Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
iScience ; 27(7): 110260, 2024 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-39055910

RESUMO

To ensure timely duplication of the entire eukaryotic genome, thousands of replication machineries (replisomes) act on genomic DNA at any time during S phase. In the final stages of this process, replisomes are unloaded from chromatin. Unloading is driven by polyubiquitylation of MCM7, a subunit of the terminated replicative helicase, and processed by p97/VCP segregase. Most of our knowledge of replication termination comes from model organisms, and little is known about how this process is executed and regulated in human somatic cells. Here we show that replisome disassembly in this system requires CUL2LRR1-driven MCM7 ubiquitylation, p97, and UBXN7 for unloading and provide evidence for "backup" mitotic replisome disassembly, demonstrating conservation of such mechanisms. Finally, we find that small-molecule inhibitors against Cullin ubiquitin ligases (CULi) and p97 (p97i) affect replisome unloading but also lead to induction of replication stress in cells, which limits their usefulness to specifically target replisome disassembly processes.

2.
Biology (Basel) ; 13(4)2024 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-38666845

RESUMO

The eukaryotic replicative helicase (CMG complex) is assembled during DNA replication initiation in a highly regulated manner, which is described in depth by other manuscripts in this Issue. During DNA replication, the replicative helicase moves through the chromatin, unwinding DNA and facilitating nascent DNA synthesis by polymerases. Once the duplication of a replicon is complete, the CMG helicase and the remaining components of the replisome need to be removed from the chromatin. Research carried out over the last ten years has produced a breakthrough in our understanding, revealing that replication termination, and more specifically replisome disassembly, is indeed a highly regulated process. This review brings together our current understanding of these processes and highlights elements of the mechanism that are conserved or have undergone divergence throughout evolution. Finally, we discuss events beyond the classic termination of DNA replication in S-phase and go over the known mechanisms of replicative helicase removal from chromatin in these particular situations.

3.
Nat Commun ; 14(1): 8293, 2023 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-38097601

RESUMO

Ubiquitin widely modifies proteins, thereby regulating most cellular functions. The complexity of ubiquitin signalling necessitates unbiased methods enabling global detection of dynamic protein ubiquitylation. Here, we describe UBIMAX (UBiquitin target Identification by Mass spectrometry in Xenopus egg extracts), which enriches ubiquitin-conjugated proteins and quantifies regulation of protein ubiquitylation under precise and adaptable conditions. We benchmark UBIMAX by investigating DNA double-strand break-responsive ubiquitylation events, identifying previously known targets and revealing the actin-organizing protein Dbn1 as a major target of DNA damage-induced ubiquitylation. We find that Dbn1 is targeted for proteasomal degradation by the SCFß-Trcp1 ubiquitin ligase, in a conserved mechanism driven by ATM-mediated phosphorylation of a previously uncharacterized ß-Trcp1 degron containing an SQ motif. We further show that this degron is sufficient to induce DNA damage-dependent protein degradation of a model substrate. Collectively, we demonstrate UBIMAX's ability to identify targets of stimulus-regulated ubiquitylation and reveal an SCFß-Trcp1-mediated ubiquitylation mechanism controlled directly by the apical DNA damage response kinases.


Assuntos
Actinas , Ubiquitina , Ubiquitina/metabolismo , Actinas/metabolismo , Ubiquitinação , Transdução de Sinais , Dano ao DNA
4.
Mol Cell ; 83(22): 4017-4031.e9, 2023 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-37820732

RESUMO

The MCM motor of the replicative helicase is loaded onto origin DNA as an inactive double hexamer before replication initiation. Recruitment of activators GINS and Cdc45 upon S-phase transition promotes the assembly of two active CMG helicases. Although work with yeast established the mechanism for origin activation, how CMG is formed in higher eukaryotes is poorly understood. Metazoan Downstream neighbor of Son (DONSON) has recently been shown to deliver GINS to MCM during CMG assembly. What impact this has on the MCM double hexamer is unknown. Here, we used cryoelectron microscopy (cryo-EM) on proteins isolated from replicating Xenopus egg extracts to identify a double CMG complex bridged by a DONSON dimer. We find that tethering elements mediating complex formation are essential for replication. DONSON reconfigures the MCM motors in the double CMG, and primordial dwarfism patients' mutations disrupting DONSON dimerization affect GINS and MCM engagement in human cells and DNA synthesis in Xenopus egg extracts.


Assuntos
Proteínas de Ciclo Celular , DNA Helicases , Proteínas Nucleares , Animais , Humanos , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Microscopia Crioeletrônica , DNA/genética , DNA/metabolismo , DNA Helicases/metabolismo , Replicação do DNA , Proteínas de Manutenção de Minicromossomo/genética , Proteínas de Manutenção de Minicromossomo/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Saccharomyces cerevisiae/genética , Ativação Enzimática
5.
Nat Commun ; 14(1): 5071, 2023 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-37604812

RESUMO

Cell division is the basis for the propagation of life and requires accurate duplication of all genetic information. DNA damage created during replication (replication stress) is a major cause of cancer, premature aging and a spectrum of other human disorders. Over the years, TRAIP E3 ubiquitin ligase has been shown to play a role in various cellular processes that govern genome integrity and faultless segregation. TRAIP is essential for cell viability, and mutations in TRAIP ubiquitin ligase activity lead to primordial dwarfism in patients. Here, we have determined the mechanism of inhibition of cell proliferation in TRAIP-depleted cells. We have taken advantage of the auxin induced degron system to rapidly degrade TRAIP within cells and to dissect the importance of various functions of TRAIP in different stages of the cell cycle. We conclude that upon rapid TRAIP degradation, specifically in S-phase, cells cease to proliferate, arrest in G2 stage of the cell cycle and undergo senescence. Our findings reveal that TRAIP works in S-phase to prevent DNA damage at transcription start sites, caused by replication-transcription conflicts.


Assuntos
Ubiquitina-Proteína Ligases , Humanos , Fase S/genética , Divisão Celular/genética , Proliferação de Células/genética , Ciclo Celular , Sobrevivência Celular , Ubiquitina-Proteína Ligases/genética
6.
Nucleic Acids Res ; 51(18): 9748-9763, 2023 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-37638758

RESUMO

Faithful cell division is the basis for the propagation of life and DNA replication must be precisely regulated. DNA replication stress is a prominent endogenous source of genome instability that not only leads to ageing, but also neuropathology and cancer development in humans. Specifically, the issues of how vertebrate cells select and activate origins of replication are of importance as, for example, insufficient origin firing leads to genomic instability and mutations in replication initiation factors lead to the rare human disease Meier-Gorlin syndrome. The mechanism of origin activation has been well characterised and reconstituted in yeast, however, an equal understanding of this process in higher eukaryotes is lacking. The firing of replication origins is driven by S-phase kinases (CDKs and DDK) and results in the activation of the replicative helicase and generation of two bi-directional replication forks. Our data, generated from cell-free Xenopus laevis egg extracts, show that DONSON is required for assembly of the active replicative helicase (CMG complex) at origins during replication initiation. DONSON has previously been shown to be essential during DNA replication, both in human cells and in Drosophila, but the mechanism of DONSON's action was unknown. Here we show that DONSON's presence is essential for replication initiation as it is required for Cdc45 and GINS association with Mcm2-7 complexes and helicase activation. To fulfil this role, DONSON interacts with the initiation factor, TopBP1, in a CDK-dependent manner. Following its initiation role, DONSON also forms a part of the replisome during the elongation stage of DNA replication. Mutations in DONSON have recently been shown to lead to the Meier-Gorlin syndrome; this novel replication initiation role of DONSON therefore provides the explanation for the phenotypes caused by DONSON mutations in patients.


Assuntos
Microtia Congênita , Transtornos do Crescimento , Micrognatismo , Patela , Humanos , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Cromatina/metabolismo , Microtia Congênita/genética , Quinases Ciclina-Dependentes/genética , Replicação do DNA/genética , Transtornos do Crescimento/genética , Micrognatismo/genética , Proteínas de Manutenção de Minicromossomo/metabolismo , Patela/anormalidades , Origem de Replicação/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética
7.
J Biol Chem ; 298(8): 102234, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35798141

RESUMO

Complex cellular processes are driven by the regulated assembly and disassembly of large multiprotein complexes. While we are beginning to understand the molecular mechanism for assembly of the eukaryotic DNA replication machinery (replisome), we still know relatively little about the regulation of its disassembly at replication termination. Recently, the first elements of this process have emerged, revealing that the replicative helicase, at the heart of the replisome, is polyubiquitylated prior to unloading and that this unloading requires p97 segregase activity. Two different E3 ubiquitin ligases have now been shown to ubiquitylate the helicase under different conditions: Cul2Lrr1 and TRAIP. Here, using Xenopus laevis egg extract cell-free system and biochemical approaches, we have found two p97 cofactors, Ubxn7 and Faf1, which can interact with p97 during replisome disassembly during S-phase. We show only Ubxn7, however, facilitates efficient replisome disassembly. Ubxn7 delivers this role through its interaction via independent domains with both Cul2Lrr1 and p97 to allow coupling between Mcm7 ubiquitylation and its removal from chromatin. Our data therefore characterize Ubxn7 as the first substrate-specific p97 cofactor regulating replisome disassembly in vertebrates and a rationale for the efficacy of the Cul2Lrr1 replisome unloading pathway in unperturbed S-phase.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Cromatina , Replicação do DNA , Fase S , Proteínas de Xenopus , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Cromatina/metabolismo , DNA Helicases/genética , DNA Helicases/metabolismo , Componente 7 do Complexo de Manutenção de Minicromossomo/metabolismo , Ubiquitinação , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Xenopus laevis/metabolismo
8.
EMBO Rep ; 22(5): e51120, 2021 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-33779025

RESUMO

Replication stress, a major cause of genome instability in cycling cells, is mainly prevented by the ATR-dependent replication stress response pathway in somatic cells. However, the replication stress response pathway in embryonic stem cells (ESCs) may be different due to alterations in cell cycle phase length. The transcription factor MYBL2, which is implicated in cell cycle regulation, is expressed a hundred to a thousand-fold more in ESCs compared with somatic cells. Here we show that MYBL2 activates ATM and suppresses replication stress in ESCs. Consequently, loss of MYBL2 or inhibition of ATM or Mre11 in ESCs results in replication fork slowing, increased fork stalling and elevated origin firing. Additionally, we demonstrate that inhibition of CDC7 activity rescues replication stress induced by MYBL2 loss and ATM inhibition, suggesting that uncontrolled new origin firing may underlie the replication stress phenotype resulting from loss/inhibition of MYBL2 and ATM. Overall, our study proposes that in addition to ATR, a MYBL2-MRN-ATM replication stress response pathway functions in ESCs to control DNA replication initiation and prevent genome instability.


Assuntos
Proteínas de Ciclo Celular , Células-Tronco Pluripotentes , Proteínas Mutadas de Ataxia Telangiectasia/genética , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Dano ao DNA , Replicação do DNA , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Células-Tronco Pluripotentes/metabolismo
9.
Biochem Soc Trans ; 48(3): 823-836, 2020 06 30.
Artigo em Inglês | MEDLINE | ID: mdl-32490508

RESUMO

DNA replication is a complex process that needs to be executed accurately before cell division in order to maintain genome integrity. DNA replication is divided into three main stages: initiation, elongation and termination. One of the key events during initiation is the assembly of the replicative helicase at origins of replication, and this mechanism has been very well described over the last decades. In the last six years however, researchers have also focused on deciphering the molecular mechanisms underlying the disassembly of the replicative helicase during termination. Similar to replisome assembly, the mechanism of replisome disassembly is strictly regulated and well conserved throughout evolution, although its complexity increases in higher eukaryotes. While budding yeast rely on just one pathway for replisome disassembly in S phase, higher eukaryotes evolved an additional mitotic pathway over and above the default S phase specific pathway. Moreover, replisome disassembly has been recently found to be a key event prior to the repair of certain DNA lesions, such as under-replicated DNA in mitosis and inter-strand cross-links (ICLs) in S phase. Although replisome disassembly in human cells has not been characterised yet, they possess all of the factors involved in these pathways in model organisms, and de-regulation of many of them are known to contribute to tumorigenesis and other pathological conditions.


Assuntos
Replicação do DNA , Neoplasias/metabolismo , Fase S , Saccharomycetales , Animais , Caenorhabditis elegans , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Sobrevivência Celular , DNA/metabolismo , Genoma , Humanos , Componente 7 do Complexo de Manutenção de Minicromossomo/metabolismo , Mitose , Mapeamento de Interação de Proteínas , Origem de Replicação , Saccharomyces cerevisiae , Saccharomycetales/metabolismo , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Xenopus laevis
10.
Life Sci Alliance ; 2(2)2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30979826

RESUMO

We have shown previously that the process of replication machinery (replisome) disassembly at the termination of DNA replication forks in the S-phase is driven through polyubiquitylation of one of the replicative helicase subunits (Mcm7) by Cul2LRR1 ubiquitin ligase. Interestingly, upon inhibition of this pathway in Caenorhabditis elegans embryos, the replisomes retained on chromatin were unloaded in the subsequent mitosis. Here, we show that this mitotic replisome disassembly pathway exists in Xenopus laevis egg extract and we determine the first elements of its regulation. The mitotic disassembly pathway depends on the formation of K6- and K63-linked ubiquitin chains on Mcm7 by TRAIP ubiquitin ligase and the activity of p97/VCP protein segregase. Unlike in lower eukaryotes, however, it does not require SUMO modifications. Importantly, we also show that this process can remove all replisomes from mitotic chromatin, including stalled ones, which indicates a wide application for this pathway over being just a "backup" for terminated replisomes. Finally, we characterise the composition of the replisome retained on chromatin until mitosis.


Assuntos
Replicação do DNA , Mitose/genética , Ubiquitina-Proteína Ligases/metabolismo , Xenopus laevis/genética , Animais , Afidicolina/farmacologia , Cafeína/farmacologia , Cromatina/metabolismo , Ciclinas/metabolismo , Ciclopentanos/farmacologia , Feminino , Masculino , Componente 7 do Complexo de Manutenção de Minicromossomo/metabolismo , Óvulo/efeitos dos fármacos , Pirimidinas/farmacologia , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Espermatozoides/efeitos dos fármacos , Enzimas de Conjugação de Ubiquitina/metabolismo , Ubiquitinação/fisiologia , Proteína com Valosina/metabolismo , Proteínas de Xenopus/metabolismo
11.
Nat Commun ; 10(1): 691, 2019 02 11.
Artigo em Inglês | MEDLINE | ID: mdl-30741925

RESUMO

Most metazoan embryos commence development with rapid, transcriptionally silent cell divisions, with genome activation delayed until the mid-blastula transition (MBT). However, a set of genes escapes global repression and gets activated before MBT. Here we describe the formation and the spatio-temporal dynamics of a pair of distinct transcription compartments, which encompasses the earliest gene expression in zebrafish. 4D imaging of pri-miR430 and zinc-finger-gene activities by a novel, native transcription imaging approach reveals transcriptional sharing of nuclear compartments, which are regulated by homologous chromosome organisation. These compartments carry the majority of nascent-RNAs and active Polymerase II, are chromatin-depleted and represent the main sites of detectable transcription before MBT. Transcription occurs during the S-phase of increasingly permissive cleavage cycles. It is proposed, that the transcription compartment is part of the regulatory architecture of embryonic nuclei and offers a transcriptionally competent environment to facilitate early escape from repression before global genome activation.


Assuntos
Ciclo Celular/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Genoma/genética , Transcrição Gênica/genética , Animais , Blastocisto/fisiologia , Blástula/diagnóstico por imagem , Blástula/fisiologia , Ciclo Celular/fisiologia , Divisão Celular , Núcleo Celular/fisiologia , Cromatina , Cromossomos , Tomografia Computadorizada Quadridimensional , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Genoma/fisiologia , MicroRNAs , Modelos Animais , Fase S/fisiologia , Análise Espaço-Temporal , Transcrição Gênica/fisiologia , Transcriptoma/genética , Peixe-Zebra/genética , Zigoto/fisiologia
12.
Nat Cell Biol ; 19(5): 468-479, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28368371

RESUMO

Replisome disassembly is the final step of DNA replication in eukaryotes, involving the ubiquitylation and CDC48-dependent dissolution of the CMG helicase (CDC45-MCM-GINS). Using Caenorhabditis elegans early embryos and Xenopus laevis egg extracts, we show that the E3 ligase CUL-2LRR-1 associates with the replisome and drives ubiquitylation and disassembly of CMG, together with the CDC-48 cofactors UFD-1 and NPL-4. Removal of CMG from chromatin in frog egg extracts requires CUL2 neddylation, and our data identify chromatin recruitment of CUL2LRR1 as a key regulated step during DNA replication termination. Interestingly, however, CMG persists on chromatin until prophase in worms that lack CUL-2LRR-1, but is then removed by a mitotic pathway that requires the CDC-48 cofactor UBXN-3, orthologous to the human tumour suppressor FAF1. Partial inactivation of lrr-1 and ubxn-3 leads to synthetic lethality, suggesting future approaches by which a deeper understanding of CMG disassembly in metazoa could be exploited therapeutically.


Assuntos
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/enzimologia , Proteínas de Transporte/metabolismo , Cromatina/enzimologia , Proteínas Culina/metabolismo , DNA/biossíntese , Mitose , Fase S , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Animais , Animais Geneticamente Modificados , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Proteínas de Transporte/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Cromatina/genética , Montagem e Desmontagem da Cromatina , Proteínas Culina/genética , DNA/genética , Genótipo , Complexos Multiproteicos , Oócitos , Fenótipo , Interferência de RNA , Fatores de Tempo , Ubiquitinação , Proteína com Valosina , Proteínas de Xenopus/genética , Proteínas de Xenopus/metabolismo , Xenopus laevis/genética , Xenopus laevis/metabolismo
13.
Adv Exp Med Biol ; 1042: 163-187, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29357058

RESUMO

Termination of DNA replication forks takes place when two replication forks coming from neighbouring origins meet each other usually in the midpoint of the replicon. At this stage, the remaining fragments of DNA have to be unwound, all remaining DNA replicated and newly synthesised strands ligated to produce continuous sister chromatids. Finally, the replication machinery has to be taken off, chromatin re-assembled, and entwisted sister chromatids resolved topologically.Over the last few decades, we have learned a lot about the assembly of the helicase and replisome and the initiation stage of DNA replication. We also know much more about the ability of forks to cope with replication stress. However, only within recent years we have gained the first glimpse of the mechanism of replication fork termination. In this chapter I will summarise the recent findings on replication termination, weigh this against the past literature and discuss relevant consequences and views for the future.


Assuntos
Cromátides/genética , Replicação do DNA/fisiologia , Eucariotos/genética , Células Eucarióticas/metabolismo , Animais , Cromátides/metabolismo , Proteínas Culina/metabolismo , Proteínas Culina/fisiologia , DNA/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/fisiologia , Humanos , Ubiquitinação
14.
Cell Cycle ; 15(16): 2183-2195, 2016 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-27327991

RESUMO

During S phase, following activation of the S phase CDKs and the DBF4-dependent kinases (DDK), double hexamers of Mcm2-7 at licensed replication origins are activated to form the core replicative helicase. Mcm10 is one of several proteins that have been implicated from work in yeasts to play a role in forming a mature replisome during the initiation process. Mcm10 has also been proposed to play a role in promoting replisome stability after initiation has taken place. The role of Mcm10 is particularly unclear in metazoans, where conflicting data has been presented. Here, we investigate the role and regulation of Mcm10 in Xenopus egg extracts. We show that Xenopus Mcm10 is recruited to chromatin late in the process of replication initiation and this requires prior action of DDKs and CDKs. We also provide evidence that Mcm10 is a CDK substrate but does not need to be phosphorylated in order to associate with chromatin. We show that in extracts depleted of more than 99% of Mcm10, the bulk of DNA replication still occurs, suggesting that Mcm10 is not required for the process of replication initiation. However, in extracts depleted of Mcm10, the replication fork elongation rate is reduced. Furthermore, the absence of Mcm10 or its phosphorylation by CDK results in instability of replisome proteins on DNA, which is particularly important under conditions of replication stress.


Assuntos
Quinases Ciclina-Dependentes/metabolismo , Replicação do DNA , Proteínas de Manutenção de Minicromossomo/metabolismo , Xenopus laevis/metabolismo , Sequência de Aminoácidos , Animais , Cromatina/metabolismo , Proteínas de Manutenção de Minicromossomo/química , Modelos Biológicos , Fosforilação , Ligação Proteica , Fase S , Especificidade por Substrato
15.
Genes (Basel) ; 6(3): 451-68, 2015 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-26121093

RESUMO

Posttranslational modification of proteins by means of attachment of a small globular protein ubiquitin (i.e., ubiquitylation) represents one of the most abundant and versatile mechanisms of protein regulation employed by eukaryotic cells. Ubiquitylation influences almost every cellular process and its key role in coordination of the DNA damage response is well established. In this review we focus, however, on the ways ubiquitylation controls the process of unperturbed DNA replication. We summarise the accumulated knowledge showing the leading role of ubiquitin driven protein degradation in setting up conditions favourable for replication origin licensing and S-phase entry. Importantly, we also present the emerging major role of ubiquitylation in coordination of the active DNA replication process: preventing re-replication, regulating the progression of DNA replication forks, chromatin re-establishment and disassembly of the replisome at the termination of replication forks.

16.
Nucleus ; 6(3): 187-96, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25835602

RESUMO

To ensure duplication of the entire genome, eukaryotic DNA replication initiates from thousands of replication origins. The replication forks move through the chromatin until they encounter forks from neighboring origins. During replication fork termination forks converge, the replisomes disassemble and topoisomerase II resolves the daughter DNA molecules. If not resolved efficiently, terminating forks result in genomic instability through the formation of pathogenic structures. Our recent findings shed light onto the mechanism of replisome disassembly upon replication fork termination. We have shown that termination-specific polyubiquitylation of the replicative helicase component - Mcm7, leads to dissolution of the active helicase in a process dependent on the p97/VCP/Cdc48 segregase. The inhibition of terminating helicase disassembly resulted in a replication termination defect. In this extended view we present hypothetical models of replication fork termination and discuss remaining and emerging questions in the DNA replication termination field.


Assuntos
Adenosina Trifosfatases/metabolismo , Proteínas de Ciclo Celular/metabolismo , Cromatina/metabolismo , Replicação do DNA , DNA Topoisomerases Tipo II/metabolismo , Proteínas de Manutenção de Minicromossomo/metabolismo , Adenosina Trifosfatases/genética , Animais , Proteínas de Ciclo Celular/genética , Cromatina/química , DNA/genética , DNA/metabolismo , DNA Polimerase I/genética , DNA Polimerase I/metabolismo , DNA Polimerase II/genética , DNA Polimerase II/metabolismo , DNA Polimerase III/genética , DNA Polimerase III/metabolismo , DNA Topoisomerases Tipo II/genética , Regulação da Expressão Gênica , Instabilidade Genômica , Humanos , Proteínas de Manutenção de Minicromossomo/genética , Poliubiquitina/genética , Poliubiquitina/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae , Transdução de Sinais , Ubiquitinação , Proteína com Valosina , Xenopus laevis/genética , Xenopus laevis/metabolismo
17.
Science ; 346(6208): 477-81, 2014 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-25342805

RESUMO

Resolution of replication forks during termination of DNA replication is essential for accurate duplication of eukaryotic genomes. Here we present evidence consistent with the idea that polyubiquitylation of a replisome component (Mcm7) leads to its disassembly at the converging terminating forks because of the action of the p97/VCP/Cdc48 protein remodeler. Using Xenopus laevis egg extract, we have shown that blocking polyubiquitylation results in the prolonged association of the active helicase with replicating chromatin. The Mcm7 subunit is the only component of the active helicase that we find polyubiquitylated during replication termination. The observed polyubiquitylation is followed by disassembly of the active helicase dependent on p97/VCP/Cdc48. Altogether, our data provide insight into the mechanism of replisome disassembly during eukaryotic DNA replication termination.


Assuntos
Adenosina Trifosfatases/metabolismo , Proteínas de Ciclo Celular/metabolismo , Replicação do DNA , Componente 7 do Complexo de Manutenção de Minicromossomo/metabolismo , Ubiquitina/metabolismo , Ubiquitinação , Animais , Cromatina/metabolismo , DNA Helicases/metabolismo , Ubiquitina/genética , Proteína com Valosina , Xenopus laevis
18.
Cell Cycle ; 12(8): 1225-32, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23518502

RESUMO

Hexameric complexes of the six related Mcm2-7 proteins form the core of the replicative helicase. Two other proteins, Mcm8 and Mcm9, with significant homology to Mcm2-7 were first shown to play distinct roles during DNA replication in Xenopus laevis egg extract. Recent work has revealed that Mcm8 and 9 form a complex that plays a role during homologous recombination in human, chicken and mouse cells. We have therefore re-examined the behavior of the Xenopus homologs of these proteins. We show that Mcm8 and Mcm9 form a dimeric complex in Xenopus egg extract. They both associate with chromatin at later stages of DNA replication, and this association is stimulated by DNA damage, suggesting that their function is analogous to the one described in higher eukaryotes. In contrast to previous reports, we do not find Mcm9 essential for loading of Mcm2-7 complex onto chromatin during origin licensing nor detect its interaction with Cdt1 origin licensing factor. Altogether, we conclude that the role Mcm8 and Mcm9 play in Xenopus egg extract is not different from recent findings in higher eukaryotes, consistent with an evolutionary conservation of their function.


Assuntos
Extratos Celulares/análise , DNA Helicases/metabolismo , Proteínas de Ligação a DNA/metabolismo , Recombinação Homóloga/fisiologia , Complexos Multiproteicos/metabolismo , Óvulo/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/metabolismo , Animais , Western Blotting , Cromatina/metabolismo , Replicação do DNA/fisiologia , Dimerização , Imunoprecipitação , Proteínas de Manutenção de Minicromossomo
19.
Mol Cell Biol ; 33(8): 1632-44, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23401855

RESUMO

The minichromosome maintenance protein homologs MCM8 and MCM9 have previously been implicated in DNA replication elongation and prereplication complex (pre-RC) formation, respectively. We found that MCM8 and MCM9 physically associate with each other and that MCM8 is required for the stability of MCM9 protein in mammalian cells. Depletion of MCM8 or MCM9 in human cancer cells or the loss of function MCM9 mutation in mouse embryo fibroblasts sensitizes cells to the DNA interstrand cross-linking (ICL) agent cisplatin. Consistent with a role in the repair of ICLs by homologous recombination (HR), knockdown of MCM8 or MCM9 significantly reduces HR repair efficiency. Chromatin immunoprecipitation analysis using human DR-GFP cells or Xenopus egg extract demonstrated that MCM8 and MCM9 proteins are rapidly recruited to DNA damage sites and promote RAD51 recruitment. Thus, these two metazoan-specific MCM homologs are new components of HR and may represent novel targets for treating cancer in combination with DNA cross-linking agents.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Dano ao DNA/genética , Proteínas de Ligação a DNA/metabolismo , Recombinação Homóloga , Rad51 Recombinase/metabolismo , Reparo de DNA por Recombinação , Animais , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Sobrevivência Celular , Imunoprecipitação da Cromatina , Cisplatino/farmacologia , Proteínas de Ligação a DNA/genética , Proteínas de Fluorescência Verde , Células HEK293 , Células HeLa , Humanos , Camundongos , Proteínas de Manutenção de Minicromossomo , Interferência de RNA , RNA Interferente Pequeno , Xenopus
20.
Methods ; 57(2): 203-13, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22521908

RESUMO

The use of cell-free extracts prepared from eggs of the South African clawed toad, Xenopus laevis, has led to many important discoveries in cell cycle research. These egg extracts recapitulate the key nuclear transitions of the eukaryotic cell cycle in vitro under apparently the same controls that exist in vivo. DNA added to the extract is first assembled into a nucleus and is then efficiently replicated. Progression of the extract into mitosis then allows the separation of paired sister chromatids. The Xenopus cell-free system is therefore uniquely suited to the study of the mechanisms, dynamics and integration of cell cycle regulated processes at a biochemical level. In this article we describe methods currently in use in our laboratory for the preparation of Xenopus egg extracts and demembranated sperm nuclei for the study of DNA replication in vitro. We also detail how DNA replication can be quantified in this system. In addition, we describe methods for isolating chromatin and chromatin-bound protein complexes from egg extracts. These recently developed and revised techniques provide a practical starting point for investigating the function of proteins involved in DNA replication.


Assuntos
Extratos Celulares/genética , Cromatina/isolamento & purificação , Replicação do DNA , Proteínas de Ligação a DNA/isolamento & purificação , Proteínas de Xenopus/isolamento & purificação , Animais , Extratos Celulares/isolamento & purificação , Núcleo Celular/metabolismo , Precipitação Química , Cromatina/genética , Cromatina/metabolismo , DNA/genética , DNA/isolamento & purificação , DNA/metabolismo , Proteínas de Ligação a DNA/metabolismo , Feminino , Masculino , Técnicas de Transferência Nuclear , Oócitos/química , Oócitos/citologia , Espermatozoides/citologia , Testículo/citologia , Ácido Tricloroacético/química , Proteínas de Xenopus/metabolismo , Xenopus laevis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...