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.
PLoS One ; 16(9): e0257688, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34591877

RESUMEN

BRCA1-associated protein 1 (BAP1) is a tumor suppressor and its loss can result in mesothelioma, uveal and cutaneous melanoma, clear cell renal cell carcinoma and bladder cancer. BAP1 is a deubiquitinating enzyme of the UCH class that has been implicated in various cellular processes like cell growth, cell cycle progression, ferroptosis, DNA damage response and ER metabolic stress response. ASXL proteins activate BAP1 by forming the polycomb repressive deubiquitinase (PR-DUB) complex which acts on H2AK119ub1. Besides the ASXL proteins, BAP1 is known to interact with an established set of additional proteins. Here, we identify novel BAP1 interacting proteins in the cytoplasm by expressing GFP-tagged BAP1 in an endogenous BAP1 deficient cell line using affinity purification followed by mass spectrometry (AP-MS) analysis. Among these novel interacting proteins are Histone acetyltransferase 1 (HAT1) and all subunits of the heptameric coat protein complex I (COPI) that is involved in vesicle formation and protein cargo binding and sorting. We validate that the HAT1 and COPI interactions occur at endogenous levels but find that this interaction with COPI is not mediated through the C-terminal KxKxx cargo sorting signals of the COPI complex.


Asunto(s)
Proteína Coat de Complejo I/metabolismo , Citoplasma/metabolismo , Histona Acetiltransferasas/metabolismo , Neoplasias/metabolismo , Proteómica/métodos , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/metabolismo , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Técnicas de Inactivación de Genes , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Espectrometría de Masas , Mutación , Proteínas Recombinantes/metabolismo
2.
iScience ; 24(5): 102435, 2021 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-34113809

RESUMEN

In an attempt to unravel functionality of the non-canonical PRC1.1 Polycomb complex in human leukemogenesis, we show that USP7 and TRIM27 are integral components of PRC1.1. USP7 interactome analyses show that PRC1.1 is the predominant Polycomb complex co-precipitating with USP7. USP7 inhibition results in PRC1.1 disassembly and loss of chromatin binding, coinciding with reduced H2AK119ub and H3K27ac levels and diminished gene transcription of active PRC1.1-controlled loci, whereas H2AK119ub marks are also lost at PRC1 loci. TRIM27 and USP7 are reciprocally required for incorporation into PRC1.1, and TRIM27 knockdown partially rescues USP7 inhibitor sensitivity. USP7 inhibitors effectively impair proliferation in AML cells in vitro, also independent of the USP7-MDM2-TP53 axis, and MLL-AF9-induced leukemia is delayed in vivo in human leukemia xenografts. We propose a model where USP7 counteracts TRIM27 E3 ligase activity, thereby maintaining PRC1.1 integrity and function. Moreover, USP7 inhibition may be a promising new strategy to treat AML patients.

3.
J Cell Biochem ; 119(12): 9781-9789, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30171711

RESUMEN

Pluripotent cells appear to be in a transient state during early development. These cells have the capability to transition into embryonic stem cells (ESCs). It has been reported that mouse pluripotent cells cultivated in chemically defined media sustain the ground state of pluripotency. Because the epigenetic pattern of pluripotent cells reflects their environment, culture under different conditions causes epigenetic changes, which could lead to genomic instability. This study focused on the DNA methylation pattern of repetitive elements (REs) and their activation levels under two ground-state conditions and assessed the genomic integrity of ESCs. We measured the methylation and expression level of REs in different media. The results indicated that although the ground-state conditions show higher REs activity, they did not lead to DNA damage; therefore, the level of genomic instability is lower under the ground-state compared with the conventional condition. Our results indicated that when choosing an optimum condition, different features of the condition must be considered to have epigenetically and genomically stable stem cells.


Asunto(s)
Metilación de ADN , Células Madre Pluripotentes/fisiología , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular/genética , Islas de CpG , Daño del ADN/genética , Genoma , Inestabilidad Genómica , Ratones , Ratones Endogámicos C57BL , Células Madre Embrionarias de Ratones , Células Madre Pluripotentes/citología , Secuencias Repetitivas de Ácidos Nucleicos , Análisis de la Célula Individual
4.
Nat Commun ; 9(1): 229, 2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29335415

RESUMEN

BRCA1-BARD1-catalyzed ubiquitination of histone H2A is an important regulator of the DNA damage response, priming chromatin for repair by homologous recombination. However, no specific deubiquitinating enzymes (DUBs) are known to antagonize this function. Here we identify ubiquitin specific protease-48 (USP48) as a H2A DUB, specific for the C-terminal BRCA1 ubiquitination site. Detailed biochemical analysis shows that an auxiliary ubiquitin, an additional ubiquitin that itself does not get cleaved, modulates USP48 activity, which has possible implications for its regulation in vivo. In cells we reveal that USP48 antagonizes BRCA1 E3 ligase function and in BRCA1-proficient cells loss of USP48 results in positioning 53BP1 further from the break site and in extended resection lengths. USP48 repression confers a survival benefit to cells treated with camptothecin and its activity acts to restrain gene conversion and mutagenic single-strand annealing. We propose that USP48 promotes genome stability by antagonizing BRCA1 E3 ligase function.


Asunto(s)
Proteína BRCA1/metabolismo , Histonas/metabolismo , Proteasas Ubiquitina-Específicas/metabolismo , Ubiquitina/metabolismo , Animales , Proteína BRCA1/genética , Secuencia de Bases , Línea Celular Tumoral , Células Cultivadas , Reparación del ADN , Células HeLa , Humanos , Cinética , Ratones Noqueados , Interferencia de ARN , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Proteasas Ubiquitina-Específicas/genética , Ubiquitinación
5.
Transcription ; 9(2): 67-74, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28976802

RESUMEN

Sma and Mad related (SMAD)-mediated Transforming Growth Factor ß (TGF-ß) and Bone Morphogenetic Protein (BMP) signaling is required for various cellular processes. The activated heterotrimeric SMAD protein complexes associate with nuclear proteins such as the histone acetyltransferases p300, PCAF and the Mixed Lineage Leukemia 4 (MLL4) subunit Pax Transactivation domain-Interacting Protein (PTIP) to regulate gene transcription. We investigated the functional role of PTIP and PTIP Interacting protein 1 (PA1) in relation to TGF-ß-activated SMAD signaling. We immunoprecipitated PTIP and PA1 with all SMAD family members to identify the TGF-ß and not BMP-specific SMADs as interacting proteins. Gene silencing experiments of MLL4 and the subunits PA1 and PTIP confirm TGF-ß-specific genes to be regulated by the MLL4 complex, which links TGF-ß signaling to transcription regulation by the MLL4 methyltransferase complex.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Activación Transcripcional , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular , Proteínas de Unión al ADN/genética , Silenciador del Gen , Células HEK293 , N-Metiltransferasa de Histona-Lisina , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Mapas de Interacción de Proteínas , Transducción de Señal , Proteínas Smad/metabolismo , Factor de Crecimiento Transformador beta/genética
6.
J Biol Chem ; 291(46): 24121-24132, 2016 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-27703004

RESUMEN

Gene-specific transcription factors (GSTFs) control gene transcription by DNA binding and specific protein complex recruitment, which regulates promoter accessibility for transcription initiation by RNA polymerase II. Mutations in the GSTFs Suppressor of Mothers Against Decapentaplegic 2 (SMAD2) and SMAD4 are frequently associated with colon and rectal carcinomas. These proteins play an important role in bone morphogenic protein (BMP) and transforming growth factor ß (TGF-ß) signaling pathways controlling cell fate and proliferation. To study the protein interactome of the SMAD protein family we generated a quantitative proteomics pipeline that allows for inducible expression of GFP-tagged SMAD proteins followed by affinity purification and quantitative mass spectrometry analysis. Data are available via ProteomeXchange with identifier PXD004529. The nuclear importin IPO5 was identified as a novel interacting protein of SMAD1. Overexpression of IPO5 in various cell lines specifically increases nuclear localization of BMP receptor-activated SMADs (R-SMADs) confirming a functional relationship between IPO5 and BMP but not TGF-ß R-SMADs. Finally, we provide evidence that variation in length of the lysine stretch of the nuclear localization sequence is a determinant for importin specificity.


Asunto(s)
Núcleo Celular/metabolismo , Proteína Smad1/metabolismo , Proteína Smad2/metabolismo , Proteína Smad4/metabolismo , beta Carioferinas/metabolismo , Receptores de Proteínas Morfogenéticas Óseas/genética , Receptores de Proteínas Morfogenéticas Óseas/metabolismo , Núcleo Celular/genética , Células HeLa , Humanos , Proteómica , Proteína Smad1/genética , Proteína Smad2/genética , Proteína Smad4/genética , beta Carioferinas/genética
7.
J Biomol Screen ; 19(2): 287-96, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24334265

RESUMEN

Posttranslational modifications of histones play an important role in the regulation of gene expression and chromatin structure in eukaryotes. The balance between chromatin factors depositing (writers) and removing (erasers) histone marks regulates the steady-state levels of chromatin modifications. Here we describe a novel microscopy-based screening method to identify proteins that regulate histone modification levels in a high-throughput fashion. We named our method CROSS, for Chromatin Regulation Ontology SiRNA Screening. CROSS is based on an siRNA library targeting the expression of 529 proteins involved in chromatin regulation. As a proof of principle, we used CROSS to identify chromatin factors involved in histone H3 methylation on either lysine-4 or lysine-27. Furthermore, we show that CROSS can be used to identify chromatin factors that affect growth in cancer cell lines. Taken together, CROSS is a powerful method to identify the writers and erasers of novel and known chromatin marks and facilitates the identification of drugs targeting epigenetic modifications.


Asunto(s)
Ensayos Analíticos de Alto Rendimiento , Histonas/genética , Microscopía , Proteínas/aislamiento & purificación , Línea Celular , Cromatina/genética , Epigénesis Genética , Histonas/metabolismo , Humanos , Lisina/genética , Metilación , Unión Proteica , Procesamiento Proteico-Postraduccional , Proteínas/genética
8.
PLoS One ; 8(7): e70035, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23894581

RESUMEN

The SGF29 protein binds to tri-methylated lysine-4 of histone H3 (H3K4me3), which is a histone modification associated with active promoters. Human SGF29 is a subunit of the histone acetyltransferase module of the SAGA (Spt-Ada-Gcn5 acetyltransferase) and ATAC (Ada-Two-A-containing 2A) co-activator complexes. Previous work revealed that the SAGA complex is recruited to endoplasmic reticulum (ER) stress target genes and required for their induction. Here, we report the involvement of SGF29 in the survival of human cells from ER stress. SGF29 knockdown results in impaired transcription of the ER stress genes GRP78 and CHOP. Besides histone H3K14 acetylation, we find that SGF29 is also required for the maintenance of H3K4me3 at these genes, which is already present prior to ER stress. Reduced levels of H3K4me3 in the absence of SGF29 correlate with a decreased association of ASH2L, which is a core component of the SET1/MLL complexes, to GFP78 and CHOP. In conclusion, our results suggest that the H3K4me3-binding protein SGF29 plays a central and dual role in the ER stress response. Prior to ER stress, the protein coordinates H3K4me3 levels, thereby maintaining a 'poised' chromatin state on ER stress target gene promoters. Following ER stress induction, SGF29 is required for increased H3K14 acetylation on these genes, which then results in full transcriptional activation, thereby promoting cell survival.


Asunto(s)
Acetiltransferasas/metabolismo , Estrés del Retículo Endoplásmico/fisiología , Histonas/metabolismo , Acetilación , Acetiltransferasas/biosíntesis , Acetiltransferasas/genética , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Línea Celular Tumoral , Supervivencia Celular/fisiología , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico/genética , Técnicas de Silenciamiento del Gen , Humanos , Lisina/metabolismo , Metilación , Osteosarcoma/genética , Osteosarcoma/metabolismo , Osteosarcoma/patología , Regiones Promotoras Genéticas , Factor de Transcripción CHOP/biosíntesis , Factor de Transcripción CHOP/genética , Factor de Transcripción CHOP/metabolismo , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA