Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
bioRxiv ; 2024 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-38260538

RESUMEN

Repair of DNA double strand breaks by the non-homologous end-joining pathway is initiated by the binding of Ku to DNA ends. Given its high affinity for ends, multiple Ku proteins load onto linear DNAs in vitro. However, in cells, Ku loading is limited to ~1-2 molecules per DNA end. The mechanisms enforcing this limit are currently unknown. Here we show that the catalytic subunit of the DNA-dependent protein kinase (DNA-PKcs), but not its protein kinase activity, is required to prevent excessive Ku entry into chromatin. Ku accumulation is further restricted by two mechanisms: a neddylation/FBXL12-dependent process which actively removes loaded Ku molecules throughout the cell cycle and a CtIP/ATM-dependent mechanism which operates in S-phase. Finally, we demonstrate that the misregulation of Ku loading leads to impaired transcription in the vicinity of DNA ends. Together our data shed light on the multiple layers of coordinated mechanisms operating to prevent Ku from invading chromatin and interfering with other DNA transactions.

2.
Nucleic Acids Res ; 51(21): 11732-11747, 2023 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-37870477

RESUMEN

The classical Non-Homologous End Joining (c-NHEJ) pathway is the predominant process in mammals for repairing endogenous, accidental or programmed DNA Double-Strand Breaks. c-NHEJ is regulated by several accessory factors, post-translational modifications, endogenous chemical agents and metabolites. The metabolite inositol-hexaphosphate (IP6) stimulates c-NHEJ by interacting with the Ku70-Ku80 heterodimer (Ku). We report cryo-EM structures of apo- and DNA-bound Ku in complex with IP6, at 3.5 Å and 2.74 Å resolutions respectively, and an X-ray crystallography structure of a Ku in complex with DNA and IP6 at 3.7 Å. The Ku-IP6 interaction is mediated predominantly via salt bridges at the interface of the Ku70 and Ku80 subunits. This interaction is distant from the DNA, DNA-PKcs, APLF and PAXX binding sites and in close proximity to XLF binding site. Biophysical experiments show that IP6 binding increases the thermal stability of Ku by 2°C in a DNA-dependent manner, stabilizes Ku on DNA and enhances XLF affinity for Ku. In cells, selected mutagenesis of the IP6 binding pocket reduces both Ku accrual at damaged sites and XLF enrolment in the NHEJ complex, which translate into a lower end-joining efficiency. Thus, this study defines the molecular bases of the IP6 metabolite stimulatory effect on the c-NHEJ repair activity.


Asunto(s)
Proteínas de Unión al ADN , Ácido Fítico , Animales , ADN/metabolismo , Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades , Proteínas de Unión al ADN/genética , Autoantígeno Ku/metabolismo , Mamíferos/genética , Humanos
3.
Sci Adv ; 9(22): eadg2834, 2023 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-37256950

RESUMEN

Nonhomologous end joining is a critical mechanism that repairs DNA double-strand breaks in human cells. In this work, we address the structural and functional role of the accessory protein PAXX [paralog of x-ray repair cross-complementing protein 4 (XRCC4) and XRCC4-like factor (XLF)] in this mechanism. Here, we report high-resolution cryo-electron microscopy (cryo-EM) and x-ray crystallography structures of the PAXX C-terminal Ku-binding motif bound to Ku70/80 and cryo-EM structures of PAXX bound to two alternate DNA-dependent protein kinase (DNA-PK) end-bridging dimers, mediated by either Ku80 or XLF. We identify residues critical for the Ku70/PAXX interaction in vitro and in cells. We demonstrate that PAXX and XLF can bind simultaneously to the Ku heterodimer and act as structural bridges in alternate forms of DNA-PK dimers. Last, we show that engagement of both proteins provides a complementary advantage for DNA end synapsis and end joining in cells.


Asunto(s)
Reparación del ADN por Unión de Extremidades , Enzimas Reparadoras del ADN , Humanos , Microscopía por Crioelectrón , ADN , Enzimas Reparadoras del ADN/genética
4.
FEBS Lett ; 596(11): 1468-1480, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35561126

RESUMEN

Arginyl-tRNA-protein transferase 1 (ATE1) catalyses N-terminal protein arginylation, a post-translational modification implicated in cell migration, invasion and the cellular stress response. Herein, we report that ATE1 is overexpressed in NRAS-mutant melanomas, while it is downregulated in BRAF-mutant melanomas. ATE1 expression was higher in metastatic tumours, compared with primary tumours. Consistent with these findings, ATE1 depletion reduced melanoma cell viability, migration and colony formation. Reduced ATE1 expression also affected cell responses to mTOR and MEK inhibitors and to serum deprivation. Among putative ATE1 substrates is the tumour suppressor AXIN1, pointing to the possibility that ATE1 may fine-tune AXIN1 function in melanoma. Our findings highlight an unexpected role for ATE1 in melanoma cell aggressiveness and suggest that ATE1 constitutes a potential new therapeutic target.


Asunto(s)
Aminoaciltransferasas , Melanoma , Aminoaciltransferasas/genética , Aminoaciltransferasas/metabolismo , Movimiento Celular , Proliferación Celular , Humanos , Melanoma/genética , Procesamiento Proteico-Postraduccional , ARN de Transferencia/metabolismo
5.
Nucleic Acids Res ; 48(17): 9710-9723, 2020 09 25.
Artículo en Inglés | MEDLINE | ID: mdl-32890395

RESUMEN

Two DNA repair pathways operate at DNA double strand breaks (DSBs): non-homologous end-joining (NHEJ), that requires two adjacent DNA ends for ligation, and homologous recombination (HR), that resects one DNA strand for invasion of a homologous duplex. Faithful repair of replicative single-ended DSBs (seDSBs) is mediated by HR, due to the lack of a second DNA end for end-joining. ATM stimulates resection at such breaks through multiple mechanisms including CtIP phosphorylation, which also promotes removal of the DNA-ends sensor and NHEJ protein Ku. Here, using a new method for imaging the recruitment of the Ku partner DNA-PKcs at DSBs, we uncover an unanticipated role of ATM in removing DNA-PKcs from seDSBs in human cells. Phosphorylation of DNA-PKcs on the ABCDE cluster is necessary not only for DNA-PKcs clearance but also for the subsequent MRE11/CtIP-dependent release of Ku from these breaks. We propose that at seDSBs, ATM activity is necessary for the release of both Ku and DNA-PKcs components of the NHEJ apparatus, and thereby prevents subsequent aberrant interactions between seDSBs accompanied by DNA-PKcs autophosphorylation and detrimental commitment to Lig4-dependent end-joining.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades/fisiología , Proteína Quinasa Activada por ADN/metabolismo , Autoantígeno Ku/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada/genética , Camptotecina/farmacología , Línea Celular , Reparación del ADN por Unión de Extremidades/efectos de los fármacos , ADN Ligasa (ATP)/genética , ADN Ligasa (ATP)/metabolismo , ADN de Cadena Simple , Proteína Quinasa Activada por ADN/genética , Humanos , Autoantígeno Ku/genética , Proteína Homóloga de MRE11/genética , Proteína Homóloga de MRE11/metabolismo , Fosforilación , Inhibidores de Topoisomerasa I/farmacología
6.
Proteomics ; 20(3-4): e1900184, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31999075

RESUMEN

It is established that short inverted repeats trigger base substitution mutagenesis in human cells. However, how the replication machinery deals with structured DNA is unknown. It has been previously reported that in human cell-free extracts, DNA primer extension using a structured single-stranded template is transiently blocked at DNA hairpins. Here, the proteomic analysis of proteins bound to the DNA template is reported and evidence that the DNA-PK complex (DNA-PKcs and the Ku heterodimer) recognizes, and is activated by, structured single-stranded DNA is provided. Hijacking the DNA-PK complex by double-stranded oligonucleotides results in a large removal of the pausing sites and an elevated DNA extension efficiency. Conversely, DNA-PKcs inhibition results in its stabilization on the template, along with other proteins acting downstream in the Non-Homologous End-Joining (NHEJ) pathway, especially the XRCC4-DNA ligase 4 complex and the cofactor PAXX. Retention of NHEJ factors to the DNA in the absence of DNA-PKcs activity correlates with additional halts of primer extension, suggesting that these proteins hinder the progression of the DNA synthesis at these sites. Overall these results raise the possibility that, upon binding to hairpins formed onto ssDNA during fork progression, the DNA-PK complex interferes with replication fork dynamics in vivo.


Asunto(s)
Reparación del ADN por Unión de Extremidades , Replicación del ADN , ADN/metabolismo , Proteoma/metabolismo , Proteómica/métodos , Extractos Celulares , ADN/química , ADN/genética , ADN Ligasa (ATP)/metabolismo , Proteína Quinasa Activada por ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Humanos , Autoantígeno Ku/metabolismo , Proteínas Nucleares/metabolismo , Unión Proteica
7.
Prog Biophys Mol Biol ; 147: 62-76, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-30851288

RESUMEN

In vertebrates, double-strand breaks in DNA are primarily repaired by Non-Homologous End-Joining (NHEJ). The ring-shaped Ku heterodimer rapidly senses and threads onto broken DNA ends forming a recruiting hub. Through protein-protein contacts eventually reinforced by protein-DNA interactions, the Ku-DNA hub attracts a series of specialized proteins with scaffolding and/or enzymatic properties. To shed light on these dynamic interplays, we review here current knowledge on proteins directly interacting with Ku and on the contact points involved, with a particular accent on the different classes of Ku-binding motifs identified in several Ku partners. An integrated structural model of the core NHEJ network at the synapsis step is proposed.


Asunto(s)
Reparación del ADN por Unión de Extremidades , ADN/genética , ADN/metabolismo , Autoantígeno Ku/metabolismo , Secuencias de Aminoácidos , Animales , Humanos
8.
Nat Struct Mol Biol ; 25(10): 971-980, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30291363

RESUMEN

The Ku70-Ku80 (Ku) heterodimer binds rapidly and tightly to the ends of DNA double-strand breaks and recruits factors of the non-homologous end-joining (NHEJ) repair pathway through molecular interactions that remain unclear. We have determined crystal structures of the Ku-binding motifs (KBM) of the NHEJ proteins APLF (A-KBM) and XLF (X-KBM) bound to a Ku-DNA complex. The two KBM motifs bind remote sites of the Ku80 α/ß domain. The X-KBM occupies an internal pocket formed by an unprecedented large outward rotation of the Ku80 α/ß domain. We observe independent recruitment of the APLF-interacting protein XRCC4 and of XLF to laser-irradiated sites via binding of A- and X-KBMs, respectively, to Ku80. Finally, we show that mutation of the X-KBM and A-KBM binding sites in Ku80 compromises both the efficiency and accuracy of end joining and cellular radiosensitivity. A- and X-KBMs may represent two initial anchor points to build the intricate interaction network required for NHEJ.


Asunto(s)
Reparación del ADN por Unión de Extremidades , Enzimas Reparadoras del ADN/química , ADN-(Sitio Apurínico o Apirimidínico) Liasa/química , Proteínas de Unión al ADN/química , Autoantígeno Ku/química , Proteínas de Unión a Poli-ADP-Ribosa/química , Secuencia Conservada , Cristalografía por Rayos X , Enzimas Reparadoras del ADN/metabolismo , Enzimas Reparadoras del ADN/fisiología , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , ADN-(Sitio Apurínico o Apirimidínico) Liasa/fisiología , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/fisiología , Humanos , Autoantígeno Ku/metabolismo , Autoantígeno Ku/fisiología , Modelos Moleculares , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/fisiología , Dominios Proteicos
9.
Mol Cancer Ther ; 16(10): 2166-2177, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28611105

RESUMEN

Poisons of topoisomerase II (TOP2) kill cancer cells by preventing religation of intermediate DNA breaks during the enzymatic process and thus by accumulating enzyme-drug-DNA complexes called TOP2 cleavage-complex (TOP2cc). F14512 is a highly cytotoxic polyamine-vectorized TOP2 inhibitor derived from etoposide and currently in clinical trials. It was shown in vitro that F14512 has acquired DNA-binding properties and that the stability of TOP2cc was strongly increased. Paradoxically, at equitoxic concentrations in cells, F14512 induced less DNA breaks than etoposide. Here, we directly compared etoposide and F14512 for their rates of TOP2cc production and resolution in human cells. We report that targeting of TOP2α and not TOP2ß impacts cell killing by F14512, contrary to etoposide that kills cells through targeting both isoforms. Then, we show that despite being more cytotoxic, F14512 is less efficient than etoposide at producing TOP2α cleavage-complex (TOP2αcc) in cells. Finally, we report that compared with TOP2αcc mediated by etoposide, those generated by F14512 persist longer in the genome, are not dependent on TDP2 for cleaning break ends from TOP2α, are channeled to a larger extent to resection-based repair processes relying on CtIP and BRCA1 and promote RAD51 recruitment to damaged chromatin. In addition to the addressing of F14512 to the polyamine transport system, the properties uncovered here would be particularly valuable for a therapeutic usage of this new anticancer compound. More generally, the concept of increasing drug cytotoxicity by switching the repair mode of the induced DNA lesions via addition of a DNA-binding moiety deserves further developments. Mol Cancer Ther; 16(10); 2166-77. ©2017 AACR.


Asunto(s)
Roturas del ADN de Doble Cadena/efectos de los fármacos , ADN-Topoisomerasas de Tipo II/genética , Neoplasias/tratamiento farmacológico , Inhibidores de Topoisomerasa II/administración & dosificación , Apoptosis/efectos de los fármacos , Proteína BRCA1/genética , Cromatina/genética , Vectores Genéticos/efectos de los fármacos , Humanos , Neoplasias/genética , Neoplasias/patología , Podofilotoxina/administración & dosificación , Podofilotoxina/análogos & derivados , Poliaminas/administración & dosificación , Recombinasa Rad51/genética
10.
EMBO Rep ; 17(4): 508-18, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26964895

RESUMEN

Ku heterodimer is a DNA binding protein with a prominent role in DNA repair. Here, we investigate whether and how Ku impacts the DNA damage response by acting as a post-transcriptional regulator of gene expression. We show that Ku represses p53 protein synthesis and p53-mediated apoptosis by binding to a bulged stem-loop structure within the p53 5' UTR However, Ku-mediated translational repression of the p53 mRNA is relieved after genotoxic stress. The underlying mechanism involves Ku acetylation which disrupts Ku-p53 mRNA interactions. These results suggest that Ku-mediated repression of p53 mRNA translation constitutes a novel mechanism linking DNA repair and mRNA translation.


Asunto(s)
Daño del ADN/fisiología , Reparación del ADN , Autoantígeno Ku/metabolismo , Biosíntesis de Proteínas , ARN Mensajero/genética , Proteína p53 Supresora de Tumor/genética , Regiones no Traducidas 5' , Acetilación , Apoptosis , Daño del ADN/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Humanos , Autoantígeno Ku/genética , Unión Proteica , ARN Mensajero/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
11.
Nucleic Acids Res ; 43(21): 10264-76, 2015 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-26350212

RESUMEN

In humans, DNA double-strand breaks (DSBs) are repaired by two mutually-exclusive mechanisms, homologous recombination or end-joining. Among end-joining mechanisms, the main process is classical non-homologous end-joining (C-NHEJ) which relies on Ku binding to DNA ends and DNA Ligase IV (Lig4)-mediated ligation. Mostly under Ku- or Lig4-defective conditions, an alternative end-joining process (A-EJ) can operate and exhibits a trend toward microhomology usage at the break junction. Homologous recombination relies on an initial MRN-dependent nucleolytic degradation of one strand at DNA ends. This process, named DNA resection generates 3' single-stranded tails necessary for homologous pairing with the sister chromatid. While it is believed from the current literature that the balance between joining and recombination processes at DSBs ends is mainly dependent on the initiation of resection, it has also been shown that MRN activity can generate short single-stranded DNA oligonucleotides (ssO) that may also be implicated in repair regulation. Here, we evaluate the effect of ssO on end-joining at DSB sites both in vitro and in cells. We report that under both conditions, ssO inhibit C-NHEJ through binding to Ku and favor repair by the Lig4-independent microhomology-mediated A-EJ process.


Asunto(s)
Antígenos Nucleares/metabolismo , Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades , ADN de Cadena Simple/metabolismo , Proteínas de Unión al ADN/metabolismo , ADN/metabolismo , Células HeLa , Humanos , Autoantígeno Ku , Oligodesoxirribonucleótidos/metabolismo
12.
Nucleic Acids Res ; 42(14): 9047-62, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25030905

RESUMEN

We previously identified the heterogeneous ribonucleoprotein SAF-A/hnRNP U as a substrate for DNA-PK, a protein kinase involved in DNA damage response (DDR). Using laser micro-irradiation in human cells, we report here that SAF-A exhibits a two-phase dynamics at sites of DNA damage, with a rapid and transient recruitment followed by a prolonged exclusion. SAF-A recruitment corresponds to its binding to Poly(ADP-ribose) while its exclusion is dependent on the activity of ATM, ATR and DNA-PK and reflects the dissociation from chromatin of SAF-A associated with ongoing transcription. Having established that SAF-A RNA-binding domain recapitulates SAF-A dynamics, we show that this domain is part of a complex comprising several mRNA biogenesis proteins of which at least two, FUS/TLS and TAFII68/TAF15, exhibit similar biphasic dynamics at sites of damage. Using an original reporter for live imaging of DNA:RNA hybrids (R-loops), we show a transient transcription-dependent accumulation of R-loops at sites of DNA damage that is prolonged upon inhibition of RNA biogenesis factors exclusion. We propose that a new component of the DDR is an active anti-R-loop mechanism operating at damaged transcribed sites which includes the exclusion of mRNA biogenesis factors such as SAF-A, FUS and TAF15.


Asunto(s)
Daño del ADN , Ribonucleoproteína Heterogénea-Nuclear Grupo U/metabolismo , Proteínas de Unión al ARN/metabolismo , Línea Celular , Núcleo Celular/metabolismo , Cromatina/metabolismo , Ribonucleoproteína Heterogénea-Nuclear Grupo U/química , Humanos , Fosfatidilinositol 3-Quinasas/metabolismo , Poli Adenosina Difosfato Ribosa/metabolismo , Estructura Terciaria de Proteína , Proteína FUS de Unión a ARN/metabolismo , Factores Asociados con la Proteína de Unión a TATA/metabolismo , Transcripción Genética
13.
DNA Repair (Amst) ; 17: 81-97, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24613763

RESUMEN

To cope with DNA double strand break (DSB) genotoxicity, cells have evolved two main repair pathways: homologous recombination which uses homologous DNA sequences as repair templates, and non-homologous Ku-dependent end-joining involving direct sealing of DSB ends by DNA ligase IV (Lig4). During the last two decades a third player most commonly named alternative end-joining (A-EJ) has emerged, which is defined as any Ku- or Lig4-independent end-joining process. A-EJ increasingly appears as a highly error-prone bricolage on DSBs and despite expanding exploration, it still escapes full characterization. In the present review, we discuss the mechanism and regulation of A-EJ as well as its biological relevance under physiological and pathological situations, with a particular emphasis on chromosomal instability and cancer. Whether or not it is a genuine DSB repair pathway, A-EJ is emerging as an important cellular process and understanding A-EJ will certainly be a major challenge for the coming years.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades , Enzimas Reparadoras del ADN/metabolismo , Neoplasias/genética , Antígenos Nucleares/metabolismo , Inestabilidad Cromosómica , ADN Ligasas/metabolismo , Proteínas de Unión al ADN/metabolismo , Humanos , Autoantígeno Ku , Modelos Genéticos
14.
J Cell Biol ; 200(2): 173-86, 2013 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-23337116

RESUMEN

Nonhomologous end joining is the primary deoxyribonucleic acid (DNA) double-strand break repair pathway in multicellular eukaryotes. To initiate repair, Ku binds DNA ends and recruits the DNA-dependent protein kinase (DNA-PK) catalytic subunit (DNA-PKcs) forming the holoenzyme. Early end synapsis is associated with kinase autophosphorylation. The XRCC4 (X4)-DNA Ligase IV (LIG4) complex (X4LIG4) executes the final ligation promoted by Cernunnos (Cer)-X4-like factor (XLF). In this paper, using a cell-free system that recapitulates end synapsis and DNA-PKcs autophosphorylation, we found a defect in both activities in human cell extracts lacking LIG4. LIG4 also stimulated the DNA-PKcs autophosphorylation in a reconstitution assay with purified components. We additionally uncovered a kinase autophosphorylation defect in LIG4-defective cells that was corrected by ectopic expression of catalytically dead LIG4. Finally, our data support a contribution of Cer-XLF to this unexpected early role of the ligation complex in end joining. We propose that productive end joining occurs by early formation of a supramolecular entity containing both DNA-PK and X4LIG4-Cer-XLF complexes on DNA ends.


Asunto(s)
Reparación del ADN por Unión de Extremidades/fisiología , ADN Ligasas/fisiología , Proteínas de Unión al ADN/fisiología , Sistema Libre de Células/metabolismo , Células Cultivadas , Daño del ADN , ADN Helicasas/metabolismo , ADN Helicasas/fisiología , ADN Ligasa (ATP) , ADN Ligasas/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Enzimas Reparadoras del ADN/fisiología , Proteína Quinasa Activada por ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Inestabilidad Genómica , Holoenzimas , Humanos , Autoantígeno Ku , Fosforilación
15.
Nucleic Acids Res ; 39(22): 9605-19, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21880593

RESUMEN

In mammalian cells, the main pathway for DNA double-strand breaks (DSBs) repair is classical non-homologous end joining (C-NHEJ). An alternative or back-up NHEJ (B-NHEJ) pathway has emerged which operates preferentially under C-NHEJ defective conditions. Although B-NHEJ appears particularly relevant to genomic instability associated with cancer, its components and regulation are still largely unknown. To get insights into this pathway, we have knocked-down Ku, the main contributor to C-NHEJ. Thus, models of human cell lines have been engineered in which the expression of Ku70/80 heterodimer can be significantly lowered by the conditional induction of a shRNA against Ku70. On Ku reduction in cells, resulting NHEJ competent protein extracts showed a shift from C- to B-NHEJ that could be reversed by addition of purified Ku protein. Using a cellular fractionation protocol after treatment with a strong DSBs inducer followed by western blotting or immunostaining, we established that, among C-NHEJ factors, Ku is the main counteracting factor against mobilization of PARP1 and the MRN complex to damaged chromatin. In addition, Ku limits PAR synthesis and single-stranded DNA production in response to DSBs. These data support the involvement of PARP1 and the MRN proteins in the B-NHEJ route for the repair of DNA DSBs.


Asunto(s)
Antígenos Nucleares/metabolismo , Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades , Proteínas de Unión al ADN/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Antígenos Nucleares/genética , Fraccionamiento Celular , Línea Celular , Cromatina/química , Cromatina/metabolismo , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Técnicas de Silenciamiento del Gen , Humanos , Autoantígeno Ku , Poli(ADP-Ribosa) Polimerasa-1 , Inhibidores de Poli(ADP-Ribosa) Polimerasas
16.
J Cell Sci ; 124(Pt 11): 1943-51, 2011 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-21576354

RESUMEN

DNA-dependent protein kinase (DNA-PK) is involved in DNA double-strand break (DSB) signalling and repair. We report that DNA-PK is activated by mild hypoxia conditions (0.1-1% O2) as shown by (1) its autophosphorylation on Ser2056, and (2) its mobilisation from a soluble nucleoplasmic compartment to a less extractable nuclear fraction. The recruitment of DNA-PK was not followed by activation and recruitment of the XRCC4-DNA-ligase-IV complex, suggesting that DSBs are not responsible for activation of DNA-PK. To unravel the mechanism of DNA-PK activation, we show that exposure of cells to trichostatin A, a histone deacetylase inhibitor, leads to DNA-PK autophosphorylation and relocalisation to DNA. Histone acetylation (mainly H3K14) is increased in hypoxic cells and treatment with anacardic acid, an inhibitor of histone acetyl transferase, prevented both histone modifications and DNA-PK activation in hypoxic conditions. Importantly, in using either silenced DNA-PK cells or cells exposed to a specific DNA-PK inhibitor (NU7026), we demonstrated that hypoxic DNA-PK activation positively regulates the key transcription factor HIF-1 and one subsequent target gene, GLUT1. Our results show that hypoxia initiates chromatin modification and consequently DNA-PK activation, which positively regulate cellular oxygen-sensing and oxygen-signalling pathways.


Asunto(s)
Cromatina/metabolismo , Proteína Quinasa Activada por ADN/metabolismo , Estrés Fisiológico , Acetilación , Adaptación Fisiológica , Aminoglicósidos/farmacología , Antígenos Nucleares/metabolismo , Hipoxia de la Célula , Línea Celular , Cromonas/farmacología , Daño del ADN , Proteína Quinasa Activada por ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/metabolismo , Enediinos/farmacología , Activación Enzimática , Transportador de Glucosa de Tipo 1/metabolismo , Histonas/metabolismo , Humanos , Ácidos Hidroxámicos/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Autoantígeno Ku , Morfolinas/farmacología , Fosforilación , Procesamiento Proteico-Postraduccional , Transducción de Señal
17.
EMBO J ; 29(9): 1573-84, 2010 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-20407424

RESUMEN

DNA-dependent protein kinase (DNA-PK) is a double-strand breaks repair complex, the subunits of which (KU and DNA-PKcs) are paradoxically present at mammalian telomeres. Telomere fusion has been reported in cells lacking these proteins, raising two questions: how is DNA-PK prevented from initiating classical ligase IV (LIG4)-dependent non-homologous end-joining (C-NHEJ) at telomeres and how is the backup end-joining (EJ) activity (B-NHEJ) that operates at telomeres under conditions of C-NHEJ deficiency controlled? To address these questions, we have investigated EJ using plasmid substrates bearing double-stranded telomeric tracks and human cell extracts with variable C-NHEJ or B-NHEJ activity. We found that (1) TRF2/RAP1 prevents C-NHEJ-mediated end fusion at the initial DNA-PK end binding and activation step and (2) DNA-PK counteracts a potent LIG4-independent EJ mechanism. Thus, telomeres are protected against EJ by a lock with two bolts. These results account for observations with mammalian models and underline the importance of alternative non-classical EJ pathways for telomere fusions in cells.


Asunto(s)
Reparación del ADN , Proteína Quinasa Activada por ADN/metabolismo , Proteínas de Unión a Telómeros/metabolismo , Telómero/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo , ADN/metabolismo , ADN Ligasa (ATP) , ADN Ligasas/metabolismo , Inestabilidad Genómica , Células HeLa , Humanos , Complejo Shelterina
18.
Cell Cycle ; 8(22): 3717-22, 2009 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-19844162

RESUMEN

Aiming to identify novel phosphorylation sites in response to DNA double-strand breaks (DSB) inducers, we have isolated a phosphorylation site on KU70. Unexpectedly, a rabbit antiserum raised against this site cross-reacted with a 120 kDa protein in cells treated by DNA DSB inducers. We identified this protein as SAF-A/hnRNP U, an abundant and essential nuclear protein containing regions binding DNA or RNA. The phosphorylation site was mapped at S59 position in a sequence context favoring a "S-hydrophobic" consensus model for DNA-PK phosphorylation site in vivo. This site was exclusively phosphorylated by DNA-PK in response to DNA DSB inducers. In addition, the extent and duration of this phosphorylation was in inverse correlation with the capacity of the cells to repair DSB by Nonhomologous End Joining. These results bring a new link between the hnRNP family and the DNA damage response. Addtionaly, the mapped phospho-site on SAF-A might serve as a potential bio-marker for DNA-PK activity in academic studies and clinical analyses of DNA-PK activators or inhibitors.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN/fisiología , Proteína Quinasa Activada por ADN/metabolismo , ADN/metabolismo , Ribonucleoproteína Heterogénea-Nuclear Grupo U/metabolismo , Proteínas Nucleares/metabolismo , Secuencia de Aminoácidos , Animales , Anticuerpos/inmunología , Antígenos Nucleares/inmunología , Antígenos Nucleares/metabolismo , Secuencia de Bases , Línea Celular , Proteínas de Unión al ADN/inmunología , Proteínas de Unión al ADN/metabolismo , Ribonucleoproteína Heterogénea-Nuclear Grupo U/genética , Humanos , Immunoblotting , Inmunoprecipitación , Autoantígeno Ku , Espectrometría de Masas , Datos de Secuencia Molecular , Fosforilación , Conejos , Análisis de Secuencia de ADN
19.
Cancer Res ; 69(20): 8120-6, 2009 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-19808974

RESUMEN

One hallmark of apoptosis is DNA degradation that first appears as high molecular weight fragments followed by extensive internucleosomal fragmentation. During apoptosis, the DNA-dependent protein kinase (DNA-PK) is activated. DNA-PK is involved in the repair of DNA double-strand breaks (DSB) and its catalytic subunit is associated with the nuclease ARTEMIS. Here, we report that, on initiation of apoptosis in human cells by agents causing DNA DSB or by staurosporine or other agents, ARTEMIS binds to apoptotic chromatin together with DNA-PK and other DSB repair proteins. ARTEMIS recruitment to chromatin showed a time and dose dependency. It required DNA-PK protein kinase activity and was blocked by antagonizing the onset of apoptosis with a pan-caspase inhibitor or on overexpression of the antiapoptotic BCL2 protein. In the absence of ARTEMIS, no defect in caspase-3, poly(ADP-ribose) polymerase-1, and XRCC4 cleavage or in H2AX phosphorylation was observed and DNA-PK catalytic subunit was still phosphorylated on S2056 in response to staurosporine. However, DNA fragmentation including high molecular weight fragmentation was delayed in ARTEMIS-deficient cells compared with cells expressing ARTEMIS. In addition, ARTEMIS enhanced the kinetics of MLL gene cleavage at a breakage cluster breakpoint that is frequently translocated in acute or therapy-related leukemias. These results show a facilitating role for ARTEMIS at least in early, site-specific chromosome breakage during apoptosis.


Asunto(s)
Apoptosis , Cromatina/metabolismo , Roturas del ADN de Doble Cadena , Fibroblastos/enzimología , Proteínas Nucleares/metabolismo , Southern Blotting , Caspasa 3/metabolismo , Células Cultivadas , Cromatina/genética , Reparación del ADN , Proteína Quinasa Activada por ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Endonucleasas , N-Metiltransferasa de Histona-Lisina , Histonas/metabolismo , Humanos , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Proteínas Nucleares/genética , Fosforilación , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Estaurosporina/farmacología
20.
Mol Cell Biol ; 29(11): 3163-72, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19332554

RESUMEN

Nonhomologous end-joining represents the major pathway used by human cells to repair DNA double-strand breaks. It relies on the XRCC4/DNA ligase IV complex to reseal DNA strands. Here we report the high-resolution crystal structure of human XRCC4 bound to the carboxy-terminal tandem BRCT repeat of DNA ligase IV. The structure differs from the homologous Saccharomyces cerevisiae complex and reveals an extensive DNA ligase IV binding interface formed by a helix-loop-helix structure within the inter-BRCT linker region, as well as significant interactions involving the second BRCT domain, which induces a kink in the tail region of XRCC4. We further demonstrate that interaction with the second BRCT domain of DNA ligase IV is necessary for stable binding to XRCC4 in cells, as well as to achieve efficient dominant-negative effects resulting in radiosensitization after ectopic overexpression of DNA ligase IV fragments in human fibroblasts. Together our findings provide unanticipated insight for understanding the physical and functional architecture of the nonhomologous end-joining ligation complex.


Asunto(s)
ADN Ligasas/química , ADN Ligasas/metabolismo , Reparación del ADN , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Secuencia de Aminoácidos , Unión Competitiva , Línea Celular , Roturas del ADN de Doble Cadena , ADN Ligasa (ATP) , Enzimas Reparadoras del ADN/metabolismo , Regulación hacia Abajo , Humanos , Datos de Secuencia Molecular , Unión Proteica , Estabilidad Proteica , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Tolerancia a Radiación , Recombinación Genética/genética , Homología Estructural de Proteína , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA