Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 657
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 175(5): 1272-1288.e20, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30343899

RESUMEN

Mammalian SWI/SNF (mSWI/SNF) ATP-dependent chromatin remodeling complexes are multi-subunit molecular machines that play vital roles in regulating genomic architecture and are frequently disrupted in human cancer and developmental disorders. To date, the modular organization and pathways of assembly of these chromatin regulators remain unknown, presenting a major barrier to structural and functional determination. Here, we elucidate the architecture and assembly pathway across three classes of mSWI/SNF complexes-canonical BRG1/BRM-associated factor (BAF), polybromo-associated BAF (PBAF), and newly defined ncBAF complexes-and define the requirement of each subunit for complex formation and stability. Using affinity purification of endogenous complexes from mammalian and Drosophila cells coupled with cross-linking mass spectrometry (CX-MS) and mutagenesis, we uncover three distinct and evolutionarily conserved modules, their organization, and the temporal incorporation of these modules into each complete mSWI/SNF complex class. Finally, we map human disease-associated mutations within subunits and modules, defining specific topological regions that are affected upon subunit perturbation.


Asunto(s)
Ensamble y Desensamble de Cromatina , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Factores de Transcripción/metabolismo , Animales , Cromatina/química , Proteínas Cromosómicas no Histona/análisis , Proteínas Cromosómicas no Histona/genética , Drosophila/metabolismo , Técnicas de Inactivación de Genes , Células HEK293 , Humanos , Espectrometría de Masas , Mutagénesis , Subunidades de Proteína/análisis , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Factores de Transcripción/análisis , Factores de Transcripción/genética
2.
Cell ; 165(7): 1686-1697, 2016 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-27212236

RESUMEN

The nucleolus and other ribonucleoprotein (RNP) bodies are membrane-less organelles that appear to assemble through phase separation of their molecular components. However, many such RNP bodies contain internal subcompartments, and the mechanism of their formation remains unclear. Here, we combine in vivo and in vitro studies, together with computational modeling, to show that subcompartments within the nucleolus represent distinct, coexisting liquid phases. Consistent with their in vivo immiscibility, purified nucleolar proteins phase separate into droplets containing distinct non-coalescing phases that are remarkably similar to nucleoli in vivo. This layered droplet organization is caused by differences in the biophysical properties of the phases-particularly droplet surface tension-which arises from sequence-encoded features of their macromolecular components. These results suggest that phase separation can give rise to multilayered liquids that may facilitate sequential RNA processing reactions in a variety of RNP bodies. PAPERCLIP.


Asunto(s)
Nucléolo Celular/química , Animales , Caenorhabditis elegans , Células Cultivadas , Proteínas Cromosómicas no Histona/análisis , Intestinos/química , Intestinos/citología , Mamíferos , Proteínas Nucleares/análisis , Nucleofosmina , Oocitos/química , Oocitos/citología , Procesamiento Postranscripcional del ARN , Ribonucleoproteínas/metabolismo , Xenopus laevis
3.
Cell ; 161(5): 1124-1137, 2015 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-26000485

RESUMEN

Mammalian mitotic chromosome morphogenesis was analyzed by 4D live-cell and snapshot deconvolution fluorescence imaging. Prophase chromosomes, whose organization was previously unknown, are revealed to comprise co-oriented sister linear loop arrays displayed along a single, peripheral, regularly kinked topoisomerase II/cohesin/condensin II axis. Thereafter, rather than smooth, progressive compaction as generally envisioned, progression to metaphase is a discontinuous process involving chromosome expansion as well as compaction. At late prophase, dependent on topoisomerase II and with concomitant cohesin release, chromosomes expand, axes split and straighten, and chromatin loops transit to a radial disposition around now-central axes. Finally, chromosomes globally compact, giving the metaphase state. These patterns are consistent with the hypothesis that the molecular events of chromosome morphogenesis are governed by accumulation and release of chromosome stress, created by chromatin compaction and expansion. Chromosome state could evolve analogously throughout the cell cycle.


Asunto(s)
Cromosomas de los Mamíferos/metabolismo , Metafase , Mitosis , Adenosina Trifosfatasas/análisis , Animales , Proteínas de Ciclo Celular/análisis , Línea Celular , Proteínas Cromosómicas no Histona/análisis , Cromosomas de los Mamíferos/química , ADN-Topoisomerasas de Tipo II/análisis , Proteínas de Unión al ADN/análisis , Ciervos , Células HeLa , Humanos , Microscopía Fluorescente , Complejos Multiproteicos/análisis , Porcinos , Cohesinas
4.
Cell ; 142(5): 810-21, 2010 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-20813266

RESUMEN

Despite many decades of study, mitotic chromosome structure and composition remain poorly characterized. Here, we have integrated quantitative proteomics with bioinformatic analysis to generate a series of independent classifiers that describe the approximately 4,000 proteins identified in isolated mitotic chromosomes. Integrating these classifiers by machine learning uncovers functional relationships between protein complexes in the context of intact chromosomes and reveals which of the approximately 560 uncharacterized proteins identified here merits further study. Indeed, of 34 GFP-tagged predicted chromosomal proteins, 30 were chromosomal, including 13 with centromere-association. Of 16 GFP-tagged predicted nonchromosomal proteins, 14 were confirmed to be nonchromosomal. An unbiased analysis of the whole chromosome proteome from genetic knockouts of kinetochore protein Ska3/Rama1 revealed that the APC/C and RanBP2/RanGAP1 complexes depend on the Ska complex for stable association with chromosomes. Our integrated analysis predicts that up to 97 new centromere-associated proteins remain to be discovered in our data set.


Asunto(s)
Proteínas Cromosómicas no Histona/análisis , Cromosomas/química , Mitosis , Proteómica/métodos , Animales , Línea Celular , Proteínas Cromosómicas no Histona/metabolismo , Cromosomas/metabolismo , Humanos , Cinetocoros/metabolismo , Huso Acromático/metabolismo
5.
Nucleic Acids Res ; 48(8): 4161-4178, 2020 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-32182352

RESUMEN

During interphase centromeres often coalesce into a small number of chromocenters, which can be visualized as distinct, DAPI dense nuclear domains. Intact chromocenters play a major role in maintaining genome stability as they stabilize the transcriptionally silent state of repetitive DNA while ensuring centromere function. Despite its biological importance, relatively little is known about the molecular composition of the chromocenter or the processes that mediate chromocenter formation and maintenance. To provide a deeper molecular insight into the composition of the chromocenter and to demonstrate the usefulness of proximity-based biotinylation as a tool to investigate those questions, we performed super resolution microscopy and proximity-based biotinylation experiments of three distinct proteins associated with the chromocenter in Drosophila. Our work revealed an intricate internal architecture of the chromocenter suggesting a complex multilayered structure of this intranuclear domain.


Asunto(s)
Proteína A Centromérica/metabolismo , Centrómero/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Drosophila/metabolismo , Interfase/genética , Adenosina Trifosfatasas/metabolismo , Animales , Biotinilación , Proteínas de Ciclo Celular/análisis , Línea Celular , Núcleo Celular/metabolismo , Proteína A Centromérica/genética , Cromatina/metabolismo , Secuenciación de Inmunoprecipitación de Cromatina , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/análisis , Proteínas Cromosómicas no Histona/genética , Proteínas de Unión al ADN/metabolismo , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Complejos Multiproteicos/metabolismo , Proteómica , Proteínas Recombinantes de Fusión/análisis , Cohesinas
6.
Int J Med Sci ; 18(2): 494-504, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33390818

RESUMEN

Background and aim: The molecular signatures of lung adenocarcinoma (LUAD) are not well understood. Centromere protein F (CENPF) has been shown to promote oncogenesis in many cancers; however, its role in LUAD has not been illustrated. We explored the role of CENPF in LUAD. Methods: CENPF expression level was investigated in public online database firstly, the prognosis of CENPF in LUAD were also assessed by Kaplan-Meier analysis. Then quantitative reverse transcription-polymerase chain reaction (qRT-PCR) was performed using 13 matched pairs of clinical LUAD tissue samples. Subsequently, the impact of CENPF expression on cell proliferation, cell cycle, apoptosis, colony formation was investigated by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT), flow cytometric analysis and colony formation assay, respectively. Finally, experimental xenograft lung cancer model of nude mice armpit of right forelimb to determine the effect of CENPF on LUAD tumorigenesis. Results: CENPF mRNA expression was significantly elevated in LUAD tissues compared with adjacent non-tumor lung tissues in Gene Expression Profiling Interactive Analysis (GEPIA) (P < 0.001). Up-regulated CENPF was remarkably positively associated with pathological stage, relapse free survival (RFS) as well as overall survival (OS) of LUAD patients. Besides, CENPF knockdown greatly suppressed A549 cell proliferation, induced S phase arrest, promoted apoptosis and decreased colony numbers of LUAD cells. Furthermore, knockdown of CENPF significantly inhibited the tumor growth of the LUAD cells in an experimental xenograft lung cancer model of nude mice armpit of right forelimb. Conclusion: Taken together, these results demonstrated that CENPF may serve as a potential biomarker of prognostic relevance and a potential therapeutic target for LUAD.


Asunto(s)
Adenocarcinoma del Pulmón/genética , Biomarcadores de Tumor/genética , Proteínas Cromosómicas no Histona/genética , Neoplasias Pulmonares/genética , Proteínas de Microfilamentos/genética , Recurrencia Local de Neoplasia/epidemiología , Adenocarcinoma del Pulmón/diagnóstico , Adenocarcinoma del Pulmón/mortalidad , Adenocarcinoma del Pulmón/patología , Animales , Biomarcadores de Tumor/análisis , Biomarcadores de Tumor/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Proteínas Cromosómicas no Histona/análisis , Proteínas Cromosómicas no Histona/metabolismo , Conjuntos de Datos como Asunto , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Estimación de Kaplan-Meier , Pulmón/patología , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/mortalidad , Neoplasias Pulmonares/patología , Ratones , Proteínas de Microfilamentos/análisis , Proteínas de Microfilamentos/metabolismo , Persona de Mediana Edad , Recurrencia Local de Neoplasia/genética , Pronóstico , Ensayos Antitumor por Modelo de Xenoinjerto
7.
J Biol Chem ; 294(2): 490-501, 2019 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-30420424

RESUMEN

Homologous recombination (HR) is a universally conserved DNA repair pathway that can result in the exchange of genetic material. In eukaryotes, HR has evolved into an essential step in meiosis. During meiosis many eukaryotes utilize a two-recombinase pathway. This system consists of Rad51 and the meiosis-specific recombinase Dmc1. Both recombinases have distinct activities during meiotic HR, despite being highly similar in sequence and having closely related biochemical activities, raising the question of how these two proteins can perform separate functions. A likely explanation for their differential regulation involves the meiosis-specific recombination proteins Hop2 and Mnd1, which are part of a highly conserved eukaryotic protein complex that participates in HR, albeit through poorly understood mechanisms. To better understand how Hop2-Mnd1 functions during HR, here we used DNA curtains in conjunction with single-molecule imaging to measure and quantify the binding of the Hop2-Mnd1 complex from Saccharomyces cerevisiae to recombination intermediates comprising Rad51- and Dmc1-ssDNA in real time. We found that yeast Hop2-Mnd1 bound rapidly to Dmc1-ssDNA filaments with high affinity and remained bound for ∼1.3 min before dissociating. We also observed that this binding interaction was highly specific for Dmc1 and found no evidence for an association of Hop2-Mnd1 with Rad51-ssDNA or RPA-ssDNA. Our findings provide new quantitative insights into the binding dynamics of Hop2-Mnd1 with the meiotic presynaptic complex. On the basis of these findings, we propose a model in which recombinase specificities for meiotic accessory proteins enhance separation of the recombinases' functions during meiotic HR.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Mapas de Interacción de Proteínas , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Ciclo Celular/análisis , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/análisis , Proteínas de Unión al ADN/análisis , Proteínas de Unión al ADN/metabolismo , Recombinación Homóloga , Meiosis , Unión Proteica , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/análisis
8.
Mol Cell ; 46(3): 260-73, 2012 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-22516971

RESUMEN

The Mis18 complex has been identified as a critical factor for the centromeric localization of a histone H3 variant, centromeric protein A (CENP-A), which is responsible for the specification of centromere identity in the chromosome. However, the functional role of Mis18 complex is largely unknown. Here, we generated Mis18α conditional knockout mice and found that Mis18α deficiency resulted in lethality at early embryonic stage with severe defects in chromosome segregation caused by mislocalization of CENP-A. Further, we demonstrate Mis18α's crucial role for epigenetic regulation of centromeric chromatin by reinforcing centromeric localization of DNMT3A/3B. Mis18α interacts with DNMT3A/3B, and this interaction is critical for maintaining DNA methylation and hence regulating epigenetic states of centromeric chromatin. Mis18α deficiency led to reduced DNA methylation, altered histone modifications, and uncontrolled noncoding transcripts in centromere region by decreased DNMT3A/3B enrichment. Together, our findings uncover the functional mechanism of Mis18α and its pivotal role in mammalian cell cycle.


Asunto(s)
Autoantígenos/metabolismo , Centrómero/genética , Proteínas Cromosómicas no Histona/metabolismo , Proteínas Cromosómicas no Histona/fisiología , Segregación Cromosómica/genética , Epigénesis Genética , Animales , Autoantígenos/análisis , Sitios de Unión , Centrómero/metabolismo , Proteína A Centromérica , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/análisis , Proteínas Cromosómicas no Histona/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN , ADN Metiltransferasa 3A , Células HeLa , Histonas/metabolismo , Humanos , Ratones , Ratones Noqueados , Mapeo de Interacción de Proteínas
9.
Nucleic Acids Res ; 46(9): 4440-4455, 2018 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-29522205

RESUMEN

Correct localization of the centromeric histone variant CenH3/CENP-A/Cse4 is an important part of faithful chromosome segregation. Mislocalization of CenH3 could affect chromosome segregation, DNA replication and transcription. CENP-A is often overexpressed and mislocalized in cancer genomes, but the underlying mechanisms are not understood. One major regulator of Cse4 deposition is Psh1, an E3 ubiquitin ligase that controls levels of Cse4 to prevent deposition into non-centromeric regions. We present evidence that Chromatin assembly factor-1 (CAF-1), an evolutionarily conserved histone H3/H4 chaperone with subunits shown previously to interact with CenH3 in flies and human cells, regulates Cse4 deposition in budding yeast. yCAF-1 interacts with Cse4 and can assemble Cse4 nucleosomes in vitro. Loss of yCAF-1 dramatically reduces the amount of Cse4 deposited into chromatin genome-wide when Cse4 is overexpressed. The incorporation of Cse4 genome-wide may have multifactorial effects on growth and gene expression. Loss of yCAF-1 can rescue growth defects and some changes in gene expression associated with Cse4 deposition that occur in the absence of Psh1-mediated proteolysis. Incorporation of Cse4 into promoter nucleosomes at transcriptionally active genes depends on yCAF-1. Overall our findings suggest CAF-1 can act as a CenH3 chaperone, regulating levels and incorporation of CenH3 in chromatin.


Asunto(s)
Factor 1 de Ensamblaje de la Cromatina/metabolismo , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Centrómero/química , Proteínas Cromosómicas no Histona/análisis , Proteínas de Unión al ADN/análisis , Eliminación de Gen , Expresión Génica , Nucleosomas/metabolismo , Regiones Promotoras Genéticas , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/análisis , Telómero , Ubiquitinación
10.
Mol Cell ; 43(4): 624-37, 2011 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-21855801

RESUMEN

The RNA exosome is a conserved degradation machinery, which obtains full activity only when associated with cofactors. The most prominent activator of the yeast nuclear exosome is the RNA helicase Mtr4p, acting in the context of the Trf4p/Air2p/Mtr4p polyadenylation (TRAMP) complex. The existence of a similar activator(s) in humans remains elusive. By establishing an interaction network of the human nuclear exosome, we identify the trimeric Nuclear Exosome Targeting (NEXT) complex, containing hMTR4, the Zn-knuckle protein ZCCHC8, and the putative RNA binding protein RBM7. ZCCHC8 and RBM7 are excluded from nucleoli, and consistently NEXT is specifically required for the exosomal degradation of promoter upstream transcripts (PROMPTs). We also detect putative homolog TRAMP subunits hTRF4-2 (Trf4p) and ZCCHC7 (Air2p) in hRRP6 and hMTR4 precipitates. However, at least ZCCHC7 function is restricted to nucleoli. Our results suggest that human nuclear exosome degradation pathways comprise modules of spatially organized cofactors that diverge from the yeast model.


Asunto(s)
Proteínas Portadoras/fisiología , Modelos Biológicos , Proteínas Nucleares/fisiología , ARN Helicasas/fisiología , Proteínas de Unión al ARN/fisiología , Ribonucleasas/metabolismo , Proteínas Portadoras/análisis , Proteínas Portadoras/metabolismo , Nucléolo Celular/enzimología , Nucléolo Celular/metabolismo , Proteínas Cromosómicas no Histona/análisis , Proteínas Cromosómicas no Histona/metabolismo , ADN Polimerasa Dirigida por ADN/análisis , ADN Polimerasa Dirigida por ADN/metabolismo , Exorribonucleasas/análisis , Exorribonucleasas/metabolismo , Exorribonucleasas/fisiología , Complejo Multienzimático de Ribonucleasas del Exosoma , Humanos , Proteínas Nucleares/análisis , Proteínas Nucleares/metabolismo , ARN Helicasas/análisis , ARN Helicasas/metabolismo , Proteínas de Unión al ARN/análisis , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción/análisis , Factores de Transcripción/metabolismo
11.
J Reprod Dev ; 65(1): 83-90, 2019 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-30606958

RESUMEN

Parthenogenesis is an activation process of oocytes that occur without the participation of sperm. Evidence suggests that normal development of embryos requires proper expression of several imprinted genes inherited from both the paternal and maternal genomes. Compared to gene expression, histone modifications and chromatin remodeling are not well-documented. In this research, by using immunofluorescence staining for several developmental-associated histone modifications, we investigated whether epigenetic impairments in parthenogenetic embryos act as constraints for proper development. At early stages, fertilized embryos exhibited high methylation of histone H3 at lysine 9 (Me-H3-K9) and Heterochromatin Protein 1 (HP1) present in the maternal chromatin, while paternal chromatin showed weaker HP1 signals. We found that at the two-cell stage in fertilized embryos, HP1, initially detected around the nucleolus, colocalized with chromocenters at one pole of the blastomere, while parthenotes showed a diffused distribution pattern of HP1 throughout the entire nucleoplasm. At the four-cell stage, methylation of histone H3 at arginine 26 (Me-H3-R26) increased at nascent RNA repression sites in fertilized embryos, while parthenotes recorded weaker signals throughout the nucleoplasm, suggesting differences in pluripotency of the ICM cells between the two types of embryos. Moreover, at the blastocyst stage, we observed that the acetylation level of histone H4 at lysine 12 (Ac-H4-K12) was significantly decreased in parthenogenetic ICM compared to that in its fertilized counterpart. To summarize, differences in epigenetic modifications correlating with paternal chromatin's capacity to regulate nascent RNA repression may contribute to aberrant development and lineage allocation in mouse parthenogenetic embryos.


Asunto(s)
Blastocisto/fisiología , Epigénesis Genética/fisiología , Partenogénesis/genética , Acetilación , Animales , Arginina/química , Cromatina/química , Cromatina/genética , Ensamble y Desensamble de Cromatina , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/análisis , Proteínas Cromosómicas no Histona/química , Desarrollo Embrionario/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Histonas/química , Histonas/metabolismo , Lisina/química , Masculino , Metilación , Ratones , Ratones Endogámicos ICR , Inyecciones de Esperma Intracitoplasmáticas
12.
Proc Natl Acad Sci U S A ; 113(12): E1625-34, 2016 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-26957605

RESUMEN

C/D box small nucleolar RNAs (SNORDs) are small noncoding RNAs, and their best-understood function is to target the methyltransferase fibrillarin to rRNA (for example, SNORD27 performs 2'-O-methylation of A27 in 18S rRNA). Unexpectedly, we found a subset of SNORDs, including SNORD27, in soluble nuclear extract made under native conditions, where fibrillarin was not detected, indicating that a fraction of the SNORD27 RNA likely forms a protein complex different from canonical snoRNAs found in the insoluble nuclear fraction. As part of this previously unidentified complex,SNORD27 regulates the alternative splicing of the transcription factor E2F7p re-mRNA through direct RNA-RNA interaction without methylating the RNA, likely by competing with U1 small nuclear ribonucleoprotein (snRNP). Furthermore, knockdown of SNORD27 activates previously "silent" exons in several other genes through base complementarity across the entire SNORD27 sequence, not just the antisense boxes. Thus, some SNORDs likely function in both rRNA and pre-mRNA processing, which increases the repertoire of splicing regulators and links both processes.


Asunto(s)
Empalme Alternativo , Precursores del ARN/metabolismo , Procesamiento Postranscripcional del ARN/fisiología , ARN Ribosómico/metabolismo , ARN Nucleolar Pequeño/fisiología , Emparejamiento Base , Secuencia de Bases , Ciclo Celular , División Celular , Fraccionamiento Celular/métodos , Núcleo Celular/química , Proteínas Cromosómicas no Histona/análisis , Factor de Transcripción E2F7/genética , Exones/genética , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Metilación , Datos de Secuencia Molecular , Oligonucleótidos Antisentido/genética , Biogénesis de Organelos , Ribonucleoproteína Nuclear Pequeña U1/metabolismo , Ribosomas/metabolismo , Solubilidad , Empalmosomas/metabolismo
13.
Int J Mol Sci ; 20(22)2019 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-31766266

RESUMEN

Gastric cancer (GC) is the second most widespread cause of cancer-related mortality worldwide. The discovery of novel biomarkers of oncoproteins can facilitate the development of therapeutic strategies for GC treatment. In this study, we identified novel biomarkers by integrating isobaric tags for relative and absolute quantitation (iTRAQ), a human plasma proteome database, and public Oncomine datasets to search for aberrantly expressed oncogene-associated proteins in GC tissues and plasma. One of the most significantly upregulated biomarkers, DEK, was selected and its expression validated. Our immunohistochemistry (IHC) (n = 92) and quantitative real-time polymerase chain reaction (qRT-PCR) (n = 72) analyses disclosed a marked increase in DEK expression in tumor tissue, compared with paired nontumor mucosa. Importantly, significantly higher preoperative plasma DEK levels were detected in GC patients than in healthy controls via enzyme-linked immunosorbent assay (ELISA). In clinicopathological analysis, higher expression of DEK in both tissue and plasma was significantly associated with advanced stage and poorer survival outcomes of GC patients. Data from receiver operating characteristic (ROC) curve analysis disclosed a better diagnostic accuracy of plasma DEK than carcinoembryonic antigen (CEA), carbohydrate antigen 19.9 (CA 19.9), and C-reactive protein (CRP), highlighting its potential as an effective plasma biomarker for GC. Plasma DEK is also more sensitive in tumor detection than the other three biomarkers. Knockdown of DEK resulted in inhibition of GC cell migration via a mechanism involving modulation of matrix metalloproteinase MMP-2/MMP-9 level and vice versa. Our results collectively support plasma DEK as a useful biomarker for making diagnosis and prognosis of GC patients.


Asunto(s)
Proteínas Cromosómicas no Histona/análisis , Proteínas Oncogénicas/análisis , Proteínas de Unión a Poli-ADP-Ribosa/análisis , Neoplasias Gástricas/patología , Anciano , Biomarcadores de Tumor/análisis , Línea Celular Tumoral , Movimiento Celular , Proteínas Cromosómicas no Histona/sangre , Femenino , Humanos , Masculino , Persona de Mediana Edad , Metástasis de la Neoplasia/diagnóstico , Metástasis de la Neoplasia/patología , Proteínas Oncogénicas/sangre , Proteínas de Unión a Poli-ADP-Ribosa/sangre , Pronóstico , Neoplasias Gástricas/sangre , Neoplasias Gástricas/diagnóstico , Análisis de Supervivencia
14.
Genes Dev ; 24(24): 2812-22, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21159821

RESUMEN

Genomic DNA is packed in chromatin fibers organized in higher-order structures within the interphase nucleus. One level of organization involves the formation of chromatin loops that may provide a favorable environment to processes such as DNA replication, transcription, and repair. However, little is known about the mechanistic basis of this structuration. Here we demonstrate that cohesin participates in the spatial organization of DNA replication factories in human cells. Cohesin is enriched at replication origins and interacts with prereplication complex proteins. Down-regulation of cohesin slows down S-phase progression by limiting the number of active origins and increasing the length of chromatin loops that correspond with replicon units. These results give a new dimension to the role of cohesin in the architectural organization of interphase chromatin, by showing its participation in DNA replication.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Cromatina/química , Proteínas Cromosómicas no Histona/fisiología , Empaquetamiento del ADN , Replicación del ADN , Proteínas de Ciclo Celular/análisis , Línea Celular , Proteínas Cromosómicas no Histona/análisis , Humanos , Interfase , Origen de Réplica , Fase S , Cohesinas
15.
Nature ; 477(7364): 354-8, 2011 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-21874020

RESUMEN

During cell division, chromosomes are segregated to nascent daughter cells by attaching to the microtubules of the mitotic spindle through the kinetochore. Kinetochores are assembled on a specialized chromatin domain called the centromere, which is characterized by the replacement of nucleosomal histone H3 with the histone H3 variant centromere protein A (CENP-A). CENP-A is essential for centromere and kinetochore formation in all eukaryotes but it is unknown how CENP-A chromatin directs centromere and kinetochore assembly. Here we generate synthetic CENP-A chromatin that recapitulates essential steps of centromere and kinetochore assembly in vitro. We show that reconstituted CENP-A chromatin when added to cell-free extracts is sufficient for the assembly of centromere and kinetochore proteins, microtubule binding and stabilization, and mitotic checkpoint function. Using chromatin assembled from histone H3/CENP-A chimaeras, we demonstrate that the conserved carboxy terminus of CENP-A is necessary and sufficient for centromere and kinetochore protein recruitment and function but that the CENP-A targeting domain--required for new CENP-A histone assembly--is not. These data show that two of the primary requirements for accurate chromosome segregation, the assembly of the kinetochore and the propagation of CENP-A chromatin, are specified by different elements in the CENP-A histone. Our unique cell-free system enables complete control and manipulation of the chromatin substrate and thus presents a powerful tool to study centromere and kinetochore assembly.


Asunto(s)
Centrómero/metabolismo , Cromatina/química , Cromatina/metabolismo , Cinetocoros/metabolismo , Animales , Autoantígenos/química , Autoantígenos/metabolismo , Extractos Celulares , Sistema Libre de Células , Proteína A Centromérica , Cromatina/genética , Proteínas Cromosómicas no Histona/análisis , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/metabolismo , Segregación Cromosómica , Secuencia Conservada , Histonas/metabolismo , Humanos , Microtúbulos/metabolismo , Mitosis , Oocitos , Estructura Terciaria de Proteína , Moldes Genéticos , Xenopus laevis
16.
Nature ; 471(7339): 480-5, 2011 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-21179089

RESUMEN

Chromatin is composed of DNA and a variety of modified histones and non-histone proteins, which have an impact on cell differentiation, gene regulation and other key cellular processes. Here we present a genome-wide chromatin landscape for Drosophila melanogaster based on eighteen histone modifications, summarized by nine prevalent combinatorial patterns. Integrative analysis with other data (non-histone chromatin proteins, DNase I hypersensitivity, GRO-Seq reads produced by engaged polymerase, short/long RNA products) reveals discrete characteristics of chromosomes, genes, regulatory elements and other functional domains. We find that active genes display distinct chromatin signatures that are correlated with disparate gene lengths, exon patterns, regulatory functions and genomic contexts. We also demonstrate a diversity of signatures among Polycomb targets that include a subset with paused polymerase. This systematic profiling and integrative analysis of chromatin signatures provides insights into how genomic elements are regulated, and will serve as a resource for future experimental investigations of genome structure and function.


Asunto(s)
Cromatina/genética , Cromatina/metabolismo , Drosophila melanogaster/genética , Animales , Línea Celular , Inmunoprecipitación de Cromatina , Proteínas Cromosómicas no Histona/análisis , Proteínas Cromosómicas no Histona/metabolismo , Desoxirribonucleasa I/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/embriología , Drosophila melanogaster/crecimiento & desarrollo , Exones/genética , Regulación de la Expresión Génica/genética , Genes de Insecto/genética , Genoma de los Insectos/genética , Histonas/química , Histonas/metabolismo , Masculino , Anotación de Secuencia Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Complejo Represivo Polycomb 1 , ARN/análisis , ARN/genética , Análisis de Secuencia , Transcripción Genética/genética
17.
Mol Cell ; 33(3): 287-98, 2009 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-19217403

RESUMEN

Mis16 and Mis18 are subunits of a protein complex required for incorporation of the histone H3 variant CenH3 (Cnp1/CENP-A) into centromeric chromatin in Schizosaccharomyces pombe and mammals. How the Mis16-Mis18 complex performs this function is unknown. Here, we report that the Mis16-Mis18 complex is required for centromere localization of Scm3(Sp), a Cnp1-binding protein related to Saccharomyces cerevisiae Scm3. Scm3(Sp) is required for centromeric localization of Cnp1, while Scm3(Sp) localizes at centromeres independently of Cnp1. Like the Mis16-Mis18 complex but unlike Cnp1, Scm3(Sp) dissociates from centromeres during mitosis. Inactivation of Scm3(Sp) or Mis18 increases centromere localization of histones H3 and H2A/H2B, which are largely absent from centromeres in wild-type cells. Whereas S. cerevisiae Scm3 is proposed to replace histone H2A/H2B in centromeric nucleosomes, the dynamic behavior of S. pombe Scm3 suggests that it acts as a Cnp1 assembly/maintenance factor that directly mediates the stable deposition of Cnp1 into centromeric chromatin.


Asunto(s)
Proteínas Portadoras/metabolismo , Centrómero/metabolismo , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo , Proteínas Portadoras/análisis , Proteínas Portadoras/genética , Proteínas Cromosómicas no Histona/análisis , Histonas/metabolismo , Mitosis , Schizosaccharomyces/genética , Schizosaccharomyces/metabolismo , Proteínas de Schizosaccharomyces pombe/análisis , Proteínas de Schizosaccharomyces pombe/genética
18.
Mol Cell ; 33(3): 299-311, 2009 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-19217404

RESUMEN

The mechanisms ensuring specific incorporation of CENP-A at centromeres are poorly understood. Mis16 and Mis18 are required for CENP-A localization at centromeres and form a complex that is conserved from fission yeast to human. Fission yeast sim1 mutants that alleviate kinetochore domain silencing are defective in Scm3(Sp), the ortholog of budding yeast Scm3(Sc). Scm3(Sp) depends on Mis16/18 for its centromere localization and like them is recruited to centromeres in late anaphase. Importantly, Scm3(Sp) coaffinity purifies with CENP-A(Cnp1) and associates with CENP-A(Cnp1) in vitro, yet localizes independently of intact CENP-A(Cnp1) chromatin and is differentially released from chromatin. While Scm3(Sc) has been proposed to form a unique hexameric nucleosome with CENP-A(Cse4) and histone H4 at budding yeast point centromeres, we favor a model in which Scm3(Sp) acts as a CENP-A(Cnp1) receptor/assembly factor, cooperating with Mis16 and Mis18 to receive CENP-A(Cnp1) from the Sim3 escort and mediate assembly of CENP-A(Cnp1) into subkinetochore chromatin.


Asunto(s)
Proteínas Portadoras/metabolismo , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Cinetocoros/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo , Schizosaccharomyces/metabolismo , Proteínas Portadoras/genética , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/análisis , Mutación , Schizosaccharomyces/genética , Proteínas de Schizosaccharomyces pombe/análisis , Proteínas de Schizosaccharomyces pombe/genética
19.
Nucleic Acids Res ; 43(18): 8694-712, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26338778

RESUMEN

Long range regulatory interactions among distal enhancers and target genes are important for tissue-specific gene expression. Genome-scale identification of these interactions in a cell line-specific manner, especially using the fewest possible datasets, is a significant challenge. We develop a novel computational approach, Regulatory Interaction Prediction for Promoters and Long-range Enhancers (RIPPLE), that integrates published Chromosome Conformation Capture (3C) data sets with a minimal set of regulatory genomic data sets to predict enhancer-promoter interactions in a cell line-specific manner. Our results suggest that CTCF, RAD21, a general transcription factor (TBP) and activating chromatin marks are important determinants of enhancer-promoter interactions. To predict interactions in a new cell line and to generate genome-wide interaction maps, we develop an ensemble version of RIPPLE and apply it to generate interactions in five human cell lines. Computational validation of these predictions using existing ChIA-PET and Hi-C data sets showed that RIPPLE accurately predicts interactions among enhancers and promoters. Enhancer-promoter interactions tend to be organized into subnetworks representing coordinately regulated sets of genes that are enriched for specific biological processes and cis-regulatory elements. Overall, our work provides a systematic approach to predict and interpret enhancer-promoter interactions in a genome-wide cell-type specific manner using a few experimentally tractable measurements.


Asunto(s)
Elementos de Facilitación Genéticos , Genómica/métodos , Modelos Genéticos , Regiones Promotoras Genéticas , Algoritmos , Factor de Unión a CCCTC , Proteínas de Ciclo Celular/análisis , Línea Celular , Cromatina/química , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/análisis , Código de Histonas , Humanos , Proteínas Represoras/análisis , Proteína de Unión a TATA-Box/análisis , Cohesinas
20.
Nucleic Acids Res ; 43(20): e132, 2015 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-26130708

RESUMEN

Sequencing DNA fragments associated with proteins following in vivo cross-linking with formaldehyde (known as ChIP-seq) has been used extensively to describe the distribution of proteins across genomes. It is not widely appreciated that this method merely estimates a protein's distribution and cannot reveal changes in occupancy between samples. To do this, we tagged with the same epitope orthologous proteins in Saccharomyces cerevisiae and Candida glabrata, whose sequences have diverged to a degree that most DNA fragments longer than 50 bp are unique to just one species. By mixing defined numbers of C. glabrata cells (the calibration genome) with S. cerevisiae samples (the experimental genomes) prior to chromatin fragmentation and immunoprecipitation, it is possible to derive a quantitative measure of occupancy (the occupancy ratio - OR) that enables a comparison of occupancies not only within but also between genomes. We demonstrate for the first time that this 'internal standard' calibration method satisfies the sine qua non for quantifying ChIP-seq profiles, namely linearity over a wide range. Crucially, by employing functional tagged proteins, our calibration process describes a method that distinguishes genuine association within ChIP-seq profiles from background noise. Our method is applicable to any protein, not merely highly conserved ones, and obviates the need for the time consuming, expensive, and technically demanding quantification of ChIP using qPCR, which can only be performed on individual loci. As we demonstrate for the first time in this paper, calibrated ChIP-seq represents a major step towards documenting the quantitative distributions of proteins along chromosomes in different cell states, which we term biological chromodynamics.


Asunto(s)
Inmunoprecipitación de Cromatina/métodos , Proteínas de Unión al ADN/análisis , Genoma Fúngico , Calibración , Candida glabrata/genética , Ciclo Celular , Proteínas de Ciclo Celular/análisis , Inmunoprecipitación de Cromatina/normas , Proteínas Cromosómicas no Histona/análisis , Proteínas Fúngicas/análisis , Secuenciación de Nucleótidos de Alto Rendimiento , Proteínas Mutantes/análisis , Saccharomyces cerevisiae/genética , Análisis de Secuencia de ADN , Cohesinas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA