Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 48(3): 514-529.e6, 2018 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-29548672

RESUMEN

Microglia as tissue macrophages contribute to the defense and maintenance of central nervous system (CNS) homeostasis. Little is known about the epigenetic signals controlling microglia function in vivo. We employed constitutive and inducible mutagenesis in microglia to delete two class I histone deacetylases, Hdac1 and Hdac2. Prenatal ablation of Hdac1 and Hdac2 impaired microglial development. Mechanistically, the promoters of pro-apoptotic and cell cycle genes were hyperacetylated in absence of Hdac1 and Hdac2, leading to increased apoptosis and reduced survival. In contrast, Hdac1 and Hdac2 were not required for adult microglia survival during homeostasis. In a mouse model of Alzheimer's disease, deletion of Hdac1 and Hdac2 in microglia, but not in neuroectodermal cells, resulted in a decrease in amyloid load and improved cognitive impairment by enhancing microglial amyloid phagocytosis. Collectively, we report a role for epigenetic factors that differentially affect microglia development, homeostasis, and disease that could potentially be utilized therapeutically.


Asunto(s)
Histona Desacetilasa 1/genética , Histona Desacetilasa 2/genética , Homeostasis , Microglía/inmunología , Microglía/metabolismo , Enfermedades Neurodegenerativas/genética , Neurogénesis/genética , Animales , Apoptosis , Proliferación Celular , Modelos Animales de Enfermedad , Epigénesis Genética , Expresión Génica , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/metabolismo , Histonas/metabolismo , Trastornos de la Memoria/genética , Trastornos de la Memoria/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Enfermedades Neurodegenerativas/psicología , Neurogénesis/inmunología , Fagocitosis/inmunología , Placa Amiloide/genética , Placa Amiloide/metabolismo , Placa Amiloide/patología , Aprendizaje Espacial , Transcriptoma
2.
Nat Immunol ; 15(5): 439-448, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24681565

RESUMEN

Molecular mechanisms that maintain lineage integrity of helper T cells are largely unknown. Here we show histone deacetylases 1 and 2 (HDAC1 and HDAC2) as crucial regulators of this process. Loss of HDAC1 and HDAC2 during late T cell development led to the appearance of major histocompatibility complex (MHC) class II-selected CD4(+) helper T cells that expressed CD8-lineage genes such as Cd8a and Cd8b1. HDAC1 and HDAC2-deficient T helper type 0 (TH0) and TH1 cells further upregulated CD8-lineage genes and acquired a CD8(+) effector T cell program in a manner dependent on Runx-CBFß complexes, whereas TH2 cells repressed features of the CD8(+) lineage independently of HDAC1 and HDAC2. These results demonstrate that HDAC1 and HDAC2 maintain integrity of the CD4 lineage by repressing Runx-CBFß complexes that otherwise induce a CD8(+) effector T cell-like program in CD4(+) T cells.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/metabolismo , Células TH1/inmunología , Animales , Diferenciación Celular/genética , Linaje de la Célula/genética , Células Cultivadas , Subunidades alfa del Factor de Unión al Sitio Principal/metabolismo , Subunidad beta del Factor de Unión al Sitio Principal/metabolismo , Citocinas/metabolismo , Citotoxicidad Inmunológica/genética , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/metabolismo , Histona Desacetilasa 1/genética , Histona Desacetilasa 2/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Unión Proteica
3.
J Biol Chem ; 300(2): 105638, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38199570

RESUMEN

The inflammasome is a large multiprotein complex that assembles in the cell cytoplasm in response to stress or pathogenic infection. Its primary function is to defend the cell and promote the secretion of pro-inflammatory cytokines, including IL-1ß and IL-18. Previous research has shown that in immortalized bone marrow-derived macrophages (iBMDMs) inflammasome assembly is dependent on the deacetylase HDAC6 and the aggresome processing pathway (APP), a cellular pathway involved in the disposal of misfolded proteins. Here we used primary BMDMs from mice in which HDAC6 is ablated or impaired and found that inflammasome activation was largely normal. We also used human peripheral blood mononuclear cells and monocyte cell lines expressing a synthetic protein blocking the HDAC6-ubiquitin interaction and impairing the APP and found that inflammasome activation was moderately affected. Finally, we used a novel HDAC6 degrader and showed that inflammasome activation was partially impaired in human macrophage cell lines with depleted HDAC6. Our results therefore show that HDAC6 importance in inflammasome activation is context-dependent.


Asunto(s)
Inflamasomas , Leucocitos Mononucleares , Animales , Humanos , Ratones , Línea Celular , Histona Desacetilasa 6/genética , Histona Desacetilasa 6/metabolismo , Inflamasomas/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Leucocitos Mononucleares/metabolismo , Macrófagos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Transporte de Proteínas/fisiología
4.
Blood ; 137(21): 2920-2934, 2021 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-33512466

RESUMEN

OBF1 is a specific coactivator of the POU family transcription factors OCT1 and OCT2. OBF1 and OCT2 are B cell-specific and indispensable for germinal center (GC) formation, but their mechanism of action is unclear. Here, we show by chromatin immunoprecipitation-sequencing that OBF1 extensively colocalizes with OCT1 and OCT2. We found that these factors also often colocalize with transcription factors of the ETS family. Furthermore, we showed that OBF1, OCT2, and OCT1 bind widely to the promoters or enhancers of genes involved in GC formation in mouse and human GC B cells. Short hairpin RNA knockdown experiments demonstrated that OCT1, OCT2, and OBF1 regulate each other and are essential for proliferation of GC-derived lymphoma cell lines. OBF1 downregulation disrupts the GC transcriptional program: genes involved in GC maintenance, such as BCL6, are downregulated, whereas genes related to exit from the GC program, such as IRF4, are upregulated. Ectopic expression of BCL6 does not restore the proliferation of GC-derived lymphoma cells depleted of OBF1 unless IRF4 is also depleted, indicating that OBF1 controls an essential regulatory node in GC differentiation.


Asunto(s)
Centro Germinal/metabolismo , Factor 1 de Transcripción de Unión a Octámeros/fisiología , Factor 2 de Transcripción de Unión a Octámeros/uso terapéutico , Transactivadores/uso terapéutico , Transcripción Genética/genética , Animales , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Ontología de Genes , Células HEK293 , Humanos , Lipopolisacáridos/farmacología , Linfoma no Hodgkin/genética , Linfoma no Hodgkin/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factor 1 de Transcripción de Unión a Octámeros/deficiencia , Factor 1 de Transcripción de Unión a Octámeros/genética , Factor 2 de Transcripción de Unión a Octámeros/deficiencia , Factor 2 de Transcripción de Unión a Octámeros/genética , Proteína Proto-Oncogénica c-ets-1/análisis , Interferencia de ARN , ARN Interferente Pequeño/genética , Proteínas Recombinantes/metabolismo , Transactivadores/deficiencia , Transactivadores/genética
5.
Mol Cell ; 55(1): 31-46, 2014 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-24882211

RESUMEN

MutS protein homolog 2 (MSH2) is a key DNA mismatch repair protein. It forms the MSH2-MSH6 (MutSα) and MSH2-MSH3 (MutSß) heterodimers, which help to ensure genomic integrity. MutSα not only recognizes and repairs mismatched nucleotides but also recognizes DNA adducts induced by DNA-damaging agents, and triggers cell-cycle arrest and apoptosis. Loss or depletion of MutSα from cells leads to microsatellite instability (MSI) and resistance to DNA damage. Although the level of MutSα can be reduced by the ubiquitin-proteasome pathway, the detailed mechanisms of this regulation remain elusive. Here we report that histone deacetylase 6 (HDAC6) sequentially deacetylates and ubiquitinates MSH2, leading to MSH2 degradation. In addition, HDAC6 significantly reduces cellular sensitivity to DNA-damaging agents and decreases cellular DNA mismatch repair activities by downregulation of MSH2. Overall, these findings reveal a mechanism by which proper levels of MutSα are maintained.


Asunto(s)
Histona Desacetilasas/fisiología , Proteína 2 Homóloga a MutS/metabolismo , Acetilación , Animales , Células Cultivadas , Células HEK293 , Células HeLa , Histona Desacetilasa 6 , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Humanos , Ratones , Estabilidad Proteica , Ubiquitinación
6.
J Biol Chem ; 295(9): 2614-2628, 2020 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-31953325

RESUMEN

Histone deacetylase 6 (HDAC6) is a multidomain cytosolic enzyme having tubulin deacetylase activity that has been unequivocally assigned to the second of the tandem catalytic domains. However, virtually no information exists on the contribution of other HDAC6 domains on tubulin recognition. Here, using recombinant protein expression, site-directed mutagenesis, fluorimetric and biochemical assays, microscale thermophoresis, and total internal reflection fluorescence microscopy, we identified the N-terminal, disordered region of HDAC6 as a microtubule-binding domain and functionally characterized it to the single-molecule level. We show that the microtubule-binding motif spans two positively charged patches comprising residues Lys-32 to Lys-58. We found that HDAC6-microtubule interactions are entirely independent of the catalytic domains and are mediated by ionic interactions with the negatively charged microtubule surface. Importantly, a crosstalk between the microtubule-binding domain and the deacetylase domain was critical for recognition and efficient deacetylation of free tubulin dimers both in vitro and in vivo Overall, our results reveal that recognition of substrates by HDAC6 is more complex than previously appreciated and that domains outside the tandem catalytic core are essential for proficient substrate deacetylation.


Asunto(s)
Histona Desacetilasa 6/metabolismo , Microtúbulos/metabolismo , Tubulina (Proteína)/metabolismo , Acetilación , Secuencia de Aminoácidos , Dominio Catalítico , Humanos , Unión Proteica , Dominios Proteicos/fisiología , Especificidad por Sustrato
7.
Nat Chem Biol ; 15(1): 51-61, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30531905

RESUMEN

Liquid-liquid phase separation (LLPS) of proteins containing intrinsically disordered regions (IDRs) has been proposed as a mechanism underlying the formation of membrane-less organelles. Tight regulation of IDR behavior is essential to ensure that LLPS only takes place when necessary. Here, we report that IDR acetylation/deacetylation regulates LLPS and assembly of stress granules (SGs), membrane-less organelles forming in response to stress. Acetylome analysis revealed that the RNA helicase DDX3X, an important component of SGs, is a novel substrate of the deacetylase HDAC6. The N-terminal IDR of DDX3X (IDR1) can undergo LLPS in vitro, and its acetylation at multiple lysine residues impairs the formation of liquid droplets. We also demonstrated that enhanced LLPS propensity through deacetylation of DDX3X-IDR1 by HDAC6 is necessary for SG maturation, but not initiation. Our analysis provides a mechanistic framework to understand how acetylation and deacetylation of IDRs regulate LLPS spatiotemporally, and impact membrane-less organelle formation in vivo.


Asunto(s)
Proteínas Intrínsecamente Desordenadas/metabolismo , ARN Helicasas/química , ARN Helicasas/metabolismo , Acetilación , Animales , Dominio Catalítico , Gránulos Citoplasmáticos/metabolismo , ARN Helicasas DEAD-box/metabolismo , Técnicas de Inactivación de Genes , Histona Desacetilasa 6/metabolismo , Humanos , Proteínas Intrínsecamente Desordenadas/química , Lisina/metabolismo , Ratones , Modelos Teóricos , Presión Osmótica , ARN Helicasas/genética
8.
J Autoimmun ; 108: 102379, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31883829

RESUMEN

Rheumatoid Arthritis (RA) represents a chronic T cell-mediated inflammatory autoimmune disease. Studies have shown that epigenetic mechanisms contribute to the pathogenesis of RA. Histone deacetylases (HDACs) represent one important group of epigenetic regulators. However, the role of individual HDAC members for the pathogenesis of arthritis is still unknown. In this study we demonstrate that mice with a T cell-specific deletion of HDAC1 (HDAC1-cKO) are resistant to the development of Collagen-induced arthritis (CIA), whereas the antibody response to collagen type II was undisturbed, indicating an unaltered T cell-mediated B cell activation. The inflammatory cytokines IL-17 and IL-6 were significantly decreased in sera of HDAC1-cKO mice. IL-6 treated HDAC1-deficient CD4+ T cells showed an impaired upregulation of CCR6. Selective inhibition of class I HDACs with the HDAC inhibitor MS-275 under Th17-skewing conditions inhibited the upregulation of chemokine receptor 6 (CCR6) in mouse and human CD4+ T cells. Accordingly, analysis of human RNA-sequencing (RNA-seq) data and histological analysis of synovial tissue samples from human RA patients revealed the existence of CD4+CCR6+ cells with enhanced HDAC1 expression. Our data indicate a key role for HDAC1 for the pathogenesis of CIA and suggest that HDAC1 and other class I HDACs might be promising targets of selective HDAC inhibitors (HDACi) for the treatment of RA.


Asunto(s)
Artritis Reumatoide/etiología , Artritis Reumatoide/metabolismo , Susceptibilidad a Enfermedades , Histona Desacetilasa 1/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Artritis Reumatoide/patología , Biomarcadores , Colágeno/efectos adversos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Histona Desacetilasa 1/genética , Humanos , Mediadores de Inflamación/metabolismo , Ratones , Ratones Noqueados , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
9.
J Autoimmun ; 86: 51-61, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28964722

RESUMEN

Multiple sclerosis (MS) is a human neurodegenerative disease characterized by the invasion of autoreactive T cells from the periphery into the CNS. Application of pan-histone deacetylase inhibitors (HDACi) ameliorates experimental autoimmune encephalomyelitis (EAE), an animal model for MS, suggesting that HDACi might be a potential therapeutic strategy for MS. However, the function of individual HDAC members in the pathogenesis of EAE is not known. In this study we report that mice with a T cell-specific deletion of HDAC1 (using the Cd4-Cre deleter strain; HDAC1-cKO) were completely resistant to EAE despite the ability of HDAC1cKO CD4+ T cells to differentiate into Th17 cells. RNA sequencing revealed STAT1 as a prominent upstream regulator of differentially expressed genes in activated HDAC1-cKO CD4+ T cells and this was accompanied by a strong increase in phosphorylated STAT1 (pSTAT1). This suggests that HDAC1 controls STAT1 activity in activated CD4+ T cells. Increased pSTAT1 levels correlated with a reduced expression of the chemokine receptors Ccr4 and Ccr6, which are important for the migration of T cells into the CNS. Finally, EAE susceptibility was restored in WT:HDAC1-cKO mixed BM chimeric mice, indicating a cell-autonomous defect. Our data demonstrate a novel pathophysiological role for HDAC1 in EAE and provide evidence that selective inhibition of HDAC1 might be a promising strategy for the treatment of MS.


Asunto(s)
Encefalomielitis Autoinmune Experimental/metabolismo , Histona Desacetilasa 1/metabolismo , Esclerosis Múltiple/metabolismo , Factor de Transcripción STAT1/metabolismo , Células Th17/fisiología , Animales , Movimiento Celular , Células Cultivadas , Quimera , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/inmunología , Histona Desacetilasa 1/genética , Humanos , Ratones , Ratones Noqueados , Esclerosis Múltiple/inmunología , Receptores CCR4/metabolismo , Receptores CCR6/metabolismo , Factor de Transcripción STAT1/genética
10.
Nat Chem Biol ; 12(9): 748-54, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27454931

RESUMEN

We report crystal structures of zebrafish histone deacetylase 6 (HDAC6) catalytic domains in tandem or as single domains in complex with the (R) and (S) enantiomers of trichostatin A (TSA) or with the HDAC6-specific inhibitor nexturastat A. The tandem domains formed, together with the inter-domain linker, an ellipsoid-shaped complex with pseudo-twofold symmetry. We identified important active site differences between both catalytic domains and revealed the binding mode of HDAC6 selective inhibitors. HDAC inhibition assays with (R)- and (S)-TSA showed that (R)-TSA was a broad-range inhibitor, whereas (S)-TSA had moderate selectivity for HDAC6. We identified a uniquely positioned α-helix and a flexible tryptophan residue in the loop joining α-helices H20 to H21 as critical for deacetylation of the physiologic substrate tubulin. Using single-molecule measurements and biochemical assays we demonstrated that HDAC6 catalytic domain 2 deacetylated α-tubulin lysine 40 in the lumen of microtubules, but that its preferred substrate was unpolymerized tubulin.


Asunto(s)
Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Ácidos Hidroxámicos/farmacología , Tubulina (Proteína)/metabolismo , Proteínas de Pez Cebra/antagonistas & inhibidores , Proteínas de Pez Cebra/metabolismo , Acetilación/efectos de los fármacos , Animales , Biocatálisis , Histona Desacetilasa 6 , Inhibidores de Histona Desacetilasas/química , Histona Desacetilasas/química , Humanos , Ácidos Hidroxámicos/química , Modelos Moleculares , Relación Estructura-Actividad , Tubulina (Proteína)/química , Pez Cebra , Proteínas de Pez Cebra/química
11.
PLoS Biol ; 13(9): e1002258, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26406915

RESUMEN

The pathogenesis of peripheral neuropathies in adults is linked to maintenance mechanisms that are not well understood. Here, we elucidate a novel critical maintenance mechanism for Schwann cell (SC)-axon interaction. Using mouse genetics, ablation of the transcriptional regulators histone deacetylases 1 and 2 (HDAC1/2) in adult SCs severely affected paranodal and nodal integrity and led to demyelination/remyelination. Expression levels of the HDAC1/2 target gene myelin protein zero (P0) were reduced by half, accompanied by altered localization and stability of neurofascin (NFasc)155, NFasc186, and loss of Caspr and septate-like junctions. We identify P0 as a novel binding partner of NFasc155 and NFasc186, both in vivo and by in vitro adhesion assay. Furthermore, we demonstrate that HDAC1/2-dependent P0 expression is crucial for the maintenance of paranodal/nodal integrity and axonal function through interaction of P0 with neurofascins. In addition, we show that the latter mechanism is impaired by some P0 mutations that lead to late onset Charcot-Marie-Tooth disease.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Enfermedad de Charcot-Marie-Tooth/genética , Proteína P0 de la Mielina/genética , Vaina de Mielina/fisiología , Factores de Crecimiento Nervioso/metabolismo , Animales , Moléculas de Adhesión Celular Neuronal/metabolismo , Enfermedad de Charcot-Marie-Tooth/enzimología , Técnicas de Inactivación de Genes , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/metabolismo , Humanos , Ratones
12.
Genes Dev ; 24(5): 455-69, 2010 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-20194438

RESUMEN

Histone deacetylases (HDACs) regulate gene expression by deacetylating histones and also modulate the acetylation of a number of nonhistone proteins, thus impinging on various cellular processes. Here, we analyzed the major class I enzymes HDAC1 and HDAC2 in primary mouse fibroblasts and in the B-cell lineage. Fibroblasts lacking both enzymes fail to proliferate in culture and exhibit a strong cell cycle block in the G1 phase that is associated with up-regulation of the CDK inhibitors p21(WAF1/CIP1) and p57(Kip2) and of the corresponding mRNAs. This regulation is direct, as in wild-type cells HDAC1 and HDAC2 are bound to the promoter regions of the p21 and p57 genes. Furthermore, analysis of the transcriptome and of histone modifications in mutant cells demonstrated that HDAC1 and HDAC2 have only partly overlapping roles. Next, we eliminated HDAC1 and HDAC2 in the B cells of conditionally targeted mice. We found that B-cell development strictly requires the presence of at least one of these enzymes: When both enzymes are ablated, B-cell development is blocked at an early stage, and the rare remaining pre-B cells show a block in G1 accompanied by the induction of apoptosis. In contrast, elimination of HDAC1 and HDAC2 in mature resting B cells has no negative impact, unless these cells are induced to proliferate. These results indicate that HDAC1 and HDAC2, by normally repressing the expression of p21 and p57, regulate the G1-to-S-phase transition of the cell cycle.


Asunto(s)
Linfocitos B , Fibroblastos , Fase G1/fisiología , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/metabolismo , Fase S/fisiología , Animales , Apoptosis/genética , Linfocitos B/citología , Linfocitos B/enzimología , Proliferación Celular , Células Cultivadas , Fibroblastos/citología , Fibroblastos/enzimología , Regulación del Desarrollo de la Expresión Génica , Histona Desacetilasa 1/genética , Histona Desacetilasa 2/genética , Ratones , Mutación/genética , Regulación hacia Arriba
13.
EMBO J ; 32(24): 3176-91, 2013 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-24240174

RESUMEN

The histone deacetylases HDAC1 and HDAC2 remove acetyl moieties from lysine residues of histones and other proteins and are important regulators of gene expression. By deleting different combinations of Hdac1 and Hdac2 alleles in the epidermis, we reveal a dosage-dependent effect of HDAC1/HDAC2 activity on epidermal proliferation and differentiation. Conditional ablation of either HDAC1 or HDAC2 in the epidermis leads to no obvious phenotype due to compensation by the upregulated paralogue. Strikingly, deletion of a single Hdac2 allele in HDAC1 knockout mice results in severe epidermal defects, including alopecia, hyperkeratosis, hyperproliferation and spontaneous tumour formation. These mice display impaired Sin3A co-repressor complex function, increased levels of c-Myc protein, p53 expression and apoptosis in hair follicles (HFs) and misregulation of HF bulge stem cells. Surprisingly, ablation of HDAC1 but not HDAC2 in a skin tumour model leads to accelerated tumour development. Our data reveal a crucial function of HDAC1/HDAC2 in the control of lineage specificity and a novel role of HDAC1 as a tumour suppressor in the epidermis.


Asunto(s)
Epidermis/crecimiento & desarrollo , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/metabolismo , Neoplasias Cutáneas/genética , Alopecia/genética , Animales , Apoptosis/genética , Linaje de la Célula , Proteínas Co-Represoras , Modelos Animales de Enfermedad , Epidermis/enzimología , Epidermis/patología , Regulación de la Expresión Génica , Genes Supresores de Tumor , Genes p53 , Folículo Piloso/patología , Histona Desacetilasa 1/genética , Histona Desacetilasa 2/genética , Queratosis/genética , Queratosis/patología , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Neoplasias Cutáneas/patología
14.
Development ; 141(3): 604-616, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24449838

RESUMEN

The histone deacetylases HDAC1 and HDAC2 are crucial regulators of chromatin structure and gene expression, thereby controlling important developmental processes. In the mouse brain, HDAC1 and HDAC2 exhibit different developmental stage- and lineage-specific expression patterns. To examine the individual contribution of these deacetylases during brain development, we deleted different combinations of Hdac1 and Hdac2 alleles in neural cells. Ablation of Hdac1 or Hdac2 by Nestin-Cre had no obvious consequences on brain development and architecture owing to compensation by the paralog. By contrast, combined deletion of Hdac1 and Hdac2 resulted in impaired chromatin structure, DNA damage, apoptosis and embryonic lethality. To dissect the individual roles of HDAC1 and HDAC2, we expressed single alleles of either Hdac1 or Hdac2 in the absence of the respective paralog in neural cells. The DNA-damage phenotype observed in double knockout brains was prevented by expression of a single allele of either Hdac1 or Hdac2. Strikingly, Hdac1(-/-)Hdac2(+/-) brains showed normal development and no obvious phenotype, whereas Hdac1(+/-)Hdac2(-/-) mice displayed impaired brain development and perinatal lethality. Hdac1(+/-)Hdac2(-/-) neural precursor cells showed reduced proliferation and premature differentiation mediated by overexpression of protein kinase C, delta, which is a direct target of HDAC2. Importantly, chemical inhibition or knockdown of protein kinase C delta was sufficient to rescue the phenotype of neural progenitor cells in vitro. Our data indicate that HDAC1 and HDAC2 have a common function in maintaining proper chromatin structures and show that HDAC2 has a unique role by controlling the fate of neural progenitors during normal brain development.


Asunto(s)
Alelos , Encéfalo/embriología , Encéfalo/enzimología , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/genética , Homología de Secuencia de Aminoácido , Acetofenonas/farmacología , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Apoptosis/genética , Benzopiranos/farmacología , Encéfalo/metabolismo , Encéfalo/patología , Proteínas Co-Represoras/metabolismo , Daño del ADN/genética , Pérdida del Embrión/enzimología , Pérdida del Embrión/patología , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Histona Desacetilasa 1/genética , Histona Desacetilasa 2/metabolismo , Ratones , Ratones Endogámicos C57BL , Fenotipo , Proteína Quinasa C-delta/antagonistas & inhibidores , Proteína Quinasa C-delta/genética , Proteína Quinasa C-delta/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
15.
J Biol Chem ; 290(7): 4215-24, 2015 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-25516595

RESUMEN

Skeletal muscle atrophy is a severe condition of muscle mass loss. Muscle atrophy is caused by a down-regulation of protein synthesis and by an increase of protein breakdown due to the ubiquitin-proteasome system and autophagy activation. Up-regulation of specific genes, such as the muscle-specific E3 ubiquitin ligase MAFbx, by FoxO transcription factors is essential to initiate muscle protein ubiquitination and degradation during atrophy. HDAC6 is a particular HDAC, which is functionally related to the ubiquitin proteasome system via its ubiquitin binding domain. We show that HDAC6 is up-regulated during muscle atrophy. HDAC6 activation is dependent on the transcription factor FoxO3a, and the inactivation of HDAC6 in mice protects against muscle wasting. HDAC6 is able to interact with MAFbx, a key ubiquitin ligase involved in muscle atrophy. Our findings demonstrate the implication of HDAC6 in skeletal muscle wasting and identify HDAC6 as a new downstream target of FoxO3a in stress response. This work provides new insights in skeletal muscle atrophy development and opens interesting perspectives on HDAC6 as a valuable marker of muscle atrophy and a potential target for pharmacological treatments.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Histona Desacetilasas/metabolismo , Músculo Esquelético/patología , Atrofia Muscular/patología , Animales , Western Blotting , Células Cultivadas , Inmunoprecipitación de Cromatina , Ensayo de Cambio de Movilidad Electroforética , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/genética , Histona Desacetilasa 6 , Histona Desacetilasas/química , Histona Desacetilasas/genética , Humanos , Inmunoprecipitación , Integrasas/metabolismo , Ratones , Ratones Noqueados , Desnervación Muscular , Músculo Esquelético/metabolismo , Atrofia Muscular/genética , Atrofia Muscular/metabolismo , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
Eur J Immunol ; 45(8): 2343-55, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26059604

RESUMEN

Immunoglobulin genes V(D)J rearrangement during early lymphopoiesis is a critical process involving sequential recombination of the heavy and light chain loci. A number of transcription factors act together with temporally activated recombinases and chromatin accessibility changes to regulate this complex process. Here, we deleted the de novo DNA methyltransferases Dnmt3a and Dnmt3b in early B cells of conditionally targeted mice, and monitored the process of V(D)J recombination. Dnmt3a and Dnmt3b deletion resulted in precocious recombination of the immunoglobulin κ light chain without impairing the differentiation of mature B cells or overall B-cell development. Ex vivo culture of IL-7 restricted early B-cell progenitors lacking Dnmt3a and Dnmt3b showed precocious Vκ-Jκ rearrangements that are limited to the proximal Vκ genes. Furthermore, B-cell progenitors deficient in Dnmt3a and Dnmt3b showed elevated levels of germline transcripts at the proximal Vκ genes, alterations in methylation patterns at Igκ enhancer sites and increased expression of the transcription factor E2A. Our data suggest that Dnmt3a and Dnmt3b are critical to regulate the onset of Igκ light chain rearrangement during early B-cell development.


Asunto(s)
Linfocitos B/inmunología , ADN (Citosina-5-)-Metiltransferasas/inmunología , Reordenamiento Génico de Cadena Ligera de Linfocito B/inmunología , Cadenas kappa de Inmunoglobulina/inmunología , Recombinación V(D)J/inmunología , Animales , Linfocitos B/citología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/inmunología , ADN (Citosina-5-)-Metiltransferasas/genética , ADN Metiltransferasa 3A , Femenino , Regulación de la Expresión Génica/inmunología , Cadenas kappa de Inmunoglobulina/genética , Ratones , Ratones Transgénicos , Células Precursoras de Linfocitos B/citología , Células Precursoras de Linfocitos B/inmunología , ADN Metiltransferasa 3B
17.
Nat Rev Immunol ; 5(6): 497-508, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15928681

RESUMEN

The development of B cells from haematopoietic stem cells proceeds along a highly ordered, yet flexible, pathway. At multiple steps along this pathway, cells are instructed by transcription factors on how to further differentiate, and several check-points have been identified. These check-points are initial commitment to lymphocytic progenitors, specification of pre-B cells, entry to the peripheral B-cell pool, maturation of B cells and differentiation into plasma cells. At each of these regulatory nodes, there are transcriptional networks that control the outcome, and much progress has recently been made in dissecting these networks. This article reviews our current understanding of this exciting field.


Asunto(s)
Linfocitos B/inmunología , Células Madre Hematopoyéticas/inmunología , Linfopoyesis/inmunología , Factores de Transcripción/inmunología , Animales , Humanos , Linfopoyesis/genética , Células Plasmáticas/inmunología
18.
J Neurosci ; 34(17): 6112-22, 2014 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-24760871

RESUMEN

Schwann cells, the myelinating glia of the peripheral nervous system (PNS), originate from multipotent neural crest cells that also give rise to other cells, including neurons, melanocytes, chondrocytes, and smooth muscle cells. The transcription factor Sox10 is required for peripheral glia specification. However, all neural crest cells express Sox10 and the mechanisms directing neural crest cells into a specific lineage are poorly understood. We show here that histone deacetylases 1 and 2 (HDAC1/2) are essential for the specification of neural crest cells into Schwann cell precursors and satellite glia, which express the early determinants of their lineage myelin protein zero (P0) and/or fatty acid binding protein 7 (Fabp7). In neural crest cells, HDAC1/2 induced expression of the transcription factor Pax3 by binding and activating the Pax3 promoter. In turn, Pax3 was required to maintain high Sox10 levels and to trigger expression of Fabp7. In addition, HDAC1/2 were bound to the P0 promoter and activated P0 transcription. Consistently, in vivo genetic deletion of HDAC1/2 in mouse neural crest cells led to strongly decreased Sox10 expression, no detectable Pax3, virtually no satellite glia, and no Schwann cell precursors in dorsal root ganglia and peripheral nerves. Similarly, in vivo ablation of Pax3 in the mouse neural crest resulted in strongly reduced expression of Sox10 and Fabp7. Therefore, by controlling the expression of Pax3 and the concerted action of Pax3 and Sox10 on their target genes, HDAC1/2 direct the specification of neural crest cells into peripheral glia.


Asunto(s)
Diferenciación Celular/fisiología , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/metabolismo , Cresta Neural/metabolismo , Células-Madre Neurales/metabolismo , Oligodendroglía/metabolismo , Células de Schwann/metabolismo , Animales , Regulación del Desarrollo de la Expresión Génica , Histona Desacetilasa 1/genética , Histona Desacetilasa 2/genética , Ratones , Cresta Neural/citología , Células-Madre Neurales/citología , Oligodendroglía/citología , Factor de Transcripción PAX3 , Factores de Transcripción Paired Box/genética , Factores de Transcripción Paired Box/metabolismo , Factores de Transcripción SOXE/genética , Factores de Transcripción SOXE/metabolismo , Células de Schwann/citología
19.
EMBO J ; 30(3): 510-23, 2011 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-21157427

RESUMEN

SRSF2 is a serine/arginine-rich protein belonging to the family of SR proteins that are crucial regulators of constitutive and alternative pre-mRNA splicing. Although it is well known that phosphorylation inside RS domain controls activity of SR proteins, other post-translational modifications regulating SRSF2 functions have not been described to date. In this study, we provide the first evidence that the acetyltransferase Tip60 acetylates SRSF2 on its lysine 52 residue inside the RNA recognition motif, and promotes its proteasomal degradation. We also demonstrate that the deacetylase HDAC6 counters this acetylation and acts as a positive regulator of SRSF2 protein level. In addition, we show that Tip60 downregulates SRSF2 phosphorylation by inhibiting the nuclear translocation of both SRPK1 and SRPK2 kinases. Finally, we demonstrate that this acetylation/phosphorylation signalling network controls SRSF2 accumulation as well as caspase-8 pre-mRNA splicing in response to cisplatin and determines whether cells undergo apoptosis or G(2)/M cell cycle arrest. Taken together, these results unravel lysine acetylation as a crucial post-translational modification regulating SRSF2 protein level and activity in response to genotoxic stress.


Asunto(s)
Diferenciación Celular/fisiología , Cisplatino/farmacología , Histona Acetiltransferasas/metabolismo , Histona Desacetilasas/metabolismo , Proteínas Nucleares/metabolismo , Ribonucleoproteínas/metabolismo , Transducción de Señal/fisiología , Acetilación , Empalme Alternativo/efectos de los fármacos , Western Blotting , Caspasa 8/genética , Caspasa 8/metabolismo , Ciclo Celular/efectos de los fármacos , Ciclo Celular/fisiología , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Cartilla de ADN/genética , Histona Desacetilasa 6 , Humanos , Inmunoprecipitación , Lisina/metabolismo , Lisina Acetiltransferasa 5 , Oligonucleótidos/genética , Fosforilación , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Empalme Serina-Arginina , Transducción de Señal/genética , Transfección
20.
Blood ; 121(11): 1930-1, 2013 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-23493769

RESUMEN

In this issue of Blood, Heideman and colleagues show that the major class I histone deacetylases (HDACs) HDAC1 and HDAC2 can act to suppress tumors in mouse thymocytes.


Asunto(s)
Dosificación de Gen/fisiología , Genes Supresores de Tumor/fisiología , Histona Desacetilasa 1/genética , Histona Desacetilasa 2/genética , Proteínas Proto-Oncogénicas c-myc/fisiología , Proteína p53 Supresora de Tumor/fisiología , Animales
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA