Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 180
Filtrar
1.
Biomed Pharmacother ; 169: 115908, 2023 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-37988849

RESUMEN

The high expression of BLM (Bloom syndrome) DNA helicase in tumors involves its strong association with cell expansion. Bisbenzylisoquinoline alkaloids own an antitumor property and have developed as candidates for anticancer drugs. This paper aimed to study the antitumor effect of fangchinoline derivative HY-2 by targeting BLM642-1290 DNA helicase, and then explore its inhibitory mechanism on proliferation of MDA-MB-435 breast cancer cells. We confirmed that the mRNA and protein levels of BLM DNA helicase in breast cancer were higher than those in normal tissues. HY-2 could inhibit the DNA binding, ATPase and DNA unwinding of BLM642-1290 DNA helicase with enzymatic assay. HY-2 could also inhibit the DNA unwinding of DNA helicase in cells. In addition, HY-2 showed an inhibiting the MDA-MB-435, MDA-MB-231, MDA-MB-436 breast cancer cells expansion. The mRNA and protein levels of BLM DNA helicase in MDA-MB-435 cells increased after HY-2 treatment, which might contribute to HY-2 inhibiting the DNA binding, ATPase and DNA unwinding of BLM DNA helicase. The mechanism of HY-2 inhibition on BLM DNA helicase was further confirmed with the effect of HY-2 on the ultraviolet spectrogram of BLM642-1290 DNA helicase and Molecular dynamics simulation of the interacting between HY-2 and BLM640-1291 DNA helicase. Our study provided some valuable clues for the exploration of HY-2 in the living body and developing it as an anticancer drug.


Asunto(s)
Antineoplásicos , Bencilisoquinolinas , Neoplasias de la Mama , Femenino , Humanos , Bencilisoquinolinas/farmacología , Neoplasias de la Mama/tratamiento farmacológico , ADN/metabolismo , RecQ Helicasas/química , RecQ Helicasas/genética , RecQ Helicasas/metabolismo , ARN Mensajero , ADN Helicasas/antagonistas & inhibidores , ADN Helicasas/metabolismo
2.
Nucleic Acids Res ; 51(9): 4363-4384, 2023 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-36942481

RESUMEN

Crossing over between homologs is critical for the stable segregation of chromosomes during the first meiotic division. Saccharomyces cerevisiae Mer3 (HFM1 in mammals) is a SF2 helicase and member of the ZMM group of proteins, that facilitates the formation of the majority of crossovers during meiosis. Here, we describe the structural organisation of Mer3 and using AlphaFold modelling and XL-MS we further characterise the previously described interaction with Mlh1-Mlh2. We find that Mer3 also forms a previously undescribed complex with the recombination regulating factors Top3 and Rmi1 and that this interaction is competitive with Sgs1BLM helicase. Using in vitro reconstituted D-loop assays we show that Mer3 inhibits the anti-recombination activity of Sgs1 helicase, but only in the presence of Dmc1. Thus we provide a mechanism whereby Mer3 interacts with a network of proteins to protect Dmc1 derived D-loops from dissolution.


Asunto(s)
ADN Helicasas , Recombinación Homóloga , Meiosis , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Proteínas de Ciclo Celular/genética , Intercambio Genético , ADN Helicasas/química , ADN Helicasas/metabolismo , Proteínas de Unión al ADN/metabolismo , Meiosis/genética , Unión Proteica , Pliegue de Proteína , RecQ Helicasas/antagonistas & inhibidores , RecQ Helicasas/química , RecQ Helicasas/metabolismo , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/enzimología , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo , Unión Competitiva
3.
Mol Genet Genomics ; 298(1): 37-47, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36264383

RESUMEN

Owing to their crucial role in genome maintenance, RecQ helicases are ubiquitous and present across organisms. Though the multiplicity of RecQ helicases is well known in higher organisms, it is rare among bacteria. The ancient cyanobacterium Nostoc sp. strain PCC7120 was found to have three annotated RecQ helicases. This study aims at understanding its structural differences and evolution through bioinformatics approach and functionality through expression analysis studies. Nostoc RecQ helicases were found to be transcriptionally regulated by LexA and DNA damage inducing stresses. Bioinformatic analysis revealed that all three RecQ helicases of Nostoc possess helicases_C and Zn+2-binding domains. Two of the helicases (AnRecQ and AnRecQ2) lacked the complete RQC and HRDC domains, and AnRecQ2 had an additional Phosphoribosyl transferase domain (Pribosyltran), also seen in RecQ-like helicase (RqlH) protein of Mycobacterium smegmatis. AnRecQ1, which was similar to most bacterial RecQ helicases, differed in having a long C-terminal tail. STRING analysis revealed that the proteins also differed in their predicted protein interactome. Phylogenetic analysis suggested that the multiple recQ genes may have been acquired through duplication and acquisition of additional domains from the smallest of the RecQ helicases (AnRecQ) to cater multiple functions required to deal with the harsh environmental conditions. In course of evolution, however, the multiplicity was lost with the modern-day bacteria and lower eukaryotes which retained fewer RecQ helicases, while further duplication of the acquired RECQ occurred in higher animals and plants to deal with cellular complexity.


Asunto(s)
Nostoc , RecQ Helicasas , Biología Computacional , Nostoc/enzimología , Filogenia , Estructura Terciaria de Proteína , RecQ Helicasas/genética , RecQ Helicasas/química , RecQ Helicasas/metabolismo
4.
Nucleic Acids Res ; 50(18): 10601-10613, 2022 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-36165957

RESUMEN

Helicases are essential for nearly all nucleic acid processes across the tree of life, yet detailed understanding of how they couple ATP hydrolysis to translocation and unwinding remains incomplete because their small (∼300 picometer), fast (∼1 ms) steps are difficult to resolve. Here, we use Nanopore Tweezers to observe single Escherichia coli RecQ helicases as they translocate on and unwind DNA at ultrahigh spatiotemporal resolution. Nanopore Tweezers simultaneously resolve individual steps of RecQ along the DNA and conformational changes of the helicase associated with stepping. Our data reveal the mechanochemical coupling between physical domain motions and chemical reactions that together produce directed motion of the helicase along DNA. Nanopore Tweezers measurements are performed under either assisting or opposing force applied directly on RecQ, shedding light on how RecQ responds to such forces in vivo. Determining the rates of translocation and physical conformational changes under a wide range of assisting and opposing forces reveals the underlying dynamic energy landscape that drives RecQ motion. We show that RecQ has a highly asymmetric energy landscape that enables RecQ to maintain velocity when encountering molecular roadblocks such as bound proteins and DNA secondary structures. This energy landscape also provides a mechanistic basis making RecQ an 'active helicase,' capable of unwinding dsDNA as fast as it translocates on ssDNA. Such an energy landscape may be a general strategy for molecular motors to maintain consistent velocity despite opposing loads or roadblocks.


Asunto(s)
RecQ Helicasas/química , Adenosina Trifosfato/metabolismo , ADN de Cadena Simple , Escherichia coli/genética , Escherichia coli/metabolismo , Nanoporos , Ácidos Nucleicos , RecQ Helicasas/metabolismo
5.
Nucleic Acids Res ; 49(20): 11834-11854, 2021 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-34751402

RESUMEN

RECQ1 is the shortest among the five human RecQ helicases comprising of two RecA like domains, a zinc-binding domain and a RecQ C-terminal domain containing the winged-helix (WH). Mutations or deletions on the tip of a ß-hairpin located in the WH domain are known to abolish the unwinding activity. Interestingly, the same mutations on the ß-hairpin of annealing incompetent RECQ1 mutant (RECQ1T1) have been reported to restore its annealing activity. In an attempt to unravel the strand annealing mechanism, we have crystallized a fragment of RECQ1 encompassing D2-Zn-WH domains harbouring mutations on the ß-hairpin. From our crystal structure data and interface analysis, we have demonstrated that an α-helix located in zinc-binding domain potentially interacts with residues of WH domain, which plays a significant role in strand annealing activity. We have shown that deletion of the α-helix or mutation of specific residues on it restores strand annealing activity of annealing deficient constructs of RECQ1. Our results also demonstrate that mutations on the α-helix induce conformational changes and affects DNA stimulated ATP hydrolysis and unwinding activity of RECQ1. Our study, for the first time, provides insight into the conformational requirements of the WH domain for efficient strand annealing by human RECQ1.


Asunto(s)
ADN de Cadena Simple/química , RecQ Helicasas/química , Sitios de Unión , ADN de Cadena Simple/metabolismo , Humanos , Simulación de Dinámica Molecular , Mutación , Unión Proteica , Conformación Proteica en Hélice alfa , RecQ Helicasas/genética , RecQ Helicasas/metabolismo , Zinc/metabolismo
6.
Nucleic Acids Res ; 49(15): 8699-8713, 2021 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-34370039

RESUMEN

The Bloom syndrome DNA helicase BLM contributes to chromosome stability through its roles in double-strand break repair by homologous recombination and DNA replication fork restart during the replication stress response. Loss of BLM activity leads to Bloom syndrome, which is characterized by extraordinary cancer risk and small stature. Here, we have analyzed the composition of the BLM complex during unperturbed S-phase and identified a direct physical interaction with the Mcm6 subunit of the minichromosome maintenance (MCM) complex. Using distinct binding sites, BLM interacts with the N-terminal domain of Mcm6 in G1 phase and switches to the C-terminal Cdt1-binding domain of Mcm6 in S-phase, with a third site playing a role for Mcm6 binding after DNA damage. Disruption of Mcm6-binding to BLM in S-phase leads to supra-normal DNA replication speed in unperturbed cells, and the helicase activity of BLM is required for this increased replication speed. Upon disruption of BLM/Mcm6 interaction, repair of replication-dependent DNA double-strand breaks is delayed and cells become hypersensitive to DNA damage and replication stress. Our findings reveal that BLM not only plays a role in the response to DNA damage and replication stress, but that its physical interaction with Mcm6 is required in unperturbed cells, most notably in S-phase as a negative regulator of replication speed.


Asunto(s)
Componente 6 del Complejo de Mantenimiento de Minicromosoma/metabolismo , RecQ Helicasas/metabolismo , Fase S/genética , Sitios de Unión , Línea Celular , Reparación del ADN , Fase G1 , Humanos , Componente 6 del Complejo de Mantenimiento de Minicromosoma/química , Mutación , Dominios y Motivos de Interacción de Proteínas , RecQ Helicasas/química
7.
Nucleic Acids Res ; 49(10): 5470-5492, 2021 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-33963870

RESUMEN

Topoisomerases are essential enzymes that regulate DNA topology. Type 1A family topoisomerases are found in nearly all living organisms and are unique in that they require single-stranded (ss)DNA for activity. These enzymes are vital for maintaining supercoiling homeostasis and resolving DNA entanglements generated during DNA replication and repair. While the catalytic cycle of Type 1A topoisomerases has been long-known to involve an enzyme-bridged ssDNA gate that allows strand passage, a deeper mechanistic understanding of these enzymes has only recently begun to emerge. This knowledge has been greatly enhanced through the combination of biochemical studies and increasingly sophisticated single-molecule assays based on magnetic tweezers, optical tweezers, atomic force microscopy and Förster resonance energy transfer. In this review, we discuss how single-molecule assays have advanced our understanding of the gate opening dynamics and strand-passage mechanisms of Type 1A topoisomerases, as well as the interplay of Type 1A topoisomerases with partner proteins, such as RecQ-family helicases. We also highlight how these assays have shed new light on the likely functional roles of Type 1A topoisomerases in vivo and discuss recent developments in single-molecule technologies that could be applied to further enhance our understanding of these essential enzymes.


Asunto(s)
ADN-Topoisomerasas de Tipo I , ADN , ADN/química , ADN-Topoisomerasas de Tipo I/química , ADN-Topoisomerasas de Tipo I/fisiología , Humanos , Estructura Molecular , RecQ Helicasas/química
8.
Biol Chem ; 402(5): 617-636, 2021 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-33567180

RESUMEN

RecQ helicases are a highly conserved class of DNA helicases that play crucial role in almost all DNA metabolic processes including replication, repair and recombination. They are able to unwind a wide variety of complex intermediate DNA structures that may result from cellular DNA transactions and hence assist in maintaining genome integrity. Interestingly, a huge number of recent reports suggest that many of the RecQ family helicases are directly or indirectly involved in regulating transcription and gene expression. On one hand, they can remove complex structures like R-loops, G-quadruplexes or RNA:DNA hybrids formed at the intersection of transcription and replication. On the other hand, emerging evidence suggests that they can also regulate transcription by directly interacting with RNA polymerase or recruiting other protein factors that may regulate transcription. This review summarizes the up to date knowledge on the involvement of three human RecQ family proteins BLM, WRN and RECQL5 in transcription regulation and management of transcription associated stress.


Asunto(s)
ADN/metabolismo , ARN/metabolismo , RecQ Helicasas/metabolismo , Helicasa del Síndrome de Werner/metabolismo , ADN/química , Humanos , ARN/química , RecQ Helicasas/química , Helicasa del Síndrome de Werner/química
9.
Structure ; 29(2): 99-113, 2021 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-33357470

RESUMEN

The Bloom syndrome complex is a DNA damage repair machine. It consists of several protein components which are functional in isolation, but interdependent in cells for the maintenance of accurate homologous recombination. Mutations to any of the genes encoding these proteins cause numerous physical and developmental markers as well as phenotypes of genome instability, infertility, and cancer predisposition. Here we review the published structural and biochemical data on each of the components of the complex: the helicase BLM, the type IA topoisomerase TOP3A, and the OB-fold-containing RMI and RPA subunits. We describe how each component contributes to function, interacts with each other, and the DNA that it manipulates/repairs.


Asunto(s)
Síndrome de Bloom/genética , ADN-Topoisomerasas de Tipo I/química , Proteínas de Unión al ADN/química , RecQ Helicasas/química , ADN-Topoisomerasas de Tipo I/metabolismo , Proteínas de Unión al ADN/metabolismo , Recombinación Homóloga , Humanos , RecQ Helicasas/metabolismo
10.
J Med Chem ; 63(17): 9752-9772, 2020 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-32697083

RESUMEN

DNA damage response (DDR) pathways are crucial for the survival of cancer cells and are attractive targets for cancer therapy. Bloom syndrome protein (BLM) is a DNA helicase that performs important roles in DDR pathways. Our previous study discovered an effective new BLM inhibitor with a quinazolinone scaffold by a screening assay. Herein, to better understand the structure-activity relationship (SAR) and biological roles of the BLM inhibitor, a series of new derivatives were designed, synthesized, and evaluated based on this scaffold. Among them, compound 9h exhibited nanomolar inhibitory activity and binding affinity for BLM. 9h could effectively disrupt BLM recruitment to DNA in cells. Furthermore, 9h inhibited the proliferation of the colorectal cell line HCT116 by significantly triggering DNA damage in the telomere region and inducing apoptosis, especially in combination with a poly (ADP-ribose) polymerase (PARP) inhibitor. This result suggested a synthetic lethal effect between the BLM and PARP inhibitors in DDR pathways.


Asunto(s)
Daño del ADN , Diseño de Fármacos , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Quinazolinonas/síntesis química , Quinazolinonas/farmacología , RecQ Helicasas/antagonistas & inhibidores , Telómero/genética , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Técnicas de Química Sintética , Sinergismo Farmacológico , Células HCT116 , Humanos , Modelos Moleculares , Conformación Proteica , Quinazolinonas/química , RecQ Helicasas/química , Relación Estructura-Actividad
11.
Biophys Chem ; 265: 106433, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32702531

RESUMEN

RecQ helicases belong to a ubiquitous family of DNA unwinding enzymes that are essential to maintain genome stability by acting at the interface between DNA replication, recombination, and repair. Humans have five different paralogues of RecQ helicases namely RecQ1, BLM, WRN, RecQ4, and RecQ5. Germ-line mutations in these helicases give rise to distinct human genetic disorders, Bloom Syndrome, Werner Syndrome, Rothmund-Thomson, RAPADILINO, and Baller-Gerold syndromes. Other than distinct clinical symptoms, all these genetic disorders show a predisposition to cancer. While the three paralogues BLM, WRN, and RecQ4 are directly associated with syndromes, loss of function of RecQ1 and RecQ5 are also emerging to be causative of various types of cancer. This review summarizes the domain architecture of RecQ helicases and the mutations that are associated with various diseases. Here we observe the occurrence of disease-causing mutations mainly in the catalytic regions of the proteins, and that some of these mutations are common between the respective disorders and cancer. Furthermore, this review discusses the results of several reports that study the role of residues with disease-causing mutations. It also overviews the research focusing on RecQ helicases as potential targets for cancer therapy.


Asunto(s)
Mutación , RecQ Helicasas/genética , Secuencia Conservada , Humanos , Neoplasias/enzimología , Dominios Proteicos , RecQ Helicasas/química , RecQ Helicasas/metabolismo
12.
Sci Rep ; 10(1): 12377, 2020 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-32704157

RESUMEN

The BLM helicase protein plays a vital role in DNA replication and the maintenance of genomic integrity. Variation in the BLM helicase gene resulted in defects in the DNA repair mechanism and was reported to be associated with Bloom syndrome (BS) and cancer. Despite extensive investigation of helicase proteins in humans, no attempt has previously been made to comprehensively analyse the single nucleotide polymorphism (SNPs) of the BLM gene. In this study, a comprehensive analysis of SNPs on the BLM gene was performed to identify, characterize and validate the pathogenic SNPs using computational approaches. We obtained SNP data from the dbSNP database version 150 and mapped these data to the genomic coordinates of the "NM_000057.3" transcript expressing BLM helicase (P54132). There were 607 SNPs mapped to missense, 29 SNPs mapped to nonsense, and 19 SNPs mapped to 3'-UTR regions. Initially, we used many consensus tools of SIFT, PROVEAN, Condel, and PolyPhen-2, which together increased the accuracy of prediction and identified 18 highly pathogenic non-synonymous SNPs (nsSNPs) out of 607 SNPs. Subsequently, these 18 high-confidence pathogenic nsSNPs were analysed for BLM protein stability, structure-function relationships and disease associations using various bioinformatics tools. These 18 mutants of the BLM protein along with the native protein were further investigated using molecular dynamics simulations to examine the structural consequences of the mutations, which might reveal their malfunction and contribution to disease. In addition, 28 SNPs were predicted as "stop gained" nonsense SNPs and one SNP was predicted as "start lost". Two SNPs in the 3'UTR were found to abolish miRNA binding and thus may enhance the expression of BLM. Interestingly, we found that BLM mRNA overexpression is associated with different types of cancers. Further investigation showed that the dysregulation of BLM is associated with poor overall survival (OS) for lung and gastric cancer patients and hence led to the conclusion that BLM has the potential to be used as an important prognostic marker for the detection of lung and gastric cancer.


Asunto(s)
Biología Computacional , Bases de Datos de Proteínas , Modelos Moleculares , Polimorfismo de Nucleótido Simple , RecQ Helicasas , Sitios de Unión , Estabilidad de Enzimas , Humanos , RecQ Helicasas/química , RecQ Helicasas/genética
13.
Biochem Biophys Res Commun ; 526(4): 993-998, 2020 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-32295713

RESUMEN

Minichromosome maintenance 8 (MCM8) is a recently identified member of the minichromosome maintenance family, which possesses helicase and ATPase activity. It interacts with MCM9 and participates in homologous recombination repair. The structure of MCM8 is unclear now. Here, we report the crystal structure of the winged-helix domain of human MCM8 (MCM8-WHD) at 1.21 Å resolution. MCM8-WHD adopts a conserved winged-helix architecture. Structure analysis and biochemical study results showed the DNA binding ability and crucial residues of MCM8-WHD. Our results are helpful to understand the function of MCM8.


Asunto(s)
Proteínas de Mantenimiento de Minicromosoma/química , Secuencia de Aminoácidos , Cristalografía por Rayos X , ADN/química , Células HEK293 , Humanos , Modelos Moleculares , Unión Proteica , Dominios Proteicos , RecQ Helicasas/química
14.
Genes (Basel) ; 11(2)2020 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-32085395

RESUMEN

With roles in DNA repair, recombination, replication and transcription, members of the RecQ DNA helicase family maintain genome integrity from bacteria to mammals. Mutations in human RecQ helicases BLM, WRN and RecQL4 cause incurable disorders characterized by genome instability, increased cancer predisposition and premature adult-onset aging. Yeast cells lacking the RecQ helicase Sgs1 share many of the cellular defects of human cells lacking BLM, including hypersensitivity to DNA damaging agents and replication stress, shortened lifespan, genome instability and mitotic hyper-recombination, making them invaluable model systems for elucidating eukaryotic RecQ helicase function. Yeast and human RecQ helicases have common DNA substrates and domain structures and share similar physical interaction partners. Here, we review the major cellular functions of the yeast RecQ helicases Sgs1 of Saccharomyces cerevisiae and Rqh1 of Schizosaccharomyces pombe and provide an outlook on some of the outstanding questions in the field.


Asunto(s)
ADN Helicasas/metabolismo , RecQ Helicasas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Schizosaccharomyces pombe/metabolismo , Schizosaccharomyces/genética , ADN Helicasas/química , ADN Helicasas/genética , Reparación del ADN , Genoma Fúngico , Inestabilidad Genómica , Humanos , Mutación , Dominios Proteicos , RecQ Helicasas/química , RecQ Helicasas/genética , Saccharomyces cerevisiae/enzimología , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Schizosaccharomyces/enzimología , Proteínas de Schizosaccharomyces pombe/química , Proteínas de Schizosaccharomyces pombe/genética
15.
J Biol Chem ; 295(51): 17646-17658, 2020 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-33454004

RESUMEN

RecQ family helicases are highly conserved from bacteria to humans and have essential roles in maintaining genome stability. Mutations in three human RecQ helicases cause severe diseases with the main features of premature aging and cancer predisposition. Most RecQ helicases shared a conserved domain arrangement which comprises a helicase core, an RecQ C-terminal domain, and an auxiliary element helicase and RNaseD C-terminal (HRDC) domain, the functions of which are poorly understood. In this study, we systematically characterized the roles of the HRDC domain in E. coli RecQ in various DNA transactions by single-molecule FRET. We found that RecQ repetitively unwinds the 3'-partial duplex and fork DNA with a moderate processivity and periodically patrols on the ssDNA in the 5'-partial duplex by translocation. The HRDC domain significantly suppresses RecQ activities in the above transactions. In sharp contrast, the HRDC domain is essential for the deep and long-time unfolding of the G4 DNA structure by RecQ. Based on the observations that the HRDC domain dynamically switches between RecA core- and ssDNA-binding modes after RecQ association with DNA, we proposed a model to explain the modulation mechanism of the HRDC domain. Our findings not only provide new insights into the activities of RecQ on different substrates but also highlight the novel functions of the HRDC domain in DNA metabolisms.


Asunto(s)
ADN/metabolismo , Escherichia coli/enzimología , G-Cuádruplex , RecQ Helicasas/metabolismo , Reparación del ADN , Transferencia Resonante de Energía de Fluorescencia , Humanos , Mutagénesis Sitio-Dirigida , Conformación de Ácido Nucleico , Unión Proteica , Dominios Proteicos , Estructura Terciaria de Proteína , RecQ Helicasas/química , RecQ Helicasas/genética , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Especificidad por Sustrato
16.
Nucleic Acids Res ; 47(21): 11225-11237, 2019 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-31544923

RESUMEN

Bloom helicase (BLM) and its orthologs are essential for the maintenance of genome integrity. BLM defects represent the underlying cause of Bloom Syndrome, a rare genetic disorder that is marked by strong cancer predisposition. BLM deficient cells accumulate extensive chromosomal aberrations stemming from dysfunctions in homologous recombination (HR). BLM participates in several HR stages and helps dismantle potentially harmful HR intermediates. However, much remains to be learned about the molecular mechanisms of these BLM-mediated regulatory effects. Here, we use DNA curtains to directly visualize the activity of BLM helicase on single molecules of DNA. Our data show that BLM is a robust helicase capable of rapidly (∼70-80 base pairs per second) unwinding extensive tracts (∼8-10 kilobases) of double-stranded DNA (dsDNA). Importantly, we find no evidence for BLM activity on single-stranded DNA (ssDNA) that is bound by replication protein A (RPA). Likewise, our results show that BLM can neither associate with nor translocate on ssDNA that is bound by the recombinase protein RAD51. Moreover, our data reveal that the presence of RAD51 also blocks BLM translocation on dsDNA substrates. We discuss our findings within the context of potential regulator roles for BLM helicase during DNA replication and repair.


Asunto(s)
ADN de Cadena Simple/metabolismo , ADN/metabolismo , RecQ Helicasas/análisis , RecQ Helicasas/metabolismo , Imagen Individual de Molécula , Emparejamiento Base , Síndrome de Bloom/genética , ADN/química , Reparación del ADN/genética , Replicación del ADN/genética , ADN de Cadena Simple/química , Recombinación Homóloga , Humanos , Modelos Moleculares , Recombinasa Rad51/metabolismo , RecQ Helicasas/química , RecQ Helicasas/genética , Proteína de Replicación A/metabolismo , Imagen Individual de Molécula/métodos
17.
Nucleic Acids Res ; 47(18): 9708-9720, 2019 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-31435650

RESUMEN

The RecQ family of helicases is highly conserved both structurally and functionally from bacteria to humans. Defects in human RecQ helicases are associated with genetic diseases that are characterized by cancer predisposition and/or premature aging. RecQ proteins exhibit 3'-5' helicase activity and play critical roles in genome maintenance. Recent advances in single-molecule techniques have revealed the reiterative unwinding behavior of RecQ helicases. However, the molecular mechanisms involved in this process remain unclear, with contradicting reports. Here, we characterized the unwinding dynamics of the Caenorhabditis elegans RecQ helicase HIM-6 using single-molecule fluorescence resonance energy transfer measurements. We found that HIM-6 exhibits reiterative DNA unwinding and the length of DNA unwound by the helicase is sharply defined at 25-31 bp. Experiments using various DNA substrates revealed that HIM-6 utilizes the mode of 'sliding back' on the translocated strand, without strand-switching for rewinding. Furthermore, we found that Caenorhabditis elegans replication protein A, a single-stranded DNA binding protein, suppresses the reiterative behavior of HIM-6 and induces unidirectional, processive unwinding, possibly through a direct interaction between the proteins. Our findings shed new light on the mechanism of DNA unwinding by RecQ family helicases and their co-operation with RPA in processing DNA.


Asunto(s)
Proteínas de Caenorhabditis elegans/genética , ADN/genética , RecQ Helicasas/genética , Proteína de Replicación A/genética , Animales , Caenorhabditis elegans/enzimología , Proteínas de Unión al ADN , Escherichia coli/genética , Humanos , RecQ Helicasas/química
18.
PLoS Genet ; 15(7): e1008266, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31276497

RESUMEN

Rothmund-Thomson syndrome (RTS) is a rare autosomal recessive disorder characterized by skin rash (poikiloderma), skeletal dysplasia, small stature, juvenile cataracts, sparse or absent hair, and predisposition to specific malignancies such as osteosarcoma and hematological neoplasms. RTS is caused by germ-line mutations in RECQL4, a RecQ helicase family member. In vitro studies have identified functions for the ATP-dependent helicase of RECQL4. However, its specific role in vivo remains unclear. To determine the physiological requirement and the biological functions of Recql4 helicase activity, we generated mice with an ATP-binding-deficient knock-in mutation (Recql4K525A). Recql4K525A/K525A mice were strikingly normal in terms of embryonic development, body weight, hematopoiesis, B and T cell development, and physiological DNA damage repair. However, mice bearing two distinct truncating mutations Recql4G522Efs and Recql4R347*, that abolished not only the helicase but also the C-terminal domain, developed a profound bone marrow failure and decrease in survival similar to a Recql4 null allele. These results demonstrate that the ATP-dependent helicase activity of Recql4 is not essential for its physiological functions and that other domains might contribute to this phenotype. Future studies need to be performed to elucidate the complex interactions of RECQL4 domains and its contribution to the development of RTS.


Asunto(s)
Adenosina Trifosfato/metabolismo , RecQ Helicasas/genética , RecQ Helicasas/metabolismo , Síndrome Rothmund-Thomson/genética , Animales , Linfocitos B/metabolismo , Sitios de Unión , Peso Corporal , Daño del ADN , Modelos Animales de Enfermedad , Desarrollo Embrionario , Técnicas de Sustitución del Gen , Hematopoyesis , Humanos , Ratones , Fenotipo , Dominios Proteicos , RecQ Helicasas/química , Linfocitos T/metabolismo
19.
Mol Microbiol ; 112(3): 854-865, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31162841

RESUMEN

The GC-rich genome of Deinococcus radiodurans contains a very high density of putative guanine quadruplex (G4) DNA motifs and its RecQ (drRecQ) was earlier characterized as a 3'→5' dsDNA helicase. We saw that N-Methyl mesoporphyrin IX (NMM), a G4 DNA binding drug affected normal growth as well as the gamma radiation resistance of the wild-type bacterium. Interestingly, NMM treatment and recQ deletion showed additive effect on normal growth but there was no effect of NMM on gamma radiation resistance of recQ mutant. The recombinant drRecQ showed ~400 times higher affinity to G4 DNA (Kd  = 11.74 ± 1.77 nM) as compared to dsDNA (Kd  = 4.88 ± 1.30 µM). drRecQ showed ATP independent helicase function on G4 DNA, which was higher than ATP-dependent helicase activity on dsDNA. Unlike wild-type cells that sparingly stained for G4 structure with Thioflavin T (ThT), recQ mutant showed very high-density of ThT fluorescence foci on DNA indicating an important role of drRecQ in regulation of G4 DNA structure dynamics in vivo. These results together suggested that drRecQ is an ATP independent G4 DNA helicase that plays an important role in the regulation of G4 DNA structure dynamics and its impact on radioresistance in D. radiodurans.


Asunto(s)
Proteínas Bacterianas/metabolismo , ADN Bacteriano/química , Deinococcus/enzimología , Deinococcus/efectos de la radiación , Regulación Bacteriana de la Expresión Génica , RecQ Helicasas/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , ADN Bacteriano/genética , ADN Bacteriano/metabolismo , Deinococcus/genética , Deinococcus/crecimiento & desarrollo , G-Cuádruplex , Viabilidad Microbiana/efectos de la radiación , RecQ Helicasas/química , RecQ Helicasas/genética , Especificidad por Sustrato
20.
Molecules ; 24(9)2019 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-31067825

RESUMEN

G-quadruplex (G4) structures are highly stable four-stranded DNA and RNA secondary structures held together by non-canonical guanine base pairs. G4 sequence motifs are enriched at specific sites in eukaryotic genomes, suggesting regulatory functions of G4 structures during different biological processes. Considering the high thermodynamic stability of G4 structures, various proteins are necessary for G4 structure formation and unwinding. In a yeast one-hybrid screen, we identified Slx9 as a novel G4-binding protein. We confirmed that Slx9 binds to G4 DNA structures in vitro. Despite these findings, Slx9 binds only insignificantly to G-rich/G4 regions in Saccharomyces cerevisiae as demonstrated by genome-wide ChIP-seq analysis. However, Slx9 binding to G4s is significantly increased in the absence of Sgs1, a RecQ helicase that regulates G4 structures. Different genetic and molecular analyses allowed us to propose a model in which Slx9 recognizes and protects stabilized G4 structures in vivo.


Asunto(s)
Proteínas de Unión al ADN/química , G-Cuádruplex , Proteínas Ribosómicas/genética , Proteínas de Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/genética , ADN Helicasas/química , ADN Helicasas/genética , Proteínas de Unión al ADN/genética , Genoma/genética , Conformación de Ácido Nucleico , Unión Proteica , RecQ Helicasas/química , RecQ Helicasas/genética , Proteínas Ribosómicas/química , Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/química , Termodinámica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...