Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros

Base de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Sci Rep ; 14(1): 7708, 2024 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-38565932

RESUMEN

Human RECQL4, a member of the RecQ helicase family, plays a role in maintaining genomic stability, but its precise function remains unclear. The N-terminus of RECQL4 has similarity to Sld2, a protein required for the firing of DNA replication origins in budding yeast. Consistent with this sequence similarity, the Xenopus laevis homolog of RECQL4 has been implicated in initiating DNA replication in egg extracts. To determine whether human RECQL4 is required for firing of DNA replication origins, we generated cells in which both RECQL4 alleles were targeted, resulting in either lack of protein expression (knock-out; KO) or expression of a full-length, mutant protein lacking helicase activity (helicase-dead; HD). Interestingly, both the RECQL4 KO and HD cells were viable and exhibited essentially identical origin firing profiles as the parental cells. Analysis of the rate of fork progression revealed increased rates in the RECQL4 KO cells, which might be indicative of decreased origin firing efficiency. Our results are consistent with human RECQL4 having a less critical role in firing of DNA replication origins, than its budding yeast homolog Sld2.


Asunto(s)
RecQ Helicasas , Origen de Réplica , Animales , Humanos , RecQ Helicasas/genética , RecQ Helicasas/metabolismo , Replicación del ADN , Xenopus laevis/metabolismo , ADN/metabolismo
2.
Nature ; 628(8007): 433-441, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38509368

RESUMEN

An important advance in cancer therapy has been the development of poly(ADP-ribose) polymerase (PARP) inhibitors for the treatment of homologous recombination (HR)-deficient cancers1-6. PARP inhibitors trap PARPs on DNA. The trapped PARPs are thought to block replisome progression, leading to formation of DNA double-strand breaks that require HR for repair7. Here we show that PARP1 functions together with TIMELESS and TIPIN to protect the replisome in early S phase from transcription-replication conflicts. Furthermore, the synthetic lethality of PARP inhibitors with HR deficiency is due to an inability to repair DNA damage caused by transcription-replication conflicts, rather than by trapped PARPs. Along these lines, inhibiting transcription elongation in early S phase rendered HR-deficient cells resistant to PARP inhibitors and depleting PARP1 by small-interfering RNA was synthetic lethal with HR deficiency. Thus, inhibiting PARP1 enzymatic activity may suffice for treatment efficacy in HR-deficient settings.


Asunto(s)
Replicación del ADN , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Poli(ADP-Ribosa) Polimerasas , Transcripción Genética , Humanos , Roturas del ADN de Doble Cadena , Replicación del ADN/efectos de los fármacos , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Reparación del ADN por Recombinación , Fase S , Transcripción Genética/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Poli(ADP-Ribosa) Polimerasa-1/metabolismo
3.
Sci Rep ; 14(1): 1117, 2024 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-38212351

RESUMEN

DNA polymerase eta (Polη) is the only translesion synthesis polymerase capable of error-free bypass of UV-induced cyclobutane pyrimidine dimers. A deficiency in Polη function is associated with the human disease Xeroderma pigmentosum variant (XPV). We hereby report the case of a 60-year-old woman known for XPV and carrying a Polη Thr191Pro variant in homozygosity. We further characterize the variant in vitro and in vivo, providing molecular evidence that the substitution abrogates polymerase activity and results in UV sensitivity through deficient damage bypass. This is the first functional molecular characterization of a missense variant of Polη, whose reported pathogenic variants have thus far been loss of function truncation or frameshift mutations. Our work allows the upgrading of Polη Thr191Pro from 'variant of uncertain significance' to 'likely pathogenic mutant', bearing direct impact on molecular diagnosis and genetic counseling. Furthermore, we have established a robust experimental approach that will allow a precise molecular analysis of further missense mutations possibly linked to XPV. Finally, it provides insight into critical Polη residues that may be targeted to develop small molecule inhibitors for cancer therapeutics.


Asunto(s)
Xerodermia Pigmentosa , Humanos , Persona de Mediana Edad , Daño del ADN , Mutación Missense , Prolina/genética , Dímeros de Pirimidina , Rayos Ultravioleta , Xerodermia Pigmentosa/genética , Xerodermia Pigmentosa/patología , Femenino
4.
STAR Protoc ; 4(1): 101970, 2023 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-36598851

RESUMEN

Cells experiencing DNA replication stress enter mitosis with under-replicated DNA, which activates a repair mechanism known as mitotic DNA synthesis (MiDAS). Here we describe a protocol to identify at genome wide and at high resolution the genomic sites where MiDAS occurs in cells exposed to aphidicolin. We use EdU incorporation to label nascent DNA in mitotic cells, followed by isolation of the EdU-labeled DNA and next-generation sequencing. For complete details on the use and execution of this protocol, please refer to Groelly et al. (2022)1 and Macheret et al. (2020).2.


Asunto(s)
Replicación del ADN , ADN , Replicación del ADN/genética , ADN/genética , ADN/metabolismo , Células Cultivadas , Mitosis/genética , Reparación del ADN
5.
J Pathol ; 259(1): 10-20, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36210634

RESUMEN

Chromatin licensing and DNA replication factor 1 (CDT1), a protein of the pre-replicative complex, is essential for loading the minichromosome maintenance complex (MCM) helicases onto the origins of DNA replication. While several studies have shown that dysregulation of CDT1 expression causes re-replication and DNA damage in cell lines, and CDT1 is highly expressed in several human cancers, whether CDT1 deregulation is sufficient to enhance tumorigenesis in vivo is currently unclear. To delineate its role in vivo, we overexpressed Cdt1 in the mouse colon and induced carcinogenesis using azoxymethane/dextran sodium sulfate (AOM/DSS). Here, we show that mice overexpressing Cdt1 develop a significantly higher number of tumors with increased tumor size, and more severe dysplastic changes (high-grade dysplasia), compared with control mice under the same treatment. These tumors exhibited an increased growth rate, while cells overexpressing Cdt1 loaded greater amounts of Mcm2 onto chromatin, demonstrating origin overlicensing. Adenomas overexpressing Cdt1 showed activation of the DNA damage response (DDR), apoptosis, formation of micronuclei, and chromosome segregation errors, indicating that aberrant expression of Cdt1 results in increased genomic and chromosomal instability in vivo, favoring cancer development. In line with these results, high-level expression of CDT1 in human colorectal cancer tissue specimens and colorectal cancer cell lines correlated significantly with increased origin licensing, activation of the DDR, and microsatellite instability (MSI). © 2022 The Pathological Society of Great Britain and Ireland.


Asunto(s)
Neoplasias Colorrectales , Replicación del ADN , Proteínas de Unión al ADN , Animales , Humanos , Ratones , Carcinogénesis/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Cromatina , Neoplasias Colorrectales/inducido químicamente , Neoplasias Colorrectales/genética , Daño del ADN , Proteínas de Unión al ADN/metabolismo
6.
Mol Cell ; 82(18): 3366-3381.e9, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-36002000

RESUMEN

Oncogene activation during tumorigenesis promotes DNA replication stress (RS), which subsequently drives the formation of cancer-associated chromosomal rearrangements. Many episodes of physiological RS likely arise due to conflicts between the DNA replication and transcription machineries operating simultaneously at the same loci. One role of the RAD51 recombinase in human cells is to protect replication forks undergoing RS. Here, we have identified a key role for RAD51 in preventing transcription-replication conflicts (TRCs) from triggering replication fork breakage. The genomic regions most affected by RAD51 deficiency are characterized by being replicated and transcribed in early S-phase and show significant overlap with loci prone to cancer-associated amplification. Consistent with a role for RAD51 in protecting against transcription-replication conflicts, many of the adverse effects of RAD51 depletion are ameliorated by inhibiting early S-phase transcription. We propose a model whereby RAD51 suppresses fork breakage and subsequent inadvertent amplification of genomic loci prone to experiencing TRCs.


Asunto(s)
Replicación del ADN , Recombinasa Rad51 , Cromosomas/metabolismo , Humanos , Recombinasa Rad51/genética , Recombinasa Rad51/metabolismo , Fase S/genética , Transcripción Genética
7.
Mol Cell ; 82(18): 3382-3397.e7, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-36002001

RESUMEN

Aberrant replication causes cells lacking BRCA2 to enter mitosis with under-replicated DNA, which activates a repair mechanism known as mitotic DNA synthesis (MiDAS). Here, we identify genome-wide the sites where MiDAS reactions occur when BRCA2 is abrogated. High-resolution profiling revealed that these sites are different from MiDAS at aphidicolin-induced common fragile sites in that they map to genomic regions replicating in the early S-phase, which are close to early-firing replication origins, are highly transcribed, and display R-loop-forming potential. Both transcription inhibition in early S-phase and RNaseH1 overexpression reduced MiDAS in BRCA2-deficient cells, indicating that transcription-replication conflicts (TRCs) and R-loops are the source of MiDAS. Importantly, the MiDAS sites identified in BRCA2-deficient cells also represent hotspots for genomic rearrangements in BRCA2-mutated breast tumors. Thus, our work provides a mechanism for how tumor-predisposing BRCA2 inactivation links transcription-induced DNA damage with mitotic DNA repair to fuel the genomic instability characteristic of cancer cells.


Asunto(s)
Replicación del ADN , Mitosis , Afidicolina/farmacología , Proteína BRCA2/genética , Sitios Frágiles del Cromosoma/genética , ADN/genética , Daño del ADN , Inestabilidad Genómica , Humanos , Mitosis/genética
8.
9.
Sci Rep ; 12(1): 2505, 2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35169179

RESUMEN

Mpro, the main protease of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is essential for the viral life cycle. Accordingly, several groups have performed in silico screens to identify Mpro inhibitors that might be used to treat SARS-CoV-2 infections. We selected more than five hundred compounds from the top-ranking hits of two very large in silico screens for on-demand synthesis. We then examined whether these compounds could bind to Mpro and inhibit its protease activity. Two interesting chemotypes were identified, which were further evaluated by characterizing an additional five hundred synthesis on-demand analogues. The compounds of the first chemotype denatured Mpro and were considered not useful for further development. The compounds of the second chemotype bound to and enhanced the melting temperature of Mpro. The most active compound from this chemotype inhibited Mpro in vitro with an IC50 value of 1 µM and suppressed replication of the SARS-CoV-2 virus in tissue culture cells. Its mode of binding to Mpro was determined by X-ray crystallography, revealing that it is a non-covalent inhibitor. We propose that the inhibitors described here could form the basis for medicinal chemistry efforts that could lead to the development of clinically relevant inhibitors.


Asunto(s)
Proteasas 3C de Coronavirus/antagonistas & inhibidores , Inhibidores de Proteasas/química , SARS-CoV-2/enzimología , Sitios de Unión , COVID-19/patología , COVID-19/virología , Proteasas 3C de Coronavirus/genética , Proteasas 3C de Coronavirus/metabolismo , Cristalografía por Rayos X , Humanos , Conformación Molecular , Simulación del Acoplamiento Molecular , Nitrilos/química , Nitrilos/metabolismo , Nitrilos/farmacología , Inhibidores de Proteasas/metabolismo , Inhibidores de Proteasas/farmacología , Quinazolinas/química , Quinazolinas/metabolismo , Quinazolinas/farmacología , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , SARS-CoV-2/aislamiento & purificación , SARS-CoV-2/fisiología , Replicación Viral/efectos de los fármacos
10.
Methods Enzymol ; 661: 283-304, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34776216

RESUMEN

Under normal conditions, the genome of eukaryotic cells is faithfully replicated during S phase. However, in cells exposed to DNA polymerase inhibitors, some regions of the genome may fail to be replicated prior to mitotic entry. To prevent chromosomal breakage and loss of genomic information, mitotic DNA synthesis (MiDAS) completes replication of the genome prior to the onset of anaphase. We have developed a protocol that allows one to map the genomic regions that are replicated by MiDAS in mammalian cells. The protocol involves incorporation of a thymidine analog in nascent DNA in mitotic cells and then capture and high throughput sequencing of the nascent DNA. With this approach, sites of MiDAS can be identified at high resolution.


Asunto(s)
Replicación del ADN , Mitosis , Animales , ADN/genética , ADN/metabolismo , Reparación del ADN , Genómica , Mamíferos/genética , Mamíferos/metabolismo , Mitosis/genética
12.
Nat Commun ; 12(1): 4919, 2021 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-34389725

RESUMEN

BRCA1 or BRCA2 germline mutations predispose to breast, ovarian and other cancers. High-throughput sequencing of tumour genomes revealed that oncogene amplification and BRCA1/2 mutations are mutually exclusive in cancer, however the molecular mechanism underlying this incompatibility remains unknown. Here, we report that activation of ß-catenin, an oncogene of the WNT signalling pathway, inhibits proliferation of BRCA1/2-deficient cells. RNA-seq analyses revealed ß-catenin-induced discrete transcriptome alterations in BRCA2-deficient cells, including suppression of CDKN1A gene encoding the CDK inhibitor p21. This accelerates G1/S transition, triggering illegitimate origin firing and DNA damage. In addition, ß-catenin activation accelerates replication fork progression in BRCA2-deficient cells, which is critically dependent on p21 downregulation. Importantly, we find that upregulated p21 expression is essential for the survival of BRCA2-deficient cells and tumours. Thus, our work demonstrates that ß-catenin toxicity in cancer cells with compromised BRCA1/2 function is driven by transcriptional alterations that cause aberrant replication and inflict DNA damage.


Asunto(s)
Proteína BRCA1/genética , Proteína BRCA2/genética , Oncogenes/genética , Transcripción Genética/genética , beta Catenina/genética , Proteína BRCA1/deficiencia , Proteína BRCA2/deficiencia , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Supervivencia Celular/genética , Células Cultivadas , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Daño del ADN , Femenino , Perfilación de la Expresión Génica/métodos , Células HeLa , Humanos , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , RNA-Seq/métodos , beta Catenina/metabolismo
13.
Cancers (Basel) ; 13(6)2021 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-33809306

RESUMEN

The genomes of many human CRCs have been sequenced, revealing a large number of genetic alterations. However, the molecular mechanisms underlying the accumulation of these alterations are still being debated. In this study, we examined colorectal tumours that developed in mice with Apclox/lox, LSL-KrasG12D, and Tp53lox/lox targetable alleles. Organoids were derived from single cells and the spectrum of mutations was determined by exome sequencing. The number of single nucleotide substitutions (SNSs) correlated with the age of the tumour, but was unaffected by the number of targeted cancer-driver genes. Thus, tumours that expressed mutant Apc, Kras, and Tp53 alleles had as many SNSs as tumours that expressed only mutant Apc. In contrast, the presence of large-scale (>10 Mb) copy number alterations (CNAs) correlated strongly with Tp53 inactivation. Comparison of the SNSs and CNAs present in organoids derived from the same tumour revealed intratumoural heterogeneity consistent with genomic lesions accumulating at significantly higher rates in tumour cells compared to normal cells. The rate of acquisition of SNSs increased from the early stages of cancer development, whereas large-scale CNAs accumulated later, after Tp53 inactivation. Thus, a significant fraction of the genomic instability present in cancer cells cannot be explained by aging processes occurring in normal cells before oncogenic transformation.

14.
iScience ; 24(2): 102021, 2021 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-33426509

RESUMEN

The unparalleled global effort to combat the continuing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic over the last year has resulted in promising prophylactic measures. However, a need still exists for cheap, effective therapeutics, and targeting multiple points in the viral life cycle could help tackle the current, as well as future, coronaviruses. Here, we leverage our recently developed, ultra-large-scale in silico screening platform, VirtualFlow, to search for inhibitors that target SARS-CoV-2. In this unprecedented structure-based virtual campaign, we screened roughly 1 billion molecules against each of 40 different target sites on 17 different potential viral and host targets. In addition to targeting the active sites of viral enzymes, we also targeted critical auxiliary sites such as functionally important protein-protein interactions.

15.
ChemRxiv ; 2020 Jul 24.
Artículo en Inglés | MEDLINE | ID: mdl-33200116

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), previously known as 2019 novel coronavirus (2019-nCoV), has spread rapidly across the globe, creating an unparalleled global health burden and spurring a deepening economic crisis. As of July 7th, 2020, almost seven months into the outbreak, there are no approved vaccines and few treatments available. Developing drugs that target multiple points in the viral life cycle could serve as a strategy to tackle the current as well as future coronavirus pandemics. Here we leverage the power of our recently developed in silico screening platform, VirtualFlow, to identify inhibitors that target SARS-CoV-2. VirtualFlow is able to efficiently harness the power of computing clusters and cloud-based computing platforms to carry out ultra-large scale virtual screens. In this unprecedented structure-based multi-target virtual screening campaign, we have used VirtualFlow to screen an average of approximately 1 billion molecules against each of 40 different target sites on 17 different potential viral and host targets in the cloud. In addition to targeting the active sites of viral enzymes, we also target critical auxiliary sites such as functionally important protein-protein interaction interfaces. This multi-target approach not only increases the likelihood of finding a potent inhibitor, but could also help identify a collection of anti-coronavirus drugs that would retain efficacy in the face of viral mutation. Drugs belonging to different regimen classes could be combined to develop possible combination therapies, and top hits that bind at highly conserved sites would be potential candidates for further development as coronavirus drugs. Here, we present the top 200 in silico hits for each target site. While in-house experimental validation of some of these compounds is currently underway, we want to make this array of potential inhibitor candidates available to researchers worldwide in consideration of the pressing need for fast-tracked drug development.

17.
Cell Res ; 30(11): 997-1008, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32561860

RESUMEN

DNA replication stress, a feature of human cancers, often leads to instability at specific genomic loci, such as the common fragile sites (CFSs). Cells experiencing DNA replication stress may also exhibit mitotic DNA synthesis (MiDAS). To understand the physiological function of MiDAS and its relationship to CFSs, we mapped, at high resolution, the genomic sites of MiDAS in cells treated with the DNA polymerase inhibitor aphidicolin. Sites of MiDAS were evident as well-defined peaks that were largely conserved between cell lines and encompassed all known CFSs. The MiDAS peaks mapped within large, transcribed, origin-poor genomic regions. In cells that had been treated with aphidicolin, these regions remained unreplicated even in late S phase; MiDAS then served to complete their replication after the cells entered mitosis. Interestingly, leading and lagging strand synthesis were uncoupled in MiDAS, consistent with MiDAS being a form of break-induced replication, a repair mechanism for collapsed DNA replication forks. Our results provide a better understanding of the mechanisms leading to genomic instability at CFSs and in cancer cells.


Asunto(s)
Sitios Frágiles del Cromosoma/genética , ADN/biosíntesis , Genoma Humano , Mitosis/genética , Análisis de Secuencia de ADN , Línea Celular Tumoral , Rotura Cromosómica , Momento de Replicación del ADN/genética , Inestabilidad Genómica , Humanos , Anotación de Secuencia Molecular , Neoplasias/genética , Origen de Réplica/genética
18.
Cancer Res ; 79(7): 1297-1298, 2019 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-30936075

RESUMEN

DNA replication stress is prevalent in human cancers, but absent in normal cells, suggesting that proteins involved in the cellular response to DNA replication stress could be potential therapeutic targets. SMARCAL1 and ZRANB3 are annealing helicases that mediate the repair of collapsed DNA replication forks. In a study in this issue of Cancer Research, Puccetti and colleagues report that mice lacking either SMARCAL1 or ZRANB3 activity have delayed development of MYC-induced B-cell lymphomas. Thus, inhibiting the response to DNA replication stress could benefit patients with cancer.See related article by Puccetti et al., p. 1612.


Asunto(s)
ADN Helicasas/genética , Replicación del ADN , Animales , Humanos , Ratones
19.
Nat Protoc ; 14(1): 51-67, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30487655

RESUMEN

A better understanding of DNA replication initiation in human cells and how this process is altered upon DNA replication stress requires the ability to study origin firing genome wide. Previously described methods of mapping DNA replication origins in higher eukaryotes rely principally on fractionation of DNA fragments based on their size and, optionally, on the presence of ribonucleotides at their 5' end. Here, we describe a protocol for EdUseq-HU, a method for mapping early S-phase replication origins. Cells, synchronized by mitotic shake-off, are released in medium containing 5-ethynyl-2'-deoxyuridine (EdU; to label nascent DNA) and hydroxyurea (HU; to limit fork progression after origin firing). After using click chemistry to tag the EdU label with a biotin conjugate that is cleavable under mild conditions, the nascent DNA is captured on streptavidin beads. One variant of EdUseq-HU allows mapping of DNA replication origins on the genome at a resolution of 10 kb, and a second variant monitors progression of replication forks. Using EdUseq-HU, the spatiotemporal program of DNA replication in human cell lines can be interrogated in <2 weeks. The protocol requires basic cell culture and molecular biology skills, as well as familiarity with the Perl programming language and the Linux operating system.


Asunto(s)
Puntos de Control del Ciclo Celular/genética , Química Clic/métodos , Replicación del ADN , ADN/genética , Técnicas de Sonda Molecular , Biotina/química , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , ADN/metabolismo , Replicación del ADN/efectos de los fármacos , Desoxiuridina/análogos & derivados , Desoxiuridina/farmacología , Genoma Humano , Células HeLa , Humanos , Hidroxiurea/farmacología , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Origen de Réplica , Programas Informáticos , Estreptavidina/química
20.
Nature ; 555(7694): 112-116, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29466339

RESUMEN

Oncogene-induced DNA replication stress contributes critically to the genomic instability that is present in cancer. However, elucidating how oncogenes deregulate DNA replication has been impeded by difficulty in mapping replication initiation sites on the human genome. Here, using a sensitive assay to monitor nascent DNA synthesis in early S phase, we identified thousands of replication initiation sites in cells before and after induction of the oncogenes CCNE1 and MYC. Remarkably, both oncogenes induced firing of a novel set of DNA replication origins that mapped within highly transcribed genes. These ectopic origins were normally suppressed by transcription during G1, but precocious entry into S phase, before all genic regions had been transcribed, allowed firing of origins within genes in cells with activated oncogenes. Forks from oncogene-induced origins were prone to collapse, as a result of conflicts between replication and transcription, and were associated with DNA double-stranded break formation and chromosomal rearrangement breakpoints both in our experimental system and in a large cohort of human cancers. Thus, firing of intragenic origins caused by premature S phase entry represents a mechanism of oncogene-induced DNA replication stress that is relevant for genomic instability in human cancer.


Asunto(s)
Replicación del ADN , Fase G1/genética , Inestabilidad Genómica/genética , Neoplasias/genética , Oncogenes/genética , Origen de Réplica/genética , Fase S/genética , Línea Celular Tumoral , Puntos de Rotura del Cromosoma , Estudios de Cohortes , Ciclina E/genética , Ciclina E/metabolismo , ADN/biosíntesis , ADN/genética , Roturas del ADN de Doble Cadena , Replicación del ADN/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Genes myc/genética , Humanos , Proteínas Oncogénicas/genética , Transcripción Genética/genética , Translocación Genética/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA