wild-type protein. The residual end joining was greater with the S/T --> D-substituted than with the S/T --> A-substituted protein. A specific inhibitor of the kinase activity of DNA-PK, KU57788, completely blocked end joining promoted by wild type as well as both mutant forms of DNA-PK, while inhibition of ATM kinase did not. The fidelity of end joining was not affected by the mutant DNA-PKcs alleles or the inhibitors. Overall, the results support a role for autophosphorylation of the 2609-2647 cluster in promoting end joining and controlling the accessibility of DNA ends, but suggest that DNA-PK-mediated phosphorylation at other sites, on either DNA-PKcs or other proteins, is at least as important as the 2609-2647 cluster in regulating end joining.


Asunto(s)
Proteína Quinasa Activada por ADN/metabolismo , Proteínas Nucleares/metabolismo , Serina/metabolismo , Treonina/metabolismo , Sustitución de Aminoácidos , Proteínas de la Ataxia Telangiectasia Mutada , Catálisis , Proteínas de Ciclo Celular/metabolismo , Extractos Celulares , Línea Celular Tumoral , ADN Ligasa (ATP) , ADN Ligasas/metabolismo , Proteína Quinasa Activada por ADN/química , Proteína Quinasa Activada por ADN/genética , Proteínas de Unión al ADN/metabolismo , Células HeLa , Humanos , Proteínas Nucleares/química , Proteínas Nucleares/genética , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Supresoras de Tumor/metabolismo
16.
Cancer Res ; 67(3): 1046-53, 2007 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-17283137

RESUMEN

The accurate joining of DNA double-strand breaks by homologous recombination repair (HRR) is critical to the long-term survival of the cell. The three major mitogen-activated protein (MAP) kinase (MAPK) signaling pathways, extracellular signal-regulated kinase (ERK), p38, and c-Jun-NH(2)-kinase (JNK), regulate cell growth, survival, and apoptosis. To determine the role of MAPK signaling in HRR, we used a human in vivo I-SceI-based repair system. First, we verified that this repair platform is amenable to pharmacologic manipulation and show that the ataxia telangiectasia mutated (ATM) kinase is critical for HRR. The ATM-specific inhibitor KU-55933 compromised HRR up to 90% in growth-arrested cells, whereas this effect was less pronounced in cycling cells. Then, using well-characterized MAPK small-molecule inhibitors, we show that ERK1/2 and JNK signaling are important positive regulators of HRR in growth-arrested cells. On the other hand, inhibition of the p38 MAPK pathway generated an almost 2-fold stimulation of HRR. When ERK1/2 signaling was stimulated by oncogenic RAF-1, an approximately 2-fold increase in HRR was observed. KU-55933 partly blocked radiation-induced ERK1/2 phosphorylation, suggesting that ATM regulates ERK1/2 signaling. Furthermore, inhibition of MAP/ERK kinase (MEK)/ERK signaling resulted in severely reduced levels of phosphorylated (S1981) ATM foci but not gamma-H2AX foci, and suppressed ATM phosphorylation levels >85% throughout the cell cycle. Collectively, these results show that MAPK signaling positively and negatively regulates HRR in human cells. More specifically, ATM-dependent signaling through the RAF/MEK/ERK pathway is critical for efficient HRR and for radiation-induced ATM activation, suggestive of a regulatory feedback loop between ERK and ATM.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Daño del ADN/fisiología , Reparación del ADN/fisiología , Proteínas de Unión al ADN/fisiología , Sistema de Señalización de MAP Quinasas/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas Serina-Treonina Quinasas/fisiología , Recombinación Genética/fisiología , Proteínas Supresoras de Tumor/fisiología , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Reparación del ADN/efectos de los fármacos , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/metabolismo , Glioma , Humanos , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/antagonistas & inhibidores , Proteína Quinasa 3 Activada por Mitógenos/genética , Morfolinas/farmacología , Fosforilación , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-raf/biosíntesis , Pironas/farmacología , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Proteínas Supresoras de Tumor/metabolismo
17.
DNA Repair (Amst) ; 68: 12-24, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29807321

RESUMEN

Polynucleotide kinase/phosphatase (PNKP) has been implicated in non-homologous end joining (NHEJ) of DNA double-strand breaks (DSBs). To assess the consequences of PNKP deficiency for NHEJ of 3'-phosphate-ended DSBs, PNKP-deficient derivatives of HCT116 and of HeLa cells were generated using CRISPR/CAS9. For both cell lines, PNKP deficiency conferred sensitivity to ionizing radiation as well as to neocarzinostatin (NCS), which specifically induces DSBs bearing protruding 3'-phosphate termini. Moreover, NCS-induced DSBs, detected as 53BP1 foci, were more persistent in PNKP -/- HCT116 cells compared to their wild-type (WT) counterparts. Surprisingly, PNKP-deficient whole-cell and nuclear extracts were biochemically competent in removing both protruding and recessed 3'-phosphates from synthetic DSB substrates, albeit much less efficiently than WT extracts, suggesting an alternative 3'-phosphatase. Measurements by ligation-mediated PCR showed that PNKP-deficient HeLa cells contained significantly more 3'-phosphate-terminated and fewer 3'-hydroxyl-terminated DSBs than parental cells 5-15 min after NCS treatment, but this difference disappeared by 1 h. These results suggest that, despite presence of an alternative 3'-phosphatase, loss of PNKP significantly sensitizes cells to 3'-phosphate-terminated DSBs, due to a 3'-dephosphorylation defect.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades , Enzimas Reparadoras del ADN/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , ADN/efectos de los fármacos , ADN/metabolismo , ADN/efectos de la radiación , Enzimas Reparadoras del ADN/metabolismo , Técnicas de Silenciamiento del Gen , Células HCT116 , Células HeLa , Humanos , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo , Radiación Ionizante , Cinostatina/toxicidad
18.
DNA Repair (Amst) ; 5(9-10): 1199-212, 2006 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-16822725

RESUMEN

Exposure of mammalian cells to agents that induce DNA double-strand breaks typically results in both reciprocal and nonreciprocal chromosome translocations. Over the past decade, breakpoint junctions of a significant number of translocations and other genomic rearrangements, both in clinical tumors and in experimental models, have been analyzed at the DNA sequence level. Based on these data, reasonable inferences regarding the biochemical mechanisms involved in translocations can be drawn. In a few cases, breakpoints have been shown to correlate with sites of double-strand cleavage by agents to which the cells or patients have been exposed, including exogenous rare-cutting endonucleases, radiomimetic compounds, and topoisomerase inhibitors. These results confirm that translocations primarily reflect misjoining of the exchanged ends of two or more double-strand breaks. Many junctions show significant loss of DNA sequence at the breakpoints, suggesting exonucleolytic degradation of DNA ends prior to joining. The size and frequency of these deletions varies widely, both between experimental systems, and among individual events in a single system. Homologous recombination between repetitive DNA sequences does not appear to be a major pathway for translocations associated with double-strand breaks. Rather, the general features of the junction sequences, particularly the high frequency small terminal deletions, the apparent splicing of DNA ends at microhomologies, and gap-filling on aligned double-strand break ends, are consistent with the known biochemical properties of the classical nonhomologous end joining pathway involving DNA-dependent protein kinase, XRCC4 and DNA ligase IV. Nevertheless, cells with deficiencies in this pathway still exhibit translocations, with grossly similar junction sequences, suggesting an alternative but less conservative end joining pathway. Although evidence for participation of specific DNA end processing enzymes in formation of translocations is largely circumstantial, likely candidates include DNA polymerases lambda and mu, Artemis nuclease, polynucleotide kinase/phosphatase, tyrosyl-DNA phosphodiesterase, DNase III, Werner syndrome protein, and the Mre11/Rad50/NBS1 complex.


Asunto(s)
Rotura Cromosómica , Daño del ADN , Enzimas Reparadoras del ADN/genética , Neoplasias de la Tiroides/genética , Translocación Genética , Animales , Secuencia de Bases , Células CHO , Cricetinae , Datos de Secuencia Molecular , Radiación Ionizante , Recombinación Genética
19.
DNA Repair (Amst) ; 5(12): 1489-94, 2006 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-16935573

RESUMEN

Hereditary spinocerebellar ataxia with axonal neuropathy (SCAN1) is caused by an inactivating mutation (H493R) in the enzyme tyrosyl-DNA phosphodiesterase (Tdp1), which removes blocked 3'-termini at DNA strand breaks. Using SCAN1 cells treated with the specific topoisomerase I (Top1) inhibitor camptothecin, we find enhanced levels of Top1 cleavage complexes (Top1cc) and defective reversal of Top1cc in SCAN1 Tdp1-deficient cells, indicating a direct involvement of Tdp1 in the repair of Top1cc. Because the defective removal of Top1cc and the hypersensitivity of SCAN1 cells to camptothecin are not affected by aphidicolin, we propose that Tdp1 is involved in the repair of Top1cc associated with transcription damage in SCAN1 cells.


Asunto(s)
Camptotecina/farmacología , Reparación del ADN , ADN-Topoisomerasas de Tipo I/metabolismo , Hidrolasas Diéster Fosfóricas/metabolismo , Ataxias Espinocerebelosas/metabolismo , Transcripción Genética , Afidicolina/farmacología , Línea Celular , Replicación del ADN/efectos de los fármacos , Humanos , Ataxias Espinocerebelosas/genética
20.
Nucleic Acids Res ; 33(1): 289-97, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-15647511

RESUMEN

Tyrosyl-DNA phosphodiesterase (TDP1) is a DNA repair enzyme that removes peptide fragments linked through tyrosine to the 3' end of DNA, and can also remove 3'-phosphoglycolates (PGs) formed by free radical-mediated DNA cleavage. To assess whether TDP1 is primarily responsible for PG removal during in vitro end joining of DNA double-strand breaks (DSBs), whole-cell extracts were prepared from lymphoblastoid cells derived either from spinocerebellar ataxia with axonal neuropathy (SCAN1) patients, who have an inactivating mutation in the active site of TDP1, or from closely matched normal controls. Whereas extracts from normal cells catalyzed conversion of 3'-PG termini, both on single-strand oligomers and on 3' overhangs of DSBs, to 3'-phosphate termini, extracts of SCAN1 cells did not process either substrate. Addition of recombinant TDP1 to SCAN1 extracts restored 3'-PG removal, allowing subsequent gap filling on the aligned DSB ends. Two of three SCAN1 lines examined were slightly more radiosensitive than normal cells, but only for fractionated radiation in plateau phase. The results suggest that the TDP1 mutation in SCAN1 abolishes the 3'-PG processing activity of the enzyme, and that there are no other enzymes in cell extracts capable of processing protruding 3'-PG termini. However, the lack of severe radiosensitivity suggests that there must be alternative, TDP1-independent pathways for repair of 3'-PG DSBs.


Asunto(s)
Daño del ADN , Glicolatos/metabolismo , Mutación , Hidrolasas Diéster Fosfóricas/genética , Hidrolasas Diéster Fosfóricas/metabolismo , Línea Celular , Humanos , Fosforilación/efectos de la radiación , Radiación Ionizante , Ataxias Espinocerebelosas/enzimología , Ataxias Espinocerebelosas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA