Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Microbiol Spectr ; 11(6): e0531622, 2023 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-37800915

RESUMEN

IMPORTANCE: Biological processes originating from the DNA and RNA can be regulated by the secondary structures present in the stretch of nucleic acids, and the G-quadruplexes are shown to regulate transcription, translation, and replication. In this study, we identified the presence of multiple G-quadruplex sites in the region (oriLyt) of Kaposi's sarcoma-associated herpesvirus (KSHV) DNA, which is essential for DNA replication during the lytic cycle. We demonstrated the roles of these G-quadruplexes through multiple biochemical and biophysical assays in controlling replication and efficient virus production. We demonstrated that KSHV achieves this by recruiting RecQ1 (helicase) at those G-quadruplex sites for efficient viral DNA replication. Analysis of the replicated DNA through nucleoside labeling and immunostaining showed a reduced initiation of DNA replication in cells with a pharmacologic stabilizer of G-quadruplexes. Overall, this study confirmed the role of the G-quadruplex in regulating viral DNA replication, which can be exploited for controlling viral DNA replication.


Asunto(s)
G-Cuádruplex , Herpesvirus Humano 8 , Herpesvirus Humano 8/genética , Replicación Viral/genética , Replicación del ADN , ADN Viral/genética , Regulación Viral de la Expresión Génica
2.
Int J Mol Sci ; 24(18)2023 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-37762489

RESUMEN

Base excision repair (BER) corrects forms of oxidative, deamination, alkylation, and abasic single-base damage that appear to have minimal effects on the helix. Since its discovery in 1974, the field has grown in several facets: mechanisms, biology and physiology, understanding deficiencies and human disease, and using BER genes as potential inhibitory targets to develop therapeutics. Within its segregation of short nucleotide (SN-) and long patch (LP-), there are currently six known global mechanisms, with emerging work in transcription- and replication-associated BER. Knockouts (KOs) of BER genes in mouse models showed that single glycosylase knockout had minimal phenotypic impact, but the effects were clearly seen in double knockouts. However, KOs of downstream enzymes showed critical impact on the health and survival of mice. BER gene deficiency contributes to cancer, inflammation, aging, and neurodegenerative disorders. Medicinal targets are being developed for single or combinatorial therapies, but only PARP and APE1 have yet to reach the clinical stage.


Asunto(s)
Medicina , Humanos , Animales , Ratones , Ratones Noqueados , Envejecimiento , Reparación del ADN , Biología
3.
Front Mol Biosci ; 9: 887758, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35782872

RESUMEN

Named the "caretakers" of the genome, RecQ helicases function in several pathways to maintain genomic stability and repair DNA. This highly conserved family of enzymes consist of five different proteins in humans: RECQL1, BLM, WRN, RECQL4, and RECQL5. Biallelic germline mutations in BLM, WRN, and RECQL4 have been linked to rare cancer-predisposing syndromes. Emerging research has also implicated somatic alterations in RecQ helicases in a variety of cancers, including hematological malignancies, breast cancer, osteosarcoma, amongst others. These alterations in RecQ helicases, particularly overexpression, may lead to increased resistance of cancer cells to conventional chemotherapy. Downregulation of these proteins may allow for increased sensitivity to chemotherapy, and, therefore, may be important therapeutic targets. Here we provide a comprehensive review of our current understanding of the role of RecQ DNA helicases in cancer and discuss the potential therapeutic opportunities in targeting these helicases.

4.
Cell Rep ; 39(9): 110879, 2022 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-35649362

RESUMEN

The MDM2 oncoprotein antagonizes the tumor suppressor p53 by physical interaction and ubiquitination. However, it also sustains the progression of DNA replication forks, even in the absence of functional p53. Here, we show that MDM2 binds, inhibits, ubiquitinates, and destabilizes poly(ADP-ribose) polymerase 1 (PARP1). When cellular MDM2 levels are increased, this leads to accelerated progression of DNA replication forks, much like pharmacological inhibition of PARP1. Conversely, overexpressed PARP1 restores normal fork progression despite elevated MDM2. Strikingly, MDM2 profoundly reduces the frequency of fork reversal, revealed as four-way junctions through electron microscopy. Depletion of RECQ1 or the primase/polymerase (PRIMPOL) reverses the MDM2-mediated acceleration of the nascent DNA elongation rate. MDM2 also increases the occurrence of micronuclei, and it exacerbates camptothecin-induced cell death. In conclusion, high MDM2 levels phenocopy PARP inhibition in modulation of fork restart, representing a potential vulnerability of cancer cells.


Asunto(s)
Replicación del ADN , Proteína p53 Supresora de Tumor , ADN/genética , Daño del ADN , ADN Primasa/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
5.
Methods ; 204: 263-268, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35231585

RESUMEN

DNA helicase RECQ1 (also known as RECQL or RECQL1) is a candidate breast cancer susceptibility gene significantly correlated with clinical outcomes of sporadic breast cancer patients. Prior studies have suggested that RECQ1 maintains genomic stability by regulating a wide variety of core cellular functions including DNA replication, DNA damage response, and transcription. However, it is unclear which, if any, of these are the primary functions of RECQ1 as related to its role in suppressing breast cancer. We describe here an unbiased integrative genomics approach that enabled us to discover a previously unknown regulatory role of RECQ1 in promoting Estrogen Receptor alpha (ERα) expression and the expression of specific ERα target genes in ER positive breast cancer cells. We discuss potential future applications of similar experimental strategies in advancing the mechanistic understanding and elucidating specific new details of genome-wide functions of RECQ1 and other RecQ helicases in maintaining genomic stability and preventing cancer.


Asunto(s)
Neoplasias de la Mama , RecQ Helicasas , Neoplasias de la Mama/genética , Receptor alfa de Estrógeno/genética , Femenino , Predisposición Genética a la Enfermedad , Inestabilidad Genómica , Humanos , RecQ Helicasas/genética
6.
Front Cell Dev Biol ; 9: 714868, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34381789

RESUMEN

Glioblastoma (GBM) is the most common aggressive primary malignant brain tumor, and patients with GBM have a median survival of 20 months. Clinical therapy resistance is a challenging barrier to overcome. Tumor genome stability maintenance during DNA replication, especially the ability to respond to replication stress, is highly correlated with drug resistance. Recently, we identified a protective role for RECQ1 under replication stress conditions. RECQ1 acts at replication forks, binds PCNA, inhibits single-strand DNA formation and nascent strand degradation in GBM cells. It is associated with the function of the PARP1 protein, promoting PARP1 recruitment to replication sites. RECQ1 is essential for DNA replication fork protection and tumor cell proliferation under replication stress conditions, and as a target of RECQ1, PARP1 effectively protects and restarts stalled replication forks, providing new insights into genomic stability maintenance and replication stress resistance. These findings indicate that tumor genome stability targeting RECQ1-PARP1 signaling may be a promising therapeutic intervention to overcome therapy resistance in GBM.

7.
Mol Cell Biol ; 41(4)2021 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-33468559

RESUMEN

Susceptibility to breast cancer is significantly increased in individuals with germ line mutations in RECQ1 (also known as RECQL or RECQL1), a gene encoding a DNA helicase essential for genome maintenance. We previously reported that RECQ1 expression predicts clinical outcomes for sporadic breast cancer patients stratified by estrogen receptor (ER) status. Here, we utilized an unbiased integrative genomics approach to delineate a cross talk between RECQ1 and ERα, a known master regulatory transcription factor in breast cancer. We found that expression of ESR1, the gene encoding ERα, is directly activated by RECQ1. More than 35% of RECQ1 binding sites were cobound by ERα genome-wide. Mechanistically, RECQ1 cooperates with FOXA1, the pioneer transcription factor for ERα, to enhance chromatin accessibility at the ESR1 regulatory regions in a helicase activity-dependent manner. In clinical ERα-positive breast cancers treated with endocrine therapy, high RECQ1 and high FOXA1 coexpressing tumors were associated with better survival. Collectively, these results identify RECQ1 as a novel cofactor for ERα and uncover a previously unknown mechanism by which RECQ1 regulates disease-driving gene expression in ER-positive breast cancer cells.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Estrógenos/metabolismo , RecQ Helicasas/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Receptor alfa de Estrógeno/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Predisposición Genética a la Enfermedad/genética , Factor Nuclear 3-alfa del Hepatocito/genética , Humanos
8.
Curr Genet ; 67(3): 369-382, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33427950

RESUMEN

DNA replication checkpoint is a cell signaling pathway that is activated in response to perturbed replication. Although it is crucial for maintaining genomic integrity and cell survival, the exact mechanism of the checkpoint signaling remains to be understood. Emerging evidence has shown that RecQ helicases, a large family of helicases that are conserved from bacteria to yeasts and humans, contribute to the replication checkpoint as sensors, adaptors, or regulation targets. Here, we highlight the multiple functions of RecQ helicases in the replication checkpoint in four model organisms and present additional evidence that fission yeast RecQ helicase Rqh1 may participate in the replication checkpoint as a sensor.


Asunto(s)
Puntos de Control del Ciclo Celular/genética , ADN Helicasas/genética , Replicación del ADN/genética , RecQ Helicasas/genética , Proteínas de Schizosaccharomyces pombe/genética , Humanos , Schizosaccharomyces/genética , Transducción de Señal/genética
9.
Mol Cell ; 81(4): 784-800.e8, 2021 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-33412112

RESUMEN

DNA replication forks use multiple mechanisms to deal with replication stress, but how the choice of mechanisms is made is still poorly understood. Here, we show that CARM1 associates with replication forks and reduces fork speed independently of its methyltransferase activity. The speeding of replication forks in CARM1-deficient cells requires RECQ1, which resolves reversed forks, and RAD18, which promotes translesion synthesis. Loss of CARM1 reduces fork reversal and increases single-stranded DNA (ssDNA) gaps but allows cells to tolerate higher replication stress. Mechanistically, CARM1 interacts with PARP1 and promotes PARylation at replication forks. In vitro, CARM1 stimulates PARP1 activity by enhancing its DNA binding and acts jointly with HPF1 to activate PARP1. Thus, by stimulating PARP1, CARM1 slows replication forks and promotes the use of fork reversal in the stress response, revealing that CARM1 and PARP1 function as a regulatory module at forks to control fork speed and the choice of stress response mechanisms.


Asunto(s)
Roturas del ADN de Cadena Simple , Replicación del ADN , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Proteína-Arginina N-Metiltransferasas/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Células HEK293 , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/genética , Proteína-Arginina N-Metiltransferasas/genética , RecQ Helicasas/genética , RecQ Helicasas/metabolismo
10.
Cell ; 172(3): 439-453.e14, 2018 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-29290468

RESUMEN

Telomere maintenance critically depends on the distinct activities of telomerase, which adds telomeric repeats to solve the end replication problem, and RTEL1, which dismantles DNA secondary structures at telomeres to facilitate replisome progression. Here, we establish that reversed replication forks are a pathological substrate for telomerase and the source of telomere catastrophe in Rtel1-/- cells. Inhibiting telomerase recruitment to telomeres, but not its activity, or blocking replication fork reversal through PARP1 inhibition or depleting UBC13 or ZRANB3 prevents the rapid accumulation of dysfunctional telomeres in RTEL1-deficient cells. In this context, we establish that telomerase binding to reversed replication forks inhibits telomere replication, which can be mimicked by preventing replication fork restart through depletion of RECQ1 or PARG. Our results lead us to propose that telomerase inappropriately binds to and inhibits restart of reversed replication forks within telomeres, which compromises replication and leads to critically short telomeres.


Asunto(s)
ADN Helicasas/genética , Replicación del ADN , Homeostasis del Telómero , Animales , Línea Celular , Células Cultivadas , ADN Helicasas/metabolismo , Glicósido Hidrolasas/metabolismo , Ratones , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , RecQ Helicasas/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo
11.
Cell Chem Biol ; 24(9): 1101-1119, 2017 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-28938088

RESUMEN

To maintain stable genomes and to avoid cancer and aging, cells need to repair a multitude of deleterious DNA lesions, which arise constantly in every cell. Processes that support genome integrity in normal cells, however, allow cancer cells to develop resistance to radiation and DNA-damaging chemotherapeutics. Chemical inhibition of the key DNA repair proteins and pharmacologically induced synthetic lethality have become instrumental in both dissecting the complex DNA repair networks and as promising anticancer agents. The difficulty in capitalizing on synthetically lethal interactions in cancer cells is that many potential targets do not possess well-defined small-molecule binding determinates. In this review, we discuss several successful campaigns to identify and leverage small-molecule inhibitors of the DNA repair proteins, from PARP1, a paradigm case for clinically successful small-molecule inhibitors, to coveted new targets, such as RAD51 recombinase, RAD52 DNA repair protein, MRE11 nuclease, and WRN DNA helicase.


Asunto(s)
Reparación del ADN , Neoplasias/tratamiento farmacológico , Bibliotecas de Moléculas Pequeñas/química , ADN Helicasas/antagonistas & inhibidores , ADN Helicasas/metabolismo , Reparación del ADN/efectos de los fármacos , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/metabolismo , Humanos , Proteína Homóloga de MRE11 , Neoplasias/patología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Recombinasa Rad51/antagonistas & inhibidores , Recombinasa Rad51/metabolismo , Proteína Recombinante y Reparadora de ADN Rad52/antagonistas & inhibidores , Proteína Recombinante y Reparadora de ADN Rad52/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Bibliotecas de Moléculas Pequeñas/uso terapéutico
12.
EMBO J ; 36(11): 1605-1622, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28373211

RESUMEN

Base excision repair (BER) is one of the most frequently used cellular DNA repair mechanisms and modulates many human pathophysiological conditions related to DNA damage. Through live cell and in vitro reconstitution experiments, we have discovered a major sub-pathway of conventional long-patch BER that involves formation of a 9-nucleotide gap 5' to the lesion. This new sub-pathway is mediated by RECQ1 DNA helicase and ERCC1-XPF endonuclease in cooperation with PARP1 poly(ADP-ribose) polymerase and RPA The novel gap formation step is employed during repair of a variety of DNA lesions, including oxidative and alkylation damage. Moreover, RECQ1 regulates PARP1 auto-(ADP-ribosyl)ation and the choice between long-patch and single-nucleotide BER, thereby modulating cellular sensitivity to DNA damage. Based on these results, we propose a revised model of long-patch BER and a new key regulation point for pathway choice in BER.


Asunto(s)
Reparación del ADN , Proteínas de Unión al ADN/metabolismo , Endonucleasas/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , RecQ Helicasas/metabolismo , Proteína de Replicación A/metabolismo , Línea Celular , ADN/metabolismo , Daño del ADN , Humanos , Modelos Biológicos
13.
Biomed Pharmacother ; 83: 1227-1232, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27565844

RESUMEN

RECQ1, the most abundant one of the human RecQ helicases family, has been identified as a prometastasis gene in breast and cervical cancers. However, the effects of RECQ1 on non-small cell lung cancer (NSCLC) and the underlying molecular mechanisms are still unclear. In the present study, RECQ1 expression (in three NSCLC cell lines and one bronchial epithelial cell line) was detected by real-time quantitative PCR (RT-qPCR). Expression of RECQ1 in A549 cells was knocked down by lentivirus-mediated RNA interference technique (RNAi). The effects of RECQ1 knockdown on cell proliferation, migration and invasion were assessed by Cell Counting Kit-8 (CCK-8) assay and transwell assays. Epithelial-mesenchymal transition (EMT)-associated proteins (E-cadherin, N-cadherin as well as vimentin) were detected by RT-qPCR and western blotting analyses. We found that RECQ1 expression was significantly higher in three NSCLC cell lines than that in a normal human bronchial epithelial cell line. Knocking down RECQ1 significantly suppressed A549 cell proliferation, migration and invasion. The expressions of the epithelial marker, E-cadherin were elevated in both mRNA and protein levels, whereas the expressions of the mesenchymal markers, N-cadherin and vimentin were decreased. Taken together, our findings suggest that RECQ1 may act as an important mediator in promoting lung cancer progression via modulation of the EMT. RECQ1 might represent a potential therapeutic target in NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Silenciador del Gen/fisiología , Neoplasias Pulmonares/genética , RecQ Helicasas/genética , Células A549 , Carcinoma de Pulmón de Células no Pequeñas/enzimología , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Humanos , Neoplasias Pulmonares/enzimología , RecQ Helicasas/metabolismo
14.
Cell Cycle ; 14(22): 3540-3, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26125302

RESUMEN

Identifying and characterizing novel genetic risk factors for BRCA1/2 negative breast cancers is highly relevant for early diagnosis and development of a management plan. Mutations in a number of DNA repair genes have been associated with genomic instability and development of breast and various other cancers. Whole exome sequencing efforts by 2 groups have led to the discovery in distinct populations of multiple breast cancer susceptibility mutations in RECQL, a gene that encodes a DNA helicase involved in homologous recombination repair and response to replication stress. RECQL pathogenic mutations were identified that truncated or disrupted the RECQL protein or introduced missense mutations in its helicase domain. RECQL mutations may serve as a useful biomarker for breast cancer. Targeting RECQL associated tumors with novel DNA repair inhibitors may provide a new strategy for anti-cancer therapy.


Asunto(s)
Neoplasias de la Mama/genética , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Mutación , RecQ Helicasas/genética , Proteína BRCA1/deficiencia , Proteína BRCA1/genética , Proteína BRCA2/deficiencia , Proteína BRCA2/genética , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Exoma , Femenino , Inestabilidad Genómica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Estructura Terciaria de Proteína , RecQ Helicasas/metabolismo , Reparación del ADN por Recombinación , Transducción de Señal
15.
J Biol Chem ; 289(29): 19928-41, 2014 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-24895130

RESUMEN

Understanding how cellular machinery deals with chromosomal genome complexity is an important question because protein bound to DNA may affect various cellular processes of nucleic acid metabolism. DNA helicases are at the forefront of such processes, yet there is only limited knowledge how they remodel protein-DNA complexes and how these mechanisms are regulated. We have determined that representative human RecQ and Fe-S cluster DNA helicases are potently blocked by a protein-DNA interaction. The Fanconi anemia group J (FANCJ) helicase partners with the single-stranded DNA-binding protein replication protein A (RPA) to displace BamHI-E111A bound to duplex DNA in a specific manner. Protein displacement was dependent on the ATPase-driven function of the helicase and unique properties of RPA. Further biochemical studies demonstrated that the shelterin proteins TRF1 and TRF2, which preferentially bind the telomeric repeat found at chromosome ends, effectively block FANCJ from unwinding the forked duplex telomeric substrate. RPA, but not the Escherichia coli single-stranded DNA-binding protein or shelterin factor Pot1, stimulated FANCJ ejection of TRF1 from the telomeric DNA substrate. FANCJ was also able to displace TRF2 from the telomeric substrate in an RPA-dependent manner. The stimulation of helicase-catalyzed protein displacement is also observed with the DNA helicase RECQ1, suggesting a conserved functional interaction of RPA-interacting helicases. These findings suggest that partnerships between RPA and interacting human DNA helicases may greatly enhance their ability to dislodge proteins bound to duplex DNA, an activity that is likely to be highly relevant to their biological roles in DNA metabolism.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , ADN/metabolismo , Proteínas del Grupo de Complementación de la Anemia de Fanconi/metabolismo , RecQ Helicasas/metabolismo , Proteína de Replicación A/metabolismo , Sustitución de Aminoácidos , Secuencia de Bases , ADN/química , ADN/genética , Desoxirribonucleasa BamHI/metabolismo , Exodesoxirribonucleasas/metabolismo , Humanos , Sustancias Macromoleculares/química , Sustancias Macromoleculares/metabolismo , Conformación de Ácido Nucleico , Unión Proteica , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteína de Replicación A/genética , Especificidad por Sustrato , Proteína 1 de Unión a Repeticiones Teloméricas/metabolismo , Helicasa del Síndrome de Werner
16.
HPB (Oxford) ; 11(5): 435-44, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19768149

RESUMEN

BACKGROUND: RECQL is a DNA helicase involved in DNA mismatch repair. The RECQL polymorphism, 3' untranslated region (UTR) A159C, was previously associated with overall survival of patients with resectable pancreatic adenocarcinoma treated with neoadjuvant chemoradiation. In the present study, we examined RECQL for somatic mutations and other polymorphisms and compared these findings with the outcome in patients who received adjuvant or neoadjuvant chemoradiation. We hypothesized that RECQL (i) would be mutated in cancer, (ii) would have polymorphisms linked to the 3'UTR A159C and that either or both events would affect function. We also hypothesized that (iii) these changes would be associated with survival in both cohorts of patients. MATERIAL AND METHODS: We sequenced RECQL's 15 exons and surrounding sequences in paired blood and tumour DNA of 39 patients. The 3'UTR A159C genotype was determined in blood DNA samples from 176 patients with resectable pancreatic adenocarcinoma treated with adjuvant (53) or neoadjuvant (123) chemoradiation. Survival was calculated using the Kaplan-Meier method, with log rank comparisons between groups. The relative impact of genotype on time to overall survival was performed using the Cox proportional hazards model. RESULTS: Somatic mutations were found in UTRs and intronic regions but not in exonic coding regions of the RECQL gene. Two single nucleotide polymorphisms (SNPs), located in introns 2 and 11, were found to be part of the same haplotype block as the RECQL A159C SNP and showed a similar association with overall survival. No short-term difference in survival between treatment strategies was found. We identified a subgroup of patients responsive to neoadjuvant therapy in which the 159 A allele conferred strikingly improved long-term survival. DISCUSSION: The RECQL 3'UTR A159C SNP is not linked with other functional SNPs within RECQL but may function as a site for regulatory molecules. The mechanism of action needs to be clarified further.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA