Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 81(4): 784-800.e8, 2021 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-33412112

RESUMEN

DNA replication forks use multiple mechanisms to deal with replication stress, but how the choice of mechanisms is made is still poorly understood. Here, we show that CARM1 associates with replication forks and reduces fork speed independently of its methyltransferase activity. The speeding of replication forks in CARM1-deficient cells requires RECQ1, which resolves reversed forks, and RAD18, which promotes translesion synthesis. Loss of CARM1 reduces fork reversal and increases single-stranded DNA (ssDNA) gaps but allows cells to tolerate higher replication stress. Mechanistically, CARM1 interacts with PARP1 and promotes PARylation at replication forks. In vitro, CARM1 stimulates PARP1 activity by enhancing its DNA binding and acts jointly with HPF1 to activate PARP1. Thus, by stimulating PARP1, CARM1 slows replication forks and promotes the use of fork reversal in the stress response, revealing that CARM1 and PARP1 function as a regulatory module at forks to control fork speed and the choice of stress response mechanisms.


Asunto(s)
Roturas del ADN de Cadena Simple , Replicación del ADN , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Células HEK293 , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/genética , Proteína-Arginina N-Metiltransferasas/genética , RecQ Helicasas/genética , RecQ Helicasas/metabolismo
2.
Genes Dev ; 33(1-2): 75-89, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30567999

RESUMEN

Numerous DNA repair and signaling proteins function at DNA damage sites to protect the genome. Here, we show that fusion of the promiscuous biotin ligase BirAR118G with RAD18 leads to localized protein biotinylation at DNA damage sites, allowing identification of ZPET (zinc finger protein proximal to RAD eighteen)/ZNF280C as a potential DNA damage response (DDR) protein. ZPET binds ssDNA and localizes to DNA double-strand breaks (DSBs) and stalled replication forks. In vitro, ZPET inhibits MRE11 binding to ssDNA. In cells, ZPET delays MRE11 binding to chromatin after DSB formation and slows DNA end resection through binding ssDNA. ZPET hinders resection independently of 53BP1 and HELB. Cells lacking ZPET displayed enhanced homologous recombination (HR), accelerated replication forks under stress, and increased resistance to DSBs and PARP inhibition. These results not only reveal ZPET as an HR repressor but also suggest that localized protein biotinylation at DNA damage sites is a useful strategy to identify DDR proteins.


Asunto(s)
Biotinilación/métodos , Daño del ADN , Reparación del ADN/genética , Proteínas de Unión al ADN/metabolismo , Recombinación Homóloga/genética , Factores de Transcripción/metabolismo , Ligasas de Carbono-Nitrógeno/genética , Línea Celular , Roturas del ADN de Doble Cadena , ADN de Cadena Simple/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Escherichia coli/genética , Técnicas de Silenciamiento del Gen , Humanos , Proteína Homóloga de MRE11/metabolismo , Unión Proteica , Transporte de Proteínas/genética , Proteínas Represoras/genética , Factores de Transcripción/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
3.
Mol Cell ; 70(6): 995-1007.e11, 2018 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-29910111

RESUMEN

Phosphotyrosine (pTyr) signaling has evolved into a key cell-to-cell communication system. Activated receptor tyrosine kinases (RTKs) initiate several pTyr-dependent signaling networks by creating the docking sites required for the assembly of protein complexes. However, the mechanisms leading to network disassembly and its consequence on signal transduction remain essentially unknown. We show that activated RTKs terminate downstream signaling via the direct phosphorylation of an evolutionarily conserved Tyr present in most SRC homology (SH) 3 domains, which are often part of key hub proteins for RTK-dependent signaling. We demonstrate that the direct EPHA4 RTK phosphorylation of adaptor protein NCK SH3s at these sites results in the collapse of signaling networks and abrogates their function. We also reveal that this negative regulation mechanism is shared by other RTKs. Our findings uncover a conserved mechanism through which RTKs rapidly and reversibly terminate downstream signaling while remaining in a catalytically active state on the plasma membrane.


Asunto(s)
Proteínas Tirosina Quinasas Receptoras/fisiología , Receptor EphA4/metabolismo , Dominios Homologos src/fisiología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Secuencia de Aminoácidos , Animales , Comunicación Celular , Drosophila/metabolismo , Células HEK293 , Células HeLa , Humanos , Ligandos , Proteínas Oncogénicas/metabolismo , Fosforilación , Fosfotirosina/metabolismo , Unión Proteica , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal/fisiología , Tirosina/metabolismo
4.
Nucleic Acids Res ; 52(7): 4002-4020, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38321934

RESUMEN

Poly(ADP-ribosylation) (PARylation) is a post-translational modification mediated by a subset of ADP-ribosyl transferases (ARTs). Although PARylation-inhibition based therapies are considered as an avenue to combat debilitating diseases such as cancer and myopathies, the role of this modification in physiological processes such as cell differentiation remains unclear. Here, we show that Tankyrase1 (TNKS1), a PARylating ART, plays a major role in myogenesis, a vital process known to drive muscle fiber formation and regeneration. Although all bona fide PARPs are expressed in muscle cells, experiments using siRNA-mediated knockdown or pharmacological inhibition show that TNKS1 is the enzyme responsible of catalyzing PARylation during myogenesis. Via this activity, TNKS1 controls the turnover of mRNAs encoding myogenic regulatory factors such as nucleophosmin (NPM) and myogenin. TNKS1 mediates these effects by targeting RNA-binding proteins such as Human Antigen R (HuR). HuR harbors a conserved TNKS-binding motif (TBM), the mutation of which not only prevents the association of HuR with TNKS1 and its PARylation, but also precludes HuR from regulating the turnover of NPM and myogenin mRNAs as well as from promoting myogenesis. Therefore, our data uncover a new role for TNKS1 as a key modulator of RBP-mediated post-transcriptional events required for vital processes such as myogenesis.


Asunto(s)
Desarrollo de Músculos , Fibras Musculares Esqueléticas , Miogenina , ARN Mensajero , Tanquirasas , Tanquirasas/metabolismo , Tanquirasas/genética , Humanos , ARN Mensajero/metabolismo , ARN Mensajero/genética , Desarrollo de Músculos/genética , Animales , Fibras Musculares Esqueléticas/metabolismo , Ratones , Miogenina/genética , Miogenina/metabolismo , Nucleofosmina , Proteína 1 Similar a ELAV/metabolismo , Proteína 1 Similar a ELAV/genética , Estabilidad del ARN/genética , Poli ADP Ribosilación/genética , Línea Celular , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Diferenciación Celular/genética , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Células HEK293
5.
FASEB J ; 38(6): e23556, 2024 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-38498348

RESUMEN

PARP-1 over-activation results in cell death via excessive PAR generation in different cell types, including neurons following brain ischemia. Glycolysis, mitochondrial function, and redox balance are key cellular processes altered in brain ischemia. Studies show that PAR generated after PARP-1 over-activation can bind hexokinase-1 (HK-1) and result in glycolytic defects and subsequent mitochondrial dysfunction. HK-1 is the neuronal hexokinase and catalyzes the first reaction of glycolysis, converting glucose to glucose-6-phosphate (G6P), a common substrate for glycolysis, and the pentose phosphate pathway (PPP). PPP is critical in maintaining NADPH and GSH levels via G6P dehydrogenase activity. Therefore, defects in HK-1 will not only decrease cellular bioenergetics but will also cause redox imbalance due to the depletion of GSH. In brain ischemia, whether PAR-mediated inhibition of HK-1 results in bioenergetics defects and redox imbalance is not known. We used oxygen-glucose deprivation (OGD) in mouse cortical neurons to mimic brain ischemia in neuronal cultures and observed that PARP-1 activation via PAR formation alters glycolysis, mitochondrial function, and redox homeostasis in neurons. We used pharmacological inhibition of PARP-1 and adenoviral-mediated overexpression of wild-type HK-1 (wtHK-1) and PAR-binding mutant HK-1 (pbmHK-1). Our data show that PAR inhibition or overexpression of HK-1 significantly improves glycolysis, mitochondrial function, redox homeostasis, and cell survival in mouse cortical neurons exposed to OGD. These results suggest that PAR binding and inhibition of HK-1 during OGD drive bioenergetic defects in neurons due to inhibition of glycolysis and impairment of mitochondrial function.


Asunto(s)
Isquemia Encefálica , Oxígeno , Ratones , Animales , Oxígeno/metabolismo , Poli Adenosina Difosfato Ribosa/metabolismo , Hexoquinasa/genética , Hexoquinasa/metabolismo , Inhibidores de Poli(ADP-Ribosa) Polimerasas/metabolismo , Glucosa/metabolismo , Isquemia Encefálica/metabolismo , Glucólisis , Neuronas/metabolismo , Oxidación-Reducción
6.
Nucleic Acids Res ; 51(20): 11056-11079, 2023 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-37823600

RESUMEN

Zinc finger (ZNF) motifs are some of the most frequently occurring domains in the human genome. It was only recently that ZNF proteins emerged as key regulators of genome integrity in mammalian cells. In this study, we report a new role for the Krüppel-type ZNF-containing protein ZNF432 as a novel poly(ADP-ribose) (PAR) reader that regulates the DNA damage response. We show that ZNF432 is recruited to DNA lesions via DNA- and PAR-dependent mechanisms. Remarkably, ZNF432 stimulates PARP-1 activity in vitro and in cellulo. Knockdown of ZNF432 inhibits phospho-DNA-PKcs and increases RAD51 foci formation following irradiation. Moreover, purified ZNF432 preferentially binds single-stranded DNA and impairs EXO1-mediated DNA resection. Consequently, the loss of ZNF432 in a cellular system leads to resistance to PARP inhibitors while its overexpression results in sensitivity. Taken together, our results support the emerging concept that ZNF-containing proteins can modulate PARylation, which can be embodied by the pivotal role of ZNF432 to finely balance the outcome of PARPi response by regulating homologous recombination.


Asunto(s)
Poli ADP Ribosilación , Poli Adenosina Difosfato Ribosa , Humanos , ADN/genética , ADN/metabolismo , Daño del ADN , Reparación del ADN , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Poli Adenosina Difosfato Ribosa/metabolismo
7.
Proc Natl Acad Sci U S A ; 118(11)2021 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-33692124

RESUMEN

Zinc finger (ZnF) proteins represent one of the largest families of human proteins, although most remain uncharacterized. Given that numerous ZnF proteins are able to interact with DNA and poly(ADP ribose), there is growing interest in understanding their mechanism of action in the maintenance of genome integrity. We now report that the ZnF protein E4F transcription factor 1 (E4F1) is an actor in DNA repair. Indeed, E4F1 is rapidly recruited, in a poly(ADP ribose) polymerase (PARP)-dependent manner, to DNA breaks and promotes ATR/CHK1 signaling, DNA-end resection, and subsequent homologous recombination. Moreover, we identify E4F1 as a regulator of the ATP-dependent chromatin remodeling SWI/SNF complex in DNA repair. E4F1 binds to the catalytic subunit BRG1/SMARCA4 and together with PARP-1 mediates its recruitment to DNA lesions. We also report that a proportion of human breast cancers show amplification and overexpression of E4F1 or BRG1 that are mutually exclusive with BRCA1/2 alterations. Together, these results reveal a function of E4F1 in the DNA damage response that orchestrates proper signaling and repair of double-strand breaks and document a molecular mechanism for its essential role in maintaining genome integrity and cell survival.


Asunto(s)
Roturas del ADN de Doble Cadena , ADN Helicasas/metabolismo , Reparación del ADN , Proteínas Nucleares/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Neoplasias de la Mama/genética , Proliferación Celular , Supervivencia Celular , Ensamble y Desensamble de Cromatina , Daño del ADN , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Recombinación Homóloga , Humanos , Unión Proteica , Proteínas Represoras/deficiencia , Transducción de Señal , Ubiquitina-Proteína Ligasas/deficiencia
8.
PLoS Genet ; 16(11): e1009183, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33137104

RESUMEN

Loss of von Hippel-Lindau protein pVHL function promotes VHL diseases, including sporadic and inherited clear cell Renal Cell Carcinoma (ccRCC). Mechanisms controlling pVHL function and regulation, including folding and stability, remain elusive. Here, we have identified the conserved cochaperone prefoldin complex in a screen for pVHL interactors. The prefoldin complex delivers non-native proteins to the chaperonin T-complex-protein-1-ring (TRiC) or Cytosolic Chaperonin containing TCP-1 (CCT) to assist folding of newly synthesized polypeptides. The pVHL-prefoldin interaction was confirmed in human cells and prefoldin knock-down reduced pVHL expression levels. Furthermore, when pVHL was expressed in Schizosaccharomyces pombe, all prefoldin mutants promoted its aggregation. We mapped the interaction of prefoldin with pVHL at the exon2-exon3 junction encoded region. Low levels of the PFDN3 prefoldin subunit were associated with poor survival in ccRCC patients harboring VHL mutations. Our results link the prefoldin complex with pVHL folding and this may impact VHL diseases progression.


Asunto(s)
Proteínas del Citoesqueleto/metabolismo , Neoplasias Renales/genética , Chaperonas Moleculares/metabolismo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/metabolismo , Chaperonina con TCP-1 , Técnicas de Silenciamiento del Gen , Células HEK293 , Células HeLa , Humanos , Estimación de Kaplan-Meier , Neoplasias Renales/mortalidad , Neoplasias Renales/patología , Chaperonas Moleculares/genética , Mutación , Unión Proteica/genética , Pliegue de Proteína , Proteolisis , Schizosaccharomyces , Proteínas de Schizosaccharomyces pombe/genética , Proteínas de Schizosaccharomyces pombe/metabolismo , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética
9.
J Virol ; 95(3)2021 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-33158944

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and other SARS-related CoVs encode 3 tandem macrodomains within nonstructural protein 3 (nsp3). The first macrodomain, Mac1, is conserved throughout CoVs and binds to and hydrolyzes mono-ADP-ribose (MAR) from target proteins. Mac1 likely counters host-mediated antiviral ADP-ribosylation, a posttranslational modification that is part of the host response to viral infections. Mac1 is essential for pathogenesis in multiple animal models of CoV infection, implicating it as a virulence factor and potential therapeutic target. Here, we report the crystal structure of SARS-CoV-2 Mac1 in complex with ADP-ribose. SARS-CoV-2, SARS-CoV, and Middle East respiratory syndrome coronavirus (MERS-CoV) Mac1 domains exhibit similar structural folds, and all 3 proteins bound to ADP-ribose with affinities in the low micromolar range. Importantly, using ADP-ribose-detecting binding reagents in both a gel-based assay and novel enzyme-linked immunosorbent assays (ELISAs), we demonstrated de-MARylating activity for all 3 CoV Mac1 proteins, with the SARS-CoV-2 Mac1 protein leading to a more rapid loss of substrate than the others. In addition, none of these enzymes could hydrolyze poly-ADP-ribose. We conclude that the SARS-CoV-2 and other CoV Mac1 proteins are MAR-hydrolases with similar functions, indicating that compounds targeting CoV Mac1 proteins may have broad anti-CoV activity.IMPORTANCE SARS-CoV-2 has recently emerged into the human population and has led to a worldwide pandemic of COVID-19 that has caused more than 1.2 million deaths worldwide. With no currently approved treatments, novel therapeutic strategies are desperately needed. All coronaviruses encode a highly conserved macrodomain (Mac1) that binds to and removes ADP-ribose adducts from proteins in a dynamic posttranslational process that is increasingly being recognized as an important factor that regulates viral infection. The macrodomain is essential for CoV pathogenesis and may be a novel therapeutic target. Thus, understanding its biochemistry and enzyme activity are critical first steps for these efforts. Here, we report the crystal structure of SARS-CoV-2 Mac1 in complex with ADP-ribose and describe its ADP-ribose binding and hydrolysis activities in direct comparison to those of SARS-CoV and MERS-CoV Mac1 proteins. These results are an important first step for the design and testing of potential therapies targeting this unique protein domain.


Asunto(s)
N-Glicosil Hidrolasas/metabolismo , SARS-CoV-2/enzimología , Proteínas no Estructurales Virales/metabolismo , Adenosina Difosfato Ribosa/química , Adenosina Difosfato Ribosa/metabolismo , Secuencia de Aminoácidos , Coronavirus/química , Coronavirus/enzimología , Coronavirus/metabolismo , Cristalografía por Rayos X , Humanos , Hidrólisis , Cinética , N-Glicosil Hidrolasas/química , Unión Proteica , Dominios Proteicos , SARS-CoV-2/química , SARS-CoV-2/metabolismo , Proteínas no Estructurales Virales/química
11.
Cogn Behav Ther ; 49(2): 97-112, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31140370

RESUMEN

Cognitive theory of obsessive-compulsive disorder (OCD) proposes that maladaptive beliefs play a pivotal role in the development and maintenance of symptoms. Clinical reports as well as recent psychometric and experimental investigations suggest that control-related beliefs in OCD may benefit from expansion to include aspects of losing control. However, currently available measures either focus on other facets of control (e.g., sense of control) or do not put emphasis on beliefs about losing control (e.g., beliefs about control over thoughts). The current study aimed to develop and validate the Beliefs About Losing Control Inventory (BALCI), a self-report measure of negative beliefs about losing control, in a sample of undergraduate participants (N= 488). An exploratory factor analysis revealed that the BALCI's 21 items capture negative beliefs about losing control over one's thoughts, behaviour, and emotions (Factor 1), beliefs about the importance of staying in control (Factor 2), and beliefs about losing control over one's body/bodily functions (Factor 3). The BALCI was also found to have good convergent and divergent validity and to be associated with elevated OCD symptoms above and beyond previously identified obsessive beliefs. Theoretical implications and recommendations for the field of cognitive-behaviour therapy are discussed.


Asunto(s)
Cultura , Trastorno Obsesivo Compulsivo/psicología , Autocontrol , Encuestas y Cuestionarios/estadística & datos numéricos , Adolescente , Adulto , Análisis Factorial , Femenino , Humanos , Masculino , Persona de Mediana Edad , Autoinforme , Adulto Joven
12.
Drug Metab Dispos ; 47(9): 949-953, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31266751

RESUMEN

Sulfotransferase 4A1 (SULT4A1), a member of cytosolic sulfotransferases (SULT), is exclusively expressed in neurons with no known function. Severe phenotype and early postnatal death in SULT4A1 knockout mice revealed that SULT4A1 is an essential neuronal protein. Localization of SULT4A1 in different cytosolic compartments, including mitochondria, suggests multiple roles for this protein. We observed that knockdown of SULT4A1 results in the accumulation of reactive oxygen species in primary cortical neurons, suggesting a potential role of SULT4A1 in regulating redox homeostasis. Expression of SULT4A1 in the human neuroblastoma SH-SY5Y cells revealed a defused but nonuniform staining pattern in the cytoplasm, with increased density around mitochondria. Subcellular fractionation of SULT4A1 expressing SH-SY5Y cells confirms the presence of SULT4A1 in mitochondrial fractions. SULT4A1 expressing cells display significant protection against H2O2-mediated defects in mitochondrial function and loss of mitochondrial membrane potential. Expression of SULT4A1 in SH-SY5Y cells also protects against H2O2-induced cell death. These data indicate that SULT4A1 protects mitochondria against oxidative damage and may serve as a potential pharmacological target in neural diseases involving mitochondrial dysfunction and oxidative stress. SIGNIFICANCE STATEMENT: Studies on SULT4A1 knockout mice suggest that SULT4A1 plays a vital role in neuronal function and survival via yet undefined mechanisms. Our data demonstrate that depletion of SULT4A1 induces oxidative stress in neurons and expression of SULT4A1 in SH-SY5Y cells protects against oxidative-stress-induced mitochondrial dysfunction and cell death. These results suggest that SULT4A1 may have a crucial protective function against mitochondrial dysfunction and oxidative stress, and may serve a potential therapeutic target in different neurological diseases involving mitochondrial dysfunction and oxidative stress.


Asunto(s)
Mitocondrias/patología , Neuronas/patología , Sulfotransferasas/metabolismo , Animales , Apoptosis , Línea Celular Tumoral , Corteza Cerebral/citología , Clonación Molecular , Humanos , Potencial de la Membrana Mitocondrial/fisiología , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Neuronas/citología , Estrés Oxidativo , Cultivo Primario de Células , Especies Reactivas de Oxígeno/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sulfotransferasas/genética
13.
J Proteome Res ; 17(7): 2542-2551, 2018 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-29812941

RESUMEN

Despite significant advances in the development of mass spectrometry-based methods for the identification of protein ADP-ribosylation, current protocols suffer from several drawbacks that preclude their widespread applicability. Given the intrinsic heterogeneous nature of poly(ADP-ribose), a number of strategies have been developed to generate simple derivatives for effective interrogation of protein databases and site-specific localization of the modified residues. Currently, the generation of spectral signatures indicative of ADP-ribosylation rely on chemical or enzymatic conversion of the modification to a single mass increment. Still, limitations arise from the lability of the poly(ADP-ribose) remnant during tandem mass spectrometry, the varying susceptibilities of different ADP-ribose-protein bonds to chemical hydrolysis, or the context dependence of enzyme-catalyzed reactions. Here, we present a chemical-based derivatization method applicable to the confident identification of site-specific ADP-ribosylation by conventional mass spectrometry on any targeted amino acid residue. Using PARP-1 as a model protein, we report that treatment of ADP-ribosylated peptides with hydrofluoric acid generates a specific +132 Da mass signature that corresponds to the decomposition of mono- and poly(ADP-ribosylated) peptides into ribose adducts as a consequence of the cleavage of the phosphorus-oxygen bonds.


Asunto(s)
ADP-Ribosilación , Ácido Fluorhídrico/química , Poli(ADP-Ribosa) Polimerasa-1/química , Adenosina Difosfato Ribosa/metabolismo , Cromatografía Liquida , Espectrometría de Masas en Tándem
14.
J Clin Psychol ; 74(10): 1791-1807, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29696644

RESUMEN

OBJECTIVES: A shorter version of the Obsessive Beliefs Questionnaire (OBQ-44) is needed to promote the use of this measure in research and increase our understanding of cognitive phenomena maintaining obsessive-compulsive disorder (OCD). Additionally, an abbreviated version of the OBQ-44 would encourage frequent monitoring of dysfunctional beliefs in intensive care settings. This study aimed to validate a nine-item version of the questionnaire (OBQ-9). METHOD: Participants seeking intensive/residential treatment for OCD (N = 311) completed relevant measures on a weekly basis and at admission and discharge. RESULTS: A confirmatory factor analysis revealed that the OBQ-9's factor structure replicated the three-factor solution of the OBQ-44. The OBQ-9 demonstrated good psychometric properties and convergent validity and was sensitive to treatment effects. Finally, the OBQ-9 subscales predicted specific OCD dimensions over and above depressive symptoms. CONCLUSION: The OBQ-9 appears to be a psychometrically sound tool for routine outcome monitoring of dysfunctional beliefs in hospital-based settings.


Asunto(s)
Trastorno Obsesivo Compulsivo/diagnóstico , Escalas de Valoración Psiquiátrica/normas , Psicometría/normas , Pensamiento/fisiología , Adolescente , Adulto , Análisis Factorial , Femenino , Humanos , Masculino , Trastorno Obsesivo Compulsivo/fisiopatología , Reproducibilidad de los Resultados , Tratamiento Domiciliario , Adulto Joven
15.
Proc Natl Acad Sci U S A ; 111(28): 10209-14, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24987120

RESUMEN

Excessive poly(ADP-ribose) (PAR) polymerase-1 (PARP-1) activation kills cells via a cell-death process designated "parthanatos" in which PAR induces the mitochondrial release and nuclear translocation of apoptosis-inducing factor to initiate chromatinolysis and cell death. Accompanying the formation of PAR are the reduction of cellular NAD(+) and energetic collapse, which have been thought to be caused by the consumption of cellular NAD(+) by PARP-1. Here we show that the bioenergetic collapse following PARP-1 activation is not dependent on NAD(+) depletion. Instead PARP-1 activation initiates glycolytic defects via PAR-dependent inhibition of hexokinase, which precedes the NAD(+) depletion in N-methyl-N-nitroso-N-nitroguanidine (MNNG)-treated cortical neurons. Mitochondrial defects are observed shortly after PARP-1 activation and are mediated largely through defective glycolysis, because supplementation of the mitochondrial substrates pyruvate and glutamine reverse the PARP-1-mediated mitochondrial dysfunction. Depleting neurons of NAD(+) with FK866, a highly specific noncompetitive inhibitor of nicotinamide phosphoribosyltransferase, does not alter glycolysis or mitochondrial function. Hexokinase, the first regulatory enzyme to initiate glycolysis by converting glucose to glucose-6-phosphate, contains a strong PAR-binding motif. PAR binds to hexokinase and inhibits hexokinase activity in MNNG-treated cortical neurons. Preventing PAR formation with PAR glycohydrolase prevents the PAR-dependent inhibition of hexokinase. These results indicate that bioenergetic collapse induced by overactivation of PARP-1 is caused by PAR-dependent inhibition of glycolysis through inhibition of hexokinase.


Asunto(s)
Corteza Cerebral/enzimología , Glucólisis/fisiología , Mitocondrias/enzimología , Proteínas del Tejido Nervioso/metabolismo , Neuronas/enzimología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Acrilamidas/farmacología , Animales , Células Cultivadas , Corteza Cerebral/citología , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Glucosa/metabolismo , Glucosa-6-Fosfato/metabolismo , Glucólisis/efectos de los fármacos , Hexoquinasa/metabolismo , Metilnitronitrosoguanidina/farmacología , Ratones , NAD/metabolismo , Neuronas/citología , Piperidinas/farmacología , Poli(ADP-Ribosa) Polimerasa-1
16.
J Cell Sci ; 125(Pt 19): 4555-66, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22767504

RESUMEN

Poly(ADP-ribose) (pADPr) is a heterogenic molecule synthesised from NAD by poly(ADP-ribose) polymerases (PARPs). Many cellular functions from genome integrity surveillance, cell cycle progression and DNA repair to apoptosis are affected by pADPr through its network of associated proteins. Using quantitative proteomics, we established a temporal map of pADPr-associated complexes upon genotoxic stress. Results suggested a strong pADPr association to many proteins involved in stress granule formation, notably the ras-GAP SH3-binding protein G3BP, as well as in the later phases of alkylation-stress-induced responses. Further investigation with dynamic imaging clearly demonstrated a pADPr-dependent initiation of stress granule assembly originating from the nucleus. The co-transfection of G3BP with poly(ADP-ribose) glycohydrolase (PARG) indicates that pADPr is involved in modulating the nuclear translocation of G3BP. Moreover, a peptide pADPr blot assay of G3BP revealed that pADPr binds to the glycine-arginine-rich domain of G3BP. Thereafter, we established a comprehensive G3BP interactome in the presence of pADPr. Our findings establish a novel function for pADPr in the formation of G3BP-induced stress granules upon genotoxic stress.


Asunto(s)
Proteínas Portadoras/metabolismo , Gránulos Citoplasmáticos/metabolismo , Daño del ADN , ADN/metabolismo , Imagenología Tridimensional/métodos , Poli Adenosina Difosfato Ribosa/metabolismo , Proteómica/métodos , Alquilación/efectos de los fármacos , Secuencia de Aminoácidos , Western Blotting , Proteínas Portadoras/química , Análisis por Conglomerados , Gránulos Citoplasmáticos/efectos de los fármacos , ADN Helicasas , Ácidos Grasos Insaturados/farmacología , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Marcaje Isotópico , Metilnitronitrosoguanidina/farmacología , Datos de Secuencia Molecular , Proteínas de Unión a Poli-ADP-Ribosa , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , Transporte de Proteínas/efectos de los fármacos , ARN Helicasas , Proteínas con Motivos de Reconocimiento de ARN , Reproducibilidad de los Resultados , Estrés Fisiológico/efectos de los fármacos , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/metabolismo , Factores de Tiempo
17.
Nucleic Acids Res ; 40(12): 5497-510, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22402492

RESUMEN

Polycomb group (PcG) proteins are involved in epigenetic silencing where they function as major determinants of cell identity, stem cell pluripotency and the epigenetic gene silencing involved in cancer development. Recently numerous PcG proteins, including CBX4, have been shown to accumulate at sites of DNA damage. However, it remains unclear whether or not CBX4 or its E3 sumo ligase activity is directly involved in the DNA damage response (DDR). Here we define a novel role for CBX4 as an early DDR protein that mediates SUMO conjugation at sites of DNA lesions. DNA damage stimulates sumoylation of BMI1 by CBX4 at lysine 88, which is required for the accumulation of BMI1 at DNA damage sites. Moreover, we establish that CBX4 recruitment to the sites of laser micro-irradiation-induced DNA damage requires PARP activity but does not require H2AX, RNF8, BMI1 nor PI-3-related kinases. The importance of CBX4 in the DDR was confirmed by the depletion of CBX4, which resulted in decreased cellular resistance to ionizing radiation. Our results reveal a direct role for CBX4 in the DDR pathway.


Asunto(s)
Daño del ADN , Reparación del ADN , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Represoras/metabolismo , Proteínas Represoras/fisiología , Sumoilación , Animales , Línea Celular , Roturas del ADN de Doble Cadena , Células HEK293 , Humanos , Ligasas , Lisina/metabolismo , Ratones , Proteínas Nucleares/química , Poli(ADP-Ribosa) Polimerasas/metabolismo , Complejo Represivo Polycomb 1 , Proteínas del Grupo Polycomb , Proteínas Inhibidoras de STAT Activados/metabolismo , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas/química , Proteínas Represoras/química , Ubiquitina-Proteína Ligasas
18.
Nucleic Acids Res ; 40(20): 10287-301, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-22941645

RESUMEN

After the generation of DNA double-strand breaks (DSBs), poly(ADP-ribose) polymerase-1 (PARP-1) is one of the first proteins to be recruited and activated through its binding to the free DNA ends. Upon activation, PARP-1 uses NAD+ to generate large amounts of poly(ADP-ribose) (PAR), which facilitates the recruitment of DNA repair factors. Here, we identify the RNA-binding protein NONO, a partner protein of SFPQ, as a novel PAR-binding protein. The protein motif being primarily responsible for PAR-binding is the RNA recognition motif 1 (RRM1), which is also crucial for RNA-binding, highlighting a competition between RNA and PAR as they share the same binding site. Strikingly, the in vivo recruitment of NONO to DNA damage sites completely depends on PAR, generated by activated PARP-1. Furthermore, we show that upon PAR-dependent recruitment, NONO stimulates nonhomologous end joining (NHEJ) and represses homologous recombination (HR) in vivo. Our results therefore place NONO after PARP activation in the context of DNA DSB repair pathway decision. Understanding the mechanism of action of proteins that act in the same pathway as PARP-1 is crucial to shed more light onto the effect of interference on PAR-mediated pathways with PARP inhibitors, which have already reached phase III clinical trials but are until date poorly understood.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades , Proteínas Asociadas a Matriz Nuclear/metabolismo , Factores de Transcripción de Octámeros/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Supervivencia Celular , Células Cultivadas , Cromatina/metabolismo , Proteínas de Unión al ADN , Células HeLa , Recombinación Homóloga , Humanos , Ratones , Proteínas Asociadas a Matriz Nuclear/antagonistas & inhibidores , Proteínas Asociadas a Matriz Nuclear/química , Factores de Transcripción de Octámeros/antagonistas & inhibidores , Factores de Transcripción de Octámeros/química , Poli(ADP-Ribosa) Polimerasa-1 , Poli Adenosina Difosfato Ribosa/metabolismo , Dominios y Motivos de Interacción de Proteínas , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/química , Radiación Ionizante
19.
Nucleic Acids Res ; 40(16): 7788-805, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22669911

RESUMEN

Upon DNA damage induction, DNA-dependent poly(ADP-ribose) polymerases (PARPs) synthesize an anionic poly(ADP-ribose) (pADPr) scaffold to which several proteins bind with the subsequent formation of pADPr-associated multiprotein complexes. We have used a combination of affinity-purification methods and proteomics approaches to isolate these complexes and assess protein dynamics with respect to pADPr metabolism. As a first approach, we developed a substrate trapping strategy by which we demonstrate that a catalytically inactive Poly(ADP-ribose) glycohydrolase (PARG) mutant can act as a physiologically selective bait for the isolation of specific pADPr-binding proteins through its macrodomain-like domain. In addition to antibody-mediated affinity-purification methods, we used a pADPr macrodomain affinity resin to recover pADPr-binding proteins and their complexes. Second, we designed a time course experiment to explore the changes in the composition of pADPr-containing multiprotein complexes in response to alkylating DNA damage-mediated PARP activation. Spectral count clustering based on GeLC-MS/MS analysis was complemented with further analyses using high precision quantitative proteomics through isobaric tag for relative and absolute quantitation (iTRAQ)- and Stable isotope labeling by amino acids in cell culture (SILAC)-based proteomics. Here, we present a valuable resource in the interpretation of systems biology of the DNA damage response network in the context of poly(ADP-ribosyl)ation and provide a basis for subsequent investigations of pADPr-binding protein candidates.


Asunto(s)
Daño del ADN , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteoma/metabolismo , Reparación del ADN , Células HEK293 , Células HeLa , Humanos , Marcaje Isotópico , Complejos Multiproteicos/aislamiento & purificación , Mapas de Interacción de Proteínas , Proteómica/métodos , Estrés Fisiológico/genética
20.
Proc Natl Acad Sci U S A ; 108(34): 14103-8, 2011 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-21825151

RESUMEN

Ubiquitin mediated protein degradation is crucial for regulation of cell signaling and protein quality control. Poly(ADP-ribose) (PAR) is a cell-signaling molecule that mediates changes in protein function through binding at PAR binding sites. Here we characterize the PAR binding protein, Iduna, and show that it is a PAR-dependent ubiquitin E3 ligase. Iduna's E3 ligase activity requires PAR binding because point mutations at Y156A and R157A eliminate Iduna's PAR binding and Iduna's E3 ligase activity. Iduna's E3 ligase activity also requires an intact really interesting new gene (RING) domain because Iduna possessing point mutations at either H54A or C60A is devoid of ubiquitination activity. Tandem affinity purification reveals that Iduna binds to a number of proteins that are either PARsylated or bind PAR including PAR polymerase-1, 2 (PARP1, 2), nucleolin, DNA ligase III, KU70, KU86, XRCC1, and histones. PAR binding to Iduna activates its E3 ligase function, and PAR binding is required for Iduna ubiquitination of PARP1, XRCC1, DNA ligase III, and KU70. Iduna's PAR-dependent ubiquitination of PARP1 targets it for proteasomal degradation. Via PAR binding and ubiquitin E3 ligase activity, Iduna protects against cell death induced by the DNA damaging agent N-methyl-N-nitro-N-nitrosoguanidine (MNNG) and rescues cells from G1 arrest and promotes cell survival after γ-irradiation. Moreover, Iduna facilitates DNA repair by reducing apurinic/apyrimidinic (AP) sites after MNNG exposure and facilitates DNA repair following γ-irradiation as assessed by the comet assay. These results define Iduna as a PAR-dependent E3 ligase that regulates cell survival and DNA repair.


Asunto(s)
Daño del ADN , Poli Adenosina Difosfato Ribosa/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Línea Celular Tumoral , Citoprotección , Humanos , Poli(ADP-Ribosa) Polimerasas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína , Especificidad por Sustrato , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/química , Ubiquitinación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA