Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Eur Biophys J ; 43(10-11): 509-16, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25119658

RESUMO

We have investigated the mobility of two EGFP-tagged DNA repair proteins, WRN and BLM. In particular, we focused on the dynamics in two locations, the nucleoli and the nucleoplasm. We found that both WRN and BLM use a "DNA-scanning" mechanism, with rapid binding-unbinding to DNA resulting in effective diffusion. In the nucleoplasm WRN and BLM have effective diffusion coefficients of 1.62 and 1.34 µm(2)/s, respectively. Likewise, the dynamics in the nucleoli are also best described by effective diffusion, but with diffusion coefficients a factor of ten lower than in the nucleoplasm. From this large reduction in diffusion coefficient we were able to classify WRN and BLM as DNA damage scanners. In addition to WRN and BLM we also classified other DNA damage proteins and found they all fall into one of two categories. Either they are scanners, similar to WRN and BLM, with very low diffusion coefficients, suggesting a scanning mechanism, or they are almost freely diffusing, suggesting that they interact with DNA only after initiation of a DNA damage response.


Assuntos
Nucléolo Celular/metabolismo , RecQ Helicases/metabolismo , Linhagem Celular Tumoral , DNA/metabolismo , Difusão , Humanos , Ligação Proteica , Transporte Proteico
2.
Nucleic Acids Res ; 40(4): 1621-35, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22013166

RESUMO

DNA decatenation mediated by Topoisomerase II is required to separate the interlinked sister chromatids post-replication. SGS1, a yeast homolog of the human RecQ family of helicases interacts with Topoisomerase II and plays a role in chromosome segregation, but this functional interaction has yet to be identified in higher organisms. Here, we report a physical and functional interaction of Topoisomerase IIα with RECQL5, one of five mammalian RecQ helicases, during DNA replication. Direct interaction of RECQL5 with Topoisomerase IIα stimulates the decatenation activity of Topoisomerase IIα. Consistent with these observations, RECQL5 co-localizes with Topoisomerase IIα during S-phase of the cell cycle. Moreover, cells with stable depletions of RECQL5 display a slow proliferation rate, a G2/M cell cycle arrest and late S-phase cycling defects. Metaphase spreads generated from RECQL5-depleted cells exhibit undercondensed and entangled chromosomes. Further, RECQL5-depleted cells activate a G2/M checkpoint and undergo apoptosis. These phenotypes are similar to those observed when Topoisomerase II catalytic activity is inhibited. These results reveal an important role for RECQL5 in the maintenance of genomic stability and a new insight into the decatenation process.


Assuntos
Antígenos de Neoplasias/metabolismo , Ciclo Celular , DNA Topoisomerases Tipo II/metabolismo , DNA Catenado/metabolismo , Proteínas de Ligação a DNA/metabolismo , RecQ Helicases/metabolismo , Apoptose , Pontos de Checagem do Ciclo Celular , Linhagem Celular , Proliferação de Células , Aberrações Cromossômicas , Exodesoxirribonucleases/metabolismo , Humanos , Metáfase/genética , RecQ Helicases/antagonistas & inibidores , Helicase da Síndrome de Werner
3.
Carcinogenesis ; 34(10): 2218-30, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23715498

RESUMO

Interstrand cross-links (ICLs) are very severe lesions as they are absolute blocks of replication and transcription. This property of interstrand cross-linking agents has been exploited clinically for the treatment of cancers and other diseases. ICLs are repaired in human cells by specialized DNA repair pathways including components of the nucleotide excision repair pathway, double-strand break repair pathway and the Fanconi anemia pathway. In this report, we identify the role of RECQL5, a member of the RecQ family of helicases, in the repair of ICLs. Using laser-directed confocal microscopy, we demonstrate that RECQL5 is recruited to ICLs formed by trioxalen (a psoralen-derived compound) and ultraviolet irradiation A. Using single-cell gel electrophoresis and proliferation assays, we identify the role of RECQL5 in the repair of ICL lesions. The domain of RECQL5 that recruits to the site of ICL was mapped to the KIX region between amino acids 500 and 650. Inhibition of transcription and of topoisomerases did not affect recruitment, which was inhibited by DNA-intercalating agents, suggesting that the DNA structure itself may be responsible for the recruitment of RECQL5 to the sites of ICLs.


Assuntos
Reagentes de Ligações Cruzadas/toxicidade , Dano ao DNA/efeitos dos fármacos , Reparo do DNA/fisiologia , Ficusina/toxicidade , RecQ Helicases/metabolismo , Linhagem Celular , DNA Topoisomerases/metabolismo , Exodesoxirribonucleases/metabolismo , Humanos , Cinética , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , RecQ Helicases/química , Inibidores da Topoisomerase/farmacologia , Transcrição Gênica , Helicase da Síndrome de Werner
4.
Nucleic Acids Res ; 38(9): 2904-16, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20081208

RESUMO

Human RECQL5 is a member of the RecQ helicase family which is implicated in genome maintenance. Five human members of the family have been identified; three of them, BLM, WRN and RECQL4 are associated with elevated cancer risk. RECQL1 and RECQL5 have not been linked to any human disorder yet; cells devoid of RECQL1 and RECQL5 display increased chromosomal instability. Here, we report the physical and functional interaction of the large isomer of RECQL5, RECQL5beta, with the human flap endonuclease 1, FEN1, which plays a critical role in DNA replication, recombination and repair. RECQL5beta dramatically stimulates the rate of FEN1 cleavage of flap DNA substrates. Moreover, we show that RECQL5beta and FEN1 interact physically and co-localize in the nucleus in response to DNA damage. Our findings, together with the previous literature on WRN, BLM and RECQL4's stimulation of FEN1, suggests that the ability of RecQ helicases to stimulate FEN1 may be a general feature of this class of enzymes. This could indicate a common role for the RecQ helicases in the processing of oxidative DNA damage.


Assuntos
Endonucleases Flap/metabolismo , RecQ Helicases/metabolismo , Linhagem Celular , Núcleo Celular/enzimologia , DNA/química , DNA/metabolismo , Clivagem do DNA , DNA de Cadeia Simples/metabolismo , Endonucleases Flap/análise , Humanos , RecQ Helicases/análise
5.
Nucleic Acids Res ; 36(4): 1380-9, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18203746

RESUMO

DNA repair is an important mechanism by which cells maintain genomic integrity. Decline in DNA repair capacity or defects in repair factors are thought to contribute to premature aging in mammals. The nematode Caenorhabditis elegans is a good model for studying longevity and DNA repair because of key advances in understanding the genetics of aging in this organism. Long-lived C. elegans mutants have been identified and shown to be resistant to oxidizing agents and UV irradiation, suggesting a genetically determined correlation between DNA repair capacity and life span. In this report, gene-specific DNA repair is compared in wild-type C. elegans and stress-resistant C. elegans mutants for the first time. DNA repair capacity is higher in long-lived C. elegans mutants than in wild-type animals. In addition, RNAi knockdown of the nucleotide excision repair gene xpa-1 increased sensitivity to UV and reduced the life span of long-lived C. elegans mutants. These findings support that DNA repair capacity correlates with longevity in C. elegans.


Assuntos
Caenorhabditis elegans/genética , Reparo do DNA , Longevidade/genética , Animais , Caenorhabditis elegans/crescimento & desenvolvimento , Caenorhabditis elegans/efeitos da radiação , Proteínas de Caenorhabditis elegans/antagonistas & inibidores , Proteínas de Caenorhabditis elegans/genética , Mutação , Estresse Oxidativo , Dímeros de Pirimidina/metabolismo , Interferência de RNA , Raios Ultravioleta
6.
Nucleic Acids Res ; 34(2): 745-54, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16449207

RESUMO

Genome instability is a characteristic of cancer and aging, and is a hallmark of the premature aging disorder Werner syndrome (WS). Evidence suggests that the Werner syndrome protein (WRN) contributes to the maintenance of genome integrity through its involvement in DNA repair. In particular, biochemical evidence indicates a role for WRN in base excision repair (BER). We have previously reported that WRN helicase activity stimulates DNA polymerase beta (pol beta) strand displacement synthesis in vitro. In this report we demonstrate that WRN exonuclease activity can act cooperatively with pol beta, a polymerase lacking 3'-5' proofreading activity. Furthermore, using small interference RNA technology, we demonstrate that WRN knockdown cells are hypersensitive to the alkylating agent methyl methanesulfonate, which creates DNA damage that is primarily repaired by the BER pathway. In addition, repair assays using whole cell extracts from WRN knockdown cells indicate a defect in long patch (LP) BER. These findings demonstrate that WRN plays a direct role in the repair of methylation-induced DNA damage, and suggest a role for both WRN helicase and exonuclease activities together with pol beta during LP BER.


Assuntos
DNA Helicases/fisiologia , DNA Polimerase beta/metabolismo , Reparo do DNA , Exodesoxirribonucleases/fisiologia , Alquilantes/toxicidade , Pareamento Incorreto de Bases , Linhagem Celular , Dano ao DNA , DNA Helicases/antagonistas & inibidores , Exodesoxirribonucleases/antagonistas & inibidores , Humanos , Metanossulfonato de Metila/toxicidade , Interferência de RNA , RecQ Helicases , Helicase da Síndrome de Werner
7.
Mol Cell Biol ; 23(23): 8601-13, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14612404

RESUMO

A defect in the Werner syndrome protein (WRN) leads to the premature aging disease Werner syndrome (WS). Hallmark features of cells derived from WS patients include genomic instability and hypersensitivity to certain DNA-damaging agents. WRN contains a highly conserved region, the RecQ conserved domain, that plays a central role in protein interactions. We searched for proteins that bound to this region, and the most prominent direct interaction was with poly(ADP-ribose) polymerase 1 (PARP-1), a nuclear enzyme that protects the genome by responding to DNA damage and facilitating DNA repair. In pursuit of a functional interaction between WRN and PARP-1, we found that WS cells are deficient in the poly(ADP-ribosyl)ation pathway after they are treated with the DNA-damaging agents H2O2 and methyl methanesulfonate. After cellular stress, PARP-1 itself becomes activated, but the poly(ADP-ribosyl)ation of other cellular proteins is severely impaired in WS cells. Overexpression of the PARP-1 binding domain of WRN strongly inhibits the poly(ADP-ribosyl)ation activity in H2O2-treated control cell lines. These results indicate that the WRN/PARP-1 complex plays a key role in the cellular response to oxidative stress and alkylating agents, suggesting a role for these proteins in the base excision DNA repair pathway.


Assuntos
Dano ao DNA , DNA Helicases/metabolismo , Poli Adenosina Difosfato Ribose/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Síndrome de Werner/metabolismo , Sítios de Ligação , Linhagem Celular , DNA Helicases/química , DNA Helicases/genética , Reparo do DNA , Exodesoxirribonucleases , Proteínas de Fluorescência Verde , Células HeLa , Humanos , Peróxido de Hidrogênio/toxicidade , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Substâncias Macromoleculares , Metanossulfonato de Metila/toxicidade , Mutação , Estresse Oxidativo , Poli(ADP-Ribose) Polimerases/química , Estrutura Terciária de Proteína , RecQ Helicases , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Síndrome de Werner/genética , Helicase da Síndrome de Werner
8.
Oncogene ; 24(32): 5026-42, 2005 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-15897889

RESUMO

The accumulation of DNA damage and mutations is considered a major cause of cancer and aging. While it is known that DNA damage can affect changes in gene expression, transcriptional regulation after DNA damage is poorly understood. We characterized the expression of 6912 genes in human primary fibroblasts after exposure to three different kinds of cellular stress that introduces DNA damage: 4-nitroquinoline-1-oxide (4NQO), gamma-irradiation, or UV-irradiation. Each type of stress elicited damage specific gene expression changes of up to 10-fold. A total of 85 genes had similar changes in expression of 3-40-fold after all three kinds of stress. We examined transcription in cells from young and old individuals and from patients with Werner syndrome (WS), a segmental progeroid condition with a high incidence of cancer, and found various age-associated transcriptional changes depending upon the type of cellular stress. Compared to young individuals, both WS and old individuals had similarly aberrant transcriptional responses to gamma- and UV-irradiation, suggesting a role for Werner protein in stress-induced gene expression. Our results suggest that aberrant DNA damage-induced gene regulation may contribute to the aging process and the premature aging in WS.


Assuntos
Envelhecimento/genética , Dano ao DNA/genética , Regulação da Expressão Gênica , Síndrome de Werner/genética , 4-Nitroquinolina-1-Óxido/farmacologia , Adulto , Idoso , Linhagem Celular , Fibroblastos/efeitos dos fármacos , Fibroblastos/fisiologia , Fibroblastos/efeitos da radiação , Raios gama , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos da radiação , Genes Precoces/efeitos dos fármacos , Genes Precoces/efeitos da radiação , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Quinolonas/farmacologia , Pele/citologia , Estresse Fisiológico , Raios Ultravioleta
9.
Nucleic Acids Res ; 30(3): 782-93, 2002 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-11809892

RESUMO

Cockayne syndrome (CS) is a rare inherited human genetic disorder characterized by UV sensitivity, developmental abnormalities and premature aging. The cellular and molecular phenotypes of CS include increased sensitivity to oxidative and UV-induced DNA lesions. The CSB protein is thought to play a pivotal role in transcription-coupled repair and CS-B cells are defective in the repair of the transcribed strand of active genes, both after exposure to UV and in the presence of oxidative DNA lesions. A previous study has indicated that a conserved helicase ATPase motif II residue is essential for the function of the CSB protein in responding to UV-induced DNA damage in a hamster cell line. Due to the limitations in studying a complex human disorder in another species, this study introduced the site-directed mutation of the ATPase motif II in the human CSB gene in an isogenic human cell line. The CSB mutant allele was tested for genetic complementation of UV-sensitive phenotypes in the human CS-B cell line CS1AN.S3.G2. In addition, the incision of an 8-oxoguanine lesion by extracts of the CS-B cell lines stably transfected with the wild-type or ATPase mutant CSB gene has been investigated. The ATPase motif II point mutation (E646Q) abolished the function of the CSB protein to complement the UV-sensitive phenotypes of survival, RNA synthesis recovery and apoptosis. Interestingly, whole-cell extract prepared from these mutant cells retained wild-type incision activity on an oligonucleotide containing a single 8-oxoguanine lesion, whereas the absence of the CSB gene altogether resulted in reduced incision activity relative to wild-type. These results suggest damage-specific functional requirements for CSB in the repair of UV-induced and oxidative lesions in human cells. The transfection of the mutant or wild-type CSB gene into the CS1AN.S3.G2 cells did not alter the expression of the subset of genes examined by cDNA array analysis.


Assuntos
Adenosina Trifosfatases/química , Síndrome de Cockayne/genética , Dano ao DNA/genética , Dano ao DNA/efeitos da radiação , DNA Helicases/química , DNA Helicases/metabolismo , Reparo do DNA/genética , Guanina/análogos & derivados , Guanina/metabolismo , Timina/análogos & derivados , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Extratos Celulares , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos da radiação , Síndrome de Cockayne/enzimologia , Citosina/análogos & derivados , Citosina/metabolismo , DNA Helicases/genética , Enzimas Reparadoras do DNA , Fibroblastos , Perfilação da Expressão Gênica , Teste de Complementação Genética , Humanos , Peróxido de Hidrogênio/farmacologia , Mutação/genética , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas de Ligação a Poli-ADP-Ribose , Estrutura Terciária de Proteína , RNA/biossíntese , Tolerância a Radiação/genética , Timina/metabolismo , Raios Ultravioleta
10.
Cell Rep ; 16(10): 2641-2650, 2016 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-27568560

RESUMO

The accumulation of damage caused by oxidative stress has been linked to aging and to the etiology of numerous age-related diseases. The longevity gene, sirtuin 6 (SIRT6), promotes genome stability by facilitating DNA repair, especially under oxidative stress conditions. Here we uncover the mechanism by which SIRT6 is activated by oxidative stress to promote DNA double-strand break (DSB) repair. We show that the stress-activated protein kinase, c-Jun N-terminal kinase (JNK), phosphorylates SIRT6 on serine 10 in response to oxidative stress. This post-translational modification facilitates the mobilization of SIRT6 to DNA damage sites and is required for efficient recruitment of poly (ADP-ribose) polymerase 1 (PARP1) to DNA break sites and for efficient repair of DSBs. Our results demonstrate a post-translational mechanism regulating SIRT6, and they provide the link between oxidative stress signaling and DNA repair pathways that may be critical for hormetic response and longevity assurance.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Estresse Oxidativo , Poli(ADP-Ribose) Polimerase-1/metabolismo , Sirtuínas/metabolismo , Adenosina Difosfato Ribose/metabolismo , Animais , Células HEK293 , Humanos , Camundongos Knockout , Modelos Biológicos , Fosforilação , Fosfosserina/metabolismo
11.
Oncogene ; 22(8): 1135-49, 2003 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-12606941

RESUMO

Cockayne syndrome (CS) is a human hereditary disease belonging to the group of segmental progerias, and the clinical phenotype is characterized by postnatal growth failure, neurological dysfunction, cachetic dwarfism, photosensitivity, sensorineural hearing loss, and retinal degradation. CS-B cells are defective in transcription-coupled DNA repair, base excision repair, transcription, and chromatin structural organization. Using array analysis, we have examined the expression profile in CS complementation group B (CS-B) fibroblasts after exposure to oxidative stress (H2O2) before and after complete complementation with the CSB gene. The following isogenic cell lines were compared: CS-B cells (CS-B null), CS-B cells complemented with wild-type CSB (CS-B wt), and a stably transformed cell line with a point mutation in the ATPase domain of CSB (CS-B ATPase mutant). In the wt rescued cells, we detected significant induction (two-fold) of 112 genes out of the 6912 analysed. The patterns suggested an induction or upregulation of genes involved in several DNA metabolic processes including DNA repair, transcription, and signal transduction. In both CS-B mutant cell lines, we found a general deficiency in transcription after oxidative stress, suggesting that the CSB protein influenced the regulation of transcription of certain genes. Of the 6912 genes, 122 were differentially regulated by more than two-fold. Evidently, the ATPase function of CSB is biologically important as the deficiencies seen in the ATPase mutant cells are very similar to those observed in the CS-B-null cells. Some major defects are in the transcription of genes involved in DNA repair, signal transduction, and ribosomal functions.


Assuntos
Síndrome de Cockayne/patologia , DNA Helicases/fisiologia , Reparo do DNA/fisiologia , Perfilação da Expressão Gênica , Estresse Oxidativo/genética , Transcrição Gênica/fisiologia , Adenosina Trifosfatases/deficiência , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/fisiologia , Northern Blotting , Linhagem Celular/efeitos dos fármacos , Linhagem Celular/enzimologia , Linhagem Celular Transformada , Síndrome de Cockayne/enzimologia , DNA Helicases/deficiência , DNA Helicases/genética , Reparo do DNA/genética , Enzimas Reparadoras do DNA , Replicação do DNA/genética , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Teste de Complementação Genética , Humanos , Peróxido de Hidrogênio/toxicidade , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas de Ligação a Poli-ADP-Ribose , Proteínas Recombinantes de Fusão/fisiologia , Reprodutibilidade dos Testes , Transdução de Sinais/genética , Transcrição Gênica/efeitos dos fármacos , Transfecção
12.
FASEB J ; 18(15): 1970-2, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15459124

RESUMO

Werner syndrome (WS) is a rare disease caused by the lack of a functional nuclear WS protein (WRN). WS is characterized by the early onset of premature aging signs and a high incidence of sarcomas. WS diploid fibroblasts have a short life span and extensive genomic instability. Mammalian cells are continuously exposed to reactive oxygen species (ROS), which represent human mutagens and are thought to be a major contributor to the aging process. Hydrogen peroxide (H2O2) is a common ROS intermediate generated by various forms of oxidative stress. In response to H2O2-induced DNA damage, normal human diploid fibroblasts follow a pathway leading to irreversible proliferation arrest and premature senescence. Here we show that in contrast to normal human fibroblasts, WS diploid fibroblasts continue proliferating after extensive H2O2-induced DNA damage and accumulate oxidative DNA lesions. A direct role of WRN in this abnormal cellular response to H2O2 is demonstrated by interfering with WRN expression in normal human fibroblasts. We propose a role for WRN in the detection and/or processing of oxidative DNA lesions and in cellular responses to H2O2 as they relate to some of the phenotypical aspects of WS cells.


Assuntos
Dano ao DNA , Peróxido de Hidrogênio/toxicidade , Síndrome de Werner/genética , Morte Celular , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Senescência Celular , DNA/metabolismo , DNA Helicases/metabolismo , Exodesoxirribonucleases , Fibroblastos/efeitos dos fármacos , Humanos , Oxirredução , RecQ Helicases , Síndrome de Werner/metabolismo , Helicase da Síndrome de Werner
13.
Aging (Albany NY) ; 6(1): 70-81, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24429382

RESUMO

WRN protein, defective in Werner syndrome (WS), a human segmental progeria, is a target of serine/threonine kinases involved in sensing DNA damage. DNA-PK phosphorylates WRN in response to DNA double strand breaks (DSBs). However, the main phosphorylation sites and functional importance of the phosphorylation of WRN has remained unclear. Here, we identify Ser-440 and -467 in WRN as major phosphorylation sites mediated by DNA-PK.In vitro, DNA-PK fails to phosphorylate a GST-WRN fragment with S440A and/or S467A substitution. In addition, full length WRN with the mutation expressed in 293T cells was not phosphorylated in response to DSBs produced by bleomycin. Accumulation of the mutant WRN at the site of laser-induced DSBs occurred with the same kinetics as wild type WRN in live HeLa cells. While the wild type WRN relocalized to the nucleoli after 24 hours recovery from etoposide-induced DSBs, the mutant WRN remained mostly in the nucleoplasm. Consistent with this, WS cells expressing the mutants exhibited less DNA repair efficiency and more sensitivity to etoposide, compared to those expressing wild type. Our findings indicate that phosphorylation of Ser-440 and -467 in WRN are important for relocalization of WRN to nucleoli, and that it is required for efficient DSB repair.


Assuntos
Nucléolo Celular/enzimologia , Quebras de DNA de Cadeia Dupla , Proteína Quinase Ativada por DNA/metabolismo , Exodesoxirribonucleases/metabolismo , Proteínas Nucleares/metabolismo , RecQ Helicases/metabolismo , Bleomicina/farmacologia , Nucléolo Celular/efeitos dos fármacos , Reparo do DNA , Proteína Quinase Ativada por DNA/genética , Relação Dose-Resposta a Droga , Etoposídeo/farmacologia , Exodesoxirribonucleases/genética , Células HEK293 , Células HeLa , Humanos , Cinética , Mutação , Proteínas Nucleares/genética , Fosforilação , Processamento de Proteína Pós-Traducional , RecQ Helicases/genética , Serina , Transfecção , Helicase da Síndrome de Werner
14.
Mol Biol Cell ; 23(21): 4273-85, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22973052

RESUMO

Human RECQL5 is a member of the RecQ helicase family, which maintains genome stability via participation in many DNA metabolic processes, including DNA repair. Human cells lacking RECQL5 display chromosomal instability. We find that cells depleted of RECQL5 are sensitive to oxidative stress, accumulate endogenous DNA damage, and increase the cellular poly(ADP-ribosyl)ate response. In contrast to the RECQ helicase family members WRN, BLM, and RECQL4, RECQL5 accumulates at laser-induced single-strand breaks in normal human cells. RECQL5 depletion affects the levels of PARP-1 and XRCC1, and our collective results suggest that RECQL5 modulates and/or directly participates in base excision repair of endogenous DNA damage, thereby promoting chromosome stability in normal human cells.


Assuntos
Dano ao DNA , RecQ Helicases/metabolismo , Dano ao DNA/genética , Reparo do DNA/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação para Baixo/genética , Técnicas de Silenciamento de Genes , Proteínas de Fluorescência Verde/metabolismo , Células HCT116 , Células HeLa , Humanos , Lasers , Modelos Biológicos , Oxirredução , Estresse Oxidativo/genética , Poli Adenosina Difosfato Ribose/metabolismo , RecQ Helicases/deficiência , Proteínas Recombinantes de Fusão/metabolismo , Proteína 1 Complementadora Cruzada de Reparo de Raio-X
15.
DNA Repair (Amst) ; 11(3): 267-77, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-22222486

RESUMO

Human exonuclease 1 (hEXO1) acts directly in diverse DNA processing events, including replication, mismatch repair (MMR), and double strand break repair (DSBR), and it was also recently described to function as damage sensor and apoptosis inducer following DNA damage. In contrast, 14-3-3 proteins are regulatory phosphorserine/threonine binding proteins involved in the control of diverse cellular events, including cell cycle checkpoint and apoptosis signaling. hEXO1 is regulated by post-translation Ser/Thr phosphorylation in a yet not fully clarified manner, but evidently three phosphorylation sites are specifically induced by replication inhibition leading to protein ubiquitination and degradation. We demonstrate direct and robust interaction between hEXO1 and six of the seven 14-3-3 isoforms in vitro, suggestive of a novel protein interaction network between DNA repair and cell cycle control. Binding experiments reveal weak affinity of the more selective isoform 14-3-3σ but both 14-3-3 isoforms η and σ significantly stimulate hEXO1 activity, indicating that these regulatory proteins exert a common regulation mode on hEXO1. Results demonstrate that binding involves the phosphorable amino acid S746 in hEXO1 and most likely a second unidentified binding motif. 14-3-3 associations do not appear to directly influence hEXO1 in vitro nuclease activity or in vitro DNA replication initiation. Moreover, specific phosphorylation variants, including hEXO1 S746A, are efficiently imported to the nucleus; to associate with PCNA in distinct replication foci and respond to DNA double strand breaks (DSBs), indicating that 14-3-3 binding does not involve regulating the subcellular distribution of hEXO1. Altogether, these results suggest that association may be related to regulation of hEXO1 availability during the DNA damage response to plausibly prevent extensive DNA resection at the damage site, as supported by recent studies.


Assuntos
Proteínas 14-3-3/metabolismo , Pontos de Checagem do Ciclo Celular , Enzimas Reparadoras do DNA/química , Enzimas Reparadoras do DNA/metabolismo , Reparo do DNA , Exodesoxirribonucleases/química , Exodesoxirribonucleases/metabolismo , Transporte Ativo do Núcleo Celular , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Núcleo Celular/metabolismo , Replicação do DNA , Células HEK293 , Células HeLa , Humanos , Camundongos , Modelos Biológicos , Dados de Sequência Molecular , Proteínas Mutantes/metabolismo , Células NIH 3T3 , Fosforilação , Antígeno Nuclear de Célula em Proliferação/metabolismo , Ligação Proteica , Mapeamento de Interação de Proteínas , Proteínas Recombinantes/metabolismo , Relação Estrutura-Atividade
16.
DNA Repair (Amst) ; 10(1): 73-86, 2011 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-20970388

RESUMO

Human exonuclease 1 (hEXO1) is implicated in DNA metabolism, including replication, recombination and repair, substantiated by its interactions with PCNA, DNA helicases BLM and WRN, and several DNA mismatch repair (MMR) proteins. We investigated the sub-nuclear localization of hEXO1 during S-phase progression and in response to laser-induced DNA double strand breaks (DSBs). We show that hEXO1 and PCNA co-localize in replication foci. This apparent interaction is sustained throughout S-phase. We also demonstrate that hEXO1 is rapidly recruited to DNA DSBs. We have identified a PCNA interacting protein (PIP-box) region on hEXO1 located in its COOH-terminal ((788)QIKLNELW(795)). This motif is essential for PCNA binding and co-localization during S-phase. Recruitment of hEXO1 to DNA DSB sites is dependent on the MMR protein hMLH1. We show that two distinct hMLH1 interaction regions of hEXO1 (residues 390-490 and 787-846) are required to direct the protein to the DNA damage site. Our results reveal that protein domains in hEXO1 in conjunction with specific protein interactions control bi-directional routing of hEXO1 between on-going DNA replication and repair processes in living cells.


Assuntos
Quebras de DNA de Cadeia Dupla/efeitos da radiação , Reparo de Erro de Pareamento de DNA/fisiologia , Enzimas Reparadoras do DNA/metabolismo , Replicação do DNA/fisiologia , Exodesoxirribonucleases/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Motivos de Aminoácidos , Substituição de Aminoácidos , Animais , DNA/genética , DNA/metabolismo , Reparo de Erro de Pareamento de DNA/efeitos da radiação , Enzimas Reparadoras do DNA/genética , Enzimas Reparadoras do DNA/efeitos da radiação , Replicação do DNA/efeitos da radiação , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Exodesoxirribonucleases/genética , Exodesoxirribonucleases/efeitos da radiação , Células HeLa , Humanos , Lasers/efeitos adversos , Camundongos , Proteína 1 Homóloga a MutL , Proteína 3 Homóloga a MutS , Células NIH 3T3 , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Antígeno Nuclear de Célula em Proliferação/genética , Antígeno Nuclear de Célula em Proliferação/metabolismo , Transporte Proteico/genética , Transporte Proteico/efeitos da radiação , RecQ Helicases/genética , RecQ Helicases/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/efeitos da radiação , Fase S , Helicase da Síndrome de Werner
17.
Aging Cell ; 9(3): 358-71, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20222902

RESUMO

Rothmund-Thomson syndrome (RTS) is an autosomal recessive hereditary disorder associated with mutation in RECQL4 gene, a member of the human RecQ helicases. The disease is characterized by genomic instability, skeletal abnormalities and predisposition to malignant tumors, especially osteosarcomas. The precise role of RECQL4 in cellular pathways is largely unknown; however, recent evidence suggests its involvement in multiple DNA metabolic pathways. This study investigates the roles of RECQL4 in DNA double-strand break (DSB) repair. The results show that RECQL4-deficient fibroblasts are moderately sensitive to gamma-irradiation and accumulate more gammaH2AX and 53BP1 foci than control fibroblasts. This is suggestive of defects in efficient repair of DSB's in the RECQL4-deficient fibroblasts. Real time imaging of live cells using laser confocal microscopy shows that RECQL4 is recruited early to laser-induced DSBs and remains for a shorter duration than WRN and BLM, indicating its distinct role in repair of DSBs. Endogenous RECQL4 also colocalizes with gammaH2AX at the site of DSBs. The RECQL4 domain responsible for its DNA damage localization has been mapped to the unique N-terminus domain between amino acids 363-492, which shares no homology to recruitment domains of WRN and BLM to the DSBs. Further, the recruitment of RECQL4 to laser-induced DNA damage is independent of functional WRN, BLM or ATM proteins. These results suggest distinct cellular dynamics for RECQL4 protein at the site of laser-induced DSB and that it might play important roles in efficient repair of DSB's.


Assuntos
Quebras de DNA de Cadeia Dupla , Reparo do DNA , DNA/metabolismo , RecQ Helicases/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos da radiação , DNA/genética , Histonas/metabolismo , Humanos , Síndrome de Rothmund-Thomson/genética , Síndrome de Rothmund-Thomson/metabolismo , Síndrome de Rothmund-Thomson/patologia
18.
Mol Cell Biochem ; 279(1-2): 75-84, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16283516

RESUMO

Chromate compounds are known human lung carcinogens. Water solubility is an important factor in the carcinogenicity of these compounds with the most potent carcinogenic compounds being water-insoluble or 'particulate'. Previously we have shown that particulate chromates dissolve extracellularly releasing chromium (Cr) and lead (Pb) ions and only the Cr ions induce genotoxicity. Pb ions have been considered to have epigenetic effects and it is thought that these may enhance the carcinogenic activity of lead chromate, perhaps by stimulating Cr-damaged cells to divide. However, this possibility has not been directly tested. Accordingly, we investigated the ability of Pb ions to stimulate human lung cells and possibly force lead chromate-damaged cells to grow. We found that at concentrations of lead chromate that induced damage, human lung cells exhibited cell cycle arrest and growth inhibition that were very similar to those observed for sodium chromate. Moreover, we found that soluble Pb ions were not growth stimulatory to human lung cells and in fact induced progressive mitotic arrest. These data indicate that lead chromate-generated Cr ions cause growth inhibition and cell cycle arrest and that Pb does not induce epigenetic effects that stimulate chromate-damaged cells to grow.


Assuntos
Proliferação de Células/efeitos dos fármacos , Cromatos , Dano ao DNA , Chumbo , Mutagênicos/toxicidade , Cátions Bivalentes , Ciclo Celular , Linhagem Celular , Cromatos/toxicidade , Relação Dose-Resposta a Droga , Fibroblastos , Glutamatos/farmacologia , Humanos , Chumbo/farmacologia , Pulmão , Compostos de Sódio , Fatores de Tempo
19.
Proc Natl Acad Sci U S A ; 100(21): 12259-64, 2003 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-14527998

RESUMO

Werner syndrome (WS) is a premature aging disorder, displaying defects in DNA replication, recombination, repair, and transcription. It has been hypothesized that several WS phenotypes are secondary consequences of aberrant gene expression and that a transcription defect may be crucial to the development of the syndrome. We used cDNA microarrays to characterize the expression of 6,912 genes and ESTs across a panel of 15 primary human fibroblast cell lines derived from young donors, old donors, and WS patients. Of the analyzed genes, 6.3% displayed significant differences in expression when either WS or old donor cells were compared with young donor cells. This result demonstrates that the WS transcription defect is specific to certain genes. Transcription alterations in WS were strikingly similar to those in normal aging: 91% of annotated genes displayed similar expression changes in WS and in normal aging, 3% were unique to WS, and 6% were unique to normal aging. We propose that a defect in the transcription of the genes as identified in this study could produce many of the complex clinical features of WS. The remarkable similarity between WS and normal aging suggests that WS causes the acceleration of a normal aging mechanism. This finding supports the use of WS as an aging model and implies that the transcription alterations common to WS and normal aging represent general events in the aging process.


Assuntos
Envelhecimento/genética , Expressão Gênica , Síndrome de Werner/genética , Envelhecimento/metabolismo , Envelhecimento/patologia , Ciclo Celular/genética , Divisão Celular/genética , Linhagem Celular , DNA/metabolismo , Etiquetas de Sequências Expressas , Perfilação da Expressão Gênica , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Oncogenes , RNA/metabolismo , Recombinação Genética , Transcrição Gênica , Síndrome de Werner/metabolismo , Síndrome de Werner/patologia
20.
Mol Cell ; 14(6): 763-74, 2004 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-15200954

RESUMO

Werner syndrome (WS) is characterized by features of premature aging and is caused by loss of the RecQ helicase protein WRN. WS fibroblasts display defects associated with telomere dysfunction, including accelerated telomere erosion and premature senescence. In yeast, RecQ helicases act in an alternative pathway for telomere lengthening (ALT) via homologous recombination. We found that WRN associates with telomeres when dissociation of telomeric D loops is likely during replication and recombination. In human ALT cells, WRN associates directly with telomeric DNA. The majority of TRF1/PCNA colocalizing foci contained WRN in live S phase ALT cells but not in telomerase-positive HeLa cells. Biochemically, the WRN helicase and 3' to 5' exonuclease act simultaneously and cooperate to release the 3' invading tail from a telomeric D loop in vitro. The telomere binding proteins TRF1 and TRF2 limit digestion by WRN. We propose roles for WRN in dissociating telomeric structures in telomerase-deficient cells.


Assuntos
DNA Helicases/metabolismo , Exonucleases/metabolismo , Telômero/química , Proteína 1 de Ligação a Repetições Teloméricas/metabolismo , Proteína 2 de Ligação a Repetições Teloméricas/metabolismo , Linhagem Celular Tumoral , DNA Helicases/análise , Exodesoxirribonucleases , Células HeLa , Humanos , Antígeno Nuclear de Célula em Proliferação/química , Antígeno Nuclear de Célula em Proliferação/fisiologia , Estrutura Terciária de Proteína , RecQ Helicases , Fase S , Proteína 1 de Ligação a Repetições Teloméricas/análise , Helicase da Síndrome de Werner
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA