Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 106
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Genes (Basel) ; 12(12)2021 11 29.
Artículo en Inglés | MEDLINE | ID: mdl-34946868

RESUMEN

RECQL4 is a member of the evolutionarily conserved RecQ family of 3' to 5' DNA helicases. RECQL4 is critical for maintaining genomic stability through its functions in DNA repair, recombination, and replication. Unlike many DNA repair proteins, RECQL4 has unique functions in many of the central DNA repair pathways such as replication, telomere, double-strand break repair, base excision repair, mitochondrial maintenance, nucleotide excision repair, and crosslink repair. Consistent with these diverse roles, mutations in RECQL4 are associated with three distinct genetic diseases, which are characterized by developmental defects and/or cancer predisposition. In this review, we provide an overview of the roles and regulation of RECQL4 during maintenance of genome homeostasis.


Asunto(s)
Inestabilidad Genómica , RecQ Helicasas/fisiología , Animales , Reparación del ADN/fisiología , Replicación del ADN/fisiología , Humanos , Mitocondrias , Proteínas de Saccharomyces cerevisiae/fisiología , Telómero/fisiología , Ubiquitinación
2.
Life Sci ; 265: 118768, 2021 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-33217443

RESUMEN

Non-small cell lung cancer (NSCLC) patients have a lower 5-year survival rate, and the distant tumor metastasis and drug resistance are the main reasons for the high mortality. RECQL5, a member of RecQ helicases family, has been linked to tumorigenesis of various cancers expect NSCLC. In the current study, analysis with the Cancer Genome Atlas (TCGA) dataset showed that the level of RECQL5 was elevated in LUAD (Lung Adenocarcinoma) and LUSC (lung squamous carcinomas), two major subtypes of NSCLC, which was confirmed by immunohistochemistry staining on Tissue array slides. The level of RECQL5 was also elevated in NSCLC cell lines. Further, Kaplan-Meier analysis of TCGA dataset suggested that the up-regulated RECQL5 was associated with poor prognosis of LUAD, but not with that of LUSC. Knockdown of RECQL5 significantly inhibited the invasion and migration of NSCLC cells, and suppressed epithelial-mesenchymal transition (EMT) as indicated by the changes of EMT-related proteins, while overexpression of RECQL5 displayed reverse effects. Lung metastasis was also suppressed by RECQL5 knockdown. Additionally, the addition of Akt inhibitor LY294002 reversed the effects of RECQL5 overexpression on cell migration, invasion and EMT. Moreover, knockdown of RECQL5 increased the apoptosis of cisplatin-resistant A549 cell line (A549/DDP) caused by cisplatin treatment. In summary, RECQL5 contributed to the metastasis of NSCLC and assisted NSCLC cells incompletely response to cisplatin therapy, and could be considered as a biomarker or clinical target for NSCLC.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Cisplatino/uso terapéutico , Neoplasias Pulmonares/metabolismo , RecQ Helicasas/metabolismo , Animales , Western Blotting , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Trasplante de Neoplasias , Reacción en Cadena en Tiempo Real de la Polimerasa , RecQ Helicasas/fisiología
3.
Life Sci Alliance ; 3(10)2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32820027

RESUMEN

Most tumors lack the G1/S phase checkpoint and are insensitive to antigrowth signals. Loss of G1/S control can severely perturb DNA replication as revealed by slow replication fork progression and frequent replication fork stalling. Cancer cells may thus rely on specific pathways that mitigate the deleterious consequences of replication stress. To identify vulnerabilities of cells suffering from replication stress, we performed an shRNA-based genetic screen. We report that the RECQL helicase is specifically essential in replication stress conditions and protects stalled replication forks against MRE11-dependent double strand break (DSB) formation. In line with these findings, knockdown of RECQL in different cancer cells increased the level of DNA DSBs. Thus, RECQL plays a critical role in sustaining DNA synthesis under conditions of replication stress and as such may represent a target for cancer therapy.


Asunto(s)
Reparación del ADN/fisiología , Replicación del ADN/fisiología , RecQ Helicasas/metabolismo , Animales , Línea Celular Tumoral , Estructuras Cromosómicas/metabolismo , ADN , Roturas del ADN de Doble Cadena , Reparación del ADN/genética , Proteínas de Unión al ADN/genética , Inestabilidad Genómica/genética , Humanos , Proteína Homóloga de MRE11/genética , Ratones , ARN Interferente Pequeño/genética , Recombinasa Rad51/genética , RecQ Helicasas/fisiología
4.
J Biol Chem ; 295(27): 8945-8957, 2020 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-32371399

RESUMEN

DNA interstrand crosslink (ICL) repair requires a complex network of DNA damage response pathways. Removal of the ICL lesions is vital, as they are physical barriers to essential DNA processes that require the separation of duplex DNA, such as replication and transcription. The Fanconi anemia (FA) pathway is the principal mechanism for ICL repair in metazoans and is coupled to DNA replication. In Saccharomyces cerevisiae, a vestigial FA pathway is present, but ICLs are predominantly repaired by a pathway involving the Pso2 nuclease, which is hypothesized to use its exonuclease activity to digest through the lesion to provide access for translesion polymerases. However, Pso2 lacks translesion nuclease activity in vitro, and mechanistic details of this pathway are lacking, especially relative to FA. We recently identified the Hrq1 helicase, a homolog of the disease-linked enzyme RecQ-like helicase 4 (RECQL4), as a component of Pso2-mediated ICL repair. Here, using genetic, biochemical, and biophysical approaches, including single-molecule FRET (smFRET)- and gel-based nuclease assays, we show that Hrq1 stimulates the Pso2 nuclease through a mechanism that requires Hrq1 catalytic activity. Importantly, Hrq1 also stimulated Pso2 translesion nuclease activity through a site-specific ICL in vitro We noted that stimulation of Pso2 nuclease activity is specific to eukaryotic RecQ4 subfamily helicases, and genetic and biochemical data suggest that Hrq1 likely interacts with Pso2 through their N-terminal domains. These results advance our understanding of FA-independent ICL repair and establish a role for the RecQ4 helicases in the repair of these detrimental DNA lesions.


Asunto(s)
Reparación del ADN/fisiología , Endodesoxirribonucleasas/metabolismo , RecQ Helicasas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , ADN/metabolismo , Daño del ADN/fisiología , Proteínas de Unión al ADN/metabolismo , RecQ Helicasas/fisiología , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/fisiología
5.
Nucleic Acids Res ; 48(10): 5467-5484, 2020 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-32329774

RESUMEN

Transcription-replication (T-R) conflicts are profound threats to genome integrity. However, whilst much is known about the existence of T-R conflicts, our understanding of the genetic and temporal nature of how cells respond to them is poorly established. Here, we address this by characterizing the early cellular response to transient T-R conflicts (TRe). This response specifically requires the DNA recombination repair proteins BLM and BRCA2 as well as a non-canonical monoubiquitylation-independent function of FANCD2. A hallmark of the TRe response is the rapid co-localization of these three DNA repair factors at sites of T-R collisions. We find that the TRe response relies on basal activity of the ATR kinase, yet it does not lead to hyperactivation of this key checkpoint protein. Furthermore, specific abrogation of the TRe response leads to DNA damage in mitosis, and promotes chromosome instability and cell death. Collectively our findings identify a new role for these well-established tumor suppressor proteins at an early stage of the cellular response to conflicts between DNA transcription and replication.


Asunto(s)
Replicación del ADN , Reparación del ADN por Recombinación , Transcripción Genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteína BRCA2/fisiología , Línea Celular , Supervivencia Celular , Quinasa 9 Dependiente de la Ciclina/metabolismo , ADN/metabolismo , Daño del ADN , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/fisiología , Humanos , Mitosis/genética , Regiones Promotoras Genéticas , ARN/metabolismo , ARN Polimerasa II/metabolismo , Empalme del ARN , RecQ Helicasas/fisiología , Ubiquitinación
6.
Genes (Basel) ; 11(2)2020 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-32098287

RESUMEN

RECQ5 belongs to the RecQ family of DNA helicases. It is conserved from Drosophila to humans and its deficiency results in genomic instability and cancer susceptibility in mice. Human RECQ5 is known for its ability to regulate homologous recombination by disrupting RAD51 nucleoprotein filaments. It also binds to RNA polymerase II (RNAPII) and negatively regulates transcript elongation by RNAPII. Here, we summarize recent studies implicating RECQ5 in the prevention and resolution of transcription-replication conflicts, a major intrinsic source of genomic instability during cancer development.


Asunto(s)
RecQ Helicasas/fisiología , Animales , ADN/genética , ADN/metabolismo , Replicación del ADN , Inestabilidad Genómica , Humanos , RecQ Helicasas/genética , RecQ Helicasas/metabolismo , Transcripción Genética/genética
7.
Mol Cell ; 77(3): 528-541.e8, 2020 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-31759821

RESUMEN

Formation of co-transcriptional R-loops underlies replication fork stalling upon head-on transcription-replication encounters. Here, we demonstrate that RAD51-dependent replication fork reversal induced by R-loops is followed by the restart of semiconservative DNA replication mediated by RECQ1 and RECQ5 helicases, MUS81/EME1 endonuclease, RAD52 strand-annealing factor, the DNA ligase IV (LIG4)/XRCC4 complex, and the non-catalytic subunit of DNA polymerase δ, POLD3. RECQ5 disrupts RAD51 filaments assembled on stalled forks after RECQ1-mediated reverse branch migration, preventing a new round of fork reversal and facilitating fork cleavage by MUS81/EME1. MUS81-dependent DNA breaks accumulate in cells lacking RAD52 or LIG4 upon induction of R-loop formation, suggesting that RAD52 acts in concert with LIG4/XRCC4 to catalyze fork religation, thereby mediating replication restart. The resumption of DNA synthesis after R-loop-associated fork stalling also requires active transcription, the restoration of which depends on MUS81, RAD52, LIG4, and the transcription elongation factor ELL. These findings provide mechanistic insights into transcription-replication conflict resolution.


Asunto(s)
Replicación del ADN/fisiología , Estructuras R-Loop/genética , Recombinasa Rad51/metabolismo , Línea Celular Tumoral , ADN Ligasas/metabolismo , ADN Polimerasa III/metabolismo , Replicación del ADN/genética , Proteínas de Unión al ADN/metabolismo , Endodesoxirribonucleasas/metabolismo , Endonucleasas/genética , Endonucleasas/metabolismo , Células HeLa , Humanos , Estructuras R-Loop/fisiología , Recombinasa Rad51/genética , Recombinasa Rad51/fisiología , Proteína Recombinante y Reparadora de ADN Rad52/metabolismo , RecQ Helicasas/metabolismo , RecQ Helicasas/fisiología , Transcripción Genética/genética
8.
Genes (Basel) ; 10(12)2019 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-31835565

RESUMEN

Genetic information of all organisms is coded in double-stranded DNA. DNA helicases are essential for unwinding this double strand when it comes to replication, repair or transcription of genetic information. In this review, we will focus on what is known about a variety of DNA helicases that are required to ensure genome stability in plants. Due to their sessile lifestyle, plants are especially exposed to harmful environmental factors. Moreover, many crop plants have large and highly repetitive genomes, making them absolutely dependent on the correct interplay of DNA helicases for safeguarding their stability. Although basic features of a number of these enzymes are conserved between plants and other eukaryotes, a more detailed analysis shows surprising peculiarities, partly also between different plant species. This is additionally of high relevance for plant breeding as a number of these helicases are also involved in crossover control during meiosis and influence the outcome of different approaches of CRISPR/Cas based plant genome engineering. Thus, gaining knowledge about plant helicases, their interplay, as well as the manipulation of their pathways, possesses the potential for improving agriculture. In the long run, this might even help us cope with the increasing obstacles of climate change threatening food security in completely new ways.


Asunto(s)
ADN Helicasas/metabolismo , Genoma de Planta/genética , Inestabilidad Genómica/fisiología , ADN/metabolismo , ADN Helicasas/genética , Reparación del ADN/fisiología , Genoma de Planta/fisiología , Inestabilidad Genómica/genética , Meiosis/genética , Plantas/enzimología , RecQ Helicasas/metabolismo , RecQ Helicasas/fisiología , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
9.
Exp Eye Res ; 178: 99-107, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30227115

RESUMEN

Age-related cataract (ARC) is the most common cause of severe visual impairment and blindness. The precise mechanisms of ARC are not completely understood, but it is well accepted that oxidative damage plays an important role in the disease pathogenesis. BLM, the key enzyme of the double-strand break repair (DSBR) pathway, is part of a family of DNA unwinding enzymes and has a crucial role in multiple steps of the DNA recombination, replication and repair processes. We have recently shown that BLM-rs1063147 is initially associated with nuclear ARC in a cross-section study. Therefore, we wanted to study the effects of BLM on ARC progression. In ARC patients, BLM transcription in lens capsules was decreased, so did the BLM protein, and after UVB irradiation, BLM mRNA and protein levels were increased in SRA01/04 cells. Upon silencing BLM in SRA01/04 cells and rat lens, cell vitality and apoptosis were altered, and the rat lens opacification was considerable. In conclusion, BLM can regulate cataract progression by influencing cell vitality and apoptosis.


Asunto(s)
Apoptosis , Catarata/fisiopatología , Células Epiteliales/fisiología , Cápsula del Cristalino/fisiopatología , RecQ Helicasas/fisiología , Animales , Western Blotting , Catarata/metabolismo , Supervivencia Celular/fisiología , Células Cultivadas , Progresión de la Enfermedad , Células Epiteliales/efectos de la radiación , Citometría de Flujo , Silenciador del Gen , Humanos , Etiquetado Corte-Fin in Situ , Cápsula del Cristalino/metabolismo , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Transfección , Rayos Ultravioleta
10.
Nucleic Acids Res ; 46(16): 8500-8515, 2018 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-30053104

RESUMEN

Most RecQ DNA helicases share a conserved domain arrangement that mediates their activities in genomic stability. This arrangement comprises a helicase motor domain, a RecQ C-terminal (RecQ-C) region including a winged-helix (WH) domain, and a 'Helicase and RNase D C-terminal' (HRDC) domain. Single-molecule real-time translocation and DNA unwinding by full-length Escherichia coli RecQ and variants lacking either the HRDC or both the WH and HRDC domains was analyzed. RecQ operated under two interconvertible kinetic modes, 'slow' and 'normal', as it unwound duplex DNA and translocated on single-stranded (ss) DNA. Consistent with a crystal structure of bacterial RecQ bound to ssDNA by base stacking, abasic sites blocked RecQ unwinding. Removal of the HRDC domain eliminates the slow mode while preserving the normal mode of activity. Unexpectedly, a RecQ variant lacking both the WH and HRDC domains retains weak helicase activity. The inclusion of E. coli ssDNA-binding protein (SSB) induces a third 'fast' unwinding mode four times faster than the normal RecQ mode and enhances the overall helicase activity (affinity, rate, and processivity). SSB stimulation was, furthermore, observed in the RecQ deletion variants, including the variant missing the WH domain. Our results support a model in which RecQ and SSB have multiple interacting modes.


Asunto(s)
ADN Bacteriano/metabolismo , Proteínas de Unión al ADN/fisiología , Proteínas de Escherichia coli/fisiología , Escherichia coli/enzimología , RecQ Helicasas/fisiología , Eliminación de Gen , Secuencias Invertidas Repetidas , Cinética , Modelos Moleculares , Pinzas Ópticas , Conformación Proteica , Dominios Proteicos , RecQ Helicasas/genética , Imagen Individual de Molécula
11.
Ann Oncol ; 29(4): 903-909, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29452344

RESUMEN

Background: Platinum-based therapy is an effective treatment for a subset of triple-negative breast cancer and ovarian cancer patients. In order to increase response rate and decrease unnecessary use, robust biomarkers that predict response to therapy are needed. Patients and methods: We performed an integrated genomic approach combining differential analysis of gene expression and DNA copy number in sensitive compared with resistant triple-negative breast cancers in two independent neoadjuvant cisplatin-treated cohorts. Functional relevance of significant hits was investigated in vitro by overexpression, knockdown and targeted inhibitor treatment. Results: We identified two genes, the Bloom helicase (BLM) and Fanconi anemia complementation group I (FANCI), that have both increased DNA copy number and gene expression in the platinum-sensitive cases. Increased level of expression of these two genes was also associated with platinum but not with taxane response in ovarian cancer. As a functional validation, we found that overexpression of BLM promotes DNA damage and induces sensitivity to cisplatin but has no effect on paclitaxel sensitivity. Conclusions: A biomarker based on the expression levels of the BLM and FANCI genes is a potential predictor of platinum sensitivity in triple-negative breast cancer and ovarian cancer.


Asunto(s)
Antineoplásicos/uso terapéutico , Daño del ADN , Neoplasias Ováricas/metabolismo , Compuestos de Platino/uso terapéutico , RecQ Helicasas/fisiología , Neoplasias de la Mama Triple Negativas/metabolismo , Femenino , Humanos , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología
12.
EMBO J ; 36(19): 2907-2919, 2017 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-28877996

RESUMEN

Alternative lengthening of telomeres (ALT) is a telomere lengthening pathway that predominates in aggressive tumors of mesenchymal origin; however, the underlying mechanism of telomere synthesis is not fully understood. Here, we show that the BLM-TOP3A-RMI (BTR) dissolvase complex is required for ALT-mediated telomere synthesis. We propose that recombination intermediates formed during strand invasion are processed by the BTR complex, initiating rapid and extensive POLD3-dependent telomere synthesis followed by dissolution, with no overall exchange of telomeric DNA. This process is counteracted by the SLX4-SLX1-ERCC4 complex, which promotes resolution of the recombination intermediate, resulting in telomere exchange in the absence of telomere extension. Our data are consistent with ALT being a conservative DNA replication process, analogous to break-induced replication, which is dependent on BTR and counteracted by SLX4 complex-mediated resolution events.


Asunto(s)
Replicación del ADN/genética , RecQ Helicasas/fisiología , Recombinasas/fisiología , Recombinación Genética/genética , Homeostasis del Telómero/genética , Células Cultivadas , ADN-Topoisomerasas de Tipo I/metabolismo , ADN-Topoisomerasas de Tipo I/fisiología , ADN Polimerasa Dirigida por ADN/metabolismo , ADN Polimerasa Dirigida por ADN/fisiología , Humanos , Complejos Multienzimáticos/metabolismo , Complejos Multienzimáticos/fisiología , RecQ Helicasas/metabolismo , Recombinasas/metabolismo , Telómero/metabolismo
13.
Leukemia ; 31(10): 2104-2113, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28186131

RESUMEN

Multiple myeloma (MM) is a plasma cell cancer with poor survival, characterized by the expansion of multiple myeloma cells (MMCs) in the bone marrow. Using a microarray-based genome-wide screen for genes responding to DNA methyltransferases (DNMT) inhibition in MM cells, we identified RECQ1 among the most downregulated genes. RecQ helicases are DNA unwinding enzymes involved in the maintenance of chromosome stability. Here we show that RECQ1 is significantly overexpressed in MMCs compared to normal plasma cells and that increased RECQ1 expression is associated with poor prognosis in three independent cohorts of patients. Interestingly, RECQ1 knockdown inhibits cells growth and induces apoptosis in MMCs. Moreover, RECQ1 depletion promotes the development of DNA double-strand breaks, as evidenced by the formation of 53BP1 foci and the phosphorylation of ataxia-telangiectasia mutated (ATM) and histone variant H2A.X (H2AX). In contrast, RECQ1 overexpression protects MMCs from melphalan and bortezomib cytotoxicity. RECQ1 interacts with PARP1 in MMCs exposed to treatment and RECQ1 depletion sensitizes MMCs to poly(ADP-ribose) polymerase (PARP) inhibitor. DNMT inhibitor treatment results in RECQ1 downregulation through miR-203 deregulation in MMC. Altogether, these data suggest that association of DNA damaging agents and/or PARP inhibitors with DNMT inhibitors may represent a therapeutic approach in patients with high RECQ1 expression associated with a poor prognosis.


Asunto(s)
ADN de Neoplasias/genética , Resistencia a Antineoplásicos/fisiología , Mieloma Múltiple/enzimología , Proteínas de Neoplasias/fisiología , RecQ Helicasas/fisiología , Bortezomib/farmacología , Ciclo Celular/efectos de los fármacos , Roturas del ADN de Doble Cadena , Daño del ADN , Metilación de ADN/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , ADN de Neoplasias/metabolismo , ADN-Citosina Metilasas/antagonistas & inhibidores , Inducción Enzimática , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Regulación Neoplásica de la Expresión Génica , Humanos , Melfalán/farmacología , MicroARNs/genética , Terapia Molecular Dirigida , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/genética , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/enzimología , Células Plasmáticas/efectos de los fármacos , Células Plasmáticas/enzimología , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética , RecQ Helicasas/antagonistas & inhibidores , RecQ Helicasas/genética , Células Tumorales Cultivadas
14.
Nucleic Acids Res ; 45(5): 2558-2570, 2017 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-27956499

RESUMEN

The DNA replication or S-phase checkpoint monitors the integrity of DNA synthesis. Replication stress or DNA damage triggers fork stalling and checkpoint signaling to activate repair pathways. Recovery from checkpoint activation is critical for cell survival following DNA damage. Recovery from the S-phase checkpoint includes inactivation of checkpoint signaling and restart of stalled replication forks. Previous studies demonstrated that degradation of Mrc1, the Saccharomyces cerevisiae ortholog of human Claspin, is facilitated by the SCFDia2 ubiquitin ligase and is important for cell cycle re-entry after DNA damage-induced S-phase checkpoint activation. Here, we show that degradation of Mrc1 facilitated by the SCFDia2 complex is critical to restart stalled replication forks during checkpoint recovery. Using DNA fiber analysis, we showed that Dia2 functions with the Sgs1 and Mph1 helicases (orthologs of human BLM and FANCM, respectively) in the recombination-mediated fork restart pathway. In addition, Dia2 physically interacts with Sgs1 upon checkpoint activation. Importantly, failure to target Mrc1 for degradation during recovery inhibits Sgs1 chromatin association, but this can be alleviated by induced proteolysis of Mrc1 after checkpoint activation. Together, these studies provide new mechanistic insights into how cells recover from activation of the S-phase checkpoint.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Replicación del ADN , Proteínas F-Box/metabolismo , RecQ Helicasas/metabolismo , Recombinación Genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Puntos de Control del Ciclo Celular , Cromatina/metabolismo , ARN Helicasas DEAD-box/metabolismo , ADN/metabolismo , Proteínas F-Box/química , Proteínas F-Box/fisiología , Dominios Proteicos , RecQ Helicasas/fisiología , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/fisiología
15.
Biophys Chem ; 225: 20-26, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-27876204

RESUMEN

DNA replication is the most vulnerable process of DNA metabolism in proliferating cells and therefore it is tightly controlled and coordinated with processes that maintain genomic stability. Human RecQ helicases are among the most important factors involved in the maintenance of replication fork integrity, especially under conditions of replication stress. RecQ helicases promote recovery of replication forks being stalled due to different replication roadblocks of either exogenous or endogenous source. They prevent generation of aberrant replication fork structures and replication fork collapse, and are involved in proper checkpoint signaling. The essential role of human RecQ helicases in the genome maintenance during DNA replication is underlined by association of defects in their function with cancer predisposition.


Asunto(s)
Replicación del ADN , RecQ Helicasas/fisiología , Inestabilidad Genómica , Humanos , Neoplasias/etiología
16.
J Reprod Dev ; 62(1): 121-5, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26598326

RESUMEN

Production of knockout mice using targeted embryonic stem cells (ESCs) is a powerful approach for investigating the function of specific genes in vivo. Although the protocol for gene targeting via homologous recombination (HR) in ESCs is already well established, the targeting efficiency varies at different target loci and is sometimes too low. It is known that knockdown of the Bloom syndrome gene, BLM, enhances HR-mediated gene targeting efficiencies in various cell lines. However, it has not yet been investigated whether this approach in ESCs is applicable for successful knockout mouse production. Therefore, we attempted to answer this question. Consistent with previous reports, Blm knockdown enhanced gene targeting efficiencies for three gene loci that we examined by 2.3-4.1-fold. Furthermore, the targeted ESC clones generated good chimeras and were successful in germline transmission. These data suggest that Blm knockdown provides a general benefit for efficient ESC-based and HR-mediated knockout mouse production.


Asunto(s)
Células Madre Embrionarias/citología , Técnicas de Silenciamiento del Gen , RecQ Helicasas/genética , RecQ Helicasas/fisiología , Animales , ADN Helicasas/genética , Marcación de Gen , Células Germinativas/metabolismo , Recombinación Homóloga , Cariotipificación , Ratones , Ratones Noqueados , Interferencia de ARN , ARN Interferente Pequeño/metabolismo
18.
J Cell Biol ; 210(2): 177-9, 2015 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-26195663

RESUMEN

Using single molecule analysis of replicated DNA (SMARD), Drosopoulos et al. (2015; J. Cell Biol. http://dx.doi.org/10.1083/jcb.201410061) report that DNA replication initiates at measurable frequency within the telomere of mouse chromosome arm 14q. They demonstrate that resolution of G4 structures on the G-rich template strand of the telomere requires some overlapping functions of BLM and WRN helicase for leading strand synthesis.


Asunto(s)
Replicación del ADN , RecQ Helicasas/fisiología , Telómero/fisiología , Animales
19.
J Cell Biol ; 210(2): 191-208, 2015 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-26195664

RESUMEN

Based on its in vitro unwinding activity on G-quadruplex (G4) DNA, the Bloom syndrome-associated helicase BLM is proposed to participate in telomere replication by aiding fork progression through G-rich telomeric DNA. Single molecule analysis of replicated DNA (SMARD) was used to determine the contribution of BLM helicase to telomere replication. In BLM-deficient cells, replication forks initiating from origins within the telomere, which copy the G-rich strand by leading strand synthesis, moved slower through the telomere compared with the adjacent subtelomere. Fork progression through the telomere was further slowed in the presence of a G4 stabilizer. Using a G4-specific antibody, we found that deficiency of BLM, or another G4-unwinding helicase, the Werner syndrome-associated helicase WRN, resulted in increased G4 structures in cells. Importantly, deficiency of either helicase led to greater increases in G4 DNA detected in the telomere compared with G4 seen genome-wide. Collectively, our findings are consistent with BLM helicase facilitating telomere replication by resolving G4 structures formed during copying of the G-rich strand by leading strand synthesis.


Asunto(s)
Replicación del ADN , RecQ Helicasas/fisiología , Telómero/fisiología , Animales , Células Cultivadas , G-Cuádruplex , Secuencia Rica en GC , Técnicas de Inactivación de Genes , Cinética , Ratones Noqueados , RecQ Helicasas/genética , RecQ Helicasas/metabolismo , Origen de Réplica , Helicasa del Síndrome de Werner
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...