Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 889
Filtrar
1.
Phytochemistry ; 216: 113885, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37806468

RESUMEN

Natural products that inhibit cell cycle progression may have potential as anticancer agents. In this study, cell cycle inhibition of microbial culture extracts was screened by fluorescent images using HeLa/Fucci2 cells. The culture extract of a fungus, Akanthomyces sp., inhibited the cell cycle progression at the S/G2/M phases, and bioassay-guided fractionation of the extract afforded three previously undescribed aphidicolin derivatives, namely akanthomins A-C, and an undescribed chromone glycoside, specifically 9-hydroxyeugenetin 9-O-ß-d-(4-O-methyl)glucopyranoside, in addition to aphidicolin. The chemical structures of these compounds were elucidated by spectroscopic analysis and chemical derivatization. Using a flow cytometer, akanthomin A and aphidicolin were found to inhibit cell cycle progression at the S phase.


Asunto(s)
Hypocreales , Afidicolina/farmacología , Ciclo Celular , División Celular , Extractos Vegetales
2.
Chromosome Res ; 31(3): 23, 2023 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-37597021

RESUMEN

Substantial background level of replication stress is a feature of embryonic and induced pluripotent stem cells (iPSCs), which can predispose to numerical and structural chromosomal instability, including recurrent aberrations of chromosome 12. In differentiated cells, replication stress-sensitive genomic regions, including common fragile sites, are widely mapped through mitotic chromosome break induction by mild aphidicolin treatment, an inhibitor of replicative polymerases. IPSCs exhibit lower apoptotic threshold and higher repair capacity hindering fragile site mapping. Caffeine potentiates genotoxic effects and abrogates G2/M checkpoint delay induced by chemical and physical mutagens. Using 5-ethynyl-2'-deoxyuridine (EdU) for replication labeling, we characterized the mitotic entry dynamics of asynchronous iPSCs exposed to aphidicolin and/or caffeine. Under the adjusted timing of replication stress exposure accounting revealed cell cycle delay, higher metaphase chromosome breakage rate was observed in iPSCs compared to primary lymphocytes. Using differential chromosome staining and subsequent locus-specific fluorescent in situ hybridization, we mapped the FRA12L fragile site spanning the large neuronal ANKS1B gene at 12q23.1, which may contribute to recurrent chromosome 12 missegregation and rearrangements in iPSCs. Publicly available data on the ANKS1B genetic alterations and their possible functional impact are reviewed. Our study provides the first evidence of common fragile site induction in iPSCs and reveals potential somatic instability of a clinically relevant gene during early human development and in vitro cell expansion.


Asunto(s)
Células Madre Pluripotentes Inducidas , Humanos , Afidicolina/farmacología , Cafeína , Cromosomas Humanos Par 12 , Hibridación Fluorescente in Situ , Péptidos y Proteínas de Señalización Intracelular
3.
Life Sci Alliance ; 6(4)2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36746532

RESUMEN

Eukaryotic genomes are duplicated from thousands of replication origins that fire sequentially forming a defined spatiotemporal pattern of replication clusters. The temporal order of DNA replication is determined by chromatin architecture and, more specifically, by chromatin contacts that are stabilized by RIF1. Here, we show that RIF1 localizes near newly synthesized DNA. In cells exposed to the DNA replication inhibitor aphidicolin, suppression of RIF1 markedly decreased the efficacy of isolation of proteins on nascent DNA, suggesting that the isolation of proteins on nascent DNA procedure is biased by chromatin topology. RIF1 was required to limit the accumulation of DNA lesions induced by aphidicolin treatment and promoted the recruitment of cohesins in the vicinity of nascent DNA. Collectively, the data suggest that the stabilization of chromatin topology by RIF1 limits replication-associated genomic instability.


Asunto(s)
Cromatina , Proteínas de Unión a Telómeros , Cromatina/genética , Afidicolina/farmacología , Proteínas de Unión a Telómeros/genética , Proteínas de Unión a Telómeros/metabolismo , ADN/metabolismo , Replicación del ADN/genética
4.
Genome Biol ; 23(1): 223, 2022 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-36266663

RESUMEN

BACKGROUND: A major driver of cancer chromosomal instability is replication stress, the slowing or stalling of DNA replication. How replication stress and genomic instability are connected is not known. Aphidicolin-induced replication stress induces breakages at common fragile sites, but the exact causes of fragility are debated, and acute genomic consequences of replication stress are not fully explored. RESULTS: We characterize DNA copy number alterations (CNAs) in single, diploid non-transformed cells, caused by one cell cycle in the presence of either aphidicolin or hydroxyurea. Multiple types of CNAs are generated, associated with different genomic regions and features, and observed copy number landscapes are distinct between aphidicolin and hydroxyurea-induced replication stress. Coupling cell type-specific analysis of CNAs to gene expression and single-cell replication timing analyses pinpointed the causative large genes of the most recurrent chromosome-scale CNAs in aphidicolin. These are clustered on chromosome 7 in RPE1 epithelial cells but chromosome 1 in BJ fibroblasts. Chromosome arm level CNAs also generate acentric lagging chromatin and micronuclei containing these chromosomes. CONCLUSIONS: Chromosomal instability driven by replication stress occurs via focal CNAs and chromosome arm scale changes, with the latter confined to a very small subset of chromosome regions, potentially heavily skewing cancer genome evolution. Different inducers of replication stress lead to distinctive CNA landscapes providing the opportunity to derive copy number signatures of specific replication stress mechanisms. Single-cell CNA analysis thus reveals the impact of replication stress on the genome, providing insights into the molecular mechanisms which fuel chromosomal instability in cancer.


Asunto(s)
Variaciones en el Número de Copia de ADN , Neoplasias , Humanos , Afidicolina/farmacología , Hidroxiurea/farmacología , Neoplasias/genética , ADN , Inestabilidad Cromosómica , Cromosomas , Cromatina
5.
Mutat Res ; 825: 111800, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36155262

RESUMEN

PTEN is a tumor suppressor protein frequently altered in various cancers. PTEN-null cells have a characteristic of rapid proliferation with an unstable genome. Replication stress is one of the causes of the accumulation of genomic instability if not sensed by the cellular signaling. Though PTEN-null cells have shown to be impaired in replication progression and stalled fork recovery, the association between the catalytic function of PTEN regulated by posttranslational modulation and cellular response to replication stress has not been studied explicitly. To understand molecular mechanism, we find that PTEN-null cells display unrestrained replication fork progression with accumulation of damaged DNA after treatment with aphidicolin which can be rescued by ectopic expression of full-length PTEN, as evident from DNA fiber assay. Moreover, the C-terminal phosphorylation (Ser 380, Thr 382/383) of PTEN is essential for its chromatin association and sensing replication stress that, in response, induce cell cycle arrest. Further, we observed that PTEN induces HP1α expression and H3K9me3 foci formation in a C-terminal phosphorylation-dependent manner. However, phosphatase dead PTEN cannot sense replication stress though it can be associated with chromatin. Together, our results suggest that DNA replication perturbation by aphidicolin enables chromatin association of PTEN through C-terminal phosphorylation, induces heterochromatin formation by stabilizing and up-regulating H3K9me3 foci and augments CHK1 activation. Thereby, PTEN prevents DNA replication fork elongation and simultaneously causes G1-S phase cell cycle arrest to limit cell proliferation in stress conditions. Thus PTEN act as stress sensing protein during replication arrest to maintain genomic stability.


Asunto(s)
Cromatina , Heterocromatina , Humanos , Fosforilación , Heterocromatina/genética , Afidicolina/farmacología , Ensamble y Desensamble de Cromatina , Inestabilidad Genómica , Fosfohidrolasa PTEN/genética
6.
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
7.
Int J Mol Sci ; 23(4)2022 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-35216252

RESUMEN

Mosaicism is the most important limitation for one-step gene editing in embryos by CRISPR/Cas9 because cuts and repairs sometimes take place after the first DNA replication of the zygote. To try to minimize the risk of mosaicism, in this study a reversible DNA replication inhibitor was used after the release of CRISPR/Cas9 in the cell. There is no previous information on the use of aphidicolin in porcine embryos, so the reversible inhibition of DNA replication and the effect on embryo development of different concentrations of this drug was first evaluated. The effect of incubation with aphidicolin was tested with CRISPR/Cas9 at different concentrations and different delivery methodologies. As a result, the reversible inhibition of DNA replication was observed, and it was concentration dependent. An optimal concentration of 0.5 µM was established and used for subsequent experiments. Following the use of this drug with CRISPR/Cas9, a halving of mosaicism was observed together with a detrimental effect on embryo development. In conclusion, the use of reversible inhibition of DNA replication offers a way to reduce mosaicism. Nevertheless, due to the reduction in embryo development, it would be necessary to reach a balance for its use to be feasible.


Asunto(s)
Afidicolina/farmacología , Sistemas CRISPR-Cas/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , Embrión de Mamíferos/efectos de los fármacos , Eucariontes/efectos de los fármacos , Animales , Animales Modificados Genéticamente , Desarrollo Embrionario/efectos de los fármacos , Edición Génica/métodos , Mosaicismo/efectos de los fármacos , Porcinos , Cigoto/efectos de los fármacos
8.
PLoS Biol ; 19(9): e3001377, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34491983

RESUMEN

Forming an embryo from a zygote poses an apparent conflict for epigenetic regulation. On the one hand, the de novo induction of cell fate identities requires the establishment and subsequent maintenance of epigenetic information to harness developmental gene expression. On the other hand, the embryo depends on cell proliferation, and every round of DNA replication dilutes preexisting histone modifications by incorporation of new unmodified histones into chromatin. Here, we investigated the possible relationship between the propagation of epigenetic information and the developmental cell proliferation during Xenopus embryogenesis. We systemically inhibited cell proliferation during the G1/S transition in gastrula embryos and followed their development until the tadpole stage. Comparing wild-type and cell cycle-arrested embryos, we show that the inhibition of cell proliferation is principally compatible with embryo survival and cellular differentiation. In parallel, we quantified by mass spectrometry the abundance of a large set of histone modification states, which reflects the developmental maturation of the embryonic epigenome. The arrested embryos developed abnormal stage-specific histone modification profiles (HMPs), in which transcriptionally repressive histone marks were overrepresented. Embryos released from the cell cycle block during neurulation reverted toward normality on morphological, molecular, and epigenetic levels. These results suggest that the cell cycle block by HUA alters stage-specific HMPs. We propose that this influence is strong enough to control developmental decisions, specifically in cell populations that switch between resting and proliferating states such as stem cells.


Asunto(s)
Epigénesis Genética , Código de Histonas , Xenopus laevis/embriología , Animales , Afidicolina/farmacología , Ciclo Celular , Proliferación Celular/efectos de los fármacos , Embrión no Mamífero/embriología , Inhibidores Enzimáticos/farmacología , Hidroxiurea/farmacología
9.
Nucleic Acids Res ; 49(13): 7507-7524, 2021 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-34181717

RESUMEN

Impaired replication progression leads to de novo copy number variant (CNV) formation at common fragile sites (CFSs). We previously showed that these hotspots for genome instability reside in late-replicating domains associated with large transcribed genes and provided indirect evidence that transcription is a factor in their instability. Here, we compared aphidicolin (APH)-induced CNV and CFS frequency between wild-type and isogenic cells in which FHIT gene transcription was ablated by promoter deletion. Two promoter-deletion cell lines showed reduced or absent CNV formation and CFS expression at FHIT despite continued instability at the NLGN1 control locus. APH treatment led to critical replication delays that remained unresolved in G2/M in the body of many, but not all, large transcribed genes, an effect that was reversed at FHIT by the promoter deletion. Altering RNase H1 expression did not change CNV induction frequency and DRIP-seq showed a paucity of R-loop formation in the central regions of large genes, suggesting that R-loops are not the primary mediator of the transcription effect. These results demonstrate that large gene transcription is a determining factor in replication stress-induced genomic instability and support models that CNV hotspots mainly result from the transcription-dependent passage of unreplicated DNA into mitosis.


Asunto(s)
Ácido Anhídrido Hidrolasas/genética , Variaciones en el Número de Copia de ADN , Replicación del ADN , Proteínas de Neoplasias/genética , Transcripción Genética , Ácido Anhídrido Hidrolasas/biosíntesis , Animales , Afidicolina/farmacología , Línea Celular , Sitios Frágiles del Cromosoma , Sitios Genéticos , Humanos , Ratones , Mutación , Proteínas de Neoplasias/biosíntesis , Regiones Promotoras Genéticas , Estructuras R-Loop , Ribonucleasa H/metabolismo , Estrés Fisiológico
10.
Int J Mol Sci ; 22(9)2021 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-34066960

RESUMEN

DNA replication timing (RT), reflecting the temporal order of origin activation, is known as a robust and conserved cell-type specific process. Upon low replication stress, the slowing of replication forks induces well-documented RT delays associated to genetic instability, but it can also generate RT advances that are still uncharacterized. In order to characterize these advanced initiation events, we monitored the whole genome RT from six independent human cell lines treated with low doses of aphidicolin. We report that RT advances are cell-type-specific and involve large heterochromatin domains. Importantly, we found that some major late to early RT advances can be inherited by the unstressed next-cellular generation, which is a unique process that correlates with enhanced chromatin accessibility, as well as modified replication origin landscape and gene expression in daughter cells. Collectively, this work highlights how low replication stress may impact cellular identity by RT advances events at a subset of chromosomal domains.


Asunto(s)
Momento de Replicación del ADN , Estrés Fisiológico , Afidicolina/farmacología , Línea Celular Tumoral , Cromatina/metabolismo , Daño del ADN , Momento de Replicación del ADN/genética , Epigénesis Genética/efectos de los fármacos , Sitios Genéticos , Código de Histonas , Humanos , Modelos Biológicos , Estrés Fisiológico/genética
11.
J Cell Biol ; 220(8)2021 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-34100862

RESUMEN

Replication stress is one of the main sources of genome instability. Although the replication stress response in eukaryotic cells has been extensively studied, almost nothing is known about the replication stress response in nucleoli. Here, we demonstrate that initial replication stress-response factors, such as RPA, TOPBP1, and ATR, are recruited inside the nucleolus in response to drug-induced replication stress. The role of TOPBP1 goes beyond the typical replication stress response; it interacts with the low-complexity nucleolar protein Treacle (also referred to as TCOF1) and forms large Treacle-TOPBP1 foci inside the nucleolus. In response to replication stress, Treacle and TOPBP1 facilitate ATR signaling at stalled replication forks, reinforce ATR-mediated checkpoint activation inside the nucleolus, and promote the recruitment of downstream replication stress response proteins inside the nucleolus without forming nucleolar caps. Characterization of the Treacle-TOPBP1 interaction mode leads us to propose that these factors can form a molecular platform for efficient stress response in the nucleolus.


Asunto(s)
Proteínas Portadoras/metabolismo , Nucléolo Celular/metabolismo , Daño del ADN , Replicación del ADN , ADN Ribosómico/biosíntesis , Proteínas de Unión al ADN/metabolismo , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Afidicolina/farmacología , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Proteínas Portadoras/genética , Nucléolo Celular/efectos de los fármacos , Nucléolo Celular/genética , ADN Ribosómico/genética , Proteínas de Unión al ADN/genética , Inestabilidad Genómica , Células HCT116 , Células HeLa , Humanos , Hidroxiurea/farmacología , Microscopía Fluorescente , Proteínas Nucleares/genética , Fosfoproteínas/genética , Unión Proteica , Transporte de Proteínas , Transducción de Señal
12.
Genetics ; 218(2)2021 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-33792683

RESUMEN

Break-induced replication (BIR) is essential for the repair of DNA double-strand breaks (DSBs) with single ends. DSBs-induced microhomology-mediated BIR (mmBIR) and template-switching can increase the risk of complex genome rearrangement. In addition, DSBs can also induce the multi-invasion-mediated DSB amplification. The mmBIR-induced genomic rearrangement has been identified in cancer cells and patients with rare diseases. However, when and how mmBIR is initiated have not been fully and deeply studied. Furthermore, it is not well understood about the conditions for initiation of multi-invasion-mediated DSB amplification. In the G2 phase oocyte of mouse, we identified a type of short-scale BIR (ssBIR) using the DNA replication indicator 5-ethynyl-2'-deoxyuridine (EdU). These ssBIRs could only be induced in the fully grown oocytes but not the growing oocytes. If the DSB oocytes were treated with Rad51 or Chek1/2 inhibitors, both EdU signals and DSB marker γH2A.X foci would decrease. In addition, the DNA polymerase inhibitor Aphidicolin could inhibit the ssBIR and another inhibitor ddATP could reduce the number of γH2A.X foci in the DSB oocytes. In conclusion, our results showed that DNA DSBs in the fully grown oocytes can initiate ssBIR and be amplified by Rad51 or DNA replication.


Asunto(s)
Roturas del ADN de Doble Cadena , Reparación del ADN/fisiología , Replicación del ADN/fisiología , Animales , Afidicolina/farmacología , Células Cultivadas , Reparación del ADN/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , ADN Polimerasa Dirigida por ADN/metabolismo , Nucleótidos de Desoxiadenina/farmacología , Didesoxinucleótidos/farmacología , Femenino , Fase G2 , Indoles/farmacología , Ratones , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Oocitos , Cultivo Primario de Células , Recombinasa Rad51/antagonistas & inhibidores , Recombinasa Rad51/metabolismo , Tetrahidroisoquinolinas/farmacología
13.
Nat Commun ; 12(1): 2455, 2021 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-33911081

RESUMEN

The mutational mechanisms underlying recurrent deletions in clonal hematopoiesis are not entirely clear. In the current study we inspect the genomic regions around recurrent deletions in myeloid malignancies, and identify microhomology-based signatures in CALR, ASXL1 and SRSF2 loci. We demonstrate that these deletions are the result of double stand break repair by a PARP1 dependent microhomology-mediated end joining (MMEJ) pathway. Importantly, we provide evidence that these recurrent deletions originate in pre-leukemic stem cells. While DNA polymerase theta (POLQ) is considered a key component in MMEJ repair, we provide evidence that pre-leukemic MMEJ (preL-MMEJ) deletions can be generated in POLQ knockout cells. In contrast, aphidicolin (an inhibitor of replicative polymerases and replication) treatment resulted in a significant reduction in preL-MMEJ. Altogether, our data indicate an association between POLQ independent MMEJ and clonal hematopoiesis and elucidate mutational mechanisms involved in the very first steps of leukemia evolution.


Asunto(s)
Hematopoyesis Clonal/genética , Reparación del ADN por Unión de Extremidades/genética , ADN Polimerasa Dirigida por ADN/genética , Leucemia Mieloide/genética , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Afidicolina/farmacología , Calreticulina/genética , Roturas del ADN de Doble Cadena , ADN Polimerasa Dirigida por ADN/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Humanos , Células Progenitoras Mieloides , Proteínas Represoras/genética , Eliminación de Secuencia/genética , Factores de Empalme Serina-Arginina/genética , ADN Polimerasa theta
14.
Nitric Oxide ; 109-110: 12-19, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33592314

RESUMEN

Aphidicolin represses DNA replication by inhibiting DNA polymerase α and δ, which leads to cell cycle arrest and cell damage. Nitric oxide (NO) generated by endothelial NO synthase (eNOS) plays an essential role in maintenance of endothelial integrity including endothelial cell (EC) survival. Previously, we reported that aphidicolin increases NO production in bovine aortic ECs (BAECs). However, the role of aphidicolin-induced NO on EC viability and its molecular mechanism remain to be elucidated. Treatment with 20 µM aphidicolin for 24 h reduced BAEC viability by ~40%, which was accompanied by increased NO production, phosphorylation of eNOS at Ser1179 (p-eNOS-Ser1179), and eNOS protein expression. The aphidicolin-increased eNOS expression and p-eNOS-Ser1179 were not altered by 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid tetrakis(acetoxymethyl ester) (BAPTA-AM), a cell permeable and specific intracellular Ca2+ chelator. Co-treatment with 2-phenyl-4, 4, 5, 5,-tetramethylimidazoline-1-oxyl 3-oxide (PTIO), an NO scavenger, or Nω-Nitro-l-arginine methyl ester hydrochloride (l-NAME), a NOS inhibitor, exacerbated aphidicolin-stimulated BAEC death. Knockdown of eNOS gene expression using siRNA aggravated aphidicolin-induced BAEC death. However, exogenous NO donors including S-nitroso-l-glutathione (GSNO) or diethylenetriamine NONOate (DETA NO) had no effect on aphidicolin-decreased BAEC viability and aggravated BAEC viability at higher doses. Interestingly, aphidicolin accumulated eNOS protein in the active form, p-eNOS-Ser1179, in the nucleus. When cells were ectopically transfected with a wild-type (WT)-eNOS gene, aphidicolin induced significant localization of the protein product in the nucleus. Additionally, aphidicolin-elicited cell death was significantly reversed in WT-eNOS gene-transfected BAECs. Furthermore, overexpression of the eNOS gene containing nuclear localization signal (NLS) but not nuclear export signal (NES) significantly attenuated aphidicolin-induced BAEC death. When G2A-eNOS mutant lacking myristoylation at Gly2 was transfected, its intracellular distribution became diffuse and included the nucleus. Finally, expression of N-myristoyltransferase 2 (NMT2) but not NMT1 significantly decreased in aphidicolin-treated BAECs. Taken together, our results suggest that aphidicolin attenuates BAEC death in part by increasing nuclear eNOS localization and NO production.


Asunto(s)
Afidicolina/farmacología , Muerte Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Células Endoteliales/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo III/metabolismo , Óxido Nítrico/metabolismo , Aciltransferasas/metabolismo , Animales , Aorta/citología , Bovinos , Supervivencia Celular/efectos de los fármacos , Células Endoteliales/metabolismo
15.
FEBS Lett ; 595(5): 595-603, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33423298

RESUMEN

We have previously demonstrated that Fanconi anemia (FA) proteins work in concert with other FA and non-FA proteins to mediate stalled replication fork restart. Previous studies suggest a connection between the FA protein FANCD2 and the non-FA protein mechanistic target of rapamycin (mTOR). A recent study showed that mTOR is involved in actin-dependent DNA replication fork restart, suggesting possible roles in the FA DNA repair pathway. In this study, we demonstrate that during replication stress mTOR interacts and cooperates with FANCD2 to provide cellular stability, mediate stalled replication fork restart, and prevent nucleolytic degradation of the nascent DNA strands. Taken together, this study unravels a novel functional cross-talk between two important mechanisms: mTOR and FA DNA repair pathways that ensure genomic stability.


Asunto(s)
Reparación del ADN/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Anemia de Fanconi/genética , Fibroblastos/metabolismo , Serina-Treonina Quinasas TOR/genética , Afidicolina/farmacología , Supervivencia Celular/efectos de los fármacos , ADN/genética , ADN/metabolismo , Anemia de Fanconi/metabolismo , Anemia de Fanconi/patología , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/deficiencia , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Genoma Humano , Inestabilidad Genómica , Humanos , Hidroxiurea/farmacología , Mitomicina/farmacología , Cultivo Primario de Células , Unión Proteica/efectos de los fármacos , Transducción de Señal , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/metabolismo
16.
Nucleic Acids Res ; 49(1): 244-256, 2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33290559

RESUMEN

The human genome contains hundreds of large, structurally diverse blocks that are insufficiently represented in the reference genome and are thus not amenable to genomic analyses. Structural diversity in the human population suggests that these blocks are unstable in the germline; however, whether or not these blocks are also unstable in the cancer genome remains elusive. Here we report that the 500 kb block called KRTAP_region_1 (KRTAP-1) on 17q12-21 recurrently demarcates the amplicon of the ERBB2 (HER2) oncogene in breast tumors. KRTAP-1 carries numerous tandemly-duplicated segments that exhibit diversity within the human population. We evaluated the fragility of the block by cytogenetically measuring the distances between the flanking regions and found that spontaneous distance outliers (i.e DNA breaks) appear more frequently at KRTAP-1 than at the representative common fragile site (CFS) FRA16D. Unlike CFSs, KRTAP-1 is not sensitive to aphidicolin. The exonuclease activity of DNA repair protein Mre11 protects KRTAP-1 from breaks, whereas CtIP does not. Breaks at KRTAP-1 lead to the palindromic duplication of the ERBB2 locus and trigger Breakage-Fusion-Bridge cycles. Our results indicate that an insufficiently investigated area of the human genome is fragile and could play a crucial role in cancer genome evolution.


Asunto(s)
Neoplasias de la Mama/genética , Sitios Frágiles del Cromosoma/genética , Reparación del ADN , Amplificación de Genes , Duplicación de Gen/genética , Genes erbB-2 , Queratinas Específicas del Pelo/fisiología , Afidicolina/farmacología , Mama/metabolismo , Neoplasias de la Mama/metabolismo , Células Cultivadas , Inestabilidad Cromosómica , Roturas del ADN , Variaciones en el Número de Copia de ADN , ADN de Neoplasias/genética , Células Epiteliales/metabolismo , Femenino , Variación Genética , Inestabilidad Genómica , Humanos , Proteína Homóloga de MRE11/fisiología , Proteínas de Neoplasias/fisiología , Secuenciación Completa del Genoma
17.
Cell Rep ; 32(12): 108179, 2020 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-32966779

RESUMEN

Fragile X syndrome (FXS) is a neurodevelopmental disorder caused by mutations in the FMR1 gene and deficiency of a functional FMRP protein. FMRP is known as a translation repressor whose nuclear function is not understood. We investigated the global impact on genome stability due to FMRP loss. Using Break-seq, we map spontaneous and replication stress-induced DNA double-strand breaks (DSBs) in an FXS patient-derived cell line. We report that the genomes of FXS cells are inherently unstable and accumulate twice as many DSBs as those from an unaffected control. We demonstrate that replication stress-induced DSBs in FXS cells colocalize with R-loop forming sequences. Exogenously expressed FMRP in FXS fibroblasts ameliorates DSB formation. FMRP, not the I304N mutant, abates R-loop-induced DSBs during programmed replication-transcription conflict. These results suggest that FMRP is a genome maintenance protein that prevents R-loop accumulation. Our study provides insights into the etiological basis for FXS.


Asunto(s)
Rotura Cromosómica , Replicación del ADN , Síndrome del Cromosoma X Frágil/genética , Genoma Humano , Estrés Fisiológico , Afidicolina/farmacología , Línea Celular , Rotura Cromosómica/efectos de los fármacos , ADN/metabolismo , Daño del ADN , Reparación del ADN/efectos de los fármacos , Replicación del ADN/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibroblastos/patología , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/metabolismo , Humanos , Modelos Biológicos , Mutación/genética , Estructuras R-Loop , ARN/metabolismo , Estrés Fisiológico/efectos de los fármacos
18.
Cell Rep ; 32(12): 108177, 2020 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-32966795

RESUMEN

Cells coordinate interphase-to-mitosis transition, but recurrent cytogenetic lesions appear at common fragile sites (CFSs), termed CFS expression, in a tissue-specific manner after replication stress, marking regions of instability in cancer. Despite such a distinct defect, no model fully provides a molecular explanation for CFSs. We show that CFSs are characterized by impaired chromatin folding, manifesting as disrupted mitotic structures visible with molecular fluorescence in situ hybridization (FISH) probes in the presence and absence of replication stress. Chromosome condensation assays reveal that compaction-resistant chromatin lesions persist at CFSs throughout the cell cycle and mitosis. Cytogenetic and molecular lesions are marked by faulty condensin loading at CFSs, a defect in condensin-I-mediated compaction, and are coincident with mitotic DNA synthesis (MIDAS). This model suggests that, in conditions of exogenous replication stress, aberrant condensin loading leads to molecular defects and CFS expression, concomitantly providing an environment for MIDAS, which, if not resolved, results in chromosome instability.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Sitios Frágiles del Cromosoma , Replicación del ADN , Proteínas de Unión al ADN/metabolismo , Complejos Multiproteicos/metabolismo , Estrés Fisiológico , Afidicolina/farmacología , Cromatina/metabolismo , ADN/biosíntesis , Replicación del ADN/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Femenino , Fase G2/efectos de los fármacos , Células HCT116 , Humanos , Masculino , Mitosis/efectos de los fármacos , Modelos Biológicos , Estrés Fisiológico/efectos de los fármacos
19.
Nat Commun ; 11(1): 3613, 2020 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-32680994

RESUMEN

Common fragile sites (CFSs) are regions susceptible to replication stress and are hotspots for chromosomal instability in cancer. Several features were suggested to underlie CFS instability, however, these features are prevalent across the genome. Therefore, the molecular mechanisms underlying CFS instability remain unclear. Here, we explore the transcriptional profile and DNA replication timing (RT) under mild replication stress in the context of the 3D genome organization. The results reveal a fragility signature, comprised of a TAD boundary overlapping a highly transcribed large gene with APH-induced RT-delay. This signature enables precise mapping of core fragility regions in known CFSs and identification of novel fragile sites. CFS stability may be compromised by incomplete DNA replication and repair in TAD boundaries core fragility regions leading to genomic instability. The identified fragility signature will allow for a more comprehensive mapping of CFSs and pave the way for investigating mechanisms promoting genomic instability in cancer.


Asunto(s)
Sitios Frágiles del Cromosoma/genética , Momento de Replicación del ADN/genética , Genoma Humano , Inestabilidad Genómica , Afidicolina/farmacología , Línea Celular , Secuenciación de Inmunoprecipitación de Cromatina , Mapeo Cromosómico/métodos , ADN/química , Momento de Replicación del ADN/efectos de los fármacos , Fibroblastos , Redes Reguladoras de Genes , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Neoplasias/genética , Conformación de Ácido Nucleico , Sensibilidad y Especificidad , Transcripción Genética/efectos de los fármacos
20.
Methods Mol Biol ; 2143: 25-39, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32524470

RESUMEN

Primary cultures of neurons of the peripheral nervous system have been successfully used for studying many aspects of neuronal development and survival, including investigations into the mechanisms of axon degeneration. In this chapter, we describe how to prepare and microinject dissociated cultures of sympathetic neurons of the superior cervical ganglion (SCG) specifically for use in highly controlled and targeted assays of axon survival and degeneration.


Asunto(s)
Axones/efectos de los fármacos , Microinyecciones/métodos , Ganglio Cervical Superior/citología , Animales , Afidicolina/farmacología , Axotomía , Colorantes Fluorescentes/administración & dosificación , Ratones , Microinyecciones/instrumentación , Microscopía Fluorescente/métodos , Microscopía de Contraste de Fase/métodos , Factor de Crecimiento Nervioso/administración & dosificación , Cultivo Primario de Células , Células Receptoras Sensoriales/fisiología , Células Receptoras Sensoriales/ultraestructura , Soluciones/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...