Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
Cells ; 13(13)2024 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-38994931

RESUMO

James German's work to establish the natural history and cancer risk associated with Bloom syndrome (BS) has had a strong influence on the generation of scientists and clinicians working to understand other RECQ deficiencies and heritable cancer predisposition syndromes. I summarize work by us and others below, inspired by James German's precedents with BS, to understand and compare BS with the other heritable RECQ deficiency syndromes with a focus on Werner syndrome (WS). What we know, unanswered questions and new opportunities are discussed, as are potential ways to treat or modify WS-associated disease mechanisms and pathways.


Assuntos
Síndrome de Bloom , RecQ Helicases , Síndrome de Werner , Humanos , RecQ Helicases/genética , RecQ Helicases/metabolismo , RecQ Helicases/deficiência , Síndrome de Bloom/genética , Síndrome de Werner/genética , História do Século XX
2.
Sci Rep ; 11(1): 12357, 2021 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-34117297

RESUMO

Tumor suppressor genes are involved in maintaining genome integrity during reproduction (e.g., meiosis). Thus, deleterious alleles in tumor suppressor-deficient mice would exhibit higher mortality during the perinatal period. A recent aging model proposes that perinatal mortality and age-related deleterious changes might define lifespan. This study aimed to quantitatively understand the relationship between reproduction and lifespan using three established tumor suppressor gene (p53, APC, and RECQL4)-deficient mouse strains with the same C57BL/6 background. Transgenic mice delivered slightly reduced numbers of 1st pups than wild-type mice [ratio: 0.81-0.93 (p = 0.1-0.61)] during a similar delivery period, which suggest that the tumor suppressor gene-deficient mice undergo relatively stable reproduction. However, the transgenic 1st pups died within a few days after birth, resulting in a further reduction in litter size at 3 weeks after delivery compared with that of wild-type mice [ratio: 0.35-0.68 (p = 0.034-0.24)] without sex differences, although the lifespan was variable. Unexpectedly, the significance of reproductive reduction in transgenic mice was decreased at the 2nd or later delivery. Because mice are easily affected by environmental factors, our data underscore the importance of defining reproductive ability through experiments on aging-related reproduction that can reveal a trade-off between fecundity and aging and identify RECQL4 as a novel pleiotropic gene.


Assuntos
Proteína da Polipose Adenomatosa do Colo/genética , Envelhecimento/genética , Fertilidade/genética , RecQ Helicases/genética , Proteína Supressora de Tumor p53/genética , Proteína da Polipose Adenomatosa do Colo/deficiência , Animais , Feminino , Pleiotropia Genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , RecQ Helicases/deficiência , Proteína Supressora de Tumor p53/deficiência
3.
Sci Rep ; 11(1): 2157, 2021 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-33495511

RESUMO

Bloom Syndrome (BS; OMIM #210900; ORPHA #125) is a rare genetic disorder that is associated with growth deficits, compromised immune system, insulin resistance, genome instability and extraordinary predisposition to cancer. Most efforts thus far have focused on understanding the role of the Bloom syndrome DNA helicase BLM as a recombination factor in maintaining genome stability and suppressing cancer. Here, we observed increased levels of reactive oxygen species (ROS) and DNA base damage in BLM-deficient cells, as well as oxidative-stress-dependent reduction in DNA replication speed. BLM-deficient cells exhibited increased mitochondrial mass, upregulation of mitochondrial transcription factor A (TFAM), higher ATP levels and increased respiratory reserve capacity. Cyclin B1, which acts in complex with cyclin-dependent kinase CDK1 to regulate mitotic entry and associated mitochondrial fission by phosphorylating mitochondrial fission protein Drp1, fails to be fully degraded in BLM-deficient cells and shows unscheduled expression in G1 phase cells. This failure to degrade cyclin B1 is accompanied by increased levels and persistent activation of Drp1 throughout mitosis and into G1 phase as well as mitochondrial fragmentation. This study identifies mitochondria-associated abnormalities in Bloom syndrome patient-derived and BLM-knockout cells and we discuss how these abnormalities may contribute to Bloom syndrome.


Assuntos
Síndrome de Bloom/enzimologia , Síndrome de Bloom/patologia , Mitocôndrias/metabolismo , Estresse Oxidativo , RecQ Helicases/deficiência , Autofagia , Ciclina B1/metabolismo , Dano ao DNA , Replicação do DNA , Proteínas de Ligação a DNA/metabolismo , Metabolismo Energético , Fibroblastos/enzimologia , Fibroblastos/patologia , Fase G1 , Humanos , Mitocôndrias/ultraestrutura , Proteínas Mitocondriais/metabolismo , Mitose , Espécies Reativas de Oxigênio/metabolismo , RecQ Helicases/metabolismo , Fatores de Transcrição/metabolismo , Regulação para Cima
4.
PLoS Genet ; 15(2): e1007942, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30735491

RESUMO

NSMCE2 is an E3 SUMO ligase and a subunit of the SMC5/6 complex that associates with the replication fork and protects against genomic instability. Here, we study the fate of collapsed replication forks generated by prolonged hydroxyurea treatment in human NSMCE2-deficient cells. Double strand breaks accumulate during rescue by converging forks in normal cells but not in NSMCE2-deficient cells. Un-rescued forks persist into mitosis, leading to increased mitotic DNA damage. Excess RAD51 accumulates and persists at collapsed forks in NSMCE2-deficient cells, possibly due to lack of BLM recruitment to stalled forks. Despite failure of BLM to accumulate at stalled forks, NSMCE2-deficient cells exhibit lower levels of hydroxyurea-induced sister chromatid exchange. In cells deficient in both NSMCE2 and BLM, hydroxyurea-induced double strand breaks and sister chromatid exchange resembled levels found in NSCME2-deficient cells. We conclude that the rescue of collapsed forks by converging forks is dependent on NSMCE2.


Assuntos
Dano ao DNA , Ligases/metabolismo , Mitose , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Replicação do DNA , Epistasia Genética , Instabilidade Genômica , Células HEK293 , Células HeLa , Humanos , Hidroxiureia/farmacologia , Ligases/deficiência , Ligases/genética , Modelos Biológicos , Rad51 Recombinase/genética , Rad51 Recombinase/metabolismo , RecQ Helicases/deficiência , RecQ Helicases/genética , RecQ Helicases/metabolismo , Troca de Cromátide Irmã/efeitos dos fármacos , Sumoilação
5.
Biochem Biophys Res Commun ; 509(2): 379-383, 2019 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-30594395

RESUMO

RecQL4 has been shown to be involved in DNA replication and repair, but its role in DNA damage checkpoint pathway has not been reported. Here, we show that RecQL4 plays an important role in the activation of ataxia telangiectasia mutated (ATM)-dependent checkpoint pathway in human cells. Cells depleted with RecQL4 or Rothmund-Thomson syndrome cells showed significant impairment in the activation of ATM and the downstream effector proteins such as checkpoint kinase 2 and p53 after DNA damage. This defect was recovered with the expression of wild type RecQL4 but not any mutant RecQL4 proteins with defective helicase activities. While RecQL4 failed to show any direct interaction with ATM, it stably interacted with the Mre11-Rad50-Nbs1 complex that is essential for the activation of ATM and was localized on the DNA damage foci. Thus, our results suggest that the helicase activity of RecQL4 plays an important role in the activation of ATM-dependent checkpoint pathway against DNA double strand breaks in human cells.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/genética , Reparo do DNA , DNA/genética , RecQ Helicases/genética , Síndrome de Rothmund-Thomson/genética , Hidrolases Anidrido Ácido , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Quinase do Ponto de Checagem 2/genética , Quinase do Ponto de Checagem 2/metabolismo , DNA/metabolismo , Quebras de DNA de Cadeia Dupla , Enzimas Reparadoras do DNA/genética , Enzimas Reparadoras do DNA/metabolismo , Replicação do DNA , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica , Teste de Complementação Genética , Humanos , Proteína Homóloga a MRE11/genética , Proteína Homóloga a MRE11/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Osteoblastos/citologia , Osteoblastos/metabolismo , Ligação Proteica , RecQ Helicases/deficiência , Síndrome de Rothmund-Thomson/metabolismo , Síndrome de Rothmund-Thomson/patologia , Transdução de Sinais , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
6.
Cell Cycle ; 17(7): 881-891, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29620483

RESUMO

'BRCAness' is a term used to describe cancer cells that behave similarly to tumors with BRCA1 or BRCA2 mutations. The BRCAness phenotype is associated with hypersensitivity to chemotherapy agents including PARP inhibitors, which are a promising class of recently-licensed anti-cancer treatments. This hypersensitivity arises because of a deficiency in the homologous recombination (HR) pathway for DNA double-strand break repair. To gain further insight into how genetic modifiers of HR contribute to the BRCAness phenotype, we created a new mouse model of BRCAness by generating mice that are deficient in BLM helicase and the Exo1 exonuclease, which are involved in the early stages of HR. We find that cells lacking BLM and Exo1 exhibit a BRCAness phenotype, with diminished HR, and hypersensitivity to PARP inhibitors. We further tested how 53BP1, an important regulator of HR, affects repair efficiency in our BRCAness model. We find that deletion of 53BP1 can relieve several of the repair deficiencies observed in cells lacking BLM and Exo1, just as it does in cells lacking BRCA1. These results substantiate the importance of BRCAness as a concept for classification of cancer cases, and further clarify the role of 53BP1 in regulation of DNA repair pathway choice in mammalian cells.


Assuntos
Enzimas Reparadoras do DNA/genética , Reparo do DNA/efeitos dos fármacos , Exodesoxirribonucleases/genética , Pontos de Checagem da Fase G2 do Ciclo Celular/genética , RecQ Helicases/genética , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/genética , Animais , Linfócitos B/efeitos dos fármacos , Linfócitos B/metabolismo , Linfócitos B/patologia , Linfócitos B/efeitos da radiação , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Proteína BRCA2/genética , Proteína BRCA2/metabolismo , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , DNA/genética , DNA/metabolismo , Quebras de DNA de Cadeia Dupla/efeitos da radiação , Enzimas Reparadoras do DNA/deficiência , Exodesoxirribonucleases/deficiência , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos da radiação , Raios gama , Deleção de Genes , Expressão Gênica , Instabilidade Genômica , Humanos , Camundongos , Camundongos Knockout , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Cultura Primária de Células , RecQ Helicases/deficiência , Troca de Cromátide Irmã , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/deficiência
7.
Biomacromolecules ; 19(3): 803-815, 2018 03 12.
Artigo em Inglês | MEDLINE | ID: mdl-29451980

RESUMO

Synthetic lethality is a molecular-targeted therapy for selective killing of cancer cells. We exploited a lethal interaction between superoxide dismutase 1 inhibition and Bloom syndrome gene product (BLM) defect for the treatment of colorectal cancer (CRC) cells (HCT 116) with a customized lung cancer screen-1-loaded nanocarrier (LCS-1-NC). The drug LCS-1 has poor aqueous solubility. To overcome its limitations, a customized NC, composed of a magnetite core coated with three polymeric shells, namely, aminocellulose (AC), branched poly(amidoamine), and paraben-PEG, was developed for encapsulating LCS-1. Encapsulation efficiency and drug loading were found to be 74% and 8.2%, respectively. LCS-1-NC exhibited sustained release, with ∼85% of drug release in 24 h. Blank NC (0.5 mg/mL) exhibited cytocompatibility toward normal cells, mainly due to the AC layer. LCS-1-NC demonstrated high killing selectivity (104 times) toward BLM-deficient HCT 116 cells over BLM-proficient HCT 116 cells. Due to enhanced efficacy of the drug using NC, the sensitivity difference for BLM-deficient cells increased to 1.7 times in comparison to that with free LCS-1. LCS-1-NC induced persistent DNA damage and apoptosis, which demonstrates that LCS-1-NC effectively and preferentially killed BLM-deficient CRC cells. This is the first report on the development of a potential drug carrier to improve the therapeutic efficacy of LCS-1 for specific killing of CRC cells having BLM defects.


Assuntos
Antineoplásicos , Materiais Revestidos Biocompatíveis , Neoplasias Colorretais/tratamento farmacológico , Portadores de Fármacos , Nanopartículas de Magnetita , RecQ Helicases/deficiência , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Materiais Revestidos Biocompatíveis/química , Materiais Revestidos Biocompatíveis/farmacocinética , Materiais Revestidos Biocompatíveis/farmacologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Portadores de Fármacos/farmacologia , Humanos , Nanopartículas de Magnetita/química , Nanopartículas de Magnetita/uso terapêutico
8.
J Cell Biol ; 216(11): 3521-3534, 2017 11 06.
Artigo em Inglês | MEDLINE | ID: mdl-28912125

RESUMO

The BLM gene product, BLM, is a RECQ helicase that is involved in DNA replication and repair of DNA double-strand breaks by the homologous recombination (HR) pathway. During HR, BLM has both pro- and anti-recombinogenic activities, either of which may contribute to maintenance of genomic integrity. We find that in cells expressing a mutant version of BRCA1, an essential HR factor, ablation of BLM rescues genomic integrity and cell survival in the presence of DNA double-strand breaks. Improved genomic integrity in these cells is linked to a substantial increase in the stability of RAD51 at DNA double-strand break sites and in the overall efficiency of HR. Ablation of BLM also rescues RAD51 foci and HR in cells lacking BRCA2 or XRCC2. These results indicate that the anti-recombinase activity of BLM is of general importance for normal retention of RAD51 at DNA break sites and regulation of HR.


Assuntos
Quebras de DNA de Cadeia Dupla , Linfócitos/enzimologia , Neoplasias/enzimologia , Rad51 Recombinase/metabolismo , RecQ Helicases/metabolismo , Reparo de DNA por Recombinação , Animais , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Proteína BRCA2/deficiência , Proteína BRCA2/genética , Linhagem Celular Tumoral , Sobrevivência Celular , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Instabilidade Genômica , Genótipo , Humanos , Linfócitos/patologia , Camundongos Knockout , Mutação , Neoplasias/genética , Neoplasias/patologia , Fenótipo , Estabilidade Proteica , Interferência de RNA , Rad51 Recombinase/genética , RecQ Helicases/deficiência , RecQ Helicases/genética , Transfecção , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/deficiência , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/genética
9.
Mol Cell Biol ; 36(23): 2877-2889, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27601585

RESUMO

Bloom syndrome (BS), an autosomal recessive disorder of the BLM gene, predisposes sufferers to various cancers. To investigate the mutator phenotype and genetic consequences of DNA double-strand breaks (DSBs) in BS cells, we developed BLM helicase-deficient human cells by disrupting the BLM gene. Cells with a loss of heterozygosity (LOH) due to homologous recombination (HR) or nonhomologous end joining (NHEJ) can be restored with or without site-directed DSB induction. BLM cells exhibited a high frequency of spontaneous interallelic HR with crossover, but noncrossover events with long-tract gene conversions also occurred. Despite the highly interallelic HR events, BLM cells predominantly produced hemizygous LOH by spontaneous deletion. These phenotypes manifested during repair of DSBs. Both NHEJ and HR appropriately repaired DSBs in BLM cells, resulting in hemizygous and homozygous LOHs, respectively. However, the magnitude of the LOH was exacerbated in BLM cells, as evidenced by large deletions and long-tract gene conversions with crossover. BLM helicase suppresses the elongation of branch migration and crossover of double Holliday junctions (HJs) during HR repair, and a deficiency in this enzyme causes collapse, abnormal elongation, and/or preferable resolution to crossover of double HJs, resulting in a large-scale LOH. This mechanism underlies the predisposition for cancer in BS.


Assuntos
Síndrome de Bloom/genética , Reparo do DNA , Perda de Heterozigosidade , RecQ Helicases/deficiência , Linhagem Celular , Quebras de DNA de Cadeia Dupla , Reparo do DNA por Junção de Extremidades , Conversão Gênica , Instabilidade Genômica , Recombinação Homóloga , Humanos
10.
DNA Repair (Amst) ; 41: 73-84, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27100209

RESUMO

Mutation of Bloom helicase (BLM) causes Bloom syndrome (BS), a rare human genetic disorder associated with genome instability, elevation of sister chromatid exchanges, and predisposition to cancer. Deficiency in BLM homologs in Drosophila and yeast brings about significantly increased rates of recombination between imperfectly matched sequences ("homeologous recombination," or HeR). To assess whether BLM deficiency provokes an increase in HeR in human cells, we transfected an HeR substrate into a BLM-null cell line derived from a BS patient. The substrate contained a thymidine kinase (tk)-neo fusion gene disrupted by the recognition site for endonuclease I-SceI, as well as a functional tk gene to serve as a potential recombination partner for the tk-neo gene. The two tk sequences on the substrate displayed 19% divergence. A double-strand break was introduced by expression of I-SceI and repair events were recovered by selection for G418-resistant clones. Among 181 events recovered, 30 were accomplished via HeR with the balance accomplished by nonhomologous end-joining. The frequency of HeR events in the BS cells was elevated significantly compared to that seen in normal human fibroblasts or in BS cells complemented for BLM expression. We conclude that BLM deficiency enables HeR in human cells.


Assuntos
Cromossomos Humanos/genética , Recombinação Homóloga , RecQ Helicases/deficiência , RecQ Helicases/genética , Sequência de Bases , Síndrome de Bloom/genética , Síndrome de Bloom/patologia , Linhagem Celular , Quebras de DNA de Cadeia Dupla , Reparo do DNA , Humanos , Mutação
11.
World J Gastroenterol ; 21(36): 10375-84, 2015 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-26420964

RESUMO

AIM: To investigate the effects of Recql5 deficiency on liver injury induced by lipopolysaccharide/D-galactosamine (LPS/D-Gal). METHODS: Liver injury was induced in wild type (WT) or Recql5-deficient mice using LPS/D-Gal, and assessed by histological, serum transaminases, and mortality analyses. Hepatocellular apoptosis was quantified by transferase dUTP nick end labeling assay and Western blot analysis of cleaved caspase-3. Liver inflammatory chemokine and cytochrome P450 expression was analyzed by quantitative reverse transcription-PCR. Neutrophil infiltration was evaluated by myeloperoxidase activity. Expression and phosphorylation of ERK, JNK, p65, and H2A.X was determined by Western blot. Oxidative stress was evaluated by measuring malondialdehyde production and nitric oxide synthase, superoxide dismutase, glutathione peroxidase, catalase, and glutathione reductase activity. RESULTS: Following LPS/D-Gal exposure, Recql5-deficient mice exhibited enhanced liver injury, as evidenced by more severe hepatic hemorrhage, higher serum aspartate transaminase and alanine transaminase levels, and lower survival rate. As compared to WT mice, Recql5-deficient mice showed an increased number of apoptotic hepatocytes and higher cleaved caspase-3 levels. Recql5-deficient mice exhibited increased DNA damage, as evidenced by increased γ-H2A.X levels. Inflammatory cytokine levels, neutrophil infiltration, and ERK phosphorylation were also significantly increased in the knockout mice. Additionally, Recql5-deficient mice exhibited increased malondialdehyde production and elevated inducible nitric oxide synthase, superoxide dismutase, glutathione peroxidase, catalase, and glutathione reductase activity, indicative of enhanced oxidative stress. Moreover, CYP450 expression was significantly downregulated in Recql5-deficient mice after LPS/D-Gal treatment. CONCLUSION: Recql5 protects the liver against LPS/D-Gal-induced injury through suppression of hepatocyte apoptosis and oxidative stress and modulation of CYP450 expression.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Galactosamina , Hepatócitos/enzimologia , Lipopolissacarídeos , Fígado/enzimologia , RecQ Helicases/metabolismo , Animais , Apoptose , Caspase 3/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/enzimologia , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Doença Hepática Induzida por Substâncias e Drogas/genética , Doença Hepática Induzida por Substâncias e Drogas/patologia , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/metabolismo , Citocinas/genética , Citocinas/metabolismo , Dano ao DNA , Modelos Animais de Doenças , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Hepatócitos/patologia , Mediadores da Inflamação/metabolismo , Fígado/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infiltração de Neutrófilos , Estresse Oxidativo , Fosforilação , RecQ Helicases/deficiência , RecQ Helicases/genética , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
12.
DNA Repair (Amst) ; 28: 73-82, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25770783

RESUMO

Formaldehyde is a reactive aldehyde that has been classified as a class I human carcinogen by the International Agency for Cancer Research. There are growing concerns over the possible adverse health effects related to the occupational and environmental human exposures to formaldehyde. Although formaldehyde-induced DNA and protein adducts have been identified, the genomic instability mechanisms and the cellular tolerance pathways associated with formaldehyde exposure are not fully characterized. This study specifically examines the role of a genome stability protein, Bloom (BLM) in limiting formaldehyde-induced cellular and genetic abnormalities. Here, we show that in the absence of BLM protein, formaldehyde-treated cells exhibited increased cellular sensitivity, an immediate cell cycle arrest, and an accumulation of chromosome radial structures. In addition, live-cell imaging experiments demonstrated that formaldehyde-treated cells are dependent on BLM for timely segregation of daughter cells. Both wild-type and BLM-deficient formaldehyde-treated cells showed an accumulation of 53BP1 and γH2AX foci indicative of DNA double-strand breaks (DSBs); however, relative to wild-type cells, the BLM-deficient cells exhibited delayed repair of formaldehyde-induced DSBs. In response to formaldehyde exposure, we observed co-localization of 53BP1 and BLM foci at the DSB repair site, where ATM-dependent accumulation of formaldehyde-induced BLM foci occurred after the recruitment of 53BP1. Together, these findings highlight the significance of functional interactions among ATM, 53BP1, and BLM proteins as responders associated with the repair and tolerance mechanisms induced by formaldehyde.


Assuntos
Reparo do DNA , Formaldeído/toxicidade , Instabilidade Genômica/efeitos dos fármacos , RecQ Helicases/metabolismo , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Pontos de Checagem do Ciclo Celular , DNA/efeitos dos fármacos , DNA/metabolismo , Quebras de DNA de Cadeia Dupla , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , RecQ Helicases/deficiência , Proteína 1 de Ligação à Proteína Supressora de Tumor p53
13.
Int J Toxicol ; 33(5): 373-81, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25228686

RESUMO

The 5 known RecQ helicases in humans (RECQ1, BLM, WRN, RECQL4, and RECQ5) have demonstrated roles in diverse genome maintenance mechanisms but their functions in safeguarding the genome from environmental toxicants are poorly understood. Here, we have evaluated a potential role of WRN (mutated in Werner syndrome) and RECQ1 (the most abundant homolog of WRN) in hydroquinone (HQ)- and benzo[a]pyrene (BaP)-induced genotoxicity. Silencing of WRN or RECQ1 expression in HeLa cells increased their sensitivity to HQ and BaP but elicited distinct DNA damage response. The RECQ1-depleted cells exhibited increased replication protein A phosphorylation, Chk1 activation, and DNA double-strand breaks (DSBs) as compared to control or WRN-depleted cells following exposure to BaP treatment. The BaP-induced DSBs in RECQ1-depleted cells were dependent on DNA-dependent protein kinase activity. Notably, loss of WRN in RECQ1-depleted cells ameliorated BaP toxicity. Collectively, our results provide first indication of nonredundant participation of WRN and RECQ1 in protection from the potentially carcinogenic effects of BaP and HQ.


Assuntos
Benzo(a)pireno/toxicidade , Exodesoxirribonucleases/deficiência , Exodesoxirribonucleases/genética , Hidroquinonas/toxicidade , Mutagênicos/toxicidade , RecQ Helicases/deficiência , RecQ Helicases/genética , Proteínas Mutadas de Ataxia Telangiectasia/genética , Proliferação de Células/efeitos dos fármacos , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Dano ao DNA , Células HeLa , Humanos , Fosforilação , Interferência de RNA , Helicase da Síndrome de Werner
14.
J Clin Invest ; 124(8): 3551-65, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24960165

RESUMO

Mutations within the gene encoding the DNA helicase RECQL4 underlie the autosomal recessive cancer-predisposition disorder Rothmund-Thomson syndrome, though it is unclear how these mutations lead to disease. Here, we demonstrated that somatic deletion of Recql4 causes a rapid bone marrow failure in mice that involves cells from across the myeloid, lymphoid, and, most profoundly, erythroid lineages. Apoptosis was markedly elevated in multipotent progenitors lacking RECQL4 compared with WT cells. While the stem cell compartment was relatively spared in RECQL4-deficent mice, HSCs from these animals were not transplantable and even selected against. The requirement for RECQL4 was intrinsic in hematopoietic cells, and loss of RECQL4 in these cells was associated with increased replicative DNA damage and failed cell-cycle progression. Concurrent deletion of p53, which rescues loss of function in animals lacking the related helicase BLM, did not rescue BM phenotypes in RECQL4-deficient animals. In contrast, hematopoietic defects in cells from Recql4Δ/Δ mice were fully rescued by a RECQL4 variant without RecQ helicase activity, demonstrating that RECQL4 maintains hematopoiesis independently of helicase activity. Together, our data indicate that RECQL4 participates in DNA replication rather than genome stability and identify RECQL4 as a regulator of hematopoiesis with a nonredundant role compared with other RecQ helicases.


Assuntos
Hematopoese/fisiologia , RecQ Helicases/genética , RecQ Helicases/metabolismo , Síndrome de Rothmund-Thomson/enzimologia , Síndrome de Rothmund-Thomson/genética , Animais , Apoptose , Transplante de Medula Óssea , Dano ao DNA , Replicação do DNA , Modelos Animais de Doenças , Instabilidade Genômica , Hematopoese/genética , Células-Tronco Hematopoéticas/enzimologia , Células-Tronco Hematopoéticas/patologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células-Tronco Multipotentes/enzimologia , Células-Tronco Multipotentes/patologia , Mutação , Fenótipo , RecQ Helicases/deficiência
15.
Cell Death Dis ; 5: e1226, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24832598

RESUMO

Cellular senescence refers to irreversible growth arrest of primary eukaryotic cells, a process thought to contribute to aging-related degeneration and disease. Deficiency of RecQ helicase RECQL4 leads to Rothmund-Thomson syndrome (RTS), and we have investigated whether senescence is involved using cellular approaches and a mouse model. We first systematically investigated whether depletion of RECQL4 and the other four human RecQ helicases, BLM, WRN, RECQL1 and RECQL5, impacts the proliferative potential of human primary fibroblasts. BLM-, WRN- and RECQL4-depleted cells display increased staining of senescence-associated ß-galactosidase (SA-ß-gal), higher expression of p16(INK4a) or/and p21(WAF1) and accumulated persistent DNA damage foci. These features were less frequent in RECQL1- and RECQL5-depleted cells. We have mapped the region in RECQL4 that prevents cellular senescence to its N-terminal region and helicase domain. We further investigated senescence features in an RTS mouse model, Recql4-deficient mice (Recql4(HD)). Tail fibroblasts from Recql4(HD) showed increased SA-ß-gal staining and increased DNA damage foci. We also identified sparser tail hair and fewer blood cells in Recql4(HD) mice accompanied with increased senescence in tail hair follicles and in bone marrow cells. In conclusion, dysfunction of RECQL4 increases DNA damage and triggers premature senescence in both human and mouse cells, which may contribute to symptoms in RTS patients.


Assuntos
Senescência Celular , Fibroblastos/enzimologia , RecQ Helicases/metabolismo , Síndrome de Rothmund-Thomson/enzimologia , Fatores Etários , Envelhecimento/genética , Envelhecimento/metabolismo , Animais , Células da Medula Óssea/enzimologia , Células da Medula Óssea/patologia , Proliferação de Células , Células Cultivadas , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Dano ao DNA , Modelos Animais de Doenças , Exodesoxirribonucleases/genética , Exodesoxirribonucleases/metabolismo , Fibroblastos/patologia , Predisposição Genética para Doença , Folículo Piloso/enzimologia , Folículo Piloso/patologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Estrutura Terciária de Proteína , Interferência de RNA , RecQ Helicases/deficiência , RecQ Helicases/genética , Síndrome de Rothmund-Thomson/genética , Síndrome de Rothmund-Thomson/patologia , Transfecção , Helicase da Síndrome de Werner
16.
Dis Model Mech ; 7(5): 583-92, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24626990

RESUMO

A major medical challenge in the elderly is osteoporosis and the high risk of fracture. Telomere dysfunction is a cause of cellular senescence and telomere shortening, which occurs with age in cells from most human tissues, including bone. Telomere defects contribute to the pathogenesis of two progeroid disorders characterized by premature osteoporosis, Werner syndrome and dyskeratosis congenital. It is hypothesized that telomere shortening contributes to bone aging. We evaluated the skeletal phenotypes of mice with disrupted telomere maintenance mechanisms as models for human bone aging, including mutants in Werner helicase (Wrn(-/-)), telomerase (Terc(-/-)) and Wrn(-/-)Terc(-/-) double mutants. Compared with young wild-type (WT) mice, micro-computerized tomography analysis revealed that young Terc(-/-) and Wrn(-/-)Terc(-/-) mice have decreased trabecular bone volume, trabecular number and trabecular thickness, as well as increased trabecular spacing. In cortical bone, young Terc(-/-) and Wrn(-/-)Terc(-/-) mice have increased cortical thinning, and increased porosity relative to age-matched WT mice. These trabecular and cortical changes were accelerated with age in Terc(-/-) and Wrn(-/-)Terc(-/-) mice compared with older WT mice. Histological quantification of osteoblasts in aged mice showed a similar number of osteoblasts in all genotypes; however, significant decreases in osteoid, mineralization surface, mineral apposition rate and bone formation rate in older Terc(-/-) and Wrn(-/-)Terc(-/-) bone suggest that osteoblast dysfunction is a prominent feature of precocious aging in these mice. Except in the Wrn(-/-) single mutant, osteoclast number did not increase in any genotype. Significant alterations in mechanical parameters (structure model index, degree of anistrophy and moment of inertia) of the Terc(-/-) and Wrn(-/-)Terc(-/-) femurs compared with WT mice were also observed. Young Wrn(-/-)Terc(-/-) mice had a statistically significant increase in bone-marrow fat content compared with young WT mice, which remained elevated in aged double mutants. Taken together, our results suggest that Terc(-/-) and Wrn(-/-)Terc(-/-) mutants recapitulate the human bone aging phenotype and are useful models for studying age-related osteoporosis.


Assuntos
Osso e Ossos/patologia , Osteoporose/patologia , Telômero/patologia , Adiposidade , Animais , Medula Óssea/patologia , Reabsorção Óssea/patologia , Contagem de Células , Modelos Animais de Doenças , Humanos , Cinética , Camundongos , Mutação/genética , Osteoblastos/metabolismo , Osteoblastos/patologia , Osteoclastos/patologia , Osteogênese , Fenótipo , RecQ Helicases/deficiência , RecQ Helicases/metabolismo , Telomerase/deficiência , Telomerase/metabolismo , Helicase da Síndrome de Werner
17.
Aging Cell ; 13(3): 573-5, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24308646

RESUMO

Werner syndrome (WS), caused by mutations at the WRN helicase gene, is a progeroid syndrome characterized by multiple features consistent with accelerated aging. Aberrant double-strand DNA damage repair leads to genomic instability and reduced replicative lifespan of somatic cells. We observed increased autophagy in WRN knockdown cells; this was further increased by short-term rapamycin treatment. Long-term rapamycin treatment resulted in improved growth rate, reduced accumulation of DNA damage foci and improved nuclear morphology; autophagy markers were reduced to near-normal levels, possibly due to clearance of damaged proteins. These data suggest that protein aggregation plays a role in the development of WS phenotypes and that the mammalian target of rapamycin complex 1 pathway is a potential therapeutic target of WS.


Assuntos
Dano ao DNA/efeitos dos fármacos , Exodesoxirribonucleases/deficiência , Fibroblastos/efeitos dos fármacos , RecQ Helicases/deficiência , Sirolimo/farmacologia , Ciclo Celular/efeitos dos fármacos , Processos de Crescimento Celular/efeitos dos fármacos , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Síndrome de Werner/genética , Síndrome de Werner/metabolismo , Síndrome de Werner/patologia , Helicase da Síndrome de Werner
18.
Cell Death Differ ; 20(11): 1498-509, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23933816

RESUMO

Werner syndrome (WS) results from dysfunction of the WRN protein, and is associated with premature aging and early death. Here we report that loss of WRN function elicits accumulation of the Yes-associated protein (YAP protein), a major effector of the Hippo tumor suppressor pathway, both experimentally and in WS-derived fibroblasts. YAP upregulation correlates with slower cell proliferation and accelerated senescence, which are partially mediated by the formation of a complex between YAP and the PML protein, whose activity promotes p53 activation. The ATM kinase is necessary for YAP and PML accumulation in WRN-depleted cells. Notably, the depletion of either YAP or PML partially impairs the induction of senescence following WRN loss. Altogether, our findings reveal that loss of WRN activity triggers the activation of an ATM-YAP-PML-p53 axis, thereby accelerating cellular senescence. The latter has features of SASP (senescence-associated secretory phenotype), whose protumorigenic properties are potentiated by YAP, PML and p53 depletion.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Exodesoxirribonucleases/metabolismo , Proteínas Nucleares/metabolismo , RecQ Helicases/metabolismo , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteínas de Ciclo Celular , Senescência Celular/fisiologia , Exodesoxirribonucleases/deficiência , Células HCT116 , Células HEK293 , Humanos , Células MCF-7 , Proteína da Leucemia Promielocítica , RecQ Helicases/deficiência , Transdução de Sinais , Transfecção , Helicase da Síndrome de Werner , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
19.
PLoS One ; 8(7): e69600, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23894508

RESUMO

Breast cancer occur both in hereditary and sporadic forms, and the later one comprises an overwhelming majority of breast cancer cases among women. Numerical and structural alterations involving chromosome 8, with loss of short arm (8p) and gain of long arm (8q), are frequently observed in breast cancer cells and tissues. In this study, we show that most of the human breast tumor cell lines examined display an over representation of 8q24, a chromosomal locus RecQL4 is regionally mapped to, and consequently, a markedly elevated level of RecQL4 expression. An increased RecQL4 mRNA level was also observed in a majority of clinical breast tumor samples (38/43) examined. shRNA-mediated RecQL4 suppression in MDA-MB453 breast cancer cells not only significantly inhibit the in vitro clonogenic survival and in vivo tumorigenicity. Further studies demonstrate that RecQL4 physically interacts with a major survival factor-survivin and its protein level affects survivin expression. Although loss of RecQL4 function due to gene mutations causally linked to occurrence of human RTS with features of premature aging and cancer predisposition, our studies provide the evidence that overexpression of RecQL4 due to gene amplification play a critical role in human breast tumor progression.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Carcinogênese/genética , Amplificação de Genes , RecQ Helicases/genética , Neoplasias da Mama/enzimologia , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Loci Gênicos/genética , Genoma Humano/genética , Humanos , Proteínas Inibidoras de Apoptose/metabolismo , RNA Interferente Pequeno/genética , RecQ Helicases/deficiência , Survivina
20.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...