Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; 300(1): 105588, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38141767

RESUMO

Proliferating cell nuclear antigen (PCNA) is a homo-trimeric clamp complex that serves as the molecular hub for various DNA transactions, including DNA synthesis and post-replicative mismatch repair. Its timely loading and unloading are critical for genome stability. PCNA loading is catalyzed by Replication factor C (RFC) and the Ctf18 RFC-like complex (Ctf18-RLC), and its unloading is catalyzed by Atad5/Elg1-RLC. However, RFC, Ctf18-RLC, and even some subcomplexes of their shared subunits are capable of unloading PCNA in vitro, leaving an ambiguity in the division of labor in eukaryotic clamp dynamics. By using a system that specifically detects PCNA unloading, we show here that Atad5-RLC, which accounts for only approximately 3% of RFC/RLCs, nevertheless provides the major PCNA unloading activity in Xenopus egg extracts. RFC and Ctf18-RLC each account for approximately 40% of RFC/RLCs, while immunodepletion of neither Rfc1 nor Ctf18 detectably affects the rate of PCNA unloading in our system. PCNA unloading is dependent on the ATP-binding motif of Atad5, independent of nicks on DNA and chromatin assembly, and inhibited effectively by PCNA-interacting peptides. These results support a model in which Atad5-RLC preferentially unloads DNA-bound PCNA molecules that are free from their interactors.


Assuntos
ATPases Associadas a Diversas Atividades Celulares , Proteínas de Ligação a DNA , Antígeno Nuclear de Célula em Proliferação , Animais , DNA , Replicação do DNA , Antígeno Nuclear de Célula em Proliferação/genética , Antígeno Nuclear de Célula em Proliferação/metabolismo , Proteína de Replicação C/genética , Proteína de Replicação C/metabolismo , Xenopus laevis/metabolismo , Oócitos , ATPases Associadas a Diversas Atividades Celulares/genética , ATPases Associadas a Diversas Atividades Celulares/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo
2.
Mol Cell ; 50(5): 661-74, 2013 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-23746350

RESUMO

Centromeres play several important roles in ensuring proper chromosome segregation. Not only do they promote kinetochore assembly for microtubule attachment, but they also support robust sister chromatid cohesion at pericentromeres and facilitate replication of centromeric DNA early in S phase. However, it is still elusive how centromeres orchestrate all these functions at the same site. Here, we show that the budding yeast Dbf4-dependent kinase (DDK) accumulates at kinetochores in telophase, facilitated by the Ctf19 kinetochore complex. This promptly recruits Sld3-Sld7 replication initiator proteins to pericentromeric replication origins so that they initiate replication early in S phase. Furthermore, DDK at kinetochores independently recruits the Scc2-Scc4 cohesin loader to centromeres in G1 phase. This enhances cohesin loading and facilitates robust pericentromeric cohesion in S phase. Thus, we have found the central mechanism by which kinetochores orchestrate early S phase DNA replication and robust sister chromatid cohesion at microtubule attachment sites.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Replicação do DNA , Cinetocoros/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular/genética , Centrômero/genética , Centrômero/metabolismo , Cromátides/genética , Cromátides/metabolismo , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Proteínas do Citoesqueleto/genética , Proteínas do Citoesqueleto/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas Serina-Treonina Quinases/genética , Fase S/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética
3.
Genes Dev ; 27(23): 2537-42, 2013 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-24298053

RESUMO

Deregulated origin licensing and rereplication promote genome instability and tumorigenesis by largely elusive mechanisms. Investigating the consequences of Early mitotic inhibitor 1 (Emi1) depletion in human cells, previously associated with rereplication, we show by DNA fiber labeling that origin reactivation occurs rapidly, well before accumulation of cells with >4N DNA, and is associated with checkpoint-blind ssDNA gaps and replication fork reversal. Massive RPA chromatin loading, formation of small chromosomal fragments, and checkpoint activation occur only later, once cells complete bulk DNA replication. We propose that deregulated origin firing leads to undetected discontinuities on newly replicated DNA, which ultimately cause breakage of rereplicating forks.


Assuntos
Quebra Cromossômica , Replicação do DNA/genética , Origem de Replicação/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , DNA/biossíntese , Proteínas F-Box/genética , Proteínas F-Box/metabolismo , Humanos , RNA Interferente Pequeno/metabolismo , Moldes Genéticos
4.
Mol Cell ; 44(1): 85-96, 2011 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-21981920

RESUMO

Faithful transmission of genomic information requires tight spatiotemporal regulation of DNA replication factors. In the licensing step of DNA replication, CDT-1 is loaded onto chromatin to subsequently promote the recruitment of additional replication factors, including CDC-45 and GINS. During the elongation step, the CDC-45/GINS complex moves with the replication fork; however, it is largely unknown how its chromatin association is regulated. Here, we show that the chaperone-like ATPase CDC-48/p97 coordinates degradation of CDT-1 with release of the CDC-45/GINS complex. C. elegans embryos lacking CDC-48 or its cofactors UFD-1/NPL-4 accumulate CDT-1 on mitotic chromatin, indicating a critical role of CDC-48 in CDT-1 turnover. Strikingly, CDC-48(UFD-1/NPL-4)-deficient embryos show persistent chromatin association of CDC-45/GINS, which is a consequence of CDT-1 stabilization. Moreover, our data confirmed a similar regulation in Xenopus egg extracts, emphasizing a conserved coordination of licensing and elongation events during eukaryotic DNA replication by CDC-48/p97.


Assuntos
Adenosina Trifosfatases/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Ciclo Celular/metabolismo , Cromatina/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Replicação do DNA , Ligases/metabolismo , Proteínas de Xenopus/metabolismo , Animais , Caenorhabditis elegans , Masculino , Mitose , Interferência de RNA , Espermatozoides/metabolismo , Técnicas do Sistema de Duplo-Híbrido , Ubiquitina/química , Ubiquitina/metabolismo , Proteína com Valosina , Xenopus laevis
5.
PLoS Genet ; 12(3): e1005872, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27010650

RESUMO

Meiotic recombination is essential for the repair of programmed double strand breaks (DSBs) to generate crossovers (COs) during meiosis. The efficient processing of meiotic recombination intermediates not only needs various resolvases but also requires proper meiotic chromosome structure. The Smc5/6 complex belongs to the structural maintenance of chromosome (SMC) family and is closely related to cohesin and condensin. Although the Smc5/6 complex has been implicated in the processing of recombination intermediates during meiosis, it is not known how Smc5/6 controls meiotic DSB repair. Here, using Caenorhabditis elegans we show that the SMC-5/6 complex acts synergistically with HIM-6, an ortholog of the human Bloom syndrome helicase (BLM) during meiotic recombination. The concerted action of the SMC-5/6 complex and HIM-6 is important for processing recombination intermediates, CO regulation and bivalent maturation. Careful examination of meiotic chromosomal morphology reveals an accumulation of inter-chromosomal bridges in smc-5; him-6 double mutants, leading to compromised chromosome segregation during meiotic cell divisions. Interestingly, we found that the lethality of smc-5; him-6 can be rescued by loss of the conserved BRCA1 ortholog BRC-1. Furthermore, the combined deletion of smc-5 and him-6 leads to an irregular distribution of condensin and to chromosome decondensation defects reminiscent of condensin depletion. Lethality conferred by condensin depletion can also be rescued by BRC-1 depletion. Our results suggest that SMC-5/6 and HIM-6 can synergistically regulate recombination intermediate metabolism and suppress ectopic recombination by controlling chromosome architecture during meiosis.


Assuntos
Proteínas de Caenorhabditis elegans/genética , Proteínas de Ciclo Celular/genética , Meiose/genética , Recombinação Genética , Animais , Caenorhabditis elegans/genética , Cromossomos/genética , Humanos , Complexos Multiproteicos/genética
6.
Proc Natl Acad Sci U S A ; 113(39): E5765-74, 2016 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-27630194

RESUMO

The replication of DNA is initiated at particular sites on the genome called replication origins (ROs). Understanding the constraints that regulate the distribution of ROs across different organisms is fundamental for quantifying the degree of replication errors and their downstream consequences. Using a simple probabilistic model, we generate a set of predictions on the extreme sensitivity of error rates to the distribution of ROs, and how this distribution must therefore be tuned for genomes of vastly different sizes. As genome size changes from megabases to gigabases, we predict that regularity of RO spacing is lost, that large gaps between ROs dominate error rates but are heavily constrained by the mean stalling distance of replication forks, and that, for genomes spanning ∼100 megabases to ∼10 gigabases, errors become increasingly inevitable but their number remains very small (three or less). Our theory predicts that the number of errors becomes significantly higher for genome sizes greater than ∼10 gigabases. We test these predictions against datasets in yeast, Arabidopsis, Drosophila, and human, and also through direct experimentation on two different human cell lines. Agreement of theoretical predictions with experiment and datasets is found in all cases, resulting in a picture of great simplicity, whereby the density and positioning of ROs explain the replication error rates for the entire range of eukaryotes for which data are available. The theory highlights three domains of error rates: negligible (yeast), tolerable (metazoan), and high (some plants), with the human genome at the extreme end of the middle domain.


Assuntos
Pareamento de Bases/genética , Replicação do DNA , Eucariotos/genética , Genoma Humano , Animais , Arabidopsis/genética , DNA/genética , Replicação do DNA/genética , Drosophila melanogaster/genética , Células HeLa , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Origem de Replicação/genética , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/metabolismo
7.
Proc Natl Acad Sci U S A ; 113(39): E5757-64, 2016 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-27516545

RESUMO

To prevent rereplication of genomic segments, the eukaryotic cell cycle is divided into two nonoverlapping phases. During late mitosis and G1 replication origins are "licensed" by loading MCM2-7 double hexamers and during S phase licensed replication origins activate to initiate bidirectional replication forks. Replication forks can stall irreversibly, and if two converging forks stall with no intervening licensed origin-a "double fork stall" (DFS)-replication cannot be completed by conventional means. We previously showed how the distribution of replication origins in yeasts promotes complete genome replication even in the presence of irreversible fork stalling. This analysis predicts that DFSs are rare in yeasts but highly likely in large mammalian genomes. Here we show that complementary strand synthesis in early mitosis, ultrafine anaphase bridges, and G1-specific p53-binding protein 1 (53BP1) nuclear bodies provide a mechanism for resolving unreplicated DNA at DFSs in human cells. When origin number was experimentally altered, the number of these structures closely agreed with theoretical predictions of DFSs. The 53BP1 is preferentially bound to larger replicons, where the probability of DFSs is higher. Loss of 53BP1 caused hypersensitivity to licensing inhibition when replication origins were removed. These results provide a striking convergence of experimental and theoretical evidence that unreplicated DNA can pass through mitosis for resolution in the following cell cycle.


Assuntos
DNA/metabolismo , Mitose , Fase S , Brônquios/citologia , Proteínas de Ciclo Celular/metabolismo , Células Epiteliais/metabolismo , Loci Gênicos , Células HeLa , Histonas/metabolismo , Humanos , Proteínas Nucleares/metabolismo , Interferência de RNA , Origem de Replicação , Proteína 1 de Ligação à Proteína Supressora de Tumor p53/metabolismo
8.
Mol Cell ; 37(1): 5-6, 2010 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-20129050

RESUMO

In this issue of Molecular Cell, Miotto and Struhl (2010) suggest that replication licensing, the loading of Mcm2-7 onto DNA, is promoted by HBO1 acetylating histone H4 at replication origins, providing a molecular view of how chromatin status influences origin usage.


Assuntos
Replicação do DNA , Histona Acetiltransferases/fisiologia , Histonas/metabolismo , Modelos Genéticos , Acetilação , Cromatina/metabolismo , Humanos
9.
Biochim Biophys Acta ; 1863(4): 673-85, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26732297

RESUMO

NF-Y is a heterotrimeric transcription factor, which plays a pioneer role in the transcriptional control of promoters containing the CCAAT-box, among which genes involved in cell cycle regulation, apoptosis and DNA damage response. The knock-down of the sequence-specific subunit NF-YA triggers defects in S-phase progression, which lead to apoptotic cell death. Here, we report that NF-Y has a critical function in DNA replication progression, independent from its transcriptional activity. NF-YA colocalizes with early DNA replication factories, its depletion affects the loading of replisome proteins to DNA, among which Cdc45, and delays the passage from early to middle-late S phase. Molecular combing experiments are consistent with a role for NF-Y in the control of fork progression. Finally, we unambiguously demonstrate a direct non-transcriptional role of NF-Y in the overall efficiency of DNA replication, specifically in the DNA elongation process, using a Xenopus cell-free system. Our findings broaden the activity of NF-Y on a DNA metabolism other than transcription, supporting the existence of specific TFs required for proper and efficient DNA replication.


Assuntos
Fator de Ligação a CCAAT/fisiologia , Replicação do DNA/genética , Animais , Fator de Ligação a CCAAT/genética , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , DNA/metabolismo , Células HCT116 , Humanos , Regiões Promotoras Genéticas , Fase S/genética , Elongação da Transcrição Genética , Transcrição Gênica , Xenopus laevis
10.
Trends Biochem Sci ; 36(8): 405-14, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21641805

RESUMO

Many replication origins that are licensed by loading MCM2-7 complexes in G1 are not normally used. Activation of these dormant origins during S phase provides a first line of defence for the genome if replication is inhibited. When replication forks fail, dormant origins are activated within regions of the genome currently engaged in replication. At the same time, DNA damage-response kinases activated by the stalled forks preferentially suppress the assembly of new replication factories, thereby ensuring that chromosomal regions experiencing replicative stress complete synthesis before new regions of the genome are replicated. Mice expressing reduced levels of MCM2-7 have fewer dormant origins, are cancer-prone and are genetically unstable, demonstrating the importance of dormant origins for preserving genome integrity. We review the function of dormant origins, the molecular mechanism of their regulation and their physiological implications.


Assuntos
Proteínas de Ciclo Celular/genética , Ciclo Celular/genética , Replicação do DNA/genética , Proteínas Nucleares/genética , Origem de Replicação/genética , Elementos Silenciadores Transcricionais/genética , Animais , Proteínas de Ligação a DNA/genética , Genes Supressores de Tumor , Camundongos , Componente 2 do Complexo de Manutenção de Minicromossomo , Componente 3 do Complexo de Manutenção de Minicromossomo , Componente 4 do Complexo de Manutenção de Minicromossomo , Componente 6 do Complexo de Manutenção de Minicromossomo , Componente 7 do Complexo de Manutenção de Minicromossomo , Fase S/genética , Processos Estocásticos
11.
Development ; 139(1): 63-74, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22096080

RESUMO

Cdt1 plays a key role in licensing DNA for replication. In the somatic cells of metazoans, both Cdt1 and its natural inhibitor geminin show reciprocal fluctuations in their protein levels owing to cell cycle-dependent proteolysis. Here, we show that the protein levels of Cdt1 and geminin are persistently high during the rapid cell cycles of the early Xenopus embryo. Immunoprecipitation of Cdt1 and geminin complexes, together with their cell cycle spatiotemporal dynamics, strongly supports the hypothesis that Cdt1 licensing activity is regulated by periodic interaction with geminin rather than its proteolysis. Overexpression of ectopic geminin slows down, but neither arrests early embryonic cell cycles nor affects endogenous geminin levels; apparent embryonic lethality is observed around 3-4 hours after mid-blastula transition. However, functional knockdown of geminin by ΔCdt1_193-447, which lacks licensing activity and degradation sequences, causes cell cycle arrest and DNA damage in affected cells. This contributes to subsequent developmental defects in treated embryos. Our results clearly show that rapidly proliferating early Xenopus embryonic cells are able to regulate replication licensing in the persistent presence of high levels of licensing proteins by relying on changing interactions between Cdt1 and geminin during the cell cycle, but not their degradation.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Replicação do DNA/fisiologia , Proteínas de Ligação a DNA/metabolismo , Embrião não Mamífero/fisiologia , Fase S/fisiologia , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriologia , Animais , Cromatina/ultraestrutura , Primers do DNA/genética , Embrião não Mamífero/metabolismo , Geminina , Processamento de Imagem Assistida por Computador , Imunoprecipitação , Microscopia de Fluorescência , Mutagênese Sítio-Dirigida , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
12.
Nucleic Acids Res ; 41(21): 9705-18, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23963700

RESUMO

During S phase, the entire genome must be precisely duplicated, with no sections of DNA left unreplicated. Here, we develop a simple mathematical model to describe the probability of replication failing due to the irreversible stalling of replication forks. We show that the probability of complete genome replication is maximized if replication origins are evenly spaced, the largest inter-origin distances are minimized, and the end-most origins are positioned close to chromosome ends. We show that origin positions in the yeast Saccharomyces cerevisiae genome conform to all three predictions thereby maximizing the probability of complete replication if replication forks stall. Origin positions in four other yeasts-Kluyveromyces lactis, Lachancea kluyveri, Lachancea waltii and Schizosaccharomyces pombe-also conform to these predictions. Equating failure rates at chromosome ends with those in chromosome interiors gives a mean per nucleotide fork stall rate of ∼5 × 10(-8), which is consistent with experimental estimates. Using this value in our theoretical predictions gives replication failure rates that are consistent with data from replication origin knockout experiments. Our theory also predicts that significantly larger genomes, such as those of mammals, will experience a much greater probability of replication failure genome-wide, and therefore will likely require additional compensatory mechanisms.


Assuntos
Genoma Fúngico , Origem de Replicação , Saccharomyces cerevisiae/genética , Cromossomos Fúngicos , Replicação do DNA , DNA Polimerase Dirigida por DNA/metabolismo , Modelos Genéticos , Complexos Multienzimáticos/metabolismo , Saccharomyces cerevisiae/metabolismo , Leveduras/genética
13.
J Biol Chem ; 288(44): 31624-34, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-24064211

RESUMO

Geminin is an important regulator of proliferation and differentiation in metazoans, which predominantly inhibits the DNA replication licensing factor Cdt1, preventing genome over-replication. We show that Geminin preferentially forms stable coiled-coil heterodimers with its homologue, Idas. In contrast to Idas-Geminin heterodimers, Idas homodimers are thermodynamically unstable and are unlikely to exist as a stable macromolecule under physiological conditions. The crystal structure of the homology regions of Idas in complex with Geminin showed a tight head-to-head heterodimeric coiled-coil. This Idas-Geminin heterodimer binds Cdt1 less strongly than Geminin-Geminin, still with high affinity (∼30 nm), but with notably different thermodynamic properties. Consistently, in Xenopus egg extracts, Idas-Geminin is less active in licensing inhibition compared with a Geminin-Geminin homodimer. In human cultured cells, ectopic expression of Idas leads to limited over-replication, which is counteracted by Geminin co-expression. The properties of the Idas-Geminin complex suggest it as the functional form of Idas and provide a possible mechanism to modulate Geminin activity.


Assuntos
Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Replicação do DNA/fisiologia , Geminina/química , Geminina/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Multimerização Proteica/fisiologia , Animais , Proteínas de Ciclo Celular/genética , Linhagem Celular , Geminina/genética , Humanos , Proteínas Nucleares/genética , Estrutura Quaternária de Proteína , Relação Estrutura-Atividade , Fatores de Transcrição , Xenopus laevis
14.
J Cell Sci ; 125(Pt 10): 2436-45, 2012 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-22366459

RESUMO

To prevent re-replication of DNA in a single cell cycle, the licensing of replication origins by Mcm2-7 is prevented during S and G2 phases. Animal cells achieve this by cell-cycle-regulated proteolysis of the essential licensing factor Cdt1 and inhibition of Cdt1 by geminin. Here we investigate the consequences of ablating geminin in synchronised human U2OS cells. Following geminin loss, cells complete an apparently normal S phase, but a proportion arrest at the G2-M boundary. When Cdt1 accumulates in these cells, DNA re-replicates, suggesting that the key role of geminin is to prevent re-licensing in G2. If cell cycle checkpoints are inhibited in cells lacking geminin, cells progress through mitosis and less re-replication occurs. Checkpoint kinases thereby amplify re-replication into an all-or-nothing response by delaying geminin-depleted cells in G2. Deep DNA sequencing revealed no preferential re-replication of specific genomic regions after geminin depletion. This is consistent with the observation that cells in G2 have lost their replication timing information. By contrast, when Cdt1 is overexpressed or is stabilised by the neddylation inhibitor MLN4924, re-replication can occur throughout S phase.


Assuntos
Pontos de Checagem do Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Células/citologia , Replicação do DNA , Fase G2 , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Células/metabolismo , Geminina , Humanos , Componente 2 do Complexo de Manutenção de Minicromossomo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fase S
15.
Mol Cell ; 41(5): 495-6, 2011 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-21362544
16.
Curr Opin Cell Biol ; 18(3): 231-9, 2006 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16650748

RESUMO

Eukaryotic DNA replication is a highly conserved process; the proteins and sequence of events that replicate animal genomes are remarkably similar to those that replicate yeast genomes. Moreover, the assembly of prereplication complexes at DNA replication origins ('DNA licensing') is regulated in all eukaryotes so that no origin fires more than once in a single cell cycle. And yet there are significant differences between species both in the selection of replication origins and in the way in which these origins are licensed to operate. Moreover, these differences impart advantages to multicellular animals and plants that facilitate their development, such as better control over endoreduplication, flexibility in origin selection, and discrimination between quiescent and proliferative states.


Assuntos
Replicação do DNA , Animais , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Divisão Celular , DNA Helicases/metabolismo , Humanos , Modelos Genéticos , Complexo de Reconhecimento de Origem/metabolismo , Plantas/genética , Origem de Replicação
17.
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
18.
Wellcome Open Res ; 8: 158, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37766844

RESUMO

Background: It has been known for many years that in metazoan cells, replication origins are organised into clusters where origins within each cluster fire near-synchronously. Despite clusters being a fundamental organising principle of metazoan DNA replication, the genomic location of origin clusters has not been documented. Methods: We synchronised human U2OS by thymidine block and release followed by L-mimosine block and release to create a population of cells progressing into S phase with a high degree of synchrony. At different times after release into S phase, cells were pulsed with EdU; the EdU-labelled DNA was then pulled down, sequenced and mapped onto the human genome. Results: The early replicating DNA showed features at a range of scales. Wavelet analysis showed that the major feature of the early replicating DNA was at a size of 500 kb, consistent with clusters of replication origins. Over the first two hours of S phase, these Replicon Cluster Domains broadened in width, consistent with their being enlarged by the progression of replication forks at their outer boundaries. The total replication signal associated with each Replicon Cluster Domain varied considerably, and this variation was reproducible and conserved over time. We provide evidence that this variability in replication signal was at least in part caused by Replicon Cluster Domains being activated at different times in different cells in the population. We also provide evidence that adjacent clusters had a statistical preference for being activated in sequence across a group, consistent with the 'domino' model of replication focus activation order observed by microscopy. Conclusions: We show that early replicating DNA is organised into Replicon Cluster Domains that behave as expected of replicon clusters observed by DNA fibre analysis. The coordinated activation of different Replicon Cluster Domains can generate the replication timing programme by which the genome is duplicated.

19.
J Biol Chem ; 286(13): 11855-64, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21282109

RESUMO

In late mitosis and G1, Mcm2-7 are assembled onto replication origins to license them for initiation in the upcoming S phase. After initiation, Mcm2-7 provide helicase activity to unwind DNA at the replication fork. Here we examine the structure of Mcm2-7 on chromatin in Xenopus egg extracts. We show that prior to replication initiation, Mcm2-7 is present at licensed replication origins in a complex with a molecular mass close to double that of the Mcm2-7 hexamer. This complex has approximately stoichiometric quantities of the 6 Mcm2-7 proteins and we conclude that it consists of a double heterohexamer. This provides a configuration potentially capable of initiating a pair of bidirectional replication forks in S phase. We also show that after initiation, Mcm2-7 associate with Cdc45 and GINS to form a relatively stable CMG (Cdc45-MCM-GINS) complex. The CMG proteins also associate less strongly with other replication proteins, consistent with the idea that a single CMG complex forms the core of the replisome.


Assuntos
Adenosina Trifosfatases/metabolismo , Proteínas de Transporte/metabolismo , Replicação do DNA/fisiologia , Complexos Multiproteicos/metabolismo , Oócitos/metabolismo , Proteínas de Xenopus/metabolismo , Adenosina Trifosfatases/genética , Animais , Proteínas de Transporte/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Sistema Livre de Células , Cromatina/metabolismo , Fase G1/fisiologia , Componente 2 do Complexo de Manutenção de Minicromossomo , Componente 7 do Complexo de Manutenção de Minicromossomo , Mitose/fisiologia , Complexos Multiproteicos/genética , Estrutura Quaternária de Proteína , Proteínas de Xenopus/genética , Xenopus laevis
20.
Biochim Biophys Acta ; 1813(6): 1129-36, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21232560

RESUMO

The facilitates chromatin transcription (FACT) complex affects nuclear DNA transactions in a chromatin context. Though the involvement of FACT in eukaryotic DNA replication has been revealed, a clear understanding of its biochemical behavior during DNA replication still remains elusive. Here, we analyzed the chromatin-binding dynamics of FACT using Xenopus egg extract cell-free system. We found that FACT has at least two distinct chromatin-binding phases: (1) a rapid chromatin-binding phase at the onset of DNA replication that did not involve origin licensing and (2) a second phase of chromatin binding that initiated after origin licensing. Intriguingly, early-binding FACT dissociated from chromatin when DNA replication was blocked by the addition of Cdc6 in the licensed state before origin firing. Cdc6-induced removal of FACT was blocked by the inhibition of origin licensing with geminin, but not by suppressing the activity of DNA polymerases, CDK, or Cdc7. Furthermore, chromatin transfer experiments revealed that impairing the later binding of FACT severely compromises DNA replication activity. Taken together, we propose that even though FACT has rapid chromatin-binding activity, the binding pattern of FACT on chromatin changes after origin licensing, which may contribute to the establishment of its functional link to the DNA replication machinery.


Assuntos
Cromatina/metabolismo , Replicação do DNA , Proteínas de Ligação a DNA/metabolismo , Proteínas de Grupo de Alta Mobilidade/metabolismo , Fatores de Elongação da Transcrição/metabolismo , Animais , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Cromatina/genética , Proteínas de Ligação a DNA/genética , Células Eucarióticas/metabolismo , Feminino , Glutationa Transferase/genética , Glutationa Transferase/metabolismo , Proteínas de Grupo de Alta Mobilidade/genética , Chaperonas de Histonas/genética , Chaperonas de Histonas/metabolismo , Humanos , Immunoblotting , Cinética , Masculino , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Oócitos/metabolismo , Ligação Proteica , Espermatozoides/metabolismo , Fatores de Tempo , Fatores de Elongação da Transcrição/genética , Xenopus laevis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA