Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
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
2.
EMBO J ; 40(22): e108234, 2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34586646

RESUMEN

DNA methylation is a fundamental epigenetic modification, important across biological processes. The maintenance methyltransferase DNMT1 is essential for lineage differentiation during development, but its functions in tissue homeostasis are incompletely understood. We show that epidermis-specific DNMT1 deletion severely disrupts epidermal structure and homeostasis, initiating a massive innate immune response and infiltration of immune cells. Mechanistically, DNA hypomethylation in keratinocytes triggered transposon derepression, mitotic defects, and formation of micronuclei. DNA release into the cytosol of DNMT1-deficient keratinocytes activated signaling through cGAS and STING, thus triggering inflammation. Our findings show that disruption of a key epigenetic mark directly impacts immune and tissue homeostasis, and potentially impacts our understanding of autoinflammatory diseases and cancer immunotherapy.


Asunto(s)
Metilación de ADN , Dermatitis/genética , Epidermis/fisiopatología , Nucleotidiltransferasas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Aberraciones Cromosómicas , Citosol/fisiología , ADN (Citosina-5-)-Metiltransferasa 1/genética , Dermatitis/inmunología , Dermatitis/patología , Humanos , Inmunidad Innata/genética , Helicasa Inducida por Interferón IFIH1/metabolismo , Queratinocitos/inmunología , Queratinocitos/metabolismo , Queratinocitos/patología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Transgénicos , Nucleotidiltransferasas/genética
3.
Nucleic Acids Res ; 51(21): 11748-11769, 2023 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-37878419

RESUMEN

Post-translational modifications of histones are important regulators of the DNA damage response (DDR). By using affinity purification mass spectrometry (AP-MS) we discovered that genetic suppressor element 1 (GSE1) forms a complex with the HDAC1/CoREST deacetylase/demethylase co-repressor complex. In-depth phosphorylome analysis revealed that loss of GSE1 results in impaired DDR, ATR signalling and γH2AX formation upon DNA damage induction. Altered profiles of ATR target serine-glutamine motifs (SQ) on DDR-related hallmark proteins point to a defect in DNA damage sensing. In addition, GSE1 knock-out cells show hampered DNA damage-induced phosphorylation on SQ motifs of regulators of histone post-translational modifications, suggesting altered histone modification. While loss of GSE1 does not affect the histone deacetylation activity of CoREST, GSE1 appears to be essential for binding of the deubiquitinase USP22 to CoREST and for the deubiquitination of H2B K120 in response to DNA damage. The combination of deacetylase, demethylase, and deubiquitinase activity makes the USP22-GSE1-CoREST subcomplex a multi-enzymatic eraser that seems to play an important role during DDR. Since GSE1 has been previously associated with cancer progression and survival our findings are potentially of high medical relevance.


Asunto(s)
Daño del ADN , Histonas , Núcleo Celular/metabolismo , Proteínas Co-Represoras/metabolismo , Enzimas Desubicuitinizantes/genética , Histonas/genética , Histonas/metabolismo , Humanos , Animales , Ratones , Línea Celular
4.
PLoS Genet ; 18(8): e1010376, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35994477

RESUMEN

The class I histone deacetylases are essential regulators of cell fate decisions in health and disease. While pan- and class-specific HDAC inhibitors are available, these drugs do not allow a comprehensive understanding of individual HDAC function, or the therapeutic potential of isoform-specific targeting. To systematically compare the impact of individual catalytic functions of HDAC1, HDAC2 and HDAC3, we generated human HAP1 cell lines expressing catalytically inactive HDAC enzymes. Using this genetic toolbox we compare the effect of individual HDAC inhibition with the effects of class I specific inhibitors on cell viability, protein acetylation and gene expression. Individual inactivation of HDAC1 or HDAC2 has only mild effects on cell viability, while HDAC3 inactivation or loss results in DNA damage and apoptosis. Inactivation of HDAC1/HDAC2 led to increased acetylation of components of the COREST co-repressor complex, reduced deacetylase activity associated with this complex and derepression of neuronal genes. HDAC3 controls the acetylation of nuclear hormone receptor associated proteins and the expression of nuclear hormone receptor regulated genes. Acetylation of specific histone acetyltransferases and HDACs is sensitive to inactivation of HDAC1/HDAC2. Over a wide range of assays, we determined that in particular HDAC1 or HDAC2 catalytic inactivation mimics class I specific HDAC inhibitors. Importantly, we further demonstrate that catalytic inactivation of HDAC1 or HDAC2 sensitizes cells to specific cancer drugs. In summary, our systematic study revealed isoform-specific roles of HDAC1/2/3 catalytic functions. We suggest that targeted genetic inactivation of particular isoforms effectively mimics pharmacological HDAC inhibition allowing the identification of relevant HDACs as targets for therapeutic intervention.


Asunto(s)
Histona Desacetilasa 1 , Inhibidores de Histona Desacetilasas , Acetilación , Histona Desacetilasa 1/genética , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/genética , Histona Desacetilasa 2/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Humanos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
5.
J Autoimmun ; 119: 102610, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33621930

RESUMEN

CD4+ T cell trafficking is a fundamental property of adaptive immunity. In this study, we uncover a novel role for histone deacetylase 1 (HDAC1) in controlling effector CD4+ T cell migration, thereby providing mechanistic insight into why a T cell-specific deletion of HDAC1 protects against experimental autoimmune encephalomyelitis (EAE). HDAC1-deficient CD4+ T cells downregulated genes associated with leukocyte extravasation. In vitro, HDAC1-deficient CD4+ T cells displayed aberrant morphology and migration on surfaces coated with integrin LFA-1 ligand ICAM-1 and showed an impaired ability to arrest on and to migrate across a monolayer of primary mouse brain microvascular endothelial cells under physiological flow. Moreover, HDAC1 deficiency reduced homing of CD4+ T cells into the intestinal epithelium and lamina propria preventing weight-loss, crypt damage and intestinal inflammation in adoptive CD4+ T cell transfer colitis. This correlated with reduced expression levels of LFA-1 integrin chains CD11a and CD18 as well as of selectin ligands CD43, CD44 and CD162 on transferred circulating HDAC1-deficient CD4+ T cells. Our data reveal that HDAC1 controls T cell-mediated autoimmunity via the regulation of CD4+ T cell trafficking into the CNS and intestinal tissues.


Asunto(s)
Autoinmunidad , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Quimiotaxis de Leucocito/inmunología , Histona Desacetilasa 1/metabolismo , Inflamación/etiología , Inflamación/metabolismo , Animales , Biomarcadores , Adhesión Celular , Quimiotaxis de Leucocito/genética , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Encefalomielitis Autoinmune Experimental/diagnóstico , Encefalomielitis Autoinmune Experimental/etiología , Encefalomielitis Autoinmune Experimental/metabolismo , Células Endoteliales , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Histona Desacetilasa 1/genética , Inmunohistoquímica , Inflamación/diagnóstico , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Noqueados
6.
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
7.
Mol Cell ; 46(4): 382-3, 2012 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-22633485

RESUMEN

In the current issue of Molecular Cell, de la Vega et al. (2012) propose an intriguing model for HIPK2 posttranslational modifications in response to oxidative stress, explaining how HIPK2 can possess both prosurvival as well as proapoptotic activities.

8.
Genes Dev ; 26(8): 857-71, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22508729

RESUMEN

Epigenetic memory mediated by Polycomb group (PcG) proteins must be maintained during cell division, but must also be flexible to allow cell fate transitions. Here we quantify dynamic chromatin-binding properties of PH::GFP and PC::GFP in living Drosophila in two cell types that undergo defined differentiation and mitosis events. Quantitative fluorescence recovery after photobleaching (FRAP) analysis demonstrates that PcG binding has a higher plasticity in stem cells than in more determined cells and identifies a fraction of PcG proteins that binds mitotic chromatin with up to 300-fold longer residence times than in interphase. Mathematical modeling examines which parameters best distinguish stem cells from differentiated cells. We identify phosphorylation of histone H3 at Ser 28 as a potential mechanism governing the extent and rate of mitotic PC dissociation in different lineages. We propose that regulation of the kinetic properties of PcG-chromatin binding is an essential factor in the choice between stability and flexibility in the establishment of cell identities.


Asunto(s)
Diferenciación Celular , Cromatina/metabolismo , Mitosis , Proteínas Represoras/metabolismo , Células Madre/citología , Animales , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citología , Recuperación de Fluorescencia tras Fotoblanqueo , Proteínas Fluorescentes Verdes/metabolismo , Histonas/metabolismo , Modelos Biológicos , Fosforilación , Proteínas del Grupo Polycomb , Serina/metabolismo , Células Madre/metabolismo
9.
J Cell Physiol ; 233(1): 530-548, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28300292

RESUMEN

Although histone acetylation is one of the most widely studied epigenetic modifications, there is still a lack of information regarding how the acetylome is regulated during brain development and pathophysiological processes. We demonstrate that the embryonic brain (E15) is characterized by an increase in H3K9 acetylation as well as decreases in the levels of HDAC1 and HDAC3. Moreover, experimental induction of H3K9 hyperacetylation led to the overexpression of NCAM in the embryonic cortex and depletion of Sox2 in the subventricular ependyma, which mimicked the differentiation processes. Inducing differentiation in HDAC1-deficient mouse ESCs resulted in early H3K9 deacetylation, Sox2 downregulation, and enhanced astrogliogenesis, whereas neuro-differentiation was almost suppressed. Neuro-differentiation of (wt) ESCs was characterized by H3K9 hyperacetylation that was associated with HDAC1 and HDAC3 depletion. Conversely, the hippocampi of schizophrenia-like animals showed H3K9 deacetylation that was regulated by an increase in both HDAC1 and HDAC3. The hippocampi of schizophrenia-like brains that were treated with the cannabinoid receptor-1 inverse antagonist AM251 expressed H3K9ac at the level observed in normal brains. Together, the results indicate that co-regulation of H3K9ac by HDAC1 and HDAC3 is important to both embryonic brain development and neuro-differentiation as well as the pathophysiology of a schizophrenia-like phenotype.


Asunto(s)
Encéfalo/enzimología , Histona Desacetilasa 1/metabolismo , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Neurogénesis , Neuronas/enzimología , Esquizofrenia/enzimología , Acetilación , Animales , Antipsicóticos/farmacología , Encéfalo/efectos de los fármacos , Encéfalo/embriología , Encéfalo/patología , Antagonistas de Receptores de Cannabinoides/farmacología , Modelos Animales de Enfermedad , Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica , Edad Gestacional , Histona Desacetilasa 1/antagonistas & inhibidores , Histona Desacetilasa 1/genética , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/genética , Acetato de Metilazoximetanol , Ratones Endogámicos C57BL , Moléculas de Adhesión de Célula Nerviosa/genética , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Neurogénesis/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/patología , Procesamiento Proteico-Postraduccional , Ratas Sprague-Dawley , Receptor Cannabinoide CB1/antagonistas & inhibidores , Receptor Cannabinoide CB1/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Esquizofrenia/inducido químicamente , Esquizofrenia/tratamiento farmacológico , Esquizofrenia/genética , Transducción de Señal , Factores de Tiempo
10.
Histochem Cell Biol ; 150(3): 255-269, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29951776

RESUMEN

Aging is associated with profound changes in the epigenome, resulting in alterations of gene expression, epigenetic landscape, and genome architecture. Class I Histone deacetylases (HDACs), consisting of HDAC1, HDAC2, HDAC3, and HDAC8, play a major role in epigenetic regulation of chromatin structure and transcriptional control, and have been implicated as key players in the pathogenesis of age-dependent diseases and disorders affecting health and longevity. Here, we report the identification of class I Hdac orthologs and their detailed spatio-temporal expression profile in the short-lived fish Nothobranchius furzeri from the onset of embryogenesis until old age covering the entire lifespan of the organism. Database search of the recently annotated N. furzeri genomes retrieved four distinct genes: two copies of hdac1 and one copy of each hdac3 and hdac8. However, no hdac2 ortholog could be identified. Phylogenetic analysis grouped the individual killifish class I Hdacs within the well-defined terminal clades. We find that upon aging, Hdac1 is significantly down-regulated in muscle, liver, and brain, and this age-dependent down-regulation in brain clearly correlates with increased mRNA levels of the cyclin-dependent kinase inhibitor cdkn1a (p21). Furthermore, this apparent reduction of class I HDACs in transcript and protein levels is mirrored in the mouse brain, highlighting an evolutionarily conserved role of class I HDACs during normal development and in the aging process.


Asunto(s)
Envejecimiento , Peces , Histona Desacetilasa 1/genética , Animales , Perfilación de la Expresión Génica , Histona Desacetilasa 1/metabolismo , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Análisis de Supervivencia
11.
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
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.
Genome Res ; 24(11): 1808-20, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25135956

RESUMEN

The selectivity of transcriptional responses to extracellular cues is reflected by the deposition of stimulus-specific chromatin marks. Although histone H3 phosphorylation is a target of numerous signaling pathways, its role in transcriptional regulation remains poorly understood. Here, for the first time, we report a genome-wide analysis of H3S28 phosphorylation in a mammalian system in the context of stress signaling. We found that this mark targets as many as 50% of all stress-induced genes, underlining its importance in signal-induced transcription. By combining ChIP-seq, RNA-seq, and mass spectrometry we identified the factors involved in the biological interpretation of this histone modification. We found that MSK1/2-mediated phosphorylation of H3S28 at stress-responsive promoters contributes to the dissociation of HDAC corepressor complexes and thereby to enhanced local histone acetylation and subsequent transcriptional activation of stress-induced genes. Our data reveal a novel function of the H3S28ph mark in the activation of mammalian genes in response to MAP kinase pathway activation.


Asunto(s)
Histonas/metabolismo , Serina/metabolismo , Estrés Fisiológico/genética , Activación Transcripcional , Células 3T3 , Acetilación , Animales , Inmunoprecipitación de Cromatina , Fibroblastos/citología , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Ontología de Genes , Estudio de Asociación del Genoma Completo , Células HeLa , Secuenciación de Nucleótidos de Alto Rendimiento , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas/genética , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosforilación , Regiones Promotoras Genéticas/genética , Proteínas Quinasas S6 Ribosómicas 90-kDa/genética , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo
15.
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
16.
Biochim Biophys Acta ; 1839(8): 711-8, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24747175

RESUMEN

Systematic analysis of histone modifications has revealed a plethora of posttranslational modifications that mediate changes in chromatin structure and gene expression. Histone phosphorylation is a transient histone modification that becomes induced by extracellular signals, DNA damage or entry into mitosis. Importantly, phosphorylation of histone proteins does lead not only to the binding of specific reader proteins but also to changes in the affinity for readers or writers of other histone modifications. This induces a cross-talk between different chromatin modifications that allows the spatio-temporal control of chromatin-associated events. In this review we will summarize the progress in our current knowledge of factors sensing reversible histone phosphorylation in different biological scenarios. This article is part of a Special Issue entitled: Molecular mechanisms of histone modification function.


Asunto(s)
Proteínas 14-3-3/metabolismo , Cromatina/química , Epigénesis Genética , Histonas/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas 14-3-3/genética , Acetilación , Cromatina/genética , Cromatina/metabolismo , Daño del ADN , Células Eucariotas/citología , Células Eucariotas/metabolismo , Histonas/genética , Humanos , Metilación , Mitosis , Fosforilación , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Transducción de Señal , Transcripción Genética
17.
Chromosoma ; 123(1-2): 67-78, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24170248

RESUMEN

The Rpd3-like members of the class I lysine deacetylase family are important regulators of chromatin structure and gene expression and have pivotal functions in the control of proliferation, differentiation and development. The highly related class I deacetylases HDAC1 and HDAC2 have partially overlapping but also isoform-specific roles in diverse biological processes, whereas HDAC3 and HDAC8 have unique functions. This review describes the role of class I KDACs in the regulation of transcription as well as their non-transcriptional functions, in particular their contributions to splicing, mitosis/meiosis, replication and DNA repair. During the past years, a number of mouse loss-of-function studies provided new insights into the individual roles of class I deacetylases in cell cycle control, differentiation and tumorigenesis. Simultaneous ablation of HDAC1 and HDAC2 or single deletion of Hdac3 severely impairs cell cycle progression in all proliferating cell types indicating that these class I deacetylases are promising targets for small molecule inhibitors as anti-tumor drugs.


Asunto(s)
Histona Desacetilasas/metabolismo , Lisina/metabolismo , Mamíferos/metabolismo , Transcripción Genética , Animales , Histona Desacetilasas/química , Histonas/metabolismo , Humanos , Especificidad por Sustrato
18.
PLoS Genet ; 8(3): e1002540, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22396659

RESUMEN

A CpG island (CGI) lies at the 5' end of the Airn macro non-protein-coding (nc) RNA that represses the flanking Igf2r promoter in cis on paternally inherited chromosomes. In addition to being modified on maternally inherited chromosomes by a DNA methylation imprint, the Airn CGI shows two unusual organization features: its position immediately downstream of the Airn promoter and transcription start site and a series of tandem direct repeats (TDRs) occupying its second half. The physical separation of the Airn promoter from the CGI provides a model to investigate if the CGI plays distinct transcriptional and epigenetic roles. We used homologous recombination to generate embryonic stem cells carrying deletions at the endogenous locus of the entire CGI or just the TDRs. The deleted Airn alleles were analyzed by using an ES cell imprinting model that recapitulates the onset of Igf2r imprinted expression in embryonic development or by using knock-out mice. The results show that the CGI is required for efficient Airn initiation and to maintain the unmethylated state of the Airn promoter, which are both necessary for Igf2r repression on the paternal chromosome. The TDRs occupying the second half of the CGI play a minor role in Airn transcriptional elongation or processivity, but are essential for methylation on the maternal Airn promoter that is necessary for Igf2r to be expressed from this chromosome. Together the data indicate the existence of a class of regulatory CGIs in the mammalian genome that act downstream of the promoter and transcription start.


Asunto(s)
Islas de CpG/genética , Metilación de ADN , Epigénesis Genética , Impresión Genómica , Regiones Promotoras Genéticas , Precursores del ARN/genética , ARN no Traducido/genética , Animales , Diferenciación Celular , Células Cultivadas , Desarrollo Embrionario , Células Madre Embrionarias , Regulación de la Expresión Génica , Recombinación Homóloga , Factor II del Crecimiento Similar a la Insulina/genética , Factor II del Crecimiento Similar a la Insulina/metabolismo , Ratones , Eliminación de Secuencia , Secuencias Repetidas en Tándem , Sitio de Iniciación de la Transcripción
19.
J Biol Chem ; 288(23): 16518-16528, 2013 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-23612983

RESUMEN

Histone deacetylase 1 (HDAC1) and HDAC2 are components of corepressor complexes that are involved in chromatin remodeling and regulation of gene expression by regulating dynamic protein acetylation. HDAC1 and -2 form homo- and heterodimers, and their activity is dependent upon dimer formation. Phosphorylation of HDAC1 and/or HDAC2 in interphase cells is required for the formation of HDAC corepressor complexes. In this study, we show that during mitosis, HDAC2 and, to a lesser extent, HDAC1 phosphorylation levels dramatically increase. When HDAC1 and -2 are displaced from the chromosome during metaphase, they dissociate from each other, but each enzyme remains in association with components of the HDAC corepressor complexes Sin3, NuRD, and CoREST as homodimers. Enzyme inhibition studies and mutational analyses demonstrated that protein kinase CK2-catalyzed phosphorylation of HDAC1 and -2 is crucial for the dissociation of these two enzymes. These results suggest that corepressor complexes, including HDAC1 or HDAC2 homodimers, might target different cellular proteins during mitosis.


Asunto(s)
Quinasa de la Caseína I/metabolismo , Cromosomas Humanos/enzimología , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/metabolismo , Mitosis/fisiología , Multimerización de Proteína/fisiología , Quinasa de la Caseína I/antagonistas & inhibidores , Quinasa de la Caseína I/genética , Cromosomas Humanos/genética , Proteínas Co-Represoras , Células HEK293 , Células HeLa , Histona Desacetilasa 1/genética , Histona Desacetilasa 2/genética , Humanos , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/genética , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Mutación , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Fosforilación/fisiología , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Complejo Correpresor Histona Desacetilasa y Sin3/genética , Complejo Correpresor Histona Desacetilasa y Sin3/metabolismo
20.
EMBO J ; 29(23): 3992-4007, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-20967026

RESUMEN

Histone deacetylase (HDAC) inhibitors induce cell cycle arrest, differentiation or apoptosis in tumour cells and are, therefore, promising anti-cancer reagents. However, the specific HDAC isoforms that mediate these effects are not yet identified. To explore the role of HDAC1 in tumourigenesis and tumour proliferation, we established an experimental teratoma model using wild-type and HDAC1-deficient embryonic stem cells. HDAC1-deficient teratomas showed no significant difference in size compared with wild-type teratomas. Surprisingly, loss of HDAC1 was not only linked to increased apoptosis, but also to significantly enhanced proliferation. Epithelial structures showed reduced differentiation as monitored by Oct3/4 expression and changed E-cadherin localization and displayed up-regulated expression of SNAIL1, a regulator of epithelial cell plasticity. Increased levels of the transcriptional regulator SNAIL1 are crucial for enhanced proliferation and reduced differentiation of HDAC1-deficient teratoma. Importantly, the analysis of human teratomas revealed a similar link between loss of HDAC1 and enhanced tumour malignancy. These findings reveal a novel role for HDAC1 in the control of tumour proliferation and identify HDAC1 as potential marker for benign teratomas.


Asunto(s)
Células Madre Embrionarias/metabolismo , Regulación Neoplásica de la Expresión Génica , Histona Desacetilasa 1/genética , Teratoma/enzimología , Animales , Apoptosis , Cadherinas/genética , Carcinoma Embrionario/enzimología , Carcinoma Embrionario/genética , Carcinoma Embrionario/patología , Línea Celular , Línea Celular Tumoral , Proliferación Celular , Células Madre Embrionarias/citología , Células Madre Embrionarias/patología , Histona Desacetilasa 1/metabolismo , Humanos , Ratones , Factor 3 de Transcripción de Unión a Octámeros/genética , Fenotipo , Factores de Transcripción de la Familia Snail , Teratoma/genética , Teratoma/patología , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA