Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 11(1): 12357, 2021 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-34117297

RESUMEN

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.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/genética , Envejecimiento/genética , Fertilidad/genética , RecQ Helicasas/genética , Proteína p53 Supresora de Tumor/genética , Proteína de la Poliposis Adenomatosa del Colon/deficiencia , Animales , Femenino , Pleiotropía Genética , Masculino , Ratones , Ratones Endogámicos C57BL , RecQ Helicasas/deficiencia , Proteína p53 Supresora de Tumor/deficiencia
2.
Sci Rep ; 11(1): 2157, 2021 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-33495511

RESUMEN

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.


Asunto(s)
Síndrome de Bloom/enzimología , Síndrome de Bloom/patología , Mitocondrias/metabolismo , Estrés Oxidativo , RecQ Helicasas/deficiencia , Autofagia , Ciclina B1/metabolismo , Daño del ADN , Replicación del ADN , Proteínas de Unión al ADN/metabolismo , Metabolismo Energético , Fibroblastos/enzimología , Fibroblastos/patología , Fase G1 , Humanos , Mitocondrias/ultraestructura , Proteínas Mitocondriales/metabolismo , Mitosis , Especies Reactivas de Oxígeno/metabolismo , RecQ Helicasas/metabolismo , Factores de Transcripción/metabolismo , Regulación hacia Arriba
3.
PLoS Genet ; 15(2): e1007942, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30735491

RESUMEN

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.


Asunto(s)
Daño del ADN , Ligasas/metabolismo , Mitosis , Roturas del ADN de Doble Cadena , Reparación del ADN , Replicación del ADN , Epistasis Genética , Inestabilidad Genómica , Células HEK293 , Células HeLa , Humanos , Hidroxiurea/farmacología , Ligasas/deficiencia , Ligasas/genética , Modelos Biológicos , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo , RecQ Helicasas/deficiencia , RecQ Helicasas/genética , RecQ Helicasas/metabolismo , Intercambio de Cromátides Hermanas/efectos de los fármacos , Sumoilación
4.
Biochem Biophys Res Commun ; 509(2): 379-383, 2019 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-30594395

RESUMEN

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.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/genética , Reparación del ADN , ADN/genética , RecQ Helicasas/genética , Síndrome Rothmund-Thomson/genética , Ácido Anhídrido Hidrolasas , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Quinasa de Punto de Control 2/genética , Quinasa de Punto de Control 2/metabolismo , ADN/metabolismo , Roturas del ADN de Doble Cadena , Enzimas Reparadoras del ADN/genética , Enzimas Reparadoras del ADN/metabolismo , Replicación del ADN , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica , Prueba de Complementación Genética , Humanos , Proteína Homóloga de MRE11/genética , Proteína Homóloga de MRE11/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Unión Proteica , RecQ Helicasas/deficiencia , Síndrome Rothmund-Thomson/metabolismo , Síndrome Rothmund-Thomson/patología , Transducción de Señal , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
5.
Cell Cycle ; 17(7): 881-891, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29620483

RESUMEN

'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.


Asunto(s)
Enzimas Reparadoras del ADN/genética , Reparación del ADN/efectos de los fármacos , Exodesoxirribonucleasas/genética , Puntos de Control de la Fase G2 del Ciclo Celular/genética , RecQ Helicasas/genética , Proteína 1 de Unión al Supresor Tumoral P53/genética , Animales , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Linfocitos B/patología , Linfocitos B/efectos de la radiación , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Proteína BRCA2/genética , Proteína BRCA2/metabolismo , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , ADN/genética , ADN/metabolismo , Roturas del ADN de Doble Cadena/efectos de la radiación , Enzimas Reparadoras del ADN/deficiencia , Exodesoxirribonucleasas/deficiencia , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de la radiación , Rayos gamma , Eliminación de Gen , Expresión Génica , Inestabilidad Genómica , Humanos , Ratones , Ratones Noqueados , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Cultivo Primario de Células , RecQ Helicasas/deficiencia , Intercambio de Cromátides Hermanas , Proteína 1 de Unión al Supresor Tumoral P53/deficiencia
6.
Biomacromolecules ; 19(3): 803-815, 2018 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-29451980

RESUMEN

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.


Asunto(s)
Antineoplásicos , Materiales Biocompatibles Revestidos , Neoplasias Colorrectales/tratamiento farmacológico , Portadores de Fármacos , Nanopartículas de Magnetita , RecQ Helicasas/deficiencia , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Línea Celular Tumoral , Materiales Biocompatibles Revestidos/química , Materiales Biocompatibles Revestidos/farmacocinética , Materiales Biocompatibles Revestidos/farmacología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Portadores de Fármacos/farmacología , Humanos , Nanopartículas de Magnetita/química , Nanopartículas de Magnetita/uso terapéutico
7.
J Cell Biol ; 216(11): 3521-3534, 2017 11 06.
Artículo en Inglés | MEDLINE | ID: mdl-28912125

RESUMEN

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.


Asunto(s)
Roturas del ADN de Doble Cadena , Linfocitos/enzimología , Neoplasias/enzimología , Recombinasa Rad51/metabolismo , RecQ Helicasas/metabolismo , Reparación del ADN por Recombinación , Animales , Proteína BRCA1/genética , Proteína BRCA1/metabolismo , Proteína BRCA2/deficiencia , Proteína BRCA2/genética , Línea Celular Tumoral , Supervivencia Celular , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Inestabilidad Genómica , Genotipo , Humanos , Linfocitos/patología , Ratones Noqueados , Mutación , Neoplasias/genética , Neoplasias/patología , Fenotipo , Estabilidad Proteica , Interferencia de ARN , Recombinasa Rad51/genética , RecQ Helicasas/deficiencia , RecQ Helicasas/genética , Transfección , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53/deficiencia , Proteína 1 de Unión al Supresor Tumoral P53/genética
8.
Mol Cell Biol ; 36(23): 2877-2889, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27601585

RESUMEN

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.


Asunto(s)
Síndrome de Bloom/genética , Reparación del ADN , Pérdida de Heterocigocidad , RecQ Helicasas/deficiencia , Línea Celular , Roturas del ADN de Doble Cadena , Reparación del ADN por Unión de Extremidades , Conversión Génica , Inestabilidad Genómica , Recombinación Homóloga , Humanos
9.
DNA Repair (Amst) ; 41: 73-84, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27100209

RESUMEN

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.


Asunto(s)
Cromosomas Humanos/genética , Recombinación Homóloga , RecQ Helicasas/deficiencia , RecQ Helicasas/genética , Secuencia de Bases , Síndrome de Bloom/genética , Síndrome de Bloom/patología , Línea Celular , Roturas del ADN de Doble Cadena , Reparación del ADN , Humanos , Mutación
10.
World J Gastroenterol ; 21(36): 10375-84, 2015 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-26420964

RESUMEN

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.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/prevención & control , Galactosamina , Hepatocitos/enzimología , Lipopolisacáridos , Hígado/enzimología , RecQ Helicasas/metabolismo , Animales , Apoptosis , Caspasa 3/metabolismo , Enfermedad Hepática Inducida por Sustancias y Drogas/enzimología , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Sistema Enzimático del Citocromo P-450/genética , Sistema Enzimático del Citocromo P-450/metabolismo , Citocinas/genética , Citocinas/metabolismo , Daño del ADN , Modelos Animales de Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hepatocitos/patología , Mediadores de Inflamación/metabolismo , Hígado/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila , Estrés Oxidativo , Fosforilación , RecQ Helicasas/deficiencia , RecQ Helicasas/genética , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
11.
DNA Repair (Amst) ; 28: 73-82, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25770783

RESUMEN

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.


Asunto(s)
Reparación del ADN , Formaldehído/toxicidad , Inestabilidad Genómica/efectos de los fármacos , RecQ Helicasas/metabolismo , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Puntos de Control del Ciclo Celular , ADN/efectos de los fármacos , ADN/metabolismo , Roturas del ADN de Doble Cadena , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , RecQ Helicasas/deficiencia , Proteína 1 de Unión al Supresor Tumoral P53
12.
Int J Toxicol ; 33(5): 373-81, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25228686

RESUMEN

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.


Asunto(s)
Benzo(a)pireno/toxicidad , Exodesoxirribonucleasas/deficiencia , Exodesoxirribonucleasas/genética , Hidroquinonas/toxicidad , Mutágenos/toxicidad , RecQ Helicasas/deficiencia , RecQ Helicasas/genética , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proliferación Celular/efectos de los fármacos , Roturas del ADN de Doble Cadena/efectos de los fármacos , Daño del ADN , Células HeLa , Humanos , Fosforilación , Interferencia de ARN , Helicasa del Síndrome de Werner
13.
J Clin Invest ; 124(8): 3551-65, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24960165

RESUMEN

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.


Asunto(s)
Hematopoyesis/fisiología , RecQ Helicasas/genética , RecQ Helicasas/metabolismo , Síndrome Rothmund-Thomson/enzimología , Síndrome Rothmund-Thomson/genética , Animales , Apoptosis , Trasplante de Médula Ósea , Daño del ADN , Replicación del ADN , Modelos Animales de Enfermedad , Inestabilidad Genómica , Hematopoyesis/genética , Células Madre Hematopoyéticas/enzimología , Células Madre Hematopoyéticas/patología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Madre Multipotentes/enzimología , Células Madre Multipotentes/patología , Mutación , Fenotipo , RecQ Helicasas/deficiencia
14.
Cell Death Dis ; 5: e1226, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24832598

RESUMEN

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.


Asunto(s)
Senescencia Celular , Fibroblastos/enzimología , RecQ Helicasas/metabolismo , Síndrome Rothmund-Thomson/enzimología , Factores de Edad , Envejecimiento/genética , Envejecimiento/metabolismo , Animales , Células de la Médula Ósea/enzimología , Células de la Médula Ósea/patología , Proliferación Celular , Células Cultivadas , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Daño del ADN , Modelos Animales de Enfermedad , Exodesoxirribonucleasas/genética , Exodesoxirribonucleasas/metabolismo , Fibroblastos/patología , Predisposición Genética a la Enfermedad , Folículo Piloso/enzimología , Folículo Piloso/patología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Estructura Terciaria de Proteína , Interferencia de ARN , RecQ Helicasas/deficiencia , RecQ Helicasas/genética , Síndrome Rothmund-Thomson/genética , Síndrome Rothmund-Thomson/patología , Transfección , Helicasa del Síndrome de Werner
15.
Dis Model Mech ; 7(5): 583-92, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24626990

RESUMEN

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.


Asunto(s)
Huesos/patología , Osteoporosis/patología , Telómero/patología , Adiposidad , Animales , Médula Ósea/patología , Resorción Ósea/patología , Recuento de Células , Modelos Animales de Enfermedad , Humanos , Cinética , Ratones , Mutación/genética , Osteoblastos/metabolismo , Osteoblastos/patología , Osteoclastos/patología , Osteogénesis , Fenotipo , RecQ Helicasas/deficiencia , RecQ Helicasas/metabolismo , Telomerasa/deficiencia , Telomerasa/metabolismo , Helicasa del Síndrome de Werner
16.
Aging Cell ; 13(3): 573-5, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24308646

RESUMEN

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.


Asunto(s)
Daño del ADN/efectos de los fármacos , Exodesoxirribonucleasas/deficiencia , Fibroblastos/efectos de los fármacos , RecQ Helicasas/deficiencia , Sirolimus/farmacología , Ciclo Celular/efectos de los fármacos , Procesos de Crecimiento Celular/efectos de los fármacos , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Síndrome de Werner/genética , Síndrome de Werner/metabolismo , Síndrome de Werner/patología , Helicasa del Síndrome de Werner
17.
Cell Death Differ ; 20(11): 1498-509, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23933816

RESUMEN

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.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Exodesoxirribonucleasas/metabolismo , Proteínas Nucleares/metabolismo , RecQ Helicasas/metabolismo , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas de Ciclo Celular , Senescencia Celular/fisiología , Exodesoxirribonucleasas/deficiencia , Células HCT116 , Células HEK293 , Humanos , Células MCF-7 , Proteína de la Leucemia Promielocítica , RecQ Helicasas/deficiencia , Transducción de Señal , Transfección , Helicasa del Síndrome de Werner , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
18.
PLoS One ; 8(7): e69600, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23894508

RESUMEN

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.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Carcinogénesis/genética , Amplificación de Genes , RecQ Helicasas/genética , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Supervivencia Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Sitios Genéticos/genética , Genoma Humano/genética , Humanos , Proteínas Inhibidoras de la Apoptosis/metabolismo , ARN Interferente Pequeño/genética , RecQ Helicasas/deficiencia , Survivin
19.
Mol Biol Cell ; 23(21): 4273-85, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22973052

RESUMEN

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.


Asunto(s)
Daño del ADN , RecQ Helicasas/metabolismo , Daño del ADN/genética , Reparación del ADN/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación hacia Abajo/genética , Técnicas de Silenciamiento del Gen , Proteínas Fluorescentes Verdes/metabolismo , Células HCT116 , Células HeLa , Humanos , Rayos Láser , Modelos Biológicos , Oxidación-Reducción , Estrés Oxidativo/genética , Poli Adenosina Difosfato Ribosa/metabolismo , RecQ Helicasas/deficiencia , Proteínas Recombinantes de Fusión/metabolismo , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos X
20.
Int J Mol Sci ; 13(5): 5866-5877, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22754337

RESUMEN

p16(Ink4a) is a tumor suppressor and a marker for cellular senescence. Previous studies have shown that p16(Ink4a) plays an important role in the response to DNA damage signals caused by telomere dysfunction. In this study, we crossed Wrn(-/-) and p16(Ink4a-/-) mice to knock out the p16(Ink4a) function in a Wrn null background. Growth curves showed that loss of p16(Ink4a) could rescue the growth barriers that are observed in Wrn(-/-) mouse embryonic fibroblasts (MEFs). By challenging the MEFs with the global genotoxin doxorubicin, we showed that loss of p16(Ink4a) did not dramatically affect the global DNA damage response of Wrn(-/-) MEFs induced by doxorubicin. However, in response to telomere dysfunction initiated by the telomere damaging protein TRF2(ΔBΔM), loss of p16(Ink4a) could partially overcome the DNA damage response by disabling p16(Ink4a) up-regulation and reducing the accumulation of γ-H2AX that is observed in Wrn(-/-) MEFs. Furthermore, in response to TRF2(ΔBΔM) overexpression, Wrn(-/-) MEFs senesced within several passages. In contrast, p16(Ink4a-/-) and p16(Ink4a-/-)Wrn(-/-) MEFs could continuously grow and lose expression of the exogenous TRF2(ΔBΔM) in their late passages. In summary, our data suggest that in the context of telomere dysfunction, loss of p16(Ink4a) function could prevent cells from senescence. These results shed light on the anti-aging strategy through regulation of p16(Ink4a) expression.


Asunto(s)
Senescencia Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Doxorrubicina/farmacología , Fibroblastos/citología , RecQ Helicasas/deficiencia , Telómero/metabolismo , Animales , Células Cultivadas , Inhibidor p16 de la Quinasa Dependiente de Ciclina/deficiencia , Daño del ADN , Embrión de Mamíferos/citología , Fibroblastos/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica , Técnicas de Sustitución del Gen , Ratones , RecQ Helicasas/genética , Proteína 2 de Unión a Repeticiones Teloméricas/genética , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Helicasa del Síndrome de Werner
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...