Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 152
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 300(1): 105588, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38141767

RESUMEN

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.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas , Proteínas de Unión al ADN , Antígeno Nuclear de Célula en Proliferación , Animales , ADN , Replicación del ADN , Antígeno Nuclear de Célula en Proliferación/genética , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteína de Replicación C/genética , Proteína de Replicación C/metabolismo , Xenopus laevis/metabolismo , Oocitos , ATPasas Asociadas con Actividades Celulares Diversas/genética , ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo
2.
Mol Cell ; 50(5): 661-74, 2013 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-23746350

RESUMEN

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.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Replicación del ADN , Cinetocoros/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular/genética , Centrómero/genética , Centrómero/metabolismo , Cromátides/genética , Cromátides/metabolismo , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Fase S/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética
3.
Genes Dev ; 27(23): 2537-42, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24298053

RESUMEN

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.


Asunto(s)
Rotura Cromosómica , Replicación del ADN/genética , Origen de Réplica/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular , ADN/biosíntesis , Proteínas F-Box/genética , Proteínas F-Box/metabolismo , Humanos , ARN Interferente Pequeño/metabolismo , Moldes Genéticos
4.
Phys Biol ; 17(4): 046002, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32320972

RESUMEN

In order to maintain functional robustness and species integrity, organisms must ensure high fidelity of the genome duplication process. This is particularly true during early development, where cell division is often occurring both rapidly and coherently. By studying the extreme limits of suppressing DNA replication failure due to double fork stall errors, we uncover a fundamental constant that describes a trade-off between genome size and architectural complexity of the developing organism. This constant has the approximate value N U ≈ 3 × 1012 basepairs, and depends only on two highly conserved molecular properties of DNA biology. We show that our theory is successful in interpreting a diverse range of data across the Eukaryota.


Asunto(s)
Replicación del ADN , ADN/biosíntesis , ADN/genética , Eucariontes/citología , Eucariontes/genética , Humanos
5.
Mol Cell ; 44(1): 85-96, 2011 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-21981920

RESUMEN

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.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Ciclo Celular/metabolismo , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Replicación del ADN , Ligasas/metabolismo , Proteínas de Xenopus/metabolismo , Animales , Caenorhabditis elegans , Masculino , Mitosis , Interferencia de ARN , Espermatozoides/metabolismo , Técnicas del Sistema de Dos Híbridos , Ubiquitina/química , Ubiquitina/metabolismo , Proteína que Contiene Valosina , Xenopus laevis
6.
PLoS Genet ; 12(3): e1005872, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27010650

RESUMEN

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.


Asunto(s)
Proteínas de Caenorhabditis elegans/genética , Proteínas de Ciclo Celular/genética , Meiosis/genética , Recombinación Genética , Animales , Caenorhabditis elegans/genética , Cromosomas/genética , Humanos , Complejos Multiproteicos/genética
7.
Proc Natl Acad Sci U S A ; 113(39): E5765-74, 2016 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-27630194

RESUMEN

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.


Asunto(s)
Emparejamiento Base/genética , Replicación del ADN , Eucariontes/genética , Genoma Humano , Animales , Arabidopsis/genética , ADN/genética , Replicación del ADN/genética , Drosophila melanogaster/genética , Células HeLa , Células Madre Embrionarias Humanas/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Origen de Réplica/genética , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo
8.
Proc Natl Acad Sci U S A ; 113(39): E5757-64, 2016 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-27516545

RESUMEN

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.


Asunto(s)
ADN/metabolismo , Mitosis , Fase S , Bronquios/citología , Proteínas de Ciclo Celular/metabolismo , Células Epiteliales/metabolismo , Sitios Genéticos , Células HeLa , Histonas/metabolismo , Humanos , Proteínas Nucleares/metabolismo , Interferencia de ARN , Origen de Réplica , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo
9.
Mol Cell ; 37(1): 5-6, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-20129050

RESUMEN

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.


Asunto(s)
Replicación del ADN , Histona Acetiltransferasas/fisiología , Histonas/metabolismo , Modelos Genéticos , Acetilación , Cromatina/metabolismo , Humanos
10.
Biochim Biophys Acta ; 1863(4): 673-85, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26732297

RESUMEN

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.


Asunto(s)
Factor de Unión a CCAAT/fisiología , Replicación del ADN/genética , Animales , Factor de Unión a CCAAT/genética , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , ADN/metabolismo , Células HCT116 , Humanos , Regiones Promotoras Genéticas , Fase S/genética , Elongación de la Transcripción Genética , Transcripción Genética , Xenopus laevis
11.
Trends Biochem Sci ; 36(8): 405-14, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21641805

RESUMEN

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.


Asunto(s)
Proteínas de Ciclo Celular/genética , Ciclo Celular/genética , Replicación del ADN/genética , Proteínas Nucleares/genética , Origen de Réplica/genética , Elementos Silenciadores Transcripcionales/genética , Animales , Proteínas de Unión al ADN/genética , Genes Supresores de Tumor , Ratones , Componente 2 del Complejo de Mantenimiento de Minicromosoma , Componente 3 del Complejo de Mantenimiento de Minicromosoma , Componente 4 del Complejo de Mantenimiento de Minicromosoma , Componente 6 del Complejo de Mantenimiento de Minicromosoma , Componente 7 del Complejo de Mantenimiento de Minicromosoma , Fase S/genética , Procesos Estocásticos
12.
Development ; 139(1): 63-74, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22096080

RESUMEN

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.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Replicación del ADN/fisiología , Proteínas de Unión al ADN/metabolismo , Embrión no Mamífero/fisiología , Fase S/fisiología , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriología , Animales , Cromatina/ultraestructura , Cartilla de ADN/genética , Embrión no Mamífero/metabolismo , Geminina , Procesamiento de Imagen Asistido por Computador , Inmunoprecipitación , Microscopía Fluorescente , Mutagénesis Sitio-Dirigida , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
13.
Nucleic Acids Res ; 41(21): 9705-18, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23963700

RESUMEN

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.


Asunto(s)
Genoma Fúngico , Origen de Réplica , Saccharomyces cerevisiae/genética , Cromosomas Fúngicos , Replicación del ADN , ADN Polimerasa Dirigida por ADN/metabolismo , Modelos Genéticos , Complejos Multienzimáticos/metabolismo , Saccharomyces cerevisiae/metabolismo , Levaduras/genética
14.
J Biol Chem ; 288(44): 31624-34, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-24064211

RESUMEN

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.


Asunto(s)
Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/metabolismo , Replicación del ADN/fisiología , Geminina/química , Geminina/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Multimerización de Proteína/fisiología , Animales , Proteínas de Ciclo Celular/genética , Línea Celular , Geminina/genética , Humanos , Proteínas Nucleares/genética , Estructura Cuaternaria de Proteína , Relación Estructura-Actividad , Factores de Transcripción , Xenopus laevis
15.
J Cell Sci ; 125(Pt 10): 2436-45, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22366459

RESUMEN

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.


Asunto(s)
Puntos de Control del Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Células/citología , Replicación del ADN , Fase G2 , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Células/metabolismo , Geminina , Humanos , Componente 2 del Complejo de Mantenimiento de Minicromosoma , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fase S
16.
Mol Cell ; 41(5): 495-6, 2011 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-21362544
17.
Curr Opin Cell Biol ; 18(3): 231-9, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16650748

RESUMEN

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.


Asunto(s)
Replicación del ADN , Animales , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , División Celular , ADN Helicasas/metabolismo , Humanos , Modelos Genéticos , Complejo de Reconocimiento del Origen/metabolismo , Plantas/genética , Origen de Réplica
18.
Methods ; 57(2): 203-13, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22521908

RESUMEN

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.


Asunto(s)
Extractos Celulares/genética , Cromatina/aislamiento & purificación , Replicación del ADN , Proteínas de Unión al ADN/aislamiento & purificación , Proteínas de Xenopus/aislamiento & purificación , Animales , Extractos Celulares/aislamiento & purificación , Núcleo Celular/metabolismo , Precipitación Química , Cromatina/genética , Cromatina/metabolismo , ADN/genética , ADN/aislamiento & purificación , ADN/metabolismo , Proteínas de Unión al ADN/metabolismo , Femenino , Masculino , Técnicas de Transferencia Nuclear , Oocitos/química , Oocitos/citología , Espermatozoides/citología , Testículo/citología , Ácido Tricloroacético/química , Proteínas de Xenopus/metabolismo , Xenopus laevis
19.
Wellcome Open Res ; 8: 158, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37766844

RESUMEN

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.

20.
J Biol Chem ; 286(13): 11855-64, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21282109

RESUMEN

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.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas Portadoras/metabolismo , Replicación del ADN/fisiología , Complejos Multiproteicos/metabolismo , Oocitos/metabolismo , Proteínas de Xenopus/metabolismo , Adenosina Trifosfatasas/genética , Animales , Proteínas Portadoras/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Sistema Libre de Células , Cromatina/metabolismo , Fase G1/fisiología , Componente 2 del Complejo de Mantenimiento de Minicromosoma , Componente 7 del Complejo de Mantenimiento de Minicromosoma , Mitosis/fisiología , Complejos Multiproteicos/genética , Estructura Cuaternaria de Proteína , Proteínas de Xenopus/genética , Xenopus laevis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA