Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Future Oncol ; 17(33): 4457-4470, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34350781

RESUMEN

Aim: To investigate the role of MCM10, a conserved replication factor, in hepatocellular carcinoma (HCC). Methods: We used data from 364 HCC patients in the Cancer Genome Atlas database and conducted in vitro experiments to confirm the role of MCM10. Results: High MCM10 expression correlated with poor HCC patient outcome and was an independent prognosticator for HCC. Time-dependent receiver operating characteristic curve analysis found that the sequential trend of MCM10 for survival was not inferior to that of the tumor node metastasis stage. The MCM10 model had a higher C-index than the non-MCM10 model, indicating that incorporating MCM10 into a multivariate model improves the model's prognostic accuracy for HCC. Genetic alterations of MCM10 prominently correlated with an unfavorable HCC outcome. Conclusion: Our findings strongly suggest using the MCM10 gene as a prognostic indicator in HCC.


Lay abstract Hepatocellular carcinoma is one of the most aggressive malignant cancers globally. Our study investigated the role of a conserved replication factor, MCM10, in hepatocellular carcinoma. We performed some bioinformatics analysis and in vitro experiments, and successfully found that MCM10 has a good predictive value for survival in patients with hepatocellular carcinoma.


Asunto(s)
Biomarcadores de Tumor/genética , Carcinoma Hepatocelular/mortalidad , Neoplasias Hepáticas/mortalidad , Proteínas de Mantenimiento de Minicromosoma/genética , Recurrencia Local de Neoplasia/epidemiología , Anciano , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/metabolismo , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/cirugía , Biología Computacional , Variaciones en el Número de Copia de ADN , Supervivencia sin Enfermedad , Femenino , Estudios de Seguimiento , Hepatectomía , Humanos , Estimación de Kaplan-Meier , Hígado/patología , Hígado/cirugía , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/cirugía , Masculino , Persona de Mediana Edad , Proteínas de Mantenimiento de Minicromosoma/análisis , Proteínas de Mantenimiento de Minicromosoma/metabolismo , Mutación , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/prevención & control , Pronóstico , Mapeo de Interacción de Proteínas , Mapas de Interacción de Proteínas/genética , Curva ROC , Estudios Retrospectivos , Medición de Riesgo/métodos
2.
J Biol Chem ; 296: 100355, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33539926

RESUMEN

The MCM8/9 complex is implicated in aiding fork progression and facilitating homologous recombination (HR) in response to several DNA damage agents. MCM9 itself is an outlier within the MCM family containing a long C-terminal extension (CTE) comprising 42% of the total length, but with no known functional components and high predicted disorder. In this report, we identify and characterize two unique motifs within the primarily unstructured CTE that are required for localization of MCM8/9 to sites of mitomycin C (MMC)-induced DNA damage. First, an unconventional "bipartite-like" nuclear localization (NLS) motif consisting of two positively charged amino acid stretches separated by a long intervening sequence is required for the nuclear import of both MCM8 and MCM9. Second, a variant of the BRC motif (BRCv) similar to that found in other HR helicases is necessary for localization to sites of MMC damage. The MCM9-BRCv directly interacts with and recruits RAD51 downstream to MMC-induced damage to aid in DNA repair. Patient lymphocytes devoid of functional MCM9 and discrete MCM9 knockout cells have a significantly impaired ability to form RAD51 foci after MMC treatment. Therefore, the disordered CTE in MCM9 is functionally important in promoting MCM8/9 activity and in recruiting downstream interactors; thus, requiring full-length MCM9 for proper DNA repair.


Asunto(s)
Antibióticos Antineoplásicos/farmacología , Daño del ADN/efectos de los fármacos , Proteínas de Mantenimiento de Minicromosoma/metabolismo , Mitomicina/farmacología , Recombinasa Rad51/metabolismo , Línea Celular Tumoral , Células HEK293 , Humanos , Proteínas de Mantenimiento de Minicromosoma/análisis , Recombinasa Rad51/análisis
3.
Nature ; 587(7833): 297-302, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33087936

RESUMEN

Minichromosome maintenance proteins (MCMs) are DNA-dependent ATPases that bind to replication origins and license them to support a single round of DNA replication. A large excess of MCM2-7 assembles on chromatin in G1 phase as pre-replication complexes (pre-RCs), of which only a fraction become the productive CDC45-MCM-GINS (CMG) helicases that are required for genome duplication1-4. It remains unclear why cells generate this surplus of MCMs, how they manage to sustain it across multiple generations, and why even a mild reduction in the MCM pool compromises the integrity of replicating genomes5,6. Here we show that, for daughter cells to sustain error-free DNA replication, their mother cells build up a nuclear pool of MCMs both by recycling chromatin-bound (parental) MCMs and by synthesizing new (nascent) MCMs. Although all MCMs can form pre-RCs, it is the parental pool that is inherently stable and preferentially matures into CMGs. By contrast, nascent MCM3-7 (but not MCM2) undergo rapid proteolysis in the cytoplasm, and their stabilization and nuclear translocation require interaction with minichromosome-maintenance complex-binding protein (MCMBP), a distant MCM paralogue7,8. By chaperoning nascent MCMs, MCMBP safeguards replicating genomes by increasing chromatin coverage with pre-RCs that do not participate on replication origins but adjust the pace of replisome movement to minimize errors during DNA replication. Consequently, although the paucity of pre-RCs in MCMBP-deficient cells does not alter DNA synthesis overall, it increases the speed and asymmetry of individual replisomes, which leads to DNA damage. The surplus of MCMs therefore increases the robustness of genome duplication by restraining the speed at which eukaryotic cells replicate their DNA. Alterations in physiological fork speed might thus explain why even a minor reduction in MCM levels destabilizes the genome and predisposes to increased incidence of tumour formation.


Asunto(s)
Replicación del ADN/genética , Genoma Humano/genética , Proteínas de Mantenimiento de Minicromosoma/biosíntesis , Proteínas de Mantenimiento de Minicromosoma/metabolismo , Transporte Activo de Núcleo Celular , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Núcleo Celular/metabolismo , Cromatina/genética , Cromatina/metabolismo , Daño del ADN , Humanos , Proteínas de Mantenimiento de Minicromosoma/análisis , Chaperonas Moleculares/química , Chaperonas Moleculares/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Estabilidad Proteica , Transporte de Proteínas
4.
Biochemistry (Mosc) ; 83(1): 69-75, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29534671

RESUMEN

Colon cancer remains one of the most common digestive system malignancies in the World. This study investigated the possible interaction between RAD51 and minichromosome maintenance proteins (MCMs) in HCT116 cells, which can serve as a model system for forming colon cancer foci. The interaction between RAD51 and MCMs was detected by mass spectrometry. Silenced MCM vectors were transfected into HTC116 cells. The expressions of RAD51 and MCMs were detected using Western blotting. Foci forming and chromatin fraction of RAD51 in HCT116 cells were also analyzed. The results showed that RAD51 directly interacted with MCM2, MCM3, MCM5, and MCM6 in colon cancer HTC116 cells. Suppression of MCM2 or MCM6 by shRNA decreased the chromatin localization of RAD51 in HTC116 cells. Moreover, silenced MCM2 or MCM6 decreased the foci forming of RAD51 in HTC116 cells. Our study suggests that the interaction between MCMs and RAD51 is essential for the chromatin localization and foci forming of RAD51 in HCT116 cell DNA damage recovery, and it may be a theoretical basis for analysis of RAD51 in tumor samples of colon cancer patients.


Asunto(s)
Neoplasias del Colon/metabolismo , Proteínas de Mantenimiento de Minicromosoma/metabolismo , Recombinasa Rad51/metabolismo , Células Cultivadas , Neoplasias del Colon/química , Células HCT116 , Células HEK293 , Humanos , Espectrometría de Masas , Proteínas de Mantenimiento de Minicromosoma/análisis , Proteínas de Mantenimiento de Minicromosoma/antagonistas & inhibidores , ARN Interferente Pequeño/farmacología , Recombinasa Rad51/análisis , Recombinasa Rad51/antagonistas & inhibidores
5.
Pediatr Blood Cancer ; 64(12)2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28598542

RESUMEN

BACKGROUND: Overexpression of minichromosome maintenance (MCM) proteins 2, 3, and 7 is associated with migration and invasion in medulloblastoma (MB). However, expression profiling of all prereplication complex (pre-RC) has not been addressed in MBs. PROCEDURE: We performed mRNA expression profiling of a large set of pre-RC elements in cell lines and tumor tissues of MB. RNAi technology was employed for functional studies in MB cell lines. RESULTS: Our data showed that most of the pre-RC components are significantly overexpressed in MB. Among all pre-RC mRNAs, MCM10 showed the highest level of expression (∼500- to 1,000-fold) in MB cell lines and tissues compared to the levels detected in cerebellum. In addition, RNAi silencing of MCM10 caused reduced cell proliferation and cell viability in MB cells. CONCLUSIONS: Taken together, our study reveals that the pre-RC is dysregulated in MB. In addition, MCM10, a member of this complex, is significantly overexpressed in MB and is required for tumor cell proliferation.


Asunto(s)
Neoplasias Cerebelosas/química , Meduloblastoma/química , Proteínas de Mantenimiento de Minicromosoma/fisiología , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Neoplasias Cerebelosas/patología , Humanos , Inmunohistoquímica , Meduloblastoma/patología , Proteínas de Mantenimiento de Minicromosoma/análisis
6.
Crit Rev Biochem Mol Biol ; 52(2): 107-144, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28094588

RESUMEN

Cellular DNA replication is initiated through the action of multiprotein complexes that recognize replication start sites in the chromosome (termed origins) and facilitate duplex DNA melting within these regions. In a typical cell cycle, initiation occurs only once per origin and each round of replication is tightly coupled to cell division. To avoid aberrant origin firing and re-replication, eukaryotes tightly regulate two events in the initiation process: loading of the replicative helicase, MCM2-7, onto chromatin by the origin recognition complex (ORC), and subsequent activation of the helicase by its incorporation into a complex known as the CMG. Recent work has begun to reveal the details of an orchestrated and sequential exchange of initiation factors on DNA that give rise to a replication-competent complex, the replisome. Here, we review the molecular mechanisms that underpin eukaryotic DNA replication initiation - from selecting replication start sites to replicative helicase loading and activation - and describe how these events are often distinctly regulated across different eukaryotic model organisms.


Asunto(s)
Replicación del ADN , Células Eucariotas/metabolismo , Animales , Cromatina/genética , Cromatina/metabolismo , Activación Enzimática , Humanos , Proteínas de Mantenimiento de Minicromosoma/análisis , Proteínas de Mantenimiento de Minicromosoma/metabolismo , Complejo de Reconocimiento del Origen/análisis , Complejo de Reconocimiento del Origen/metabolismo , Origen de Réplica
7.
Nucleic Acids Res ; 43(22): e150, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26250117

RESUMEN

Passage through the Retinoblastoma protein (RB1)-dependent restriction point and the loading of minichromosome maintenance proteins (MCMs) are two crucial events in G1-phase that help maintain genome integrity. Deregulation of these processes can cause uncontrolled proliferation and cancer development. Both events have been extensively characterized individually, but their relative timing and inter-dependence remain less clear. Here, we describe a novel method to simultaneously measure MCM loading and passage through the restriction point. We exploit that the RB1 protein is anchored in G1-phase but is released when hyper-phosphorylated at the restriction point. After extracting cells with salt and detergent before fixation we can simultaneously measure, by flow cytometry, the loading of MCMs onto chromatin and RB1 binding to determine the order of the two events in individual cells. We have used this method to examine the relative timing of the two events in human cells. Whereas in BJ fibroblasts released from G0-phase MCM loading started mainly after the restriction point, in a significant fraction of exponentially growing BJ and U2OS osteosarcoma cells MCMs were loaded in G1-phase with RB1 anchored, demonstrating that MCM loading can also start before the restriction point. These results were supported by measurements in synchronized U2OS cells.


Asunto(s)
Puntos de Control de la Fase G1 del Ciclo Celular , Proteínas de Mantenimiento de Minicromosoma/análisis , Línea Celular Tumoral , Células Cultivadas , Citometría de Flujo , Humanos , Nocodazol/farmacología , Fase de Descanso del Ciclo Celular , Proteína de Retinoblastoma/metabolismo , Análisis de la Célula Individual
8.
Methods Mol Biol ; 1170: 529-37, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24906334

RESUMEN

S-CDK and DDK protein kinases initiate DNA replication at replication origins. Prior to the activation of these kinases, origins must become competent for replication by loading MCM2-7 DNA helicase on chromatin. This process is known as replication licensing or pre-replicative complex (pre-RC) formation. After the onset of S phase, however, licensing is inhibited to prevent re-replication of DNA. In this chapter, we describe a method to analyze origin licensing by imaging the chromatin-bound licensing factor MCM2-7. In a normal cell cycle, MCM2-7 is loaded at the end of mitosis or early G1 phase. As S phase progresses, MCM2-7 is dissociated from the replicated regions. When DNA replication is completed, cells in G2 phase have no chromatin-bound MCM2-7. The analysis of chromatin-bound MCM2-7 in each cell provides an insight into cell cycle stage and condition for cell cycle.


Asunto(s)
Cromatina/metabolismo , Técnica del Anticuerpo Fluorescente/métodos , Microscopía Fluorescente/métodos , Proteínas de Mantenimiento de Minicromosoma/metabolismo , Ciclo Celular , Células HeLa , Humanos , Proteínas de Mantenimiento de Minicromosoma/análisis , Imagen Óptica/métodos , Unión Proteica , Fijación del Tejido/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...