Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
2.
Cells ; 12(22)2023 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-37998365

RESUMEN

In metazoans, the largest sirtuin, SIRT1, is a nuclear protein implicated in epigenetic modifications, circadian signaling, DNA recombination, replication, and repair. Our previous studies have demonstrated that SIRT1 binds replication origins and inhibits replication initiation from a group of potential initiation sites (dormant origins). We studied the effects of aging and SIRT1 activity on replication origin usage and the incidence of transcription-replication collisions (creating R-loop structures) in adult human cells obtained at different time points during chronological aging and in cancer cells. In primary, untransformed cells, SIRT1 activity declined and the prevalence of R-loops rose with chronological aging. Both the reduction in SIRT1 activity and the increased abundance of R-loops were also observed during the passage of primary cells in culture. All cells, regardless of donor age or transformation status, reacted to the short-term, acute chemical inhibition of SIRT1 with the activation of excessive replication initiation events coincident with an increased prevalence of R-loops. However, cancer cells activated dormant replication origins, genome-wide, during long-term proliferation with mutated or depleted SIRT1, whereas, in primary cells, the aging-associated SIRT1-mediated activation of dormant origins was restricted to rDNA loci. These observations suggest that chronological aging and the associated decline in SIRT1 activity relax the regulatory networks that protect cells against excess replication and that the mechanisms protecting from replication-transcription collisions at the rDNA loci manifest as differentially enhanced sensitivities to SIRT1 decline and chronological aging.


Asunto(s)
Estructuras R-Loop , Sirtuina 1 , Humanos , ADN Ribosómico/genética , Sirtuina 1/genética , Sirtuina 1/metabolismo , Replicación del ADN/genética , Envejecimiento/genética
3.
Cell Commun Signal ; 21(1): 219, 2023 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-37612584

RESUMEN

BACKGROUND: Megakaryocytes (MKs) are platelet precursors, which arise from hematopoietic stem cells (HSCs). While MK lineage commitment and differentiation are accompanied by changes in gene expression, many factors that modulate megakaryopoiesis remain to be uncovered. Replication initiation determinant protein (RepID) which has multiple histone-code reader including bromodomain, cryptic Tudor domain and WD40 domains and Cullin 4-RING E3 ubiquitin ligase complex (CRL4) recruited to chromatin mediated by RepID have potential roles in gene expression changes via epigenetic regulations. We aimed to investigate whether RepID-CRL4 participates in transcriptional changes required for MK differentiation. METHODS: The PCR array was performed using cDNAs derived from RepID-proficient or RepID-deficient K562 erythroleukemia cell lines. Correlation between RepID and DAB2 expression was examined in the Cancer Cell Line Encyclopedia (CCLE) through the CellMinerCDB portal. The acceleration of MK differentiation in RepID-deficient K562 cells was determined by estimating cell sizes as well as counting multinucleated cells known as MK phenotypes, and by qRT-PCR analysis to validate transcripts of MK markers using phorbol 12-myristate 13-acetate (PMA)-mediated MK differentiation condition. Interaction between CRL4 and histone methylation modifying enzymes were investigated using BioGRID database, immunoprecipitation and proximity ligation assay. Alterations of expression and chromatin binding affinities of RepID, CRL4 and histone methylation modifying enzymes were investigated using subcellular fractionation followed by immunoblotting. RepID-CRL4-JARID1A-based epigenetic changes on DAB2 promoter were analyzed by chromatin-immunoprecipitation and qPCR analysis. RESULTS: RepID-deficient K562 cells highly expressing MK markers showed accelerated MKs differentiation exhibiting increases in cell size, lobulated nuclei together with reaching maximum levels of MK marker expression earlier than RepID-proficient K562 cells. Recovery of WD40 domain-containing RepID constructs in RepID-deficient background repressed DAB2 expression. CRL4A formed complex with histone H3K4 demethylase JARID1A in soluble nucleus and loaded to the DAB2 promoter in a RepID-dependent manner during proliferation condition. RepID, CRL4A, and JARID1A were dissociated from the chromatin during MK differentiation, leading to euchromatinization of the DAB2 promoter. CONCLUSION: This study uncovered a role for the RepID-CRL4A-JARID1A pathway in the regulation of gene expression for MK differentiation, which can form the basis for the new therapeutic approaches to induce platelet production. Video Abstract.


Asunto(s)
Núcleo Celular , Histonas , Proteínas de Ciclo Celular , Diferenciación Celular , Cromatina , Dominio Tudor
4.
Res Sq ; 2023 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-37461562

RESUMEN

Background Megakaryocytes (MKs) are platelet precursors, which arise from hematopoietic stem cells (HSCs). While MK lineage commitment and differentiation are accompanied by changes in gene expression, many factors that modulate megakaryopoiesis remain to be uncovered. Replication origin binding protein (RepID) which has multiple histone-code reader including bromodomain, cryptic Tudor domain and WD40 domains and Cullin 4-RING ubiquitin ligase complex (CRL4) recruited to chromatin mediated by RepID have potential roles in gene expression changes via epigenetic regulations. We aimed to investigate whether RepID-CRL4 participates in transcriptional changes required for MK differentiation. Methods The PCR array was performed using cDNAs derived from RepID-proficient or RepID-deficient K562 erythroleukemia cell lines. Correlation between RepID and DAB2 expression was examined in the Cancer Cell Line Encyclopedia (CCLE) through the CellMinerCDB portal. The acceleration of MK differentiation in RepID-deficient K562 cells was determined by estimating cell sizes as well as counting multinucleated cells known as MK phenotypes, and by qRT-PCR analysis to validate transcripts of MK markers using phorbol 12-myristate 13-acetate (PMA)-mediated MK differentiation condition. Interaction between CRL4 and histone methylation modifying enzymes were investigated using BioGRID database, immunoprecipitation and proximity ligation assay. Alterations of expression and chromatin binding affinities of RepID, CRL4 and histone methylation modifying enzymes were investigated using subcellular fractionation followed by immunoblotting. RepID-CRL4-JARID1A-based epigenetic changes on DAB2 promoter were analyzed by chromatin-immunoprecipitation and qPCR analysis. Results RepID-deficient K562 cells highly expressing MK markers showed accelerated MKs differentiation exhibiting increases in cell size, lobulated nuclei together with reaching maximum levels of MK marker expression earlier than RepID-proficient K562 cells. Recovery of WD40 domain-containing RepID constructs in RepID-deficient background repressed DAB2 expression. CRL4A formed complex with histone H3K4 demethylase JARID1A in soluble nucleus and loaded to the DAB2 promoter in a RepID-dependent manner during proliferation condition. RepID, CRL4A, and JARID1A were dissociated from the chromatin during MK differentiation, leading to euchromatinization of the DAB2 promoter. Conclusion This study uncovered a role for the RepID-CRL4A-JARID1A pathway in the regulation of gene expression for MK differentiation, which can form the basis for the new therapeutic approaches to induce platelet production.

5.
EMBO Mol Med ; 15(8): e17313, 2023 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-37491889

RESUMEN

Small-cell lung cancer (SCLC) is the most lethal type of lung cancer. Specifically, MYC-driven non-neuroendocrine SCLC is particularly resistant to standard therapies. Lurbinectedin was recently approved for the treatment of relapsed SCLC, but combinatorial approaches are needed to increase the depth and duration of responses to lurbinectedin. Using high-throughput screens, we found inhibitors of ataxia telangiectasia mutated and rad3 related (ATR) as the most effective agents for augmenting lurbinectedin efficacy. First-in-class ATR inhibitor berzosertib synergized with lurbinectedin in multiple SCLC cell lines, organoid, and in vivo models. Mechanistically, ATR inhibition abrogated S-phase arrest induced by lurbinectedin and forced cell cycle progression causing mitotic catastrophe and cell death. High CDKN1A/p21 expression was associated with decreased synergy due to G1 arrest, while increased levels of ERCC5/XPG were predictive of increased combination efficacy. Importantly, MYC-driven non-neuroendocrine tumors which are resistant to first-line therapies show reduced CDKN1A/p21 expression and increased ERCC5/XPG indicating they are primed for response to lurbinectedin-berzosertib combination. The combination is being assessed in a clinical trial NCT04802174.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Neoplasias Pulmonares , Carcinoma Pulmonar de Células Pequeñas , Humanos , Recurrencia Local de Neoplasia , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/patología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo
6.
Mol Cancer Ther ; 22(8): 985-995, 2023 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-37216280

RESUMEN

Schlafen 11 (SLFN11) is an increasingly prominent predictive biomarker and a molecular sensor for a wide range of clinical drugs: topoisomerases, PARP and replication inhibitors, and platinum derivatives. To expand the spectrum of drugs and pathways targeting SLFN11, we ran a high-throughput screen with 1,978 mechanistically annotated, oncology-focused compounds in two isogenic pairs of SLFN11-proficient and -deficient cells (CCRF-CEM and K562). We identified 29 hit compounds that selectively kill SLFN11-proficient cells, including not only previously known DNA-targeting agents, but also the neddylation inhibitor pevonedistat (MLN-4924) and the DNA polymerase α inhibitor AHPN/CD437, which both induced SLFN11 chromatin recruitment. By inactivating cullin-ring E3 ligases, pevonedistat acts as an anticancer agent partly by inducing unscheduled re-replication through supraphysiologic accumulation of CDT1, an essential factor for replication initiation. Unlike the known DNA-targeting agents and AHPN/CD437 that recruit SLFN11 onto chromatin in 4 hours, pevonedistat recruited SLFN11 at late time points (24 hours). While pevonedistat induced unscheduled re-replication in SLFN11-deficient cells after 24 hours, the re-replication was largely blocked in SLFN11-proficient cells. The positive correlation between sensitivity to pevonedistat and SLFN11 expression was also observed in non-isogenic cancer cells in three independent cancer cell databases (NCI-60, CTRP: Cancer Therapeutics Response Portal and GDSC: Genomic of Drug Sensitivity in Cancer). The present study reveals that SLFN11 not only detects stressed replication but also inhibits unscheduled re-replication induced by pevonedistat, thereby enhancing its anticancer efficacy. It also suggests SLFN11 as a potential predictive biomarker for pevonedistat in ongoing and future clinical trials.


Asunto(s)
Antineoplásicos , Neoplasias , Humanos , Antineoplásicos/farmacología , Ciclopentanos/farmacología , Línea Celular Tumoral , Cromatina/genética , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Proteínas Nucleares/genética
7.
Cancer Res Commun ; 2(6): 503-517, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-36381660

RESUMEN

Endogenous replication stress is a major driver of genomic instability. Current assessments of replication stress are low throughput precluding its comprehensive assessment across tumors. Here we develop and validate a transcriptional profile of replication stress by leveraging established cellular characteristics that portend replication stress. The repstress gene signature defines a subset of tumors across lineages characterized by activated oncogenes, aneuploidy, extrachromosomal DNA amplification, immune evasion, high genomic instability, and poor survival, and importantly predicts response to agents targeting replication stress more robustly than previously reported transcriptomic measures of replication stress. Repstress score profiles the dual roles of replication stress during tumorigenesis and in established cancers and defines distinct molecular subtypes within cancers that may be more vulnerable to drugs targeting this dependency. Altogether, our study provides a molecular profile of replication stress, providing novel biological insights of the replication stress phenotype, with clinical implications.


Asunto(s)
Replicación del ADN , Neoplasias , Humanos , Replicación del ADN/genética , Oncogenes/genética , Neoplasias/genética , Transformación Celular Neoplásica/genética , Inestabilidad Genómica/genética
8.
Oncotarget ; 13: 986-1002, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36093297

RESUMEN

Classical MCL (cMCL) constitutes 6-8% of all B cell NHL. Despite recent advances, MCL is incurable except with allogeneic stem cell transplant. Blastic mantle cell lymphoma (bMCL) is a rarer subtype of cMCL associated with an aggressive clinical course and poor treatment response, frequent relapse and poor outcomes. We treated 13 bMCL patients with combined epigenetic and immunotherapy treatment consisting of vorinostat, cladribine and rituximab (SCR). We report an increased OS greater than 40 months with several patients maintaining durable remissions without relapse for longer than 5 years. This is remarkably better then current treatment regimens which in bMCL range from 14.5-24 months with conventional chemotherapy regimens. We demonstrate that the G/A870 CCND1 polymorphism is predictive of blastic disease, nuclear localization of cyclinD1 and response to SCR therapy. The major resistance mechanisms to SCR therapy are loss of CD20 expression and evasion of treatment by sanctuary in the CNS. These data indicate that administration of epigenetic agents improves efficacy of anti-CD20 immunotherapies. This approach is promising in the treatment of MCL and potentially other previously treatment refractory cancers.


Asunto(s)
Epigénesis Genética , Inmunoterapia , Linfoma de Células del Manto , Adulto , Antígenos CD20/inmunología , Cladribina , Humanos , Factores Inmunológicos/uso terapéutico , Linfoma de Células del Manto/tratamiento farmacológico , Linfoma de Células del Manto/terapia , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/terapia , Rituximab/uso terapéutico , Vorinostat/uso terapéutico
9.
Nat Commun ; 12(1): 5966, 2021 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-34645815

RESUMEN

The BRCA2 tumor suppressor protects genome integrity by promoting homologous recombination-based repair of DNA breaks, stability of stalled DNA replication forks and DNA damage-induced cell cycle checkpoints. BRCA2 deficient cells display the radio-resistant DNA synthesis (RDS) phenotype, however the mechanism has remained elusive. Here we show that cells without BRCA2 are unable to sufficiently restrain DNA replication fork progression after DNA damage, and the underrestrained fork progression is due primarily to Primase-Polymerase (PRIMPOL)-mediated repriming of DNA synthesis downstream of lesions, leaving behind single-stranded DNA gaps. Moreover, we find that BRCA2 associates with the essential DNA replication factor MCM10 and this association suppresses PRIMPOL-mediated repriming and ssDNA gap formation, while having no impact on the stability of stalled replication forks. Our findings establish an important function for BRCA2, provide insights into replication fork control during the DNA damage response, and may have implications in tumor suppression and therapy response.


Asunto(s)
Proteína BRCA2/genética , ADN Primasa/genética , ADN de Neoplasias/genética , ADN de Cadena Simple/genética , ADN Polimerasa Dirigida por ADN/genética , Proteínas de Mantenimiento de Minicromosoma/genética , Enzimas Multifuncionales/genética , Reparación del ADN por Recombinación , Proteína BRCA2/antagonistas & inhibidores , Proteína BRCA2/metabolismo , Línea Celular Tumoral , Supervivencia Celular , Daño del ADN , ADN Helicasas/antagonistas & inhibidores , ADN Helicasas/genética , ADN Helicasas/metabolismo , ADN Primasa/antagonistas & inhibidores , ADN Primasa/metabolismo , Replicación del ADN , ADN de Neoplasias/metabolismo , ADN de Cadena Simple/metabolismo , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , ADN Polimerasa Dirigida por ADN/metabolismo , Regulación Neoplásica de la Expresión Génica , Inestabilidad Genómica , Células HEK293 , Células HeLa , Humanos , Proteínas de Mantenimiento de Minicromosoma/antagonistas & inhibidores , Proteínas de Mantenimiento de Minicromosoma/metabolismo , Enzimas Multifuncionales/antagonistas & inhibidores , Enzimas Multifuncionales/metabolismo , Osteoblastos/metabolismo , Osteoblastos/patología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
10.
Nat Commun ; 12(1): 3448, 2021 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-34103496

RESUMEN

Safeguards against excess DNA replication are often dysregulated in cancer, and driving cancer cells towards over-replication is a promising therapeutic strategy. We determined DNA synthesis patterns in cancer cells undergoing partial genome re-replication due to perturbed regulatory interactions (re-replicating cells). These cells exhibited slow replication, increased frequency of replication initiation events, and a skewed initiation pattern that preferentially reactivated early-replicating origins. Unlike in cells exposed to replication stress, which activated a novel group of hitherto unutilized (dormant) replication origins, the preferred re-replicating origins arose from the same pool of potential origins as those activated during normal growth. Mechanistically, the skewed initiation pattern reflected a disproportionate distribution of pre-replication complexes on distinct regions of licensed chromatin prior to replication. This distinct pattern suggests that circumventing the strong inhibitory interactions that normally prevent excess DNA synthesis can occur via at least two pathways, each activating a distinct set of replication origins.


Asunto(s)
Replicación del ADN , Origen de Réplica , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Ciclopentanos/farmacología , Replicación del ADN/efectos de los fármacos , Replicación del ADN/genética , Genoma Humano , Humanos , Mitosis/efectos de los fármacos , Modelos Biológicos , Pirimidinas/farmacología , Origen de Réplica/genética
11.
J Biol Chem ; 295(50): 17169-17186, 2020 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-33028635

RESUMEN

We have observed overexpression of PACS-1, a cytosolic sorting protein in primary cervical tumors. Absence of exonic mutations and overexpression at the RNA level suggested a transcriptional and/or posttranscriptional regulation. University of California Santa Cruz genome browser analysis of PACS-1 micro RNAs (miR), revealed two 8-base target sequences at the 3' terminus for hsa-miR-34a and hsa-miR-449a. Quantitative RT-PCR and Northern blotting studies showed reduced or loss of expression of the two microRNAs in cervical cancer cell lines and primary tumors, indicating dysregulation of these two microRNAs in cervical cancer. Loss of PACS-1 with siRNA or exogenous expression of hsa-miR-34a or hsa-miR-449a in HeLa and SiHa cervical cancer cell lines resulted in DNA damage response, S-phase cell cycle arrest, and reduction in cell growth. Furthermore, the siRNA studies showed that loss of PACS-1 expression was accompanied by increased nuclear γH2AX expression, Lys382-p53 acetylation, and genomic instability. PACS-1 re-expression through LNA-hsa-anti-miR-34a or -449a or through PACS-1 cDNA transfection led to the reversal of DNA damage response and restoration of cell growth. Release of cells post 24-h serum starvation showed PACS-1 nuclear localization at G1-S phase of the cell cycle. Our results therefore indicate that the loss of hsa-miR-34a and hsa-miR-449a expression in cervical cancer leads to overexpression of PACS-1 and suppression of DNA damage response, resulting in the development of chemo-resistant tumors.


Asunto(s)
Daño del ADN , Resistencia a Antineoplásicos , MicroARNs/metabolismo , ARN Neoplásico/metabolismo , Neoplasias del Cuello Uterino/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Femenino , Fase G1 , Células HeLa , Humanos , MicroARNs/genética , ARN Neoplásico/genética , Puntos de Control de la Fase S del Ciclo Celular , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/patología , Proteínas de Transporte Vesicular/genética
12.
Mol Med Rep ; 22(5): 3715-3722, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32901867

RESUMEN

The imbalance induced by inhibition of bone mesenchymal stem cell (BMSC) osteogenic differentiation results in osteoporosis (OP); however, the underlying regulatory mechanism is not completely understood. Long non­coding RNAs (lncRNAs) serve crucial roles in osteogenic differentiation; therefore, investigating their regulatory role in the process of osteogenic differentiation may identify a promising therapeutic target for OP. The expression of small nucleolar RNA host gene 1 (SNHG1), Dickkopf 1 (DKK1), microRNA (miR)­101, RUNX family transcription factor 2 (RUNX2), osteopontin (OPN) and osteocalin (OCN) were detected via reverse transcription­quantitative PCR. The protein expression levels of DKK1, ß­catenin, RUNX2, OPN, OCN, osterix and collagen type I α1 chain were analyzed by performing western blotting. The osteoblastic phenotype was assessed by conducting alkaline phosphatase activity detection and Alizarin Red staining. The interaction between SNHG1 and miR­101 was validated by bioinformatics and luciferase assays. The regulatory role of SNHG1 in BMSC osteogenic differentiation was assessed. SNHG1 expression was downregulated in a time­dependent manner during the process of osteogenic differentiation. SNHG1 overexpression inhibited osteogenic differentiation compared with the pcDNA group. The results indicated that SNHG1 and DKK1 directly interacted with miR­101. Moreover, SNHG1 regulated the Wnt/ß­catenin signaling pathway to inhibit osteogenic differentiation via the miR­101/DKK1 axis. The present study indicated that lncRNA SNHG1 could attenuate BMSC osteogenic differentiation via the miR­101/DKK1 axis as a competitive endogenous RNA. Therefore, the present study furthered the current understanding of the potential mechanism underlying lncRNAs in in osteogenic differentiation.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/genética , Células Madre Mesenquimatosas/citología , MicroARNs/genética , ARN Largo no Codificante/genética , Diferenciación Celular , Células Cultivadas , Regulación de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Células Madre Mesenquimatosas/metabolismo , Osteogénesis , Fenotipo , Factores de Tiempo , Vía de Señalización Wnt
13.
Nat Commun ; 11(1): 24, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-31911655

RESUMEN

The spindle assembly checkpoint (SAC) prevents premature chromosome segregation by inactivating the anaphase promoting complex/cyclosome (APC/C) until all chromosomes are properly attached to mitotic spindles. Here we identify a role for Cullin-RING ubiquitin ligase complex 4 (CRL4), known for modulating DNA replication, as a crucial mitotic regulator that triggers the termination of the SAC and enables chromosome segregation. CRL4 is recruited to chromatin by the replication origin binding protein RepID/DCAF14/PHIP. During mitosis, CRL4 dissociates from RepID and replaces it with RB Binding Protein 7 (RBBP7), which ubiquitinates the SAC mediator BUB3 to enable mitotic exit. During interphase, BUB3 is protected from CRL4-mediated degradation by associating with promyelocytic leukemia (PML) nuclear bodies, ensuring its availability upon mitotic onset. Deficiencies in RepID, CRL4 or RBBP7 delay mitotic exit, increase genomic instability and enhance sensitivity to paclitaxel, a microtubule stabilizer and anti-tumor drug.


Asunto(s)
Anafase , Proteínas de Ciclo Celular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Metafase , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas de Ciclo Celular/genética , Línea Celular , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Mitosis , Proteínas de Unión a Poli-ADP-Ribosa/genética , Proteína de la Leucemia Promielocítica/genética , Proteína de la Leucemia Promielocítica/metabolismo , Unión Proteica , Proteolisis , Proteína 7 de Unión a Retinoblastoma/genética , Proteína 7 de Unión a Retinoblastoma/metabolismo , Huso Acromático/metabolismo , Ubiquitina-Proteína Ligasas/genética
14.
Hum Mol Genet ; 28(5): 842-857, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30445628

RESUMEN

The mammary gland undergoes fast cell proliferation during early pregnancy, yet the mechanism to ensure genome integrity during this highly proliferative stage is largely unknown. We show that pregnancy triggers replicative stresses leading to genetic instability in mice carrying a mammary specific disruption of breast cancer associated gene-1 (BRCA1). The fast cell proliferation was correlated with enhanced expression of most genes encoding replisomes, which are positively regulated by estrogen/ERα signaling but negatively regulated by BRCA1. Our further analysis revealed two parallel signaling pathways, which are mediated by ATR-CHK1 and WEE1-MCM2 and are responsible for regulating DNA replication checkpoint. Upon DNA damage, BRCA1 deficiency markedly enhances DNA replication initiation and preferably impairs DNA replication checkpoint mediated by ATR and CHK1. Meanwhile, DNA damage also activates WEE1-MCM2 signaling, which inhibits DNA replication initiation and enables BRCA1-deficient cells to avoid further genomic instability. Finally, we demonstrated that overriding this defense by WEE1 inhibition in combination with cisplatin, which causes DNA damage, serves as a promising therapeutic approach for killing BRCA1-deficient cancer cells.


Asunto(s)
Proteína BRCA1/genética , Proteínas de Ciclo Celular/metabolismo , Replicación del ADN , Estrógenos/metabolismo , Inestabilidad Genómica , Componente 2 del Complejo de Mantenimiento de Minicromosoma/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Transducción de Señal , Antineoplásicos Inmunológicos/farmacología , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Secuencia de Bases , Sitios de Unión , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Estrógenos/agonistas , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Fosforilación , Embarazo , Regiones Promotoras Genéticas , Transducción de Señal/efectos de los fármacos
15.
Curr Protoc Microbiol ; 51(1): e61, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30129235

RESUMEN

Human papillomaviruses (HPVs) are frequently integrated in HPV-associated cancers. HPV genomes can be integrated in three patterns: A single integrated HPV genome (type I), multiple, tandemly integrated HPV genomes (type II), and multiple, tandemly integrated HPV genomes interspersed with host DNA (type III). Analysis of the organization of type II and type III integration sites is complicated by their repetitive nature, as sequences of individual repeats are difficult to distinguish from each other. This article presents a method for directly visualizing HPV integration sites using molecular combing combined with fluorescent in situ hybridization, also known as fiber-FISH. In this technique, genomic DNA is stretched across a glass coverslip and individual integrated HPV sequences are detected and directly visualized by in situ hybridization with a resolution of ∼1 kb. Fiber-FISH allows comprehensive characterization of the genomic organization of HPV integration sites containing type II and type III integration. © 2018 by John Wiley & Sons, Inc.


Asunto(s)
ADN Viral/genética , Hibridación Fluorescente in Situ/métodos , Biología Molecular/métodos , Papillomaviridae/fisiología , Integración Viral , ADN Viral/análisis , Humanos , Papillomaviridae/genética
16.
Nat Commun ; 9(1): 2782, 2018 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-30018425

RESUMEN

Cell cycle progression in mammals is modulated by two ubiquitin ligase complexes, CRL4 and SCF, which facilitate degradation of chromatin substrates involved in the regulation of DNA replication. One member of the CRL4 complex, the WD-40 containing protein RepID (DCAF14/PHIP), selectively binds and activates a group of replication origins. Here we show that RepID recruits the CRL4 complex to chromatin prior to DNA synthesis, thus playing a crucial architectural role in the proper licensing of chromosomes for replication. In the absence of RepID, cells rely on the alternative ubiquitin ligase, SKP2-containing SCF, to progress through the cell cycle. RepID depletion markedly increases cellular sensitivity to SKP2 inhibitors, which triggered massive genome re-replication. Both RepID and SKP2 interact with distinct, non-overlapping groups of replication origins, suggesting that selective interactions of replication origins with specific CRL components execute the DNA replication program and maintain genomic stability by preventing re-initiation of DNA replication.


Asunto(s)
Cromatina/química , Proteínas Cullin/genética , Replicación del ADN , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Quinasas Asociadas a Fase-S/genética , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Línea Celular Tumoral , Cromatina/metabolismo , Proteínas Cullin/metabolismo , Ciclopentanos/farmacología , Expresión Génica , Células HCT116 , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Células K562 , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Unión Proteica , Transporte de Proteínas , Pirimidinas/farmacología , Origen de Réplica , Proteínas Quinasas Asociadas a Fase-S/antagonistas & inhibidores , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Factor de Células Madre/genética , Factor de Células Madre/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
17.
PLoS Genet ; 14(1): e1007179, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29364907

RESUMEN

Integration of human papillomavirus (HPV) genomes into cellular chromatin is common in HPV-associated cancers. Integration is random, and each site is unique depending on how and where the virus integrates. We recently showed that tandemly integrated HPV16 could result in the formation of a super-enhancer-like element that drives transcription of the viral oncogenes. Here, we characterize the chromatin landscape and genomic architecture of this integration locus to elucidate the mechanisms that promoted de novo super-enhancer formation. Using next-generation sequencing and molecular combing/fiber-FISH, we show that ~26 copies of HPV16 are integrated into an intergenic region of chromosome 2p23.2, interspersed with 25 kb of amplified, flanking cellular DNA. This interspersed, co-amplified viral-host pattern is frequent in HPV-associated cancers and here we designate it as Type III integration. An abundant viral-cellular fusion transcript encoding the viral E6/E7 oncogenes is expressed from the integration locus and the chromatin encompassing both the viral enhancer and a region in the adjacent amplified cellular sequences is strongly enriched in the super-enhancer markers H3K27ac and Brd4. Notably, the peak in the amplified cellular sequence corresponds to an epithelial-cell-type specific enhancer. Thus, HPV16 integration generated a super-enhancer-like element composed of tandem interspersed copies of the viral upstream regulatory region and a cellular enhancer, to drive high levels of oncogene expression.


Asunto(s)
Regulación Viral de la Expresión Génica , Genes Virales , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/patogenicidad , Factores de Transcripción/metabolismo , Integración Viral/fisiología , Células Cultivadas , Elementos de Facilitación Genéticos , Células HCT116 , Células HeLa , Células Hep G2 , Interacciones Huésped-Patógeno/genética , Células Endoteliales de la Vena Umbilical Humana , Papillomavirus Humano 16/metabolismo , Humanos , Células K562 , Virus Oncogénicos/genética , Virus Oncogénicos/patogenicidad , Papillomaviridae/genética , Papillomaviridae/metabolismo , Papillomaviridae/patogenicidad , Unión Proteica , Multimerización de Proteína , Regulación hacia Arriba/genética
18.
Nucleic Acids Res ; 45(13): 7807-7824, 2017 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-28549174

RESUMEN

Chromatin structure affects DNA replication patterns, but the role of specific chromatin modifiers in regulating the replication process is yet unclear. We report that phosphorylation of the human SIRT1 deacetylase on Threonine 530 (T530-pSIRT1) modulates DNA synthesis. T530-pSIRT1 associates with replication origins and inhibits replication from a group of 'dormant' potential replication origins, which initiate replication only when cells are subject to replication stress. Although both active and dormant origins bind T530-pSIRT1, active origins are distinguished from dormant origins by their unique association with an open chromatin mark, histone H3 methylated on lysine 4. SIRT1 phosphorylation also facilitates replication fork elongation. SIRT1 T530 phosphorylation is essential to prevent DNA breakage upon replication stress and cells harboring SIRT1 that cannot be phosphorylated exhibit a high prevalence of extrachromosomal elements, hallmarks of perturbed replication. These observations suggest that SIRT1 phosphorylation modulates the distribution of replication initiation events to insure genomic stability.


Asunto(s)
Replicación del ADN , Inestabilidad Genómica , Origen de Réplica , Sirtuina 1/metabolismo , Línea Celular , Roturas del ADN , Replicación del ADN/genética , Células HCT116 , Humanos , Células K562 , Células MCF-7 , Modelos Biológicos , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , ARN Interferente Pequeño/genética , Sirtuina 1/antagonistas & inhibidores , Sirtuina 1/genética , Treonina/química , Quinasas DyrK
19.
Artículo en Inglés | MEDLINE | ID: mdl-27168766

RESUMEN

BACKGROUND: Eukaryotic genome duplication starts at discrete sequences (replication origins) that coordinate cell cycle progression, ensure genomic stability and modulate gene expression. Origins share some sequence features, but their activity also responds to changes in transcription and cellular differentiation status. RESULTS: To identify chromatin states and histone modifications that locally mark replication origins, we profiled origin distributions in eight human cell lines representing embryonic and differentiated cell types. Consistent with a role of chromatin structure in determining origin activity, we found that cancer and non-cancer cells of similar lineages exhibited highly similar replication origin distributions. Surprisingly, our study revealed that DNase hypersensitivity, which often correlates with early replication at large-scale chromatin domains, did not emerge as a strong local determinant of origin activity. Instead, we found that two distinct sets of chromatin modifications exhibited strong local associations with two discrete groups of replication origins. The first origin group consisted of about 40,000 regions that actively initiated replication in all cell types and preferentially colocalized with unmethylated CpGs and with the euchromatin markers, H3K4me3 and H3K9Ac. The second group included origins that were consistently active in cells of a single type or lineage and preferentially colocalized with the heterochromatin marker, H3K9me3. Shared origins replicated throughout the S-phase of the cell cycle, whereas cell-type-specific origins preferentially replicated during late S-phase. CONCLUSIONS: These observations are in line with the hypothesis that differentiation-associated changes in chromatin and gene expression affect the activation of specific replication origins.

20.
Int J Biol Sci ; 10(10): 1193-202, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25516717

RESUMEN

SIRT1, the mammalian homolog of yeast Sir2, is a founding member of a family of 7 protein and histone deacetylases that are involved in numerous biological functions. Previous studies revealed that SIRT1 deficiency results in genome instability, which eventually leads to cancer formation, yet the underlying mechanism is unclear. To investigate this, we conducted a proteomics study and found that SIRT1 interacted with many proteins involved in replication fork protection and origin firing. We demonstrated that loss of SIRT1 resulted in increased replication origin firing, asymmetric fork progression, defective intra-S-phase checkpoint, and chromosome damage. Mechanistically, SIRT1 deacetylates and affects the activity of TopBP1, which plays an essential role in DNA replication fork protection and replication origin firing. Our study demonstrated that ectopic over-expression of the deacetylated form of TopBP1 in SIRT1 mutant cells repressed replication origin firing, while the acetylated form of TopBP1 lost this function. Thus, SIRT1 acts upstream of TopBP1 and plays an essential role in maintaining genome stability by modulating DNA replication fork initiation and the intra-S-phase cell cycle checkpoint.


Asunto(s)
Proteínas Portadoras/metabolismo , Inestabilidad Genómica/genética , Origen de Réplica/fisiología , Puntos de Control de la Fase S del Ciclo Celular/fisiología , Sirtuina 1/metabolismo , Acetilación , Animales , Western Blotting , Bromodesoxiuridina , Análisis Citogenético , Vectores Genéticos/genética , Células HEK293 , Humanos , Inmunoprecipitación , Lentivirus , Espectrometría de Masas , Ratones , Ratones Noqueados , ARN Interferente Pequeño/genética , Sirtuina 1/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA