Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 81(4): 801-810.e3, 2021 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-33385326

RESUMEN

DNA-dependent protein kinase (DNA-PK), like all phosphatidylinositol 3-kinase-related kinases (PIKKs), is composed of conserved FAT and kinase domains (FATKINs) along with solenoid structures made of HEAT repeats. These kinases are activated in response to cellular stress signals, but the mechanisms governing activation and regulation remain unresolved. For DNA-PK, all existing structures represent inactive states with resolution limited to 4.3 Å at best. Here, we report the cryoelectron microscopy (cryo-EM) structures of DNA-PKcs (DNA-PK catalytic subunit) bound to a DNA end or complexed with Ku70/80 and DNA in both inactive and activated forms at resolutions of 3.7 Å overall and 3.2 Å for FATKINs. These structures reveal the sequential transition of DNA-PK from inactive to activated forms. Most notably, activation of the kinase involves previously unknown stretching and twisting within individual solenoid segments and loosens DNA-end binding. This unprecedented structural plasticity of helical repeats may be a general regulatory mechanism of HEAT-repeat proteins.


Asunto(s)
Reparación del ADN por Unión de Extremidades , Proteína Quinasa Activada por ADN/química , Autoantígeno Ku/química , Complejos Multiproteicos/química , Microscopía por Crioelectrón , Proteína Quinasa Activada por ADN/genética , Activación Enzimática , Células HEK293 , Células HeLa , Humanos , Complejos Multiproteicos/genética , Complejos Multiproteicos/ultraestructura
2.
Nucleic Acids Res ; 50(13): 7697-7720, 2022 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-35801871

RESUMEN

Artemis nuclease and DNA-dependent protein kinase catalytic subunit (DNA-PKcs) are key components in nonhomologous DNA end joining (NHEJ), the major repair mechanism for double-strand DNA breaks. Artemis activation by DNA-PKcs resolves hairpin DNA ends formed during V(D)J recombination. Artemis deficiency disrupts development of adaptive immunity and leads to radiosensitive T- B- severe combined immunodeficiency (RS-SCID). An activated state of Artemis in complex with DNA-PK was solved by cryo-EM recently, which showed Artemis bound to the DNA. Here, we report that the pre-activated form (basal state) of the Artemis:DNA-PKcs complex is stable on an agarose-acrylamide gel system, and suitable for cryo-EM structural analysis. Structures show that the Artemis catalytic domain is dynamically positioned externally to DNA-PKcs prior to ABCDE autophosphorylation and show how both the catalytic and regulatory domains of Artemis interact with the N-HEAT and FAT domains of DNA-PKcs. We define a mutually exclusive binding site for Artemis and XRCC4 on DNA-PKcs and show that an XRCC4 peptide disrupts the Artemis:DNA-PKcs complex. All of the findings are useful in explaining how a hypomorphic L3062R missense mutation of DNA-PKcs could lead to insufficient Artemis activation, hence RS-SCID. Our results provide various target site candidates to design disruptors for Artemis:DNA-PKcs complex formation.


Asunto(s)
Proteína Quinasa Activada por ADN/química , Proteínas de Unión al ADN/química , Endonucleasas/química , Reparación del ADN , Proteína Quinasa Activada por ADN/genética , Proteína Quinasa Activada por ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Endonucleasas/metabolismo , Humanos , Proteínas Nucleares/metabolismo , Inmunodeficiencia Combinada Grave/genética
3.
Cell Mol Biol Lett ; 25: 2, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31988640

RESUMEN

This review focuses on DNA-dependent protein kinase (DNA-PK), which is the key regulator of canonical non-homologous end-joining (NHEJ), the predominant mechanism of DNA double-strand break (DSB) repair in mammals. DNA-PK consists of the DNA-binding Ku70/80 heterodimer and the catalytic subunit DNA-PKcs. They assemble at DNA ends, forming the active DNA-PK complex, which initiates NHEJ-mediated DSB repair. Paradoxically, both Ku and DNA-PKcs are associated with telomeres, and they play crucial roles in protecting the telomere against fusions. Herein, we discuss possible mechanisms and contributions of Ku and DNA-PKcs in telomere regulation.


Asunto(s)
Proteína Quinasa Activada por ADN/metabolismo , Ribonucleoproteína Nuclear Heterogénea A1/metabolismo , Telomerasa/metabolismo , Proteínas de Unión a Telómeros/metabolismo , Telómero/metabolismo , Animales , Reparación del ADN por Unión de Extremidades/genética , ADN-Topoisomerasas de Tipo II/metabolismo , Proteína Quinasa Activada por ADN/química , Proteína Quinasa Activada por ADN/genética , Humanos , Autoantígeno Ku/metabolismo , Complejo Shelterina , Telómero/genética
4.
Nucleic Acids Res ; 46(4): 1847-1859, 2018 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-29309644

RESUMEN

The DNA-dependent protein kinase (DNA-PK), consisting of the DNA binding Ku70/80 heterodimer and the catalytic subunit DNA-PKcs, has been well characterized in the non-homologous end-joining mechanism for DNA double strand break (DSB) repair and radiation resistance. Besides playing a role in DSB repair, DNA-PKcs is required for the cellular response to replication stress and participates in the ATR-Chk1 signaling pathway. However, the mechanism through which DNA-PKcs is recruited to stalled replication forks is still unclear. Here, we report that the apoptosis mediator p53-induced protein with a death domain (PIDD) is required to promote DNA-PKcs activity in response to replication stress. PIDD is known to interact with PCNA upon UV-induced replication stress. Our results demonstrate that PIDD is required to recruit DNA-PKcs to stalled replication forks through direct binding to DNA-PKcs at the N' terminal region. Disruption of the interaction between DNA-PKcs and PIDD not only compromises the ATR association and regulation of DNA-PKcs, but also the ATR signaling pathway, intra-S-phase checkpoint and cellular resistance to replication stress. Taken together, our results indicate that PIDD, but not the Ku heterodimer, mediates the DNA-PKcs activity at stalled replication forks and facilitates the ATR signaling pathway in the cellular response to replication stress.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Replicación del ADN , Proteína Quinasa Activada por ADN/metabolismo , Proteínas Adaptadoras de Señalización del Receptor del Dominio de Muerte/metabolismo , Proteínas Nucleares/metabolismo , Secuencias de Aminoácidos , Animales , Línea Celular , Cricetinae , Proteína Quinasa Activada por ADN/química , Humanos , Autoantígeno Ku/fisiología , Proteínas Nucleares/química , Puntos de Control de la Fase S del Ciclo Celular , Transducción de Señal , Estrés Fisiológico , Rayos Ultravioleta
5.
Proc Natl Acad Sci U S A ; 114(28): 7367-7372, 2017 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-28652322

RESUMEN

DNA-dependent protein kinase (DNA-PK) is a large protein complex central to the nonhomologous end joining (NHEJ) DNA-repair pathway. It comprises the DNA-PK catalytic subunit (DNA-PKcs) and the heterodimer of DNA-binding proteins Ku70 and Ku80. Here, we report the cryo-electron microscopy (cryo-EM) structures of human DNA-PKcs at 4.4-Å resolution and the DNA-PK holoenzyme at 5.8-Å resolution. The DNA-PKcs structure contains three distinct segments: the N-terminal region with an arm and a bridge, the circular cradle, and the head that includes the kinase domain. Two perpendicular apertures exist in the structure, which are sufficiently large for the passage of dsDNA. The DNA-PK holoenzyme cryo-EM map reveals density for the C-terminal globular domain of Ku80 that interacts with the arm of DNA-PKcs. The Ku80-binding site is adjacent to the previously identified density for the DNA-binding region of the Ku70/Ku80 complex, suggesting concerted DNA interaction by DNA-PKcs and the Ku complex.


Asunto(s)
Microscopía por Crioelectrón , Proteína Quinasa Activada por ADN/química , Proteínas Nucleares/química , Catálisis , Dominio Catalítico , Cristalografía por Rayos X , Roturas del ADN de Doble Cadena , Reparación del ADN , Proteína Quinasa Activada por ADN/genética , Células HeLa , Humanos , Autoantígeno Ku/química , Proteínas Nucleares/genética , Unión Proteica
6.
Proc Jpn Acad Ser B Phys Biol Sci ; 96(5): 171-179, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32389917

RESUMEN

It is generally thought that younger people are more susceptible to cancer development after exposure to ionizing radiation in reference to epidemiological studies and animal experiments. However, little is known about the age-dependent alteration in DNA repair ability. In the present study, we examined the expression levels of proteins involved in the repair of DNA double-strand breaks through non-homologous end joining (NHEJ), i.e., DNA-dependent protein kinase catalytic subunit (DNA-PKcs), X-ray repair cross-complementing 4 (XRCC4) and XRCC4-like factor (XLF). We found that the expression of DNA-PKcs in brain tissues was higher in neonatal mice (1 week after birth) than in young adult mice (7 weeks after birth). In association with this, DNA double-strand breaks were repaired more rapidly in the brain tissues of neonatal mice than in those of young adult mice. The current results suggested a possible role for DNA-PKcs protecting developing brain tissues from DNA double-strand breaks.


Asunto(s)
Encéfalo/metabolismo , Dominio Catalítico , Proteína Quinasa Activada por ADN/química , Proteína Quinasa Activada por ADN/metabolismo , Regulación Enzimológica de la Expresión Génica , Animales , Animales Recién Nacidos , Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades , Reparación del ADN , Ratones
7.
J Biol Chem ; 292(34): 13914-13924, 2017 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-28696258

RESUMEN

In humans, nonhomologous DNA end-joining (NHEJ) is the major pathway by which DNA double-strand breaks are repaired. Recognition of each broken DNA end by the DNA repair protein Ku is the first step in NHEJ, followed by the iterative binding of nucleases, DNA polymerases, and the XRCC4-DNA ligase IV (X4-LIV) complex in an order influenced by the configuration of the two DNA ends at the break site. The endonuclease Artemis improves joining efficiency by functioning in a complex with DNA-dependent protein kinase, catalytic subunit (DNA-PKcs) that carries out endonucleolytic cleavage of 5' and 3' overhangs. Previously, we observed that X4-LIV alone can stimulate Artemis activity on 3' overhangs, but this DNA-PKcs-independent endonuclease activity of Artemis awaited confirmation. Here, using in vitro nuclease and ligation assays, we find that stimulation of Artemis nuclease activity by X4-LIV and the efficiency of blunt-end ligation are determined by structural configurations at the DNA end. Specifically, X4-LIV stimulated Artemis to cut near the end of 3' overhangs without the involvement of other NHEJ proteins. Of note, this ligase complex is not able to stimulate Artemis activity at hairpins or at 5' overhangs. We also found that X4-LIV and DNA-PKcs interfere with one another with respect to stimulating Artemis activity at 3' overhangs, favoring the view that these NHEJ proteins are sequentially rather than concurrently recruited to DNA ends. These data suggest specific functional and positional relationships among these components that explain genetic and molecular features of NHEJ and V(D)J recombination within cells.


Asunto(s)
ADN Ligasa (ATP)/metabolismo , Proteínas de Unión al ADN/metabolismo , Endonucleasas/metabolismo , Modelos Moleculares , Reparación del ADN por Recombinación , Recombinación V(D)J , Animales , Línea Celular , ADN/química , ADN/metabolismo , Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades , ADN Ligasa (ATP)/química , ADN Ligasa (ATP)/genética , Proteína Quinasa Activada por ADN/química , Proteína Quinasa Activada por ADN/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Endonucleasas/química , Células HeLa , Humanos , Cinética , Mariposas Nocturnas , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Conformación de Ácido Nucleico , Multimerización de Proteína , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Células Sf9 , Especificidad por Sustrato
8.
Proteins ; 86(10): 1055-1063, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30019773

RESUMEN

The Ets-1 oncoprotein is a transcription factor that promotes target gene expression in specific biological processes. Typically, Ets-1 activity is low in healthy cells, but elevated levels of expression have been found in cancerous cells, specifically related to tumor progression. Like the vast majority of the cellular effectors, Ets-1 does not act alone but in association with partners. Given the important role that is attributed to Ets-1 in major human diseases, it is crucial to identify its partners and characterize their interactions. In this context, two DNA-repair enzymes, PARP-1 and DNA-PK, have been identified recently as interaction partners of Ets-1. We here identify their binding mode by means of protein docking. The results identify the interacting surface between Ets-1 and the two DNA-repair enzymes centered on the α-helix H1 of the ETS domain, leaving α-helix H3 available to bind DNA. The models highlight a hydrophobic patch on Ets-1 at the center of the interaction interface that includes three tryptophans (Trp338, Trp356, and Trp361). We rationalize the binding mode using a series of computational analyses, including alanine scanning, molecular dynamics simulation, and residue centrality analysis. Our study constitutes a first but important step in the characterization, at the molecular level, of the interaction between an oncoprotein and DNA-repair enzymes.


Asunto(s)
Enzimas Reparadoras del ADN/metabolismo , Mapas de Interacción de Proteínas , Proteína Proto-Oncogénica c-ets-1/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Enzimas Reparadoras del ADN/química , Proteína Quinasa Activada por ADN/química , Proteína Quinasa Activada por ADN/metabolismo , Humanos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Poli(ADP-Ribosa) Polimerasa-1/química , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Unión Proteica , Conformación Proteica , Conformación Proteica en Hélice alfa , Proteína Proto-Oncogénica c-ets-1/química , Alineación de Secuencia
9.
J Biol Chem ; 291(53): 26987-27006, 2016 12 30.
Artículo en Inglés | MEDLINE | ID: mdl-27875301

RESUMEN

DNA double-strand break (DSB) repair by non-homologous end joining (NHEJ) in human cells is initiated by Ku heterodimer binding to a DSB, followed by recruitment of core NHEJ factors including DNA-dependent protein kinase catalytic subunit (DNA-PKcs), XRCC4-like factor (XLF), and XRCC4 (X4)-DNA ligase IV (L4). Ku also interacts with accessory factors such as aprataxin and polynucleotide kinase/phosphatase-like factor (APLF). Yet, how these factors interact to tether, process, and ligate DSB ends while allowing regulation and chromatin interactions remains enigmatic. Here, small angle X-ray scattering (SAXS) and mutational analyses show APLF is largely an intrinsically disordered protein that binds Ku, Ku/DNA-PKcs (DNA-PK), and X4L4 within an extended flexible NHEJ core complex. X4L4 assembles with Ku heterodimers linked to DNA-PKcs via flexible Ku80 C-terminal regions (Ku80CTR) in a complex stabilized through APLF interactions with Ku, DNA-PK, and X4L4. Collective results unveil the solution architecture of the six-protein complex and suggest cooperative assembly of an extended flexible NHEJ core complex that supports APLF accessibility while possibly providing flexible attachment of the core complex to chromatin. The resulting dynamic tethering furthermore, provides geometric access of L4 catalytic domains to the DNA ends during ligation and of DNA-PKcs for targeted phosphorylation of other NHEJ proteins as well as trans-phosphorylation of DNA-PKcs on the opposing DSB without disrupting the core ligation complex. Overall the results shed light on evolutionary conservation of Ku, X4, and L4 activities, while explaining the observation that Ku80CTR and DNA-PKcs only occur in a subset of higher eukaryotes.


Asunto(s)
Reparación del ADN por Unión de Extremidades/genética , ADN Ligasa (ATP)/metabolismo , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Proteína Quinasa Activada por ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Autoantígeno Ku/metabolismo , Proteínas Nucleares/metabolismo , Western Blotting , Reactivos de Enlaces Cruzados , Roturas del ADN de Doble Cadena , ADN Ligasa (ATP)/química , ADN Ligasa (ATP)/genética , ADN-(Sitio Apurínico o Apirimidínico) Liasa/química , ADN-(Sitio Apurínico o Apirimidínico) Liasa/genética , Proteína Quinasa Activada por ADN/química , Proteína Quinasa Activada por ADN/genética , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Células HeLa , Humanos , Inmunoprecipitación , Autoantígeno Ku/química , Autoantígeno Ku/genética , Modelos Moleculares , Proteínas Nucleares/química , Proteínas Nucleares/genética , Fosforilación , Proteínas de Unión a Poli-ADP-Ribosa , Unión Proteica , Conformación Proteica , Dispersión del Ángulo Pequeño , Difracción de Rayos X
10.
J Biol Chem ; 290(52): 30713-25, 2015 Dec 25.
Artículo en Inglés | MEDLINE | ID: mdl-26559969

RESUMEN

In vertebrate species, the innate immune system down-regulates protein translation in response to viral infection through the action of the double-stranded RNA (dsRNA)-activated protein kinase (PKR). In some teleost species another protein kinase, Z-DNA-dependent protein kinase (PKZ), plays a similar role but instead of dsRNA binding domains, PKZ has Zα domains. These domains recognize the left-handed conformer of dsDNA and dsRNA known as Z-DNA/Z-RNA. Cyprinid herpesvirus 3 infects common and koi carp, which have PKZ, and encodes the ORF112 protein that itself bears a Zα domain, a putative competitive inhibitor of PKZ. Here we present the crystal structure of ORF112-Zα in complex with an 18-bp CpG DNA repeat, at 1.5 Å. We demonstrate that the bound DNA is in the left-handed conformation and identify key interactions for the specificity of ORF112. Localization of ORF112 protein in stress granules induced in Cyprinid herpesvirus 3-infected fish cells suggests a functional behavior similar to that of Zα domains of the interferon-regulated, nucleic acid surveillance proteins ADAR1 and DAI.


Asunto(s)
ADN de Forma Z/metabolismo , Proteína Quinasa Activada por ADN/química , Proteína Quinasa Activada por ADN/metabolismo , Enfermedades de los Peces/virología , Virus ARN/enzimología , Proteínas Virales/química , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Carpas , Secuencia Conservada , ADN de Forma Z/química , ADN de Forma Z/genética , Proteína Quinasa Activada por ADN/genética , Interferones/genética , Interferones/metabolismo , Modelos Moleculares , Conformación de Ácido Nucleico , Poxviridae/química , Poxviridae/enzimología , Poxviridae/genética , Unión Proteica , Estructura Terciaria de Proteína , Virus ARN/química , Virus ARN/genética , ARN Bicatenario/química , ARN Bicatenario/genética , ARN Bicatenario/metabolismo , Proteínas Virales/genética
11.
Nature ; 463(7277): 118-21, 2010 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-20023628

RESUMEN

Broken chromosomes arising from DNA double-strand breaks result from endogenous events such as the production of reactive oxygen species during cellular metabolism, as well as from exogenous sources such as ionizing radiation. Left unrepaired or incorrectly repaired they can lead to genomic changes that may result in cell death or cancer. DNA-dependent protein kinase (DNA-PK), a holoenzyme that comprises the DNA-PK catalytic subunit (DNA-PKcs) and the heterodimer Ku70/Ku80, has a major role in non-homologous end joining-the main pathway in mammals used to repair double-strand breaks. DNA-PKcs is a serine/threonine protein kinase comprising a single polypeptide chain of 4,128 amino acids and belonging to the phosphatidylinositol-3-OH kinase (PI(3)K)-related protein family. DNA-PKcs is involved in the sensing and transmission of DNA damage signals to proteins such as p53, setting off events that lead to cell cycle arrest. It phosphorylates a wide range of substrates in vitro, including Ku70/Ku80, which is translocated along DNA. Here we present the crystal structure of human DNA-PKcs at 6.6 A resolution, in which the overall fold is clearly visible, to our knowledge, for the first time. The many alpha-helical HEAT repeats (helix-turn-helix motifs) facilitate bending and allow the polypeptide chain to fold into a hollow circular structure. The carboxy-terminal kinase domain is located on top of this structure, and a small HEAT repeat domain that probably binds DNA is inside. The structure provides a flexible cradle to promote DNA double-strand-break repair.


Asunto(s)
Proteína Quinasa Activada por ADN/química , Secuencias Hélice-Giro-Hélice , Proteínas Nucleares/química , Antígenos Nucleares/química , Dominio Catalítico , Cristalografía por Rayos X , ADN/metabolismo , Roturas del ADN de Doble Cadena , Proteína Quinasa Activada por ADN/metabolismo , Proteínas de Unión al ADN/química , Células HeLa , Humanos , Autoantígeno Ku , Modelos Moleculares , Proteínas Nucleares/metabolismo , Pliegue de Proteína , Estructura Secundaria de Proteína
12.
Nucleic Acids Res ; 42(18): 11487-501, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25223785

RESUMEN

Non-homologous end-joining (NHEJ) and homologous recombination (HR) are the two prominent pathways responsible for the repair of DNA double-strand breaks (DSBs). NHEJ is not restricted to a cell-cycle stage, whereas HR is active primarily in the S/G2 phases suggesting there are cell cycle-specific mechanisms that play a role in the choice between NHEJ and HR. Here we show NHEJ is attenuated in S phase via modulation of the autophosphorylation status of the NHEJ factor DNA-PKcs at serine 2056 by the pro-HR factor BRCA1. BRCA1 interacts with DNA-PKcs in a cell cycle-regulated manner and this interaction is mediated by the tandem BRCT domain of BRCA1, but surprisingly in a phospho-independent manner. BRCA1 attenuates DNA-PKcs autophosphorylation via directly blocking the ability of DNA-PKcs to autophosphorylate. Subsequently, blocking autophosphorylation of DNA-PKcs at the serine 2056 phosphorylation cluster promotes HR-required DNA end processing and loading of HR factors to DSBs and is a possible mechanism by which BRCA1 promotes HR.


Asunto(s)
Proteína BRCA1/metabolismo , Proteína Quinasa Activada por ADN/metabolismo , Fase S , Proteína BRCA1/química , Línea Celular , Roturas del ADN de Doble Cadena , Proteína Quinasa Activada por ADN/química , Células HeLa , Humanos , Fosforilación , Estructura Terciaria de Proteína , Tolerancia a Radiación , Reparación del ADN por Recombinación , Fase S/genética , Serina/metabolismo
13.
Mol Biol (Mosk) ; 49(1): 67-74, 2015.
Artículo en Ruso | MEDLINE | ID: mdl-25916111

RESUMEN

Apurinic/apyrimidinic (AP) sites are some of the most frequent lesions in genomic DNA. It is widely accepted that, irrespective of their origin, AP sites are further processed by the base excision repair (BER) machinery, being the central intermediate of this process. Under special conditions, proteins, which recognize AP sites, are able to form covalent adducts with DNA. By combination of the cross-linking technique with mass-spectrometry analysis, Ku antigen (Ku)--the central player in nonhomologous end joining (NHEJ), the pathway of double-strand break (DSB) repair--was identified as a protein reactive to AP sites. Moreover, Ku was shown to be a 5'-dRP/AP lyase that acts near DSBs in NHEJ. The recent studies have demonstrated involvement of Ku in the different stages of BER. Here, Ku roles in NHEJ and BER pathways of DNA repair are overviewed.


Asunto(s)
Antígenos Nucleares/genética , Reparación del ADN por Unión de Extremidades/genética , Reparación del ADN/genética , Proteínas de Unión al ADN/genética , Animales , Antígenos Nucleares/química , Ácido Apurínico/química , Ácido Apurínico/genética , Dominio Catalítico/genética , Aductos de ADN/genética , Proteína Quinasa Activada por ADN/química , Proteína Quinasa Activada por ADN/genética , Proteínas de Unión al ADN/química , Autoantígeno Ku , Ratones , Complejos Multiproteicos/química , Complejos Multiproteicos/genética , Polinucleótidos/química , Polinucleótidos/genética , Multimerización de Proteína/genética
14.
J Biol Chem ; 288(10): 7037-46, 2013 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-23322783

RESUMEN

DNA-dependent protein kinase (DNA-PK) plays an essential role in the repair of DNA double-stranded breaks (DSBs) mediated by the nonhomologous end-joining pathway. DNA-PK is a holoenzyme consisting of a DNA-binding (Ku70/Ku80) and catalytic (DNA-PKcs) subunit. DNA-PKcs is a serine/threonine protein kinase that is recruited to DSBs via Ku70/80 and is activated once the kinase is bound to the DSB ends. In this study, two large, distinct fragments of DNA-PKcs, consisting of the N terminus (amino acids 1-2713), termed N-PKcs, and the C terminus (amino acids 2714-4128), termed C-PKcs, were produced to determine the role of each terminal region in regulating the activity of DNA-PKcs. N-PKcs but not C-PKcs interacts with the Ku-DNA complex and is required for the ability of DNA-PKcs to localize to DSBs. C-PKcs has increased basal kinase activity compared with DNA-PKcs, suggesting that the N-terminal region of DNA-PKcs keeps basal activity low. The kinase activity of C-PKcs is not stimulated by Ku70/80 and DNA, further supporting that the N-terminal region is required for binding to the Ku-DNA complex and full activation of kinase activity. Collectively, the results show the N-terminal region mediates the interaction between DNA-PKcs and the Ku-DNA complex and is required for its DSB-induced enzymatic activity.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN , Proteína Quinasa Activada por ADN/metabolismo , Proteínas Nucleares/metabolismo , Animales , Antígenos Nucleares/química , Antígenos Nucleares/genética , Antígenos Nucleares/metabolismo , Western Blotting , Células CHO , Cricetinae , Cricetulus , ADN/genética , ADN/metabolismo , Proteína Quinasa Activada por ADN/química , Proteína Quinasa Activada por ADN/genética , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Activación Enzimática , Células HeLa , Humanos , Autoantígeno Ku , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/genética , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Unión Proteica , Multimerización de Proteína , Células Sf9
15.
Breast Cancer Res Treat ; 146(2): 309-20, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24972688

RESUMEN

DNA-dependent protein kinase catalytic subunit (DNA-PKcs), a serine threonine kinase belonging to the PIKK family (phosphoinositide 3-kinase-like-family of protein kinase), is a critical component of the non-homologous end-joining pathway required for the repair of DNA double-strand breaks. DNA-PKcs may be involved in breast cancer pathogenesis. We evaluated clinicopathological significance of DNA-PKcs protein expression in 1,161 tumours and DNA-PKcs mRNA expression in 1,950 tumours. We correlated DNA-PKcs to markers of aggressive phenotypes, DNA repair, apoptosis, cell cycle regulation and survival. Low DNA-PKcs protein expression was associated with higher tumour grade, higher mitotic index, tumour de-differentiation and tumour type (ps < 0.05). The absence of BRCA1, low XRCC1, low SMUG1, low APE1 and low Polß was also more likely in low DNA-PKcs expressing tumours (ps < 0.05). Low DNA-PKcs protein expression was significantly associated with worse breast cancer-specific survival (BCSS) in univariate and multivariate analysis (ps < 0.01). At the mRNA level, similarly, low DNA-PKcs was associated with poor BCSS. In patients with ER-positive tumours who received endocrine therapy, low DNA-PKcs (protein and mRNA) was associated with poor survival. In ER-negative patients, low DNA-PKcs mRNA remains significantly associated with adverse outcome. Our study suggests that low DNA-PKcs expression may have prognostic and predictive significance in breast cancers.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/mortalidad , Dominio Catalítico/genética , Proteína Quinasa Activada por ADN/genética , Expresión Génica , Dominios y Motivos de Interacción de Proteínas/genética , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/diagnóstico , Línea Celular Tumoral , Proteína Quinasa Activada por ADN/química , Proteína Quinasa Activada por ADN/metabolismo , Femenino , Humanos , Persona de Mediana Edad , Clasificación del Tumor , Estadificación de Neoplasias , Pronóstico , Carga Tumoral
16.
Nucleic Acids Res ; 40(7): 2964-73, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22167471

RESUMEN

The DNA-dependent protein kinase (DNA-PK) was identified as an activity and as its three component polypeptides 25 and 15 years ago, respectively. It has been exhaustively characterized as being absolutely dependent on free double stranded DNA ends (to which it is directed by its regulatory subunit, Ku) for its activation as a robust nuclear serine/threonine protein kinase. Here, we report the unexpected finding of robust DNA-PKcs activation by N-terminal constraint, independent of either DNA or its regulatory subunit Ku. These data suggest that an N-terminal conformational change (likely induced by DNA binding) induces enzymatic activation.


Asunto(s)
Proteína Quinasa Activada por ADN/química , Proteína Quinasa Activada por ADN/metabolismo , ADN/metabolismo , Animales , Antígenos Nucleares/fisiología , Células CHO , Dominio Catalítico , Cricetinae , Cricetulus , Proteínas de Unión al ADN/fisiología , Activación Enzimática , Humanos , Autoantígeno Ku , Fosforilación , Cloruro de Sodio/química , Proteína p53 Supresora de Tumor/metabolismo
17.
Nucleic Acids Res ; 40(9): 4168-77, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22223246

RESUMEN

The DNA-dependent protein kinase (DNA-PK) and Poly(ADP-ribose) polymerase-1 (PARP1) are critical enzymes that reduce genomic damage caused by DNA lesions. They are both activated by DNA strand breaks generated by physiological and environmental factors, and they have been shown to interact. Here, we report in vivo evidence that DNA-PK and PARP1 are equally necessary for rapid repair. We purified a DNA-PK/PARP1 complex loaded on DNA and performed electron microscopy and single particle analysis on its tetrameric and dimer-of-tetramers forms. By comparison with the DNA-PK holoenzyme and fitting crystallographic structures, we see that the PARP1 density is in close contact with the Ku subunit. Crucially, PARP1 binding elicits substantial conformational changes in the DNA-PK synaptic dimer assembly. Taken together, our data support a functional, in-pathway role for DNA-PK and PARP1 in double-strand break (DSB) repair. We also propose a NHEJ model where protein-protein interactions alter substantially the architecture of DNA-PK dimers at DSBs, to trigger subsequent interactions or enzymatic reactions.


Asunto(s)
Reparación del ADN , Proteína Quinasa Activada por ADN/ultraestructura , Proteínas Nucleares/ultraestructura , Poli(ADP-Ribosa) Polimerasas/ultraestructura , Animales , Células Cultivadas , Roturas del ADN de Doble Cadena , Proteína Quinasa Activada por ADN/química , Proteína Quinasa Activada por ADN/fisiología , Proteínas de Unión al ADN/fisiología , Dimerización , Ratones , Proteínas Nucleares/química , Proteínas Nucleares/fisiología , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/química , Poli(ADP-Ribosa) Polimerasas/fisiología
18.
Prog Biophys Mol Biol ; 186: 1-13, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38036101

RESUMEN

The proteins and protein assemblies involved in DNA repair have been the focus of a multitude of structural studies for the past few decades. Historically, the structures of these protein complexes have been resolved by X-ray crystallography. However, more recently with the advancements in cryo-electron microscopy (cryo-EM) ranging from optimising the methodology for sample preparation to the development of improved electron detectors, the focus has shifted from X-ray crystallography to cryo-EM. This methodological transition has allowed for the structural determination of larger, more complex protein assemblies involved in DNA repair pathways and has subsequently led to a deeper understanding of the mechanisms utilised by these fascinating molecular machines. Here, we review some of the key structural advancements that have been gained in the study of non-homologous end joining (NHEJ) by the use of cryo-EM, with a focus on assemblies composed of DNA-PKcs and Ku70/80 (Ku) and the various methodologies utilised to obtain these structures.


Asunto(s)
Reparación del ADN por Unión de Extremidades , Reparación del ADN , Microscopía por Crioelectrón , Proteína Quinasa Activada por ADN/química , Cristalografía por Rayos X , ADN/metabolismo
19.
J Biol Chem ; 287(12): 9230-9, 2012 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-22298787

RESUMEN

DNA-dependent protein kinase (DNA-PK) becomes activated in response to DNA double strand breaks, initiating repair by the non-homologous end joining pathway. DNA·PK complexes with the regulatory subunit SAPSR1 (R1) of protein phosphatase-6 (PP6). Knockdown of either R1 or PP6c prevents DNA-PK activation in response to ionizing radiation-induced DNA damage and radiosensitizes glioblastoma cells. Here, we demonstrate that R1 is necessary for and bridges the interaction between DNA-PK and PP6c. Using R1 deletion mutants, DNA-PK binding was mapped to two distinct regions of R1 spanning residues 1-326 and 522-700. Either region expressed alone was sufficient to bind DNA-PK, but only deletion of residues 1-326, not 522-700, eliminated interaction of R1 with DNA-PK. We assign 1-326 as the dominant domain and 522-700 as the supporting region. These results demonstrate that R1 acts as a bidentate anchor to DNA-PK and recruits PP6c. Targeting the dominant interface with small molecule or peptidomimetic inhibitors could specifically prevent activation of DNA-PK and thereby sensitize cells to ionizing radiation and other genotoxic agents.


Asunto(s)
Proteína Quinasa Activada por ADN/metabolismo , Fosfoproteínas Fosfatasas/metabolismo , Secuencias de Aminoácidos , Línea Celular , Proteína Quinasa Activada por ADN/química , Proteína Quinasa Activada por ADN/genética , Humanos , Fosfoproteínas Fosfatasas/química , Fosfoproteínas Fosfatasas/genética , Unión Proteica
20.
EMBO J ; 28(23): 3667-80, 2009 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-19851285

RESUMEN

Human tyrosyl-DNA phosphodiesterase (TDP1) hydrolyzes the phosphodiester bond at a DNA 3' end linked to a tyrosyl moiety. This type of linkage is found at stalled topoisomerase I (Top1)-DNA covalent complexes, and TDP1 has been implicated in the repair of such complexes. Here we show that Top1-associated DNA double-stranded breaks (DSBs) induce the phosphorylation of TDP1 at S81. This phosphorylation is mediated by the protein kinases: ataxia-telangiectasia-mutated (ATM) and DNA-dependent protein kinase (DNA-PK). Phosphorylated TDP1 forms nuclear foci that co-localize with those of phosphorylated histone H2AX (gammaH2AX). Both Top1-induced replication- and transcription-mediated DNA damages induce TDP1 phosphorylation. Furthermore, we show that S81 phosphorylation stabilizes TDP1, induces the formation of XRCC1 (X-ray cross-complementing group 1)-TDP1 complexes and enhances the mobilization of TDP1 to DNA damage sites. Finally, we provide evidence that TDP1-S81 phosphorylation promotes cell survival and DNA repair in response to CPT-induced DSBs. Together; our findings provide a new mechanism for TDP1 post-translational regulation by ATM and DNA-PK.


Asunto(s)
Proteínas de Ciclo Celular/química , Reparación del ADN , Proteína Quinasa Activada por ADN/química , Proteínas de Unión al ADN/química , Hidrolasas Diéster Fosfóricas/metabolismo , Proteínas Serina-Treonina Quinasas/química , Proteínas Supresoras de Tumor/química , Ataxia Telangiectasia/enzimología , Ataxia Telangiectasia/genética , Proteínas de la Ataxia Telangiectasia Mutada , Carnitina O-Palmitoiltransferasa/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/fisiología , Supervivencia Celular/genética , Roturas del ADN de Doble Cadena , Reparación del ADN/genética , Proteína Quinasa Activada por ADN/metabolismo , Proteína Quinasa Activada por ADN/fisiología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/fisiología , Humanos , Hidrolasas Diéster Fosfóricas/química , Hidrolasas Diéster Fosfóricas/fisiología , Fosforilación/genética , Procesamiento Proteico-Postraduccional , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/fisiología , Serina/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas Supresoras de Tumor/fisiología , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA