Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 9(1): 10075, 2019 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-31296950

RESUMO

The prime function of nucleoli is ribogenesis, however, several other, non-canonical functions have recently been identified, including a role in genotoxic stress response. Upon DNA damage, numerous proteins shuttle dynamically between the nucleolus and the nucleoplasm, yet the underlying molecular mechanisms are incompletely understood. Here, we demonstrate that PARP1 and PARylation contribute to genotoxic stress-induced nucleolar-nucleoplasmic shuttling of key genome maintenance factors in HeLa cells. Our work revealed that the RECQ helicase, WRN, translocates from nucleoli to the nucleoplasm upon treatment with the oxidizing agent H2O2, the alkylating agent 2-chloroethyl ethyl sulfide (CEES), and the topoisomerase inhibitor camptothecin (CPT). We show that after treatment with H2O2 and CEES, but not CPT, WRN translocation was dependent on PARP1 protein, yet independent of its enzymatic activity. In contrast, nucleolar-nucleoplasmic translocation of the base excision repair protein, XRCC1, was dependent on both PARP1 protein and its enzymatic activity. Furthermore, gossypol, which inhibits PARP1 activity by disruption of PARP1-protein interactions, abolishes nucleolar-nucleoplasmic shuttling of WRN, XRCC1 and PARP1, indicating the involvement of further upstream factors. In conclusion, this study highlights a prominent role of PARP1 in the DNA damage-induced nucleolar-nucleoplasmic shuttling of genome maintenance factors in HeLa cells in a toxicant and protein-specific manner.


Assuntos
Nucléolo Celular/metabolismo , Núcleo Celular/metabolismo , Poli(ADP-Ribose) Polimerase-1/metabolismo , Helicase da Síndrome de Werner/metabolismo , Proteína 1 Complementadora Cruzada de Reparo de Raio-X/metabolismo , Camptotecina/metabolismo , Dano ao DNA , Gossipol/metabolismo , Células HeLa , Humanos , Peróxido de Hidrogênio/metabolismo , Gás de Mostarda/análogos & derivados , Gás de Mostarda/metabolismo , Poli(ADP-Ribose) Polimerase-1/genética , Ligação Proteica , Transporte Proteico
2.
Nucleic Acids Res ; 46(2): 804-822, 2018 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-29216372

RESUMO

The post-translational modification poly(ADP-ribosyl)ation (PARylation) plays key roles in genome maintenance and transcription. Both non-covalent poly(ADP-ribose) binding and covalent PARylation control protein functions, however, it is unknown how the two modes of modification crosstalk mechanistically. Employing the tumor suppressor p53 as a model substrate, this study provides detailed insights into the interplay between non-covalent and covalent PARylation and unravels its functional significance in the regulation of p53. We reveal that the multifunctional C-terminal domain (CTD) of p53 acts as the central hub in the PARylation-dependent regulation of p53. Specifically, p53 bound to auto-PARylated PARP1 via highly specific non-covalent PAR-CTD interaction, which conveyed target specificity for its covalent PARylation by PARP1. Strikingly, fusing the p53-CTD to a protein that is normally not PARylated, renders this a target for covalent PARylation as well. Functional studies revealed that the p53-PAR interaction had substantial implications on molecular and cellular levels. Thus, PAR significantly influenced the complex p53-DNA binding properties and controlled p53 functions, with major implications on the p53-dependent interactome, transcription, and replication-associated recombination. Remarkably, this mechanism potentially also applies to other PARylation targets, since a bioinformatics analysis revealed that CTD-like regions are highly enriched in the PARylated proteome.


Assuntos
Poli(ADP-Ribose) Polimerase-1/metabolismo , Poli ADP Ribosilação , Processamento de Proteína Pós-Traducional , Proteína Supressora de Tumor p53/metabolismo , Linhagem Celular Tumoral , Humanos , Células K562 , Poli(ADP-Ribose) Polimerase-1/genética , Ligação Proteica , Domínios Proteicos , Proteína Supressora de Tumor p53/química , Proteína Supressora de Tumor p53/genética
3.
Nucleic Acids Res ; 44(21): 10386-10405, 2016 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-27694308

RESUMO

Genotoxic stress activates PARP1, resulting in the post-translational modification of proteins with poly(ADP-ribose) (PAR). We genetically deleted PARP1 in one of the most widely used human cell systems, i.e. HeLa cells, via TALEN-mediated gene targeting. After comprehensive characterization of these cells during genotoxic stress, we analyzed structure-function relationships of PARP1 by reconstituting PARP1 KO cells with a series of PARP1 variants. Firstly, we verified that the PARP1\E988K mutant exhibits mono-ADP-ribosylation activity and we demonstrate that the PARP1\L713F mutant is constitutively active in cells. Secondly, both mutants exhibit distinct recruitment kinetics to sites of laser-induced DNA damage, which can potentially be attributed to non-covalent PARP1-PAR interaction via several PAR binding motifs. Thirdly, both mutants had distinct functional consequences in cellular patho-physiology, i.e. PARP1\L713F expression triggered apoptosis, whereas PARP1\E988K reconstitution caused a DNA-damage-induced G2 arrest. Importantly, both effects could be rescued by PARP inhibitor treatment, indicating distinct cellular consequences of constitutive PARylation and mono(ADP-ribosyl)ation. Finally, we demonstrate that the cancer-associated PARP1 SNP variant (V762A) as well as a newly identified inherited PARP1 mutation (F304L\V762A) present in a patient with pediatric colorectal carcinoma exhibit altered biochemical and cellular properties, thereby potentially supporting human carcinogenesis. Together, we establish a novel cellular model for PARylation research, by revealing strong structure-function relationships of natural and artificial PARP1 variants.


Assuntos
Poli(ADP-Ribose) Polimerase-1/química , Poli(ADP-Ribose) Polimerase-1/metabolismo , Nucleases dos Efetores Semelhantes a Ativadores de Transcrição/química , Nucleases dos Efetores Semelhantes a Ativadores de Transcrição/metabolismo , Animais , Linhagem Celular , Dano ao DNA , Técnicas de Inativação de Genes , Marcação de Genes , Variação Genética , Células HeLa , Humanos , NAD/metabolismo , Poli(ADP-Ribose) Polimerase-1/genética , Ligação Proteica , Conformação Proteica , Proteínas Recombinantes , Deleção de Sequência , Relação Estrutura-Atividade
4.
Mol Cell Biol ; 35(23): 3974-89, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26391948

RESUMO

Poly(ADP-ribose) (PAR) polymerase 1 (PARP1) catalyzes the poly(ADP-ribosyl)ation (PARylation) of proteins, a posttranslational modification which forms the nucleic acid-like polymer PAR. PARP1 and PAR are integral players in the early DNA damage response, since PARylation orchestrates the recruitment of repair proteins to sites of damage. Human RecQ helicases are DNA unwinding proteins that are critical responders to DNA damage, but how their recruitment and activities are regulated by PARPs and PAR is poorly understood. Here we report that all human RecQ helicases interact with PAR noncovalently. Furthermore, we define the effects that PARP1, PARylated PARP1, and PAR have on RECQL5 and WRN, using both in vitro and in vivo assays. We show that PARylation is involved in the recruitment of RECQL5 and WRN to laser-induced DNA damage and that RECQL5 and WRN have differential responses to PARylated PARP1 and PAR. Furthermore, we show that the loss of RECQL5 or WRN resulted in increased sensitivity to PARP inhibition. In conclusion, our results demonstrate that PARP1 and PAR actively, and in some instances differentially, regulate the activities and cellular localization of RECQL5 and WRN, suggesting that PARylation acts as a fine-tuning mechanism to coordinate their functions in time and space during the genotoxic stress response.


Assuntos
Exodesoxirribonucleases/metabolismo , Poli Adenosina Difosfato Ribose/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Mapas de Interação de Proteínas , RecQ Helicases/metabolismo , Adenosina Trifosfatases/metabolismo , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Ativação Enzimática/efeitos dos fármacos , Células HEK293 , Células HeLa , Humanos , Poli(ADP-Ribose) Polimerase-1 , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Helicase da Síndrome de Werner
5.
Ageing Res Rev ; 23(Pt A): 12-28, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25555679

RESUMO

Genome instability represents a primary hallmark of aging and cancer. RecQL helicases (i.e., RECQL1, WRN, BLM, RECQL4, RECQL5) as well as poly(ADP-ribose) polymerases (PARPs, in particular PARP1) represent two central quality control systems to preserve genome integrity in mammalian cells. Consistently, both enzymatic families have been linked to mechanisms of aging and carcinogenesis in mice and humans. This is in accordance with clinical and epidemiological findings demonstrating that defects in three RecQL helicases, i.e., WRN, BLM, RECQL4, are related to human progeroid and cancer predisposition syndromes, i.e., Werner, Bloom, and Rothmund Thomson syndrome, respectively. Moreover, PARP1 hypomorphy is associated with a higher risk for certain types of cancer. On a molecular level, RecQL helicases and PARP1 are involved in the control of DNA repair, telomere maintenance, and replicative stress. Notably, over the last decade, it became apparent that all five RecQL helicases physically or functionally interact with PARP1 and/or its enzymatic product poly(ADP-ribose) (PAR). Furthermore, a profound body of evidence revealed that the cooperative function of RECQLs and PARP1 represents an important factor for maintaining genome integrity. In this review, we summarize the status quo of this molecular cooperation and discuss open questions that provide a basis for future studies to dissect the cooperative functions of RecQL helicases and PARP1 in aging and carcinogenesis.


Assuntos
Envelhecimento/genética , Genoma Humano/genética , Instabilidade Genômica/genética , Instabilidade Genômica/fisiologia , Poli(ADP-Ribose) Polimerases/genética , RecQ Helicases/genética , Envelhecimento/fisiologia , Animais , DNA/genética , Genoma Humano/fisiologia , Humanos , Camundongos , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/fisiologia , RecQ Helicases/fisiologia
6.
Med Sci Sports Exerc ; 47(3): 593-600, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25003772

RESUMO

INTRODUCTION: In some endurance sports, athletes complete several competitions within a short period, resulting in accumulated fatigue. It is unclear whether fatigued athletes choose the same pacing pattern (PP) as when they have recovered. PURPOSE: This study aimed to analyze effects of fatigue on PP of cyclists during a 40-km time trial (TT). METHODS: Twenty-three male cyclists (28.8 ± 7.6 yr) completed three 40-km TT on a cycle ergometer. TT were conducted before (TT1) and after (TT2) a 6-d training period. A third TT was carried out after 72 h of recovery (TT3). Training days consisted of two cycling sessions: mornings, 1 h at 95% of lactate threshold or 3 × 5 × 30 s all-out sprint; afternoons, 3 h at 80% individual anaerobic threshold. Four-kilometer split times (min) and RPE were recorded during TT. RESULTS: Performance decreased from TT1 to TT2 (65.7 ± 3.5 vs 66.7 ± 3.3 min; P < 0.05) and increased from TT2 to TT3 (66.7 ± 3.3 vs 65.5 ± 3.3 min; P < 0.01). PP showed a significant difference between TT1 and TT2 (P < 0.001) as well as between TT2 and TT3 (P < 0.01). PP in TT1 and TT3 showed no significant difference (P > 0.05). In TT1 and TT3, cyclists started faster in the first 4 km compared with TT2. RPE course showed no significant difference between TT (P > 0.05). CONCLUSIONS: Fatigue reversibly changes the PP of cyclists during a 40-km TT. Participants reduced their power output until premature exhaustion seemed very unlikely. This supports the assumption that pacing includes a combination of anticipation and feedback mechanisms.


Assuntos
Ciclismo/fisiologia , Fadiga/etiologia , Educação Física e Treinamento , Adulto , Antecipação Psicológica/fisiologia , Retroalimentação Fisiológica , Frequência Cardíaca , Humanos , Ácido Láctico/sangue , Masculino , Consumo de Oxigênio , Percepção/fisiologia , Esforço Físico , Respiração , Adulto Jovem
7.
Cell Metab ; 20(5): 840-855, 2014 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-25440059

RESUMO

Cockayne syndrome (CS) is an accelerated aging disorder characterized by progressive neurodegeneration caused by mutations in genes encoding the DNA repair proteins CS group A or B (CSA or CSB). Since dietary interventions can alter neurodegenerative processes, Csb(m/m) mice were given a high-fat, caloric-restricted, or resveratrol-supplemented diet. High-fat feeding rescued the metabolic, transcriptomic, and behavioral phenotypes of Csb(m/m) mice. Furthermore, premature aging in CS mice, nematodes, and human cells results from aberrant PARP activation due to deficient DNA repair leading to decreased SIRT1 activity and mitochondrial dysfunction. Notably, ß-hydroxybutyrate levels are increased by the high-fat diet, and ß-hydroxybutyrate, PARP inhibition, or NAD(+) supplementation can activate SIRT1 and rescue CS-associated phenotypes. Mechanistically, CSB can displace activated PARP1 from damaged DNA to limit its activity. This study connects two emerging longevity metabolites, ß-hydroxybutyrate and NAD(+), through the deacetylase SIRT1 and suggests possible interventions for CS.


Assuntos
Senilidade Prematura/dietoterapia , Senilidade Prematura/etiologia , Síndrome de Cockayne/complicações , Dieta Hiperlipídica , NAD/metabolismo , Sirtuína 1/metabolismo , Ácido 3-Hidroxibutírico/metabolismo , Senilidade Prematura/metabolismo , Senilidade Prematura/patologia , Animais , Linhagem Celular , Síndrome de Cockayne/metabolismo , Síndrome de Cockayne/patologia , Ativação Enzimática , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/metabolismo
8.
FEBS J ; 281(16): 3625-41, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24953096

RESUMO

Poly(ADP-ribose) (PAR) is a complex and reversible post-translational modification that controls protein function and localization through covalent modification of, or noncovalent binding to target proteins. Previously, we and others characterized the noncovalent, high-affinity binding of the key nucleotide excision repair (NER) protein XPA to PAR. In the present study, we address the functional relevance of this interaction. First, we confirm that pharmacological inhibition of cellular poly(ADP-ribosyl)ation (PARylation) impairs NER efficacy. Second, we demonstrate that the XPA-PAR interaction is mediated by specific basic amino acids within a highly conserved PAR-binding motif, which overlaps the DNA damage-binding protein 2 (DDB2) and transcription factor II H (TFIIH) interaction domains of XPA. Third, biochemical studies reveal a mutual regulation of PARP1 and XPA functions showing that, on the one hand, the XPA-PAR interaction lowers the DNA binding affinity of XPA, whereas, on the other hand, XPA itself strongly stimulates PARP1 enzymatic activity. Fourth, microirradiation experiments in U2OS cells demonstrate that PARP inhibition alters the recruitment properties of XPA-green fluorescent protein to sites of laser-induced DNA damage. In conclusion, our results reveal that XPA and PARP1 regulate each other in a reciprocal and PAR-dependent manner, potentially acting as a fine-tuning mechanism for the spatio-temporal regulation of the two factors during NER.


Assuntos
Poli Adenosina Difosfato Ribose/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Processamento de Proteína Pós-Traducional , Proteína de Xeroderma Pigmentoso Grupo A/metabolismo , Sequência de Aminoácidos , Sequência Consenso , Reparo do DNA , Células HeLa , Humanos , Cinética , Dados de Sequência Molecular , Poli(ADP-Ribose) Polimerase-1 , Poli(ADP-Ribose) Polimerases/química , Ligação Proteica , Células Sf9 , Proteína de Xeroderma Pigmentoso Grupo A/química
9.
ACS Chem Biol ; 8(1): 179-88, 2013 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-23082994

RESUMO

Werner syndrome is a premature aging disorder that is caused by defects in the Werner protein (WRN). WRN is a member of the RecQ helicase family and possesses helicase and exonuclease activities. It is involved in various aspects of DNA metabolism such as DNA repair, telomere maintenance, and replication. Poly(ADP-ribose) polymerase 1 (PARP1) is also involved in these processes by catalyzing the formation of the nucleic-acid-like biopolymer poly(ADP-ribose) (PAR). It was previously shown that WRN interacts with PARP1 and that WRN activity is inhibited by PARP1. Using several bioanalytical approaches, here we demonstrate that the enzymatic product of PARP1, i.e., PAR, directly interacts with WRN physically and functionally. First, WRN binds HPLC-size-fractionated short and long PAR in a noncovalent manner. Second, we identified and characterized a PAR-binding motif (PBM) within the WRN sequence and showed that several basic and hydrophobic amino acids are of critical importance for mediating the PAR binding. Third, PAR-binding inhibits the DNA-binding, the helicase and the exonuclease activities of WRN in a concentration-dependent manner. On the basis of our results we propose that the transient nature of PAR produced by living cells would provide a versatile and swiftly reacting control system for WRN's function. More generally, our work underscores the important role of noncovalent PAR-protein interactions as a regulatory mechanism of protein function.


Assuntos
Exodesoxirribonucleases/química , Poli Adenosina Difosfato Ribose/química , RecQ Helicases/química , Síndrome de Werner , Sequência de Aminoácidos , Sítios de Ligação , Eletroforese em Gel de Poliacrilamida , Exodesoxirribonucleases/metabolismo , Dados de Sequência Molecular , Poli Adenosina Difosfato Ribose/metabolismo , Estrutura Terciária de Proteína , RecQ Helicases/metabolismo , Alinhamento de Sequência , Helicase da Síndrome de Werner
10.
Mol Pharmacol ; 83(2): 470-80, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23188717

RESUMO

G-quadruplexes are higher-order nucleic acid structures that can form in G-rich telomeres and promoter regions of oncogenes. Telomeric quadruplex stabilization by small molecules can lead to telomere uncapping, followed by DNA damage response and senescence, as well as chromosomal fusions leading to deregulation of mitosis, followed by apoptosis and downregulation of oncogene expression. We report here on investigations into the mechanism of action of tetra-substituted naphthalene diimide ligands on the basis of cell biologic data together with a National Cancer Institute COMPARE study. We conclude that four principal mechanisms of action are implicated for these compounds: 1) telomere uncapping with subsequent DNA damage response and senescence; 2) inhibition of transcription/translation of oncogenes; 3) genomic instability through telomeric DNA end fusions, resulting in mitotic catastrophe and apoptosis; and 4) induction of chromosomal instability by telomere aggregate formation.


Assuntos
Antineoplásicos/farmacologia , Dano ao DNA , Quadruplex G , Imidas/farmacologia , Naftalenos/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , DNA/efeitos dos fármacos , DNA/metabolismo , Humanos , Ligantes , Células MCF-7 , Mitose/efeitos dos fármacos , Oncogenes/efeitos dos fármacos , Telômero/efeitos dos fármacos , Telômero/metabolismo
11.
Mech Ageing Dev ; 133(8): 575-9, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22766507

RESUMO

Werner syndrome is a disorder characterized by a premature aging phenotype. The disease is caused by mutations in the WRN gene which encodes a DNA helicase/exonuclease which is involved in multiple aspects of DNA metabolism. Current methods mostly rely on radiometric techniques to assess WRN exonuclease activity. Here we present an alternative, quantitative approach based on non-radioactive isotope dilution mass spectrometry (LC-MS/MS). A oligoduplex substrate mimicking the telomeric sequence was used for method development. Released nucleotides, which correlate with the degree of oligoduplex degradation, were dephosphorylated, purified, and quantified by LC-MS/MS. Heavy-isotope-labeled internal standards were used to account for technical variability. The method was validated in terms of reproducibility, time-course and concentration-dependency of the reaction. As shown in this study, the LC-MS/MS method can assess exonuclease activity of WRN mutants, WRN's substrate and strand specificity, and modulatory effects of WRN interaction partners and posttranslational modifications. Moreover, it can be used to analyze the selectivity and processivity of WRN exonuclease and allows the screening of small molecules for WRN exonuclease inhibitors. Importantly, this approach can easily be adapted to study nucleases other than WRN. This is of general interest, because exonucleases are key players in DNA metabolism and aging mechanisms.


Assuntos
DNA/química , Exodesoxirribonucleases/química , Espectrometria de Massas , RecQ Helicases/química , Telômero/química , Envelhecimento/genética , Envelhecimento/metabolismo , DNA/genética , DNA/metabolismo , Exodesoxirribonucleases/genética , Exodesoxirribonucleases/metabolismo , Humanos , Marcação por Isótopo , Mutação , RecQ Helicases/genética , RecQ Helicases/metabolismo , Telômero/genética , Telômero/metabolismo , Helicase da Síndrome de Werner
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...