Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Mol Cell ; 81(17): 3526-3541.e8, 2021 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-34186021

RESUMEN

BAP1 is mutated or deleted in many cancer types, including mesothelioma, uveal melanoma, and cholangiocarcinoma. It is the catalytic subunit of the PR-DUB complex, which removes PRC1-mediated H2AK119ub1, essential for maintaining transcriptional repression. However, the precise relationship between BAP1 and Polycombs remains elusive. Using embryonic stem cells, we show that BAP1 restricts H2AK119ub1 deposition to Polycomb target sites. This increases the stability of Polycomb with their targets and prevents diffuse accumulation of H2AK119ub1 and H3K27me3. Loss of BAP1 results in a broad increase in H2AK119ub1 levels that is primarily dependent on PCGF3/5-PRC1 complexes. This titrates PRC2 away from its targets and stimulates H3K27me3 accumulation across the genome, leading to a general chromatin compaction. This provides evidence for a unifying model that resolves the apparent contradiction between BAP1 catalytic activity and its role in vivo, uncovering molecular vulnerabilities that could be useful for BAP1-related pathologies.


Asunto(s)
Cromatina/metabolismo , Proteínas del Grupo Polycomb/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Animales , Línea Celular/metabolismo , Cromatina/genética , Cromatina/fisiología , Células Madre Embrionarias/metabolismo , Heterocromatina , Histonas/metabolismo , Humanos , Ratones , Células Madre Embrionarias de Ratones/metabolismo , Complejo Represivo Polycomb 1/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Proteínas del Grupo Polycomb/genética , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/fisiología , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/fisiología , Ubiquitinación
2.
Mol Cell ; 77(4): 840-856.e5, 2020 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-31883952

RESUMEN

Polycomb group proteins (PcGs) maintain transcriptional repression to preserve cellular identity in two distinct repressive complexes, PRC1 and PRC2, that modify histones by depositing H2AK119ub1 and H3K27me3, respectively. PRC1 and PRC2 exist in different variants and show a complex regulatory cross-talk. However, the contribution that H2AK119ub1 plays in mediating PcG repressive functions remains largely controversial. Using a fully catalytic inactive RING1B mutant, we demonstrated that H2AK119ub1 deposition is essential to maintain PcG-target gene repression in embryonic stem cells (ESCs). Loss of H2AK119ub1 induced a rapid displacement of PRC2 activity and a loss of H3K27me3 deposition. This preferentially affected PRC2.2 variant with respect to PRC2.1, destabilizing canonical PRC1 activity. Finally, we found that variant PRC1 forms can sense H2AK119ub1 deposition, which contributes to their stabilization specifically at sites where this modification is highly enriched. Overall, our data place H2AK119ub1 deposition as a central hub that mounts PcG repressive machineries to preserve cell transcriptional identity.


Asunto(s)
Regulación de la Expresión Génica , Histonas/metabolismo , Complejo Represivo Polycomb 1/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Transcripción Genética , Ubiquitinación , Línea Celular , Cromatina/metabolismo , Células Madre Embrionarias/metabolismo , Mutación Missense , Complejo Represivo Polycomb 1/genética , Complejo Represivo Polycomb 2/fisiología , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
3.
Mol Cell ; 74(5): 1037-1052.e7, 2019 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-31029542

RESUMEN

Polycomb repressive complexes 1 and 2 (PRC1 and PRC2) control cell identity by establishing facultative heterochromatin repressive domains at common sets of target genes. PRC1, which deposits H2Aub1 through the E3 ligases RING1A/B, forms six biochemically distinct subcomplexes depending on the assembled PCGF protein (PCGF1-PCGF6); however, it is yet unclear whether these subcomplexes have also specific activities. Here we show that PCGF1 and PCGF2 largely compensate for each other, while other PCGF proteins have high levels of specificity for distinct target genes. PCGF2 associates with transcription repression, whereas PCGF3 and PCGF6 associate with actively transcribed genes. Notably, PCGF3 and PCGF6 complexes can assemble and be recruited to several active sites independently of RING1A/B activity (therefore, of PRC1). For chromatin recruitment, the PCGF6 complex requires the combinatorial activities of its MGA-MAX and E2F6-DP1 subunits, while PCGF3 requires an interaction with the USF1 DNA binding transcription factor.


Asunto(s)
Complejo Represivo Polycomb 1/genética , Transcripción Genética , Ubiquitina-Proteína Ligasas/genética , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Cromatina/genética , Proteínas de Unión al ADN/genética , Factor de Transcripción E2F6/genética , Heterocromatina/genética , Ratones , Células Madre Embrionarias de Ratones/metabolismo , Complejo Represivo Polycomb 2/genética , Proteínas del Grupo Polycomb/genética , Proteínas Represoras/genética , Factor de Transcripción DP1/genética , Factores de Transcripción/genética , Factores Estimuladores hacia 5'/genética
4.
Trends Genet ; 38(4): 333-352, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34426021

RESUMEN

Cell identity is tightly controlled by specific transcriptional programs which require post-translational modifications of histones. These histone modifications allow the establishment and maintenance of active and repressed chromatin domains. Histone H2A lysine 119 ubiquitination (H2AK119ub1) has an essential role in building repressive chromatin domains during development. It is regulated by the counteracting activities of the Polycomb repressive complex 1 (PRC1) and the Polycomb repressive-deubiquitinase (PR-DUB) complexes, two multi-subunit ensembles that write and erase this modification, respectively. We have catalogued the recurrent genetic alterations in subunits of the PRC1 and PR-DUB complexes in both neurodevelopmental disorders and cancer. These genetic lesions are often shared across disorders, and we highlight common mechanisms of H2AK119ub1 dysregulation and how they affect development in multiple disease contexts.


Asunto(s)
Discapacidades del Desarrollo , Histonas , Neoplasias , Complejo Represivo Polycomb 1 , Proteínas del Grupo Polycomb , Ubiquitinación , Niño , Cromatina/genética , Discapacidades del Desarrollo/genética , Histonas/genética , Histonas/metabolismo , Humanos , Neoplasias/genética , Complejo Represivo Polycomb 1/genética , Complejo Represivo Polycomb 1/metabolismo , Proteínas del Grupo Polycomb/genética , Proteínas del Grupo Polycomb/metabolismo
5.
Nucleic Acids Res ; 49(2): 791-804, 2021 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-33398338

RESUMEN

The proteolytic cleavage of histone tails, also termed histone clipping, has been described as a mechanism for permanent removal of post-translational modifications (PTMs) from histone proteins. Such activity has been ascribed to ensure regulatory function in key cellular processes such as differentiation, senescence and transcriptional control, for which different histone-specific proteases have been described. However, all these studies were exclusively performed using cell lines cultured in vitro and no clear evidence that histone clipping is regulated in vivo has been reported. Here we show that histone H3 N-terminal tails undergo extensive cleavage in the differentiated cells of the villi in mouse intestinal epithelium. Combining biochemical methods, 3D organoid cultures and in vivo approaches, we demonstrate that intestinal H3 clipping is the result of multiple proteolytic activities. We identified Trypsins and Cathepsin L as specific H3 tail proteases active in small intestinal differentiated cells and showed that their proteolytic activity is differentially affected by the PTM pattern of histone H3 tails. Together, our findings provide in vivo evidence of H3 tail proteolysis in mammalian tissues, directly linking H3 clipping to cell differentiation.


Asunto(s)
Enterocitos/metabolismo , Histonas/metabolismo , Intestino Delgado/citología , Células de Paneth/metabolismo , Péptido Hidrolasas/metabolismo , Procesamiento Proteico-Postraduccional , Células Madre/metabolismo , Animales , Catepsina L/metabolismo , Diferenciación Celular , Homeostasis , Mucosa Intestinal/citología , Ratones , Microvellosidades/ultraestructura , Nucleosomas/metabolismo , Nucleosomas/ultraestructura , Organoides , Dominios Proteicos , Tripsina/metabolismo
6.
EMBO Rep ; 21(9): e50446, 2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32749065

RESUMEN

Melanoma progression is generally associated with increased transcriptional activity mediated by the Yes-associated protein (YAP). Mechanical signals from the extracellular matrix are sensed by YAP, which then activates the expression of proliferative genes, promoting melanoma progression and drug resistance. Which extracellular signals induce mechanotransduction, and how this is mediated, is not completely understood. Here, using secretome analyses, we reveal the extracellular accumulation of amyloidogenic proteins, i.e. premelanosome protein (PMEL), in metastatic melanoma, together with proteins that assist amyloid maturation into fibrils. We also confirm the accumulation of amyloid-like aggregates, similar to those detected in Alzheimer disease, in metastatic cell lines, as well as in human melanoma biopsies. Mechanistically, beta-secretase 2 (BACE2) regulates the maturation of these aggregates, which in turn induce YAP activity. We also demonstrate that recombinant PMEL fibrils are sufficient to induce mechanotransduction, triggering YAP signaling. Finally, we demonstrate that BACE inhibition affects cell proliferation and increases drug sensitivity, highlighting the importance of amyloids for melanoma survival, and the use of beta-secretase inhibitors as potential therapeutic approach for metastatic melanoma.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Melanoma , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Amiloidogénicas , Humanos , Mecanotransducción Celular , Melanoma/tratamiento farmacológico , Melanoma/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
7.
Dev Cell ; 59(3): 368-383.e7, 2024 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-38228142

RESUMEN

Cell fate is determined by specific transcription programs that are essential for tissue homeostasis and regeneration. The E3-ligases RING1A and B represent the core activity of the Polycomb repressive complex 1 (PRC1) that deposits repressive histone H2AK119 mono-ubiquitination (H2AK119ub1), which is essential for mouse intestinal homeostasis by preserving stem cell functions. However, the specific role of different PRC1 forms, which are defined by the six distinct PCGF1-6 paralogs, remains largely unexplored in vivo. We report that PCGF6 regulates mouse intestinal Tuft cell differentiation independently of H2AK119ub1 deposition. We show that PCGF6 chromatin occupancy expands outside Polycomb repressive domains, associating with unique promoter and distal regulatory elements. This occurs in the absence of RING1A/B and involves MGA-mediated E-BOX recognition and specific H3K9me2 promoter deposition. PCGF6 inactivation induces an epithelial autonomous accumulation of Tuft cells that was not phenocopied by RING1A/B loss. This involves direct PCGF6 association with a Tuft cell differentiation program that identified Polycomb-independent properties of PCGF6 in adult tissues homeostasis.


Asunto(s)
Complejo Represivo Polycomb 1 , Células en Penacho , Animales , Ratones , Diferenciación Celular/fisiología , Proteínas del Grupo Polycomb , Ubiquitina-Proteína Ligasas
8.
bioRxiv ; 2023 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-36865140

RESUMEN

The maintenance of gene expression patterns during metazoan development is achieved by the actions of Polycomb group (PcG) complexes. An essential modification marking silenced genes is monoubiquitination of histone H2A lysine 119 (H2AK119Ub) deposited by the E3 ubiquitin ligase activity of the non-canonical Polycomb Repressive Complex 1. The Polycomb Repressive Deubiquitinase (PR-DUB) complex cleaves monoubiquitin from histone H2A lysine 119 (H2AK119Ub) to restrict focal H2AK119Ub at Polycomb target sites and to protect active genes from aberrant silencing. BAP1 and ASXL1, subunits that form active PR-DUB, are among the most frequently mutated epigenetic factors in human cancers, underscoring their biological importance. How PR-DUB achieves specificity for H2AK119Ub to regulate Polycomb silencing is unknown, and the mechanisms of most of the mutations in BAP1 and ASXL1 found in cancer have not been established. Here we determine a cryo-EM structure of human BAP1 bound to the ASXL1 DEUBAD domain in complex with a H2AK119Ub nucleosome. Our structural, biochemical, and cellular data reveal the molecular interactions of BAP1 and ASXL1 with histones and DNA that are critical for remodeling the nucleosome and thus establishing specificity for H2AK119Ub. These results further provide a molecular explanation for how >50 mutations in BAP1 and ASXL1 found in cancer can dysregulate H2AK119Ub deubiquitination, providing new insight into understanding cancer etiology. One Sentence Summary: We reveal the molecular mechanism of nucleosomal H2AK119Ub deubiquitination by human BAP1/ASXL1.

9.
Sci Adv ; 9(32): eadg9832, 2023 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-37556531

RESUMEN

Histone H2A lysine 119 (H2AK119Ub) is monoubiquitinated by Polycomb repressive complex 1 and deubiquitinated by Polycomb repressive deubiquitinase complex (PR-DUB). PR-DUB cleaves H2AK119Ub to restrict focal H2AK119Ub at Polycomb target sites and to protect active genes from aberrant silencing. The PR-DUB subunits (BAP1 and ASXL1) are among the most frequently mutated epigenetic factors in human cancers. How PR-DUB establishes specificity for H2AK119Ub over other nucleosomal ubiquitination sites and how disease-associated mutations of the enzyme affect activity are unclear. Here, we determine a cryo-EM structure of human BAP1 and the ASXL1 DEUBAD in complex with a H2AK119Ub nucleosome. Our structural, biochemical, and cellular data reveal the molecular interactions of BAP1 and ASXL1 with histones and DNA that are critical for restructuring the nucleosome and thus establishing specificity for H2AK119Ub. These results further provide a molecular explanation for how >50 mutations in BAP1 and ASXL1 found in cancer can dysregulate H2AK119Ub deubiquitination, providing insight into understanding cancer etiology.


Asunto(s)
Proteínas de Drosophila , Neoplasias , Humanos , Histonas/genética , Nucleosomas , Lisina , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/metabolismo , Proteínas del Grupo Polycomb/genética , Proteínas de Drosophila/genética , Neoplasias/genética , Proteínas Represoras/genética , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
10.
Sci Rep ; 10(1): 16809, 2020 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-33033354

RESUMEN

Both onco-suppressor PREP1 and the oncogene MEIS1 bind to PBX1. This interaction stabilizes the two proteins and allows their translocation into the nucleus and thus their transcriptional activity. Here, we have combined cross-linking mass-spectrometry and systematic mutagenesis to detail the binding geometry of the PBX1-PREP1 (and PBX1-MEIS1) complexes, under native in vivo conditions. The data confirm the existence of two distinct interaction sites within the PBC domain of PBX1 and unravel differences among the highly similar binding sites of MEIS1 and PREP1. The HR2 domain has a fundamental role in binding the PBC-B domain of PBX1 in both PREP1 and MEIS1. The HR1 domain of MEIS1, however, seem to play a less stringent role in PBX1 interaction with respect to that of PREP1. This difference is also reflected by the different binding affinity of the two proteins to PBX1. Although partial, this analysis provides for the first time some ideas on the tertiary structure of the complexes not available before. Moreover, the extensive mutagenic analysis of PREP1 identifies the role of individual hydrophobic HR1 and HR2 residues, both in vitro and in vivo.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Factor de Transcripción 1 de la Leucemia de Células Pre-B/metabolismo , Mapeo de Interacción de Proteínas , Células A549 , Sitios de Unión , Clonación Molecular , Ensayo de Inmunoadsorción Enzimática , Humanos , Espectrometría de Masas , Mutagénesis , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/metabolismo , Mapeo de Interacción de Proteínas/métodos
11.
Biochim Biophys Acta Gen Subj ; 1863(2): 456-465, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30465816

RESUMEN

BACKGROUND: Human Sp140 protein is a leukocyte-specific member of the speckled protein (Sp) family (Sp100, Sp110, Sp140, Sp140L), a class of multi-domain nuclear proteins involved in intrinsic immunity and transcriptional regulation. Sp140 regulates macrophage transcriptional program and is implicated in several haematologic malignancies. Little is known about Sp140 structural domains and its post-translational modifications. METHODS: We used mass spectrometry and biochemical experiments to investigate endogenous Sp140 SUMOylation in Burkitt's Lymphoma cells and Sp140 SUMOylation sites in HEK293T cells, FLAG-Sp140 transfected and His6-SUMO-1T95K infected. NMR spectroscopy and in vitro SUMOylation reactions were applied to investigate the role of Sp140 PHD finger in the SUMOylation of the adjacent BRD. RESULTS: Endogenous Sp140 is a SUMO-1 target, whereby FLAG-Sp140 harbors at least 13 SUMOylation sites distributed along the protein sequence, including the BRD. NMR experiments prove direct binding of the SUMO E2 ligase Ubc9 and SUMO-1 to PHD-BRDSp140. In vitro SUMOylation reactions show that the PHDSp140 behaves as SUMO E3 ligase, assisting intramolecular SUMOylation of the adjacent BRD. CONCLUSIONS: Sp140 is multi-SUMOylated and its PHD finger works as versatile protein-protein interaction platform promoting intramolecular SUMOylation of the adjacent BRD. Thus, combinatorial association of Sp140 chromatin binding domains generates a multifaceted interaction scaffold, whose function goes beyond the canonical histone recognition. GENERAL SIGNIFICANCE: The addition of Sp140 to the increasing lists of multi-SUMOylated proteins opens new perspectives for molecular studies on Sp140 transcriptional activity, where SUMOylation could represent a regulatory route and a docking surface for the recruitment and assembly of leukocyte-specific transcription regulators.


Asunto(s)
Antígenos Nucleares/química , Antígenos Nucleares/metabolismo , Dedos de Zinc PHD , Dominios Proteicos , Sumoilación , Factores de Transcripción/química , Factores de Transcripción/metabolismo , Células HEK293 , Humanos , Unión Proteica
13.
FEBS J ; 281(1): 216-31, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24267382

RESUMEN

Sp140 is a nuclear leukocyte-specific protein involved in primary biliary cirrhosis and a risk factor in chronic lymphocytic leukemia. The presence of several chromatin related modules such as plant homeodomain (PHD), bromodomain and SAND domain suggests a role in chromatin-mediated regulation of gene expression; however, its real function is still elusive. Herein we present the solution structure of Sp140-PHD finger and investigate its role as epigenetic reader in vitro. Sp140-PHD presents an atypical PHD finger fold which does not bind to histone H3 tails but is recognized by peptidylprolyl isomerase Pin1. Pin1 specifically binds to a phosphopeptide corresponding to the L3 loop of Sp140-PHD and catalyzes cis-trans isomerization of a pThr-Pro bond. Moreover co-immunoprecipitation experiments demonstrate FLAG-Sp140 interaction with endogenous Pin1 in vivo. Overall these data include Sp140 in the list of the increasing number of Pin1 binders and expand the regulatory potential of PHD fingers as versatile structural platforms for diversified interactions.


Asunto(s)
Antígenos Nucleares/química , Antígenos Nucleares/metabolismo , Histonas/metabolismo , Isomerasa de Peptidilprolil/metabolismo , Pliegue de Proteína , Factores de Transcripción/química , Factores de Transcripción/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Western Blotting , Cromatina/genética , Células HEK293 , Humanos , Inmunoprecipitación , Espectroscopía de Resonancia Magnética , Datos de Secuencia Molecular , Peptidilprolil Isomerasa de Interacción con NIMA , Fosforilación , Unión Proteica , Conformación Proteica , Homología de Secuencia de Aminoácido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA