Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
EMBO J ; 42(20): e110844, 2023 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-37661798

RESUMEN

Homologous recombination (HR) is a prominent DNA repair pathway maintaining genome integrity. Mutations in many HR genes lead to cancer predisposition. Paradoxically, the implication of the pivotal HR factor RAD51 on cancer development remains puzzling. Particularly, no RAD51 mouse models are available to address the role of RAD51 in aging and carcinogenesis in vivo. We engineered a mouse model with an inducible dominant-negative form of RAD51 (SMRad51) that suppresses RAD51-mediated HR without stimulating alternative mutagenic repair pathways. We found that in vivo expression of SMRad51 led to replicative stress, systemic inflammation, progenitor exhaustion, premature aging and reduced lifespan, but did not trigger tumorigenesis. Expressing SMRAD51 in a breast cancer predisposition mouse model (PyMT) decreased the number and the size of tumors, revealing an anti-tumor activity of SMRAD51. We propose that these in vivo phenotypes result from chronic endogenous replication stress caused by HR decrease, which preferentially targets progenitors and tumor cells. Our work underlines the importance of RAD51 activity for progenitor cell homeostasis, preventing aging and more generally for the balance between cancer and aging.


Asunto(s)
Neoplasias , Recombinasa Rad51 , Animales , Ratones , Envejecimiento/genética , Carcinogénesis/genética , Transformación Celular Neoplásica , Daño del ADN , Reparación del ADN , Recombinación Homóloga , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo
2.
Mol Cell ; 70(3): 449-461.e5, 2018 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-29727617

RESUMEN

Hard-to-replicate regions of chromosomes (e.g., pericentromeres, centromeres, and telomeres) impede replication fork progression, eventually leading, in the event of replication stress, to chromosome fragility, aging, and cancer. Our knowledge of the mechanisms controlling the stability of these regions is essentially limited to telomeres, where fragility is counteracted by the shelterin proteins. Here we show that the shelterin subunit TRF2 ensures progression of the replication fork through pericentromeric heterochromatin, but not centromeric chromatin. In a process involving its N-terminal basic domain, TRF2 binds to pericentromeric Satellite III sequences during S phase, allowing the recruitment of the G-quadruplex-resolving helicase RTEL1 to facilitate fork progression. We also show that TRF2 is required for the stability of other heterochromatic regions localized throughout the genome, paving the way for future research on heterochromatic replication and its relationship with aging and cancer.


Asunto(s)
Replicación del ADN/genética , Genoma/genética , Heterocromatina/genética , Telómero/genética , Proteína 2 de Unión a Repeticiones Teloméricas/genética , Línea Celular Tumoral , Centrómero/genética , Cromatina/genética , ADN Helicasas/genética , G-Cuádruplex , Células HeLa , Humanos , Fase S/genética
3.
Nat Rev Genet ; 18(9): 535-550, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28714480

RESUMEN

The interplay between replication stress and the S phase checkpoint is a key determinant of genome maintenance, and has a major impact on human diseases, notably, tumour initiation and progression. Recent studies have yielded insights into sequence-dependent and sequence-independent sources of endogenous replication stress. These stresses result in nuclease-induced DNA damage, checkpoint activation and genome-wide replication fork slowing. Several hypotheses have been proposed to account for the mechanisms involved in this complex response. Recent results have shown that the slowing of the replication forks most commonly results from DNA precursor starvation. By concomitantly increasing the density of replication initiation, the cell elicits an efficient compensatory strategy to avoid mitotic anomalies and the inheritance of damage over cell generations.


Asunto(s)
Daño del ADN , Replicación del ADN , Animales , Ciclo Celular , Células/metabolismo , Desoxirribonucleótidos/metabolismo , Humanos , Conformación de Ácido Nucleico , Transcripción Genética
4.
Int J Mol Sci ; 24(13)2023 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-37445805

RESUMEN

Over the last decade, CDK4/6 inhibitors (palbociclib, ribociclib and abemaciclib) have emerged as promising anticancer drugs. Numerous studies have demonstrated that CDK4/6 inhibitors efficiently block the pRb-E2F pathway and induce cell cycle arrest in pRb-proficient cells. Based on these studies, the inhibitors have been approved by the FDA for treatment of advanced hormonal receptor (HR) positive breast cancers in combination with hormonal therapy. However, some evidence has recently shown unexpected effects of the inhibitors, underlining a need to characterize the effects of CDK4/6 inhibitors beyond pRb. Our study demonstrates how palbociclib impairs origin firing in the DNA replication process in pRb-deficient cell lines. Strikingly, despite the absence of pRb, cells treated with palbociclib synthesize less DNA while showing no cell cycle arrest. Furthermore, this CDK4/6 inhibitor treatment disturbs the temporal program of DNA replication and reduces the density of replication forks. Cells treated with palbociclib show a defect in the loading of the Pre-initiation complex (Pre-IC) proteins on chromatin, indicating a reduced initiation of DNA replication. Our findings highlight hidden effects of palbociclib on the dynamics of DNA replication and of its cytotoxic consequences on cell viability in the absence of pRb. This study provides a potential therapeutic application of palbociclib in combination with other drugs to target genomic instability in pRB-deficient cancers.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama , Humanos , Femenino , Origen de Réplica , Inhibidores de Proteínas Quinasas/uso terapéutico , Quinasa 4 Dependiente de la Ciclina , Quinasa 6 Dependiente de la Ciclina , Neoplasias de la Mama/tratamiento farmacológico , Proteínas Inhibidoras de las Quinasas Dependientes de la Ciclina , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico
5.
Nucleic Acids Res ; 46(3): 1280-1294, 2018 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-29253234

RESUMEN

Common Chromosomal Fragile Sites (CFSs) are specific genomic regions prone to form breaks on metaphase chromosomes in response to replication stress. Moreover, CFSs are mutational hotspots in cancer genomes, showing that the mutational mechanisms that operate at CFSs are highly active in cancer cells. Orthologs of human CFSs are found in a number of other mammals, but the extent of CFS conservation beyond the mammalian lineage is unclear. Characterization of CFSs from distantly related organisms can provide new insight into the biology underlying CFSs. Here, we have mapped CFSs in an avian cell line. We find that, overall the most significant CFSs coincide with extremely large conserved genes, from which very long transcripts are produced. However, no significant correlation between any sequence characteristics and CFSs is found. Moreover, we identified putative early replicating fragile sites (ERFSs), which is a distinct class of fragile sites and we developed a fluctuation analysis revealing high mutation rates at the CFS gene PARK2, with deletions as the most prevalent mutation. Finally, we show that avian homologs of the human CFS genes despite their fragility have resisted the general intron size reduction observed in birds suggesting that CFSs have a conserved biological function.


Asunto(s)
Proteínas Aviares/genética , Linfocitos B/metabolismo , Sitios Frágiles del Cromosoma , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Transcripción Genética , Ubiquitina-Proteína Ligasas/genética , Animales , Proteínas Aviares/metabolismo , Linfocitos B/patología , Sitios de Unión , Línea Celular Transformada , Pollos , Mapeo Cromosómico , Secuencia Conservada , Replicación del ADN , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Ontología de Genes , Metafase , Anotación de Secuencia Molecular , Mutación , Unión Proteica , Ubiquitina-Proteína Ligasas/metabolismo
6.
Genes Chromosomes Cancer ; 58(5): 305-316, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30387289

RESUMEN

Some regions of the genome, notably common fragile sites (CFSs), are hypersensitive to replication stress and often involved in the generation of gross chromosome rearrangements in cancer cells. CFSs nest within very large genes and display cell-type-dependent instability. Fragile or not, large genes tend to replicate late in S-phase. A number of data now show that transcription perturbs replication completion across the body of large genes, particularly upon replication stress. However, the molecular mechanisms by which transcription elicits such under-replication and subsequent instability remain unclear. We present here our view of the mechanisms responsible for CFS under-replication and those allowing the cells to cope with this problem in G2 and mitosis. We notably focus on the major role played by the FANC proteins in the protection of CFSs from S phase up to late mitosis. We finally discuss a possible rationale for the conservation of large genes across vertebrate evolution.


Asunto(s)
Sitios Frágiles del Cromosoma , Fase S/genética , Telofase/genética , Animales , Evolución Molecular , Humanos
7.
PLoS Genet ; 12(5): e1006007, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27135742

RESUMEN

Replications forks are routinely hindered by different endogenous stresses. Because homologous recombination plays a pivotal role in the reactivation of arrested replication forks, defects in homologous recombination reveal the initial endogenous stress(es). Homologous recombination-defective cells consistently exhibit a spontaneously reduced replication speed, leading to mitotic extra centrosomes. Here, we identify oxidative stress as a major endogenous source of replication speed deceleration in homologous recombination-defective cells. The treatment of homologous recombination-defective cells with the antioxidant N-acetyl-cysteine or the maintenance of the cells at low O2 levels (3%) rescues both the replication fork speed, as monitored by single-molecule analysis (molecular combing), and the associated mitotic extra centrosome frequency. Reciprocally, the exposure of wild-type cells to H2O2 reduces the replication fork speed and generates mitotic extra centrosomes. Supplying deoxynucleotide precursors to H2O2-exposed cells rescued the replication speed. Remarkably, treatment with N-acetyl-cysteine strongly expanded the nucleotide pool, accounting for the replication speed rescue. Remarkably, homologous recombination-defective cells exhibit a high level of endogenous reactive oxygen species. Consistently, homologous recombination-defective cells accumulate spontaneous γH2AX or XRCC1 foci that are abolished by treatment with N-acetyl-cysteine or maintenance at 3% O2. Finally, oxidative stress stimulated homologous recombination, which is suppressed by supplying deoxynucleotide precursors. Therefore, the cellular redox status strongly impacts genome duplication and transmission. Oxidative stress should generate replication stress through different mechanisms, including DNA damage and nucleotide pool imbalance. These data highlight the intricacy of endogenous replication and oxidative stresses, which are both evoked during tumorigenesis and senescence initiation, and emphasize the importance of homologous recombination as a barrier against spontaneous genetic instability triggered by the endogenous oxidative/replication stress axis.


Asunto(s)
Replicación del ADN/genética , Recombinación Homóloga/genética , Mitosis/genética , Estrés Oxidativo/genética , Acetilcisteína/farmacología , Animales , Células CHO , Centrosoma/efectos de los fármacos , Cricetulus , Daño del ADN/genética , Reparación del ADN/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Redes Reguladoras de Genes/genética , Histonas/genética , Peróxido de Hidrógeno/farmacología , Estrés Oxidativo/efectos de los fármacos , Imagen Individual de Molécula , Proteína 1 de Reparación por Escisión del Grupo de Complementación Cruzada de las Lesiones por Rayos X
8.
Nature ; 470(7332): 120-3, 2011 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-21258320

RESUMEN

Common fragile sites have long been identified by cytogeneticists as chromosomal regions prone to breakage upon replication stress. They are increasingly recognized to be preferential targets for oncogene-induced DNA damage in pre-neoplastic lesions and hotspots for chromosomal rearrangements in various cancers. Common fragile site instability was attributed to the fact that they contain sequences prone to form secondary structures that may impair replication fork movement, possibly leading to fork collapse resulting in DNA breaks. Here we show, in contrast to this view, that the fragility of FRA3B--the most active common fragile site in human lymphocytes--does not rely on fork slowing or stalling but on a paucity of initiation events. Indeed, in lymphoblastoid cells, but not in fibroblasts, initiation events are excluded from a FRA3B core extending approximately 700 kilobases, which forces forks coming from flanking regions to cover long distances in order to complete replication. We also show that origins of the flanking regions fire in mid-S phase, leaving the site incompletely replicated upon fork slowing. Notably, FRA3B instability is specific to cells showing this particular initiation pattern. The fact that both origin setting and replication timing are highly plastic in mammalian cells explains the tissue specificity of common fragile site instability we observed. Thus, we propose that common fragile sites correspond to the latest initiation-poor regions to complete replication in a given cell type. For historical reasons, common fragile sites have been essentially mapped in lymphocytes. Therefore, common fragile site contribution to chromosomal rearrangements in tumours should be reassessed after mapping fragile sites in the cell type from which each tumour originates.


Asunto(s)
Ácido Anhídrido Hidrolasas/genética , Sitios Frágiles del Cromosoma/genética , Fragilidad Cromosómica/fisiología , Replicación del ADN/fisiología , Proteínas de Neoplasias/genética , Origen de Réplica/genética , Línea Celular , Rotura Cromosómica , Fragilidad Cromosómica/genética , Replicación del ADN/genética , Fibroblastos , Genes Supresores de Tumor , Sitios Genéticos/genética , Humanos , Linfocitos/metabolismo , Modelos Biológicos , Especificidad de Órganos
9.
Proc Natl Acad Sci U S A ; 111(2): 763-8, 2014 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-24347643

RESUMEN

Homologous recombination deficient (HR(-)) mammalian cells spontaneously display reduced replication fork (RF) movement and mitotic extra centrosomes. We show here that these cells present a complex mitotic phenotype, including prolonged metaphase arrest, anaphase bridges, and multipolar segregations. We then asked whether the replication and the mitotic phenotypes are interdependent. First, we determined low doses of hydroxyurea that did not affect the cell cycle distribution or activate CHK1 phosphorylation but did slow the replication fork movement of wild-type cells to the same level than in HR(-) cells. Remarkably, these low hydroxyurea doses generated the same mitotic defects (and to the same extent) in wild-type cells as observed in unchallenged HR(-) cells. Reciprocally, supplying nucleotide precursors to HR(-) cells suppressed both their replication deceleration and mitotic extra centrosome phenotypes. Therefore, subtle replication stress that escapes to surveillance pathways and, thus, fails to prevent cells from entering mitosis alters metaphase progression and centrosome number, resulting in multipolar mitosis. Importantly, multipolar mitosis results in global unbalanced chromosome segregation involving the whole genome, even fully replicated chromosomes. These data highlight the cross-talk between chromosome replication and segregation, and the importance of HR at the interface of these two processes for protection against general genome instability.


Asunto(s)
Inestabilidad Cromosómica/fisiología , Replicación del ADN/fisiología , Recombinación Homóloga/fisiología , Mitosis/fisiología , Animales , Afidicolina , Línea Celular , Centrosoma/fisiología , Segregación Cromosómica/fisiología , Cricetinae , Cricetulus , Citometría de Flujo , Hidroxiurea/metabolismo , Microscopía por Video , Estadísticas no Paramétricas , Imagen de Lapso de Tiempo
10.
Biochem Biophys Res Commun ; 469(4): 960-6, 2016 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-26721438

RESUMEN

The mammalian maintenance methyltransferase DNMT1 [DNA (cytosine-5-)-methyltransferase 1] mediates the inheritance of the DNA methylation pattern during replication. Previous studies have shown that depletion of DNMT1 causes a severe growth defect and apoptosis in differentiated cells. However, the detailed mechanisms behind this phenomenon remain poorly understood. Here we show that conditional ablation of Dnmt1 in murine embryonic fibroblasts (MEFs) resulted in an aberrant DNA replication program showing an accumulation of late-S phase replication and causing severely defective growth. Furthermore, we found that the catalytic activity and replication focus targeting sequence of DNMT1 are required for a proper DNA replication program. Taken together, our findings suggest that the maintenance of DNA methylation by DNMT1 plays a critical role in proper regulation of DNA replication in mammalian cells.


Asunto(s)
Daño del ADN/genética , Metilación de ADN/genética , Replicación del ADN/genética , Proteínas de Mantenimiento de Minicromosoma/genética , Origen de Réplica/genética , Proteínas Represoras/genética , Animales , Proliferación Celular/genética , Células Cultivadas , Fibroblastos/fisiología , Genes cdc/genética , Ratones
11.
Nucleic Acids Res ; 42(21): 13194-205, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25378339

RESUMEN

The mechanisms of gene amplification in tumour cells are poorly understood and the relationship between extrachromosomal DNA molecules, named double minutes (dmins), and intrachromosomal homogeneously staining regions (hsr) is not documented at nucleotide resolution. Using fluorescent in situ hybridization and whole genome sequencing, we studied a xenografted human oligodendroglioma where the co-amplification of the EGFR and MYC loci was present in the form of dmins at early passages and of an hsr at later passages. The amplified regions underwent multiple rearrangements and deletions during the formation of the dmins and their transformation into hsr. In both forms of amplification, non-homologous end-joining and microhomology-mediated end-joining rather than replication repair mechanisms prevailed in fusions. Small fragments, some of a few tens of base pairs, were associated in contigs. They came from clusters of breakpoints localized hundreds of kilobases apart in the amplified regions. The characteristics of some pairs of junctions suggest that at least some fragments were not fused randomly but could result from the concomitant repair of neighbouring breakpoints during the interaction of remote DNA sequences. This characterization at nucleotide resolution of the transition between extra- and intrachromosome amplifications highlights a hitherto uncharacterized organization of the amplified regions suggesting the involvement of new mechanisms in their formation.


Asunto(s)
Amplificación de Genes , Oligodendroglioma/genética , Animales , Aberraciones Cromosómicas , Puntos de Rotura del Cromosoma , Cromosomas Humanos , Genes erbB-1 , Genes myc , Humanos , Ratones
12.
PLoS Genet ; 9(7): e1003643, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23874235

RESUMEN

Breaks at common fragile sites (CFS) are a recognized source of genome instability in pre-neoplastic lesions, but how such checkpoint-proficient cells escape surveillance and continue cycling is unknown. Here we show, in lymphocytes and fibroblasts, that moderate replication stresses like those inducing breaks at CFSs trigger chromatin loading of sensors and mediators of the ATR pathway but fail to activate Chk1 or p53. Consistently, we found that cells depleted of ATR, but not of Chk1, accumulate single-stranded DNA upon Mre11-dependent resection of collapsed forks. Partial activation of the pathway under moderate stress thus takes steps against fork disassembly but tolerates S-phase progression and mitotic onset. We show that fork protection by ATR is crucial to CFS integrity, specifically in the cell type where a given site displays paucity in backup replication origins. Tolerance to mitotic entry with under-replicated CFSs therefore results in chromosome breaks, providing a pool of cells committed to further instability.


Asunto(s)
Cromatina/genética , Sitios Frágiles del Cromosoma/genética , Inestabilidad Genómica/genética , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1) , Replicación del ADN/genética , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Linfocitos/citología , Linfocitos/metabolismo , Mitosis/genética , Proteínas Quinasas/genética , Origen de Réplica/genética , Transducción de Señal , Proteína p53 Supresora de Tumor/genética
13.
Trends Genet ; 28(1): 22-32, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22094264

RESUMEN

Common fragile sites (CFSs) are large chromosomal regions prone to breakage upon replication stress that are considered a driving force of oncogenesis. CFSs were long believed to contain sequences blocking fork progression, thus impeding replication completion and leading to DNA breaks upon chromosome condensation. However, recent studies show that delayed completion of DNA replication instead depends on a regional paucity in initiation events. Because the distribution and the timing of these events are cell type dependent, different chromosomal regions can be committed to fragility in different cell types. These new data reveal the epigenetic nature of CFSs and open the way to a reevaluation of the role played by these sites in the formation of chromosome rearrangements found in tumors from different tissues.


Asunto(s)
Sitios Frágiles del Cromosoma , Inestabilidad Genómica , Animales , ADN/genética , ADN/metabolismo , Replicación del ADN , Epigénesis Genética , Humanos , Transcripción Genética
14.
Cell Mol Life Sci ; 71(23): 4489-94, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25248392

RESUMEN

Common fragile sites (CFSs) are large chromosomal regions long identified by conventional cytogenetics as sequences prone to breakage in cells subjected to replication stress. The interest in CFSs came from their key role in the formation of DNA damage, resulting in chromosomal rearrangements. The instability of CFSs was notably correlated with the appearance of genome instability in precancerous lesions and during tumor progression. Identification of the molecular mechanisms responsible for their instability therefore represents a major challenge. A number of data show that breaks result from mitotic entry before replication completion but the mechanisms responsible for such delayed replication of CFSs and relaxed checkpoint surveillance are still debated. In addition, clues to the molecular events leading to breakage just start to emerge. We present here the results of recent reports addressing these questions.


Asunto(s)
Sitios Frágiles del Cromosoma , Inestabilidad Genómica , Neoplasias/genética , Animales , Ciclo Celular , Daño del ADN , Replicación del ADN , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias/patología
15.
Nucleic Acids Res ; 41(17): 8210-9, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23821669

RESUMEN

The mechanisms of formation of intrachromosomal amplifications in tumours are still poorly understood. By using quantitative polymerase chain reaction, DNA sequencing, chromosome walking, in situ hybridization on metaphase chromosomes and whole-genome analysis, we studied two cancer cell lines containing an MYC oncogene amplification with acquired copies ectopically inserted in rearranged chromosomes 17. These intrachromosomal amplifications result from the integration of extrachromosomal DNA molecules. Replication stress could explain the formation of the double-strand breaks involved in their insertion and in the rearrangements of the targeted chromosomes. The sequences of the junctions indicate that homologous recombination was not involved in their formation and support a non-homologous end-joining process. The replication stress-inducible common fragile sites present in the amplicons may have driven the intrachromosomal amplifications. Mechanisms associating break-fusion-bridge cycles and/or chromosome fragmentation may have led to the formation of the uncovered complex structures. To our knowledge, this is the first characterization of an intrachromosomal amplification site at nucleotide resolution.


Asunto(s)
Duplicación Cromosómica , Amplificación de Genes , Neoplasias/genética , Línea Celular Tumoral , Sitios Frágiles del Cromosoma , Cromosomas Humanos Par 17 , Genes myc , Humanos , Hibridación Fluorescente in Situ , Polimorfismo de Nucleótido Simple
16.
Nature ; 455(7212): 557-60, 2008 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-18716622

RESUMEN

Genome stability requires one, and only one, DNA duplication at each S phase. The mechanisms preventing origin firing on newly replicated DNA are well documented, but much less is known about the mechanisms controlling the spacing of initiation events(2,3), namely the completion of DNA replication. Here we show that origin use in Chinese hamster cells depends on both the movement of the replication forks and the organization of chromatin loops. We found that slowing the replication speed triggers the recruitment of latent origins within minutes, allowing the completion of S phase in a timely fashion. When slowly replicating cells are shifted to conditions of fast fork progression, although the decrease in the overall number of active origins occurs within 2 h, the cells still have to go through a complete cell cycle before the efficiency specific to each origin is restored. We observed a strict correlation between replication speed during a given S phase and the size of chromatin loops in the next G1 phase. Furthermore, we found that origins located at or near sites of anchorage of chromatin loops in G1 are activated preferentially in the following S phase. These data suggest a mechanism of origin programming in which replication speed determines the spacing of anchorage regions of chromatin loops, that, in turn, controls the choice of initiation sites.


Asunto(s)
Cromatina/metabolismo , Replicación del ADN/fisiología , Movimiento , Origen de Réplica/genética , Animales , Línea Celular , Cromatina/genética , Cricetinae , Cricetulus , ADN/biosíntesis , ADN/genética , Fase G1 , Matriz Nuclear/metabolismo , Fase S , Factores de Tiempo
17.
Nucleic Acids Res ; 39(8): 3141-55, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21148149

RESUMEN

Genome-scale mapping of pre-replication complex proteins has not been reported in mammalian cells. Poor enrichment of these proteins at specific sites may be due to dispersed binding, poor epitope availability or cell cycle stage-specific binding. Here, we have mapped sites of biotin-tagged ORC and MCM protein binding in G1-synchronized populations of Chinese hamster cells harboring amplified copies of the dihydrofolate reductase (DHFR) locus, using avidin-affinity purification of biotinylated chromatin followed by high-density microarray analysis across the DHFR locus. We have identified several sites of significant enrichment for both complexes distributed throughout the previously identified initiation zone. Analysis of the frequency of initiations across stretched DNA fibers from the DHFR locus confirmed a broad zone of de-localized initiation activity surrounding the sites of ORC and MCM enrichment. Mapping positions of mononucleosomal DNA empirically and computing nucleosome-positioning information in silico revealed that ORC and MCM map to regions of low measured and predicted nucleosome occupancy. Our results demonstrate that specific sites of ORC and MCM enrichment can be detected within a mammalian initiation zone, and suggest that initiation zones may be regions of generally low nucleosome occupancy where flexible nucleosome positioning permits flexible pre-RC assembly sites.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Nucleosomas/metabolismo , Origen de Réplica , Tetrahidrofolato Deshidrogenasa/genética , Animales , Sitios de Unión , Biotinilación , Células CHO , Ligasas de Carbono-Nitrógeno/metabolismo , Cromatina/química , Cricetinae , Cricetulus , Proteínas de Escherichia coli/metabolismo , Fase G1 , Proteínas Represoras/metabolismo
18.
Nat Struct Mol Biol ; 30(4): 539-550, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37024657

RESUMEN

Genome integrity requires replication to be completed before chromosome segregation. The DNA-replication checkpoint (DRC) contributes to this coordination by inhibiting CDK1, which delays mitotic onset. Under-replication of common fragile sites (CFSs), however, escapes surveillance, resulting in mitotic chromosome breaks. Here we asked whether loose DRC activation induced by modest stresses commonly used to destabilize CFSs could explain this leakage. We found that tightening DRC activation or CDK1 inhibition stabilizes CFSs in human cells. Repli-Seq and molecular combing analyses showed a burst of replication initiations implemented in mid S-phase across a subset of late-replicating sequences, including CFSs, while the bulk genome was unaffected. CFS rescue and extra-initiations required CDC6 and CDT1 availability in S-phase, implying that CDK1 inhibition permits mistimed origin licensing and firing. In addition to delaying mitotic onset, tight DRC activation therefore supports replication completion of late origin-poor domains at risk of under-replication, two complementary roles preserving genome stability.


Asunto(s)
Proteínas de Ciclo Celular , Replicación del ADN , Humanos , Fase S , Sitios Frágiles del Cromosoma/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , ADN
19.
Hum Mol Genet ; 19(7): 1276-85, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20056677

RESUMEN

Accumulation of extrachromosomal DNA molecules (double minute) is often responsible for gene amplification in cancers, but the mechanisms leading to their formation are still largely unknown. By using quantitative PCR, chromosome walking, in situ hybridization on metaphase chromosomes and whole genome analysis, we studied a glioma containing four extrachromosomally amplified loci (7p11, 1q32.1, 5p15 and 9p2). Complex extrachromosomal DNA molecules were formed by the fusion of several syntenic or non-syntenic DNA fragments from 7p11, 5p15 to 9p2. Fragments ranged from a few base pairs to megabase pairs. Scars of the amplification process remained at the original locus in the form of deletions or chromosome rearrangements. Chromosome fragmentation, due to replication stress, could explain this complex situation. In contrast, at 1q32.1, the initial extrachromosomal DNA molecule resulted from the circularization of a single fragment associated with an intrachromosomal deletion including, but larger than, the amplified sequence. The nature of the sequences involved in these rearrangements suggests that a V(D)J-like illegitimate recombination contributes to its formation.


Asunto(s)
Herencia Extracromosómica , Amplificación de Genes , Glioma/genética , Neoplasias Encefálicas/genética , Deleción Cromosómica , Estructuras Cromosómicas , Cromosomas Humanos Par 1 , Humanos , Hibridación de Ácido Nucleico
20.
EMBO Rep ; 11(9): 698-704, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20671737

RESUMEN

In eukaryotes, only a fraction of replication origins fire at each S phase. Local histone acetylation was proposed to control firing efficiency of origins, but conflicting results were obtained. We report that local histone acetylation does not reflect origin efficiencies along the adenosine monophosphate deaminase 2 locus in mammalian fibroblasts. Reciprocally, modulation of origin efficiency does not affect acetylation. However, treatment with a deacetylase inhibitor changes the initiation pattern. We demonstrate that this treatment alters pyrimidine biosynthesis and decreases fork speed, which recruits latent origins. Our findings reconcile results that seemed inconsistent and reveal an unsuspected effect of deacetylase inhibitors on replication dynamics.


Asunto(s)
Replicación del ADN , Histonas/metabolismo , Nucleótidos/metabolismo , Origen de Réplica , Acetilación , Animales , Células Cultivadas , Fibroblastos/citología , Fibroblastos/fisiología , Inhibidores de Histona Desacetilasas/metabolismo , Histona Desacetilasas/metabolismo , Humanos , Ácidos Hidroxámicos/metabolismo , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA