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













Base de datos
Intervalo de año de publicación
1.
Cell Death Dis ; 12(8): 779, 2021 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-34365463

RESUMEN

The PD-L1 overexpression is an important event of immune escape and metastasis in triple-negative breast cancer (TNBC), but the molecular mechanism remains to be determined. Interferon gamma (IFNγ) represents a major driving force behind PD-L1 expression in tumor microenvironment, and histone deacetylase 2 (HDAC2) is required for IFN signaling. Here, we investigated the regulation of HDAC2 on the IFNγ-induced PD-L1 expression in TNBC cells. We found the HDAC2 and PD-L1 expression in TNBC was significantly higher than that in non-TNBC, and HDAC2 was positively correlated with PD-L1 expression. HDAC2 promoted PD-L1 induction by upregulating the phosphorylation of JAK1, JAK2, and STAT1, as well as the translocation of STAT1 to the nucleus and the recruitment of STAT1 to the PD-L1 promoter. Meanwhile, HDAC2 was recruited to the PD-L1 promoter by STAT1, and HDAC2 knockout compromised IFNγ-induced upregulation of H3K27, H3K9 acetylation, and the BRD4 recruitment in PD-L1 promoter. In addition, significant inhibition of proliferation, colony formation, migration, and cell cycle of TNBC cells were observed following knockout of HDAC2 in vitro. Furthermore, HDAC2 knockout reduced IFNγ-induced PD-L1 expression, lymphocyte infiltration, and retarded tumor growth and metastasis in the breast cancer mouse models. This study may provide evidence that HDAC2 promotes IFNγ-induced PD-L1 expression, suggesting a way for enhanced antitumor immunity when targeting the HDAC2 in TNBC.


Asunto(s)
Antígeno B7-H1/genética , Regulación hacia Abajo/genética , Regulación Neoplásica de la Expresión Génica , Técnicas de Inactivación de Genes , Histona Desacetilasa 2/deficiencia , Evasión Inmune , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/inmunología , Acetilación/efectos de los fármacos , Animales , Antígeno B7-H1/metabolismo , Secuencia de Bases , Ciclo Celular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Femenino , Histona Desacetilasa 2/metabolismo , Histonas/metabolismo , Humanos , Evasión Inmune/genética , Interferón gamma/farmacología , Janus Quinasa 2/metabolismo , Linfocitos Infiltrantes de Tumor/inmunología , Ratones Endogámicos BALB C , Ratones Desnudos , Regiones Promotoras Genéticas/genética , Factor de Transcripción STAT1/metabolismo , Transducción de Señal/efectos de los fármacos , Ensayo de Tumor de Célula Madre
2.
Cells ; 10(2)2021 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-33498747

RESUMEN

We have previously reported that histone deacetylase epigenetic regulator Hdac1 and Hdac2 deletion in intestinal epithelial cells (IEC) disrupts mucosal tissue architecture and barrier, causing chronic inflammation. In this study, proteome and transcriptome analysis revealed the importance of signaling pathways induced upon genetic IEC-Hdac1 and Hdac2 deletion. Indeed, Gene Ontology biological process analysis of enriched deficient IEC RNA and proteins identified common pathways, including lipid metabolic and oxidation-reduction process, cell adhesion, and antigen processing and presentation, related to immune responses, correlating with dysregulation of major histocompatibility complex (MHC) class II genes. Top upstream regulators included regulators associated with environmental sensing pathways to xenobiotics, microbial and diet-derived ligands, and endogenous metabolites. Proteome analysis revealed mTOR signaling IEC-specific defects. In addition to mTOR, the STAT and Notch pathways were dysregulated specifically in jejunal IEC. To determine the impact of pathway dysregulation on mutant jejunum alterations, we treated mutant mice with Tofacitinib, a JAK inhibitor. Treatment with the inhibitor partially corrected proliferation and tight junction defects, as well as niche stabilization by increasing Paneth cell numbers. Thus, IEC-specific histone deacetylases 1 (HDAC1) and 2 (HDAC2) support intestinal homeostasis by regulating survival and translation processes, as well as differentiation and metabolic pathways. HDAC1 and HDAC2 may play an important role in the regulation of IEC-specific inflammatory responses by controlling, directly or indirectly, the JAK/STAT pathway. IEC-specific JAK/STAT pathway deregulation may be, at least in part, responsible for intestinal homeostasis disruption in mutant mice.


Asunto(s)
Células Epiteliales/metabolismo , Histona Desacetilasa 1/deficiencia , Histona Desacetilasa 2/deficiencia , Homeostasis , Intestinos/citología , Quinasas Janus/metabolismo , Factores de Transcripción STAT/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Eliminación de Gen , Histona Desacetilasa 1/genética , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/genética , Histona Desacetilasa 2/metabolismo , Homeostasis/efectos de los fármacos , Recuento de Linfocitos , Ratones Endogámicos C57BL , Ratones Transgénicos , Organoides/efectos de los fármacos , Organoides/crecimiento & desarrollo , Células de Paneth/efectos de los fármacos , Células de Paneth/metabolismo , Piperidinas/farmacología , Pirimidinas/farmacología , Linfocitos T/efectos de los fármacos
3.
J Exp Clin Cancer Res ; 37(1): 296, 2018 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-30509303

RESUMEN

BACKGROUND: Cancer stem cells (CSCs) play a key role in cancer initiation, progression and chemoresistance. Epigenetic alterations have been identified as prominent factors that contribute to the CSCs phenotype. Here, we investigated the effects of the HDAC inhibitor valproic acid (VPA) and the demethylating agent, 5'azacytidine (DAC) on the stem phenotype of MG63 and Saos2 osteosarcoma cell lines. METHODS: Saos2 and MG63 cells were treated with DAC and VPA, alone and in combination. Untreated and treated cells were examined for stemness phenotype by cytometry and real-time PCR. Sarcospheres and colonies formation were also evaluated. Moreover, histone modification and methylation were tested by flow cytomery and western blotting. HDAC2 depleted cells were examined for stemness phenotype and their ability to generate tumors in NOD/SCID IL2R-gamma-0 (NSG) mice. HDAC2 expression on human osteosarcoma tissues was evaluated. RESULTS: We found that DAC and VPA induce an increased expression of stem markers including CD133, OCT4, SOX2 and NANOG, and an increased ability in sarcospheres and colonies formation efficiency. Interestingly, we showed that DAC and VPA treatment decreased repressive histone markers, while increased the active ones. These histone modifications were also associated with an increase of acetylation of histones H3, a decrease of DNA global methylation, HDAC2 and DNMT3a. Furthermore, HDAC2 silenced-MG63 and Saos2 cells acquired a stem phenotype, and promoted in vivo tumorigenesis. In human osteosarcoma tissues, HDAC2 was strongly expressed in nucleus. CONCLUSIONS: Collectively, our results suggest that VPA and DAC induce an expansion of osteosarcoma CSCs, and we report for the first time that HDAC2 is a key factor regulating both CSCs phenotype and in vivo cancer growth. In conclusion, we have identified HDAC2 as a potential therapeutic target in human osteosarcoma treatment.


Asunto(s)
Neoplasias Óseas/enzimología , Histona Desacetilasa 2/deficiencia , Células Madre Neoplásicas/enzimología , Osteosarcoma/enzimología , Animales , Azacitidina/farmacología , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Línea Celular Tumoral , Metilación de ADN , Xenoinjertos , Histona Desacetilasa 2/antagonistas & inhibidores , Histona Desacetilasa 2/metabolismo , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Osteosarcoma/genética , Osteosarcoma/patología , Transfección , Ácido Valproico/farmacología
4.
Stem Cell Reports ; 10(4): 1369-1383, 2018 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-29641990

RESUMEN

The first hematopoietic stem and progenitor cells are generated during development from hemogenic endothelium (HE) through trans-differentiation. The molecular mechanisms underlying this endothelial-to-hematopoietic transition (EHT) remain poorly understood. Here, we explored the role of the epigenetic regulators HDAC1 and HDAC2 in the emergence of these first blood cells in vitro and in vivo. Loss of either of these epigenetic silencers through conditional genetic deletion reduced hematopoietic transition from HE, while combined deletion was incompatible with blood generation. We investigated the molecular basis of HDAC1 and HDAC2 requirement and identified TGF-ß signaling as one of the pathways controlled by HDAC1 and HDAC2. Accordingly, we experimentally demonstrated that activation of this pathway in HE cells reinforces hematopoietic development. Altogether, our results establish that HDAC1 and HDAC2 modulate TGF-ß signaling and suggest that stimulation of this pathway in HE cells would be beneficial for production of hematopoietic cells for regenerative therapies.


Asunto(s)
Células Endoteliales/citología , Células Endoteliales/metabolismo , Hematopoyesis , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo , Animales , Benzamidas/farmacología , Diferenciación Celular/efectos de los fármacos , Dioxoles/farmacología , Células Endoteliales/efectos de los fármacos , Eliminación de Gen , Hemangioblastos/citología , Hematopoyesis/efectos de los fármacos , Histona Desacetilasa 1/deficiencia , Histona Desacetilasa 2/deficiencia , Inhibidores de Histona Desacetilasas/farmacología , Ratones , Transducción de Señal/efectos de los fármacos
5.
Nat Neurosci ; 20(9): 1247-1259, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28783139

RESUMEN

Antipsychotic drugs remain the standard for schizophrenia treatment. Despite their effectiveness in treating hallucinations and delusions, prolonged exposure to antipsychotic medications leads to cognitive deficits in both schizophrenia patients and animal models. The molecular mechanisms underlying these negative effects on cognition remain to be elucidated. Here we demonstrate that chronic antipsychotic drug exposure increases nuclear translocation of NF-κB in both mouse and human frontal cortex, a trafficking event triggered via 5-HT2A-receptor-dependent downregulation of the NF-κB repressor IκBα. This upregulation of NF-κB activity led to its increased binding at the Hdac2 promoter, thereby augmenting Hdac2 transcription. Deletion of HDAC2 in forebrain pyramidal neurons prevented the negative effects of antipsychotic treatment on synaptic remodeling and cognition. Conversely, virally mediated activation of NF-κB signaling decreased cortical synaptic plasticity via HDAC2. Together, these observations may aid in developing therapeutic strategies to improve the outcome of schizophrenia treatment.


Asunto(s)
Antipsicóticos/efectos adversos , Trastornos del Conocimiento/inducido químicamente , Trastornos del Conocimiento/metabolismo , Histona Desacetilasa 2/metabolismo , FN-kappa B/metabolismo , Sinapsis/metabolismo , Animales , Antipsicóticos/toxicidad , Trastornos del Conocimiento/genética , Lóbulo Frontal/efectos de los fármacos , Lóbulo Frontal/metabolismo , Células HEK293 , Histona Desacetilasa 2/deficiencia , Histona Desacetilasa 2/genética , Humanos , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , FN-kappa B/genética , Sinapsis/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos , Activación Transcripcional/fisiología
6.
Nat Commun ; 8: 14272, 2017 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-28139683

RESUMEN

The peripheral nervous system (PNS) regenerates after injury. However, regeneration is often compromised in the case of large lesions, and the speed of axon reconnection to their target is critical for successful functional recovery. After injury, mature Schwann cells (SCs) convert into repair cells that foster axonal regrowth, and redifferentiate to rebuild myelin. These processes require the regulation of several transcription factors, but the driving mechanisms remain partially understood. Here we identify an early response to nerve injury controlled by histone deacetylase 2 (HDAC2), which coordinates the action of other chromatin-remodelling enzymes to induce the upregulation of Oct6, a key transcription factor for SC development. Inactivating this mechanism using mouse genetics allows earlier conversion into repair cells and leads to faster axonal regrowth, but impairs remyelination. Consistently, short-term HDAC1/2 inhibitor treatment early after lesion accelerates functional recovery and enhances regeneration, thereby identifying a new therapeutic strategy to improve PNS regeneration after lesion.


Asunto(s)
Benzamidas/farmacología , Histona Desacetilasa 1/genética , Histona Desacetilasa 2/genética , Inhibidores de Histona Desacetilasas/farmacología , Regeneración Nerviosa/efectos de los fármacos , Traumatismos de los Nervios Periféricos/tratamiento farmacológico , Pirimidinas/farmacología , Células de Schwann/efectos de los fármacos , Animales , Axones/efectos de los fármacos , Axones/metabolismo , Proteína 2 de la Respuesta de Crecimiento Precoz/genética , Proteína 2 de la Respuesta de Crecimiento Precoz/metabolismo , Regulación de la Expresión Génica , Genes Reporteros , Histona Desacetilasa 1/antagonistas & inhibidores , Histona Desacetilasa 1/deficiencia , Histona Desacetilasa 2/antagonistas & inhibidores , Histona Desacetilasa 2/deficiencia , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Luciferasas/genética , Luciferasas/metabolismo , Ratones , Ratones Noqueados , Regeneración Nerviosa/genética , Factor de Transcripción PAX3/genética , Factor de Transcripción PAX3/metabolismo , Traumatismos de los Nervios Periféricos/genética , Traumatismos de los Nervios Periféricos/metabolismo , Traumatismos de los Nervios Periféricos/patología , Recuperación de la Función/efectos de los fármacos , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Células de Schwann/metabolismo , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
7.
J Cell Physiol ; 231(2): 436-48, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26174178

RESUMEN

The intestinal epithelium responds to and transmits signals from the microbiota and the mucosal immune system to insure intestinal homeostasis. These interactions are in part conveyed by epigenetic modifications, which respond to environmental changes. Protein acetylation is an epigenetic signal regulated by histone deacetylases, including Hdac1 and Hdac2. We have previously shown that villin-Cre-inducible intestinal epithelial cell (IEC)-specific Hdac1 and Hdac2 deletions disturb intestinal homeostasis. To determine the role of Hdac1 and Hdac2 in the regulation of IEC function and the establishment of the dual knockout phenotype, we have generated villin-Cre murine models expressing one Hdac1 allele without Hdac2, or one Hdac2 allele without Hdac1. We have also investigated the effect of short-term deletion of both genes in naphtoflavone-inducible Ah-Cre and tamoxifen-inducible villin-Cre(ER) mice. Mice with one Hdac1 allele displayed normal tissue architecture, but increased sensitivity to DSS-induced colitis. In contrast, mice with one Hdac2 allele displayed intestinal architecture defects, increased proliferation, decreased goblet cell numbers as opposed to Paneth cells, increased immune cell infiltration associated with fibrosis, and increased sensitivity to DSS-induced colitis. In comparison to dual knockout mice, intermediary activation of Notch, mTOR, and Stat3 signaling pathways was observed. While villin-Cre(ER) Hdac1 and Hdac2 deletions led to an impaired epithelium and differentiation defects, Ah-Cre-mediated deletion resulted in blunted proliferation associated with the induction of a DNA damage response. Our results suggest that IEC determination and intestinal homeostasis are highly dependent on Hdac1 and Hdac2 activity levels, and that changes in the IEC acetylome may alter the mucosal environment.


Asunto(s)
Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/metabolismo , Mucosa Intestinal/enzimología , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Proliferación Celular/genética , Proliferación Celular/fisiología , Colitis/enzimología , Colitis/genética , Colitis/patología , Daño del ADN , Modelos Animales de Enfermedad , Células Epiteliales/enzimología , Células Caliciformes/citología , Células Caliciformes/enzimología , Histona Desacetilasa 1/deficiencia , Histona Desacetilasa 1/genética , Histona Desacetilasa 2/deficiencia , Histona Desacetilasa 2/genética , Homeostasis , Inmunidad Mucosa , Mucosa Intestinal/anomalías , Mucosa Intestinal/citología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Notch/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo
8.
Sci Rep ; 5: 13396, 2015 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-26300473

RESUMEN

Methamphetamine (METH) produces increases in the expression of immediate early genes (IEGs) and of histone deacetylase 2 (HDAC2) in the rat nucleus accumbens (NAc). Here, we tested whether HDAC2 deletion influenced the effects of METH on IEG expression in the NAc. Microarray analyses showed no baseline differences in IEG expression between wild-type (WT) and HDAC2 knockout (KO) mice. Quantitative-PCR analysis shows that an acute METH injection produced time-dependent increases in mRNA levels of several IEGs in both genotypes. Interestingly, HDAC2KO mice displayed greater METH-induced increases in Egr1 and Egr2 mRNA levels measured at one hour post-injection. The levels of Fosb, Fra2, Egr1, and Egr3 mRNAs stayed elevated in the HDAC2KO mice 2 hours after the METH injection whereas these mRNAs had normalized in the WT mice. In WT mice, METH caused increased HDAC2 recruitment to the promoters some IEGs at 2 hours post injection. METH-induced prolonged increases in Fosb, Fra2, Egr1, and Egr3 mRNA levels in HDAC2KO mice were associated with increased enrichment of phosphorylated CREB (pCREB) on the promoters of these genes. Based on our observations, we hypothesize that HDAC2 may regulate the expression of these genes, in part, by prolonging the actions of pCREB in the mouse NAc.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Eliminación de Gen , Regulación de la Expresión Génica/efectos de los fármacos , Genes Inmediatos-Precoces , Histona Desacetilasa 2/deficiencia , Metanfetamina/farmacología , Núcleo Accumbens/metabolismo , Animales , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Histona Desacetilasa 2/metabolismo , Masculino , Ratones Noqueados , Núcleo Accumbens/efectos de los fármacos , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Unión Proteica/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Tiempo , Factor de Transcripción AP-1/metabolismo
9.
ASN Neuro ; 7(3)2015.
Artículo en Inglés | MEDLINE | ID: mdl-26129908

RESUMEN

Histones deacetylases (HDACs), besides their function as epigenetic regulators, deacetylate and critically regulate the activity of nonhistone targets. In particular, HDACs control partially the proapoptotic activity of p53 by balancing its acetylation state. HDAC inhibitors have revealed neuroprotective properties in different models, but the exact mechanisms of action remain poorly understood. We have generated a conditional knockout mouse model targeting retinal ganglion cells (RGCs) to investigate specifically the functional role of HDAC1 and HDAC2 in an acute model of optic nerve injury. Our results demonstrate that combined HDAC1 and HDAC2 ablation promotes survival of axotomized RGCs. Based on global gene expression analyses, we identified the p53-PUMA apoptosis-inducing axis to be strongly activated in axotomized mouse RGCs. Specific HDAC1/2 ablation inhibited this apoptotic pathway by impairing the crucial acetylation status of p53 and reducing PUMA expression, thereby contributing to the ensuing enhanced neuroprotection due to HDAC1/2 depletion. HDAC1/2 inhibition and the affected downstream signaling components emerge as specific targets for developing therapeutic strategies in neuroprotection.


Asunto(s)
Supervivencia Celular/fisiología , Genes p53 , Histona Desacetilasa 1/deficiencia , Histona Desacetilasa 2/deficiencia , Neuroprotección , Traumatismos del Nervio Óptico/enzimología , Células Ganglionares de la Retina/enzimología , Células Ganglionares de la Retina/fisiología , Acetilación , Enfermedad Aguda , Animales , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Axotomía , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Histona Desacetilasa 1/genética , Histona Desacetilasa 2/genética , Sistema de Señalización de MAP Quinasas , Ratones Noqueados , Traumatismos del Nervio Óptico/patología , Células Ganglionares de la Retina/patología , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
10.
PLoS One ; 9(8): e102684, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25136952

RESUMEN

Selective histone deacetylase (HDAC) inhibitors have emerged as a potential anti-latency therapy for persistent human immunodeficiency virus type 1 (HIV-1) infection. We utilized a combination of small molecule inhibitors and short hairpin (sh)RNA-mediated gene knockdown strategies to delineate the key HDAC(s) to be targeted for selective induction of latent HIV-1 expression. Individual depletion of HDAC3 significantly induced expression from the HIV-1 promoter in the 2D10 latency cell line model. However, depletion of HDAC1 or -2 alone or in combination did not significantly induce HIV-1 expression. Co-depletion of HDAC2 and -3 resulted in a significant increase in expression from the HIV-1 promoter. Furthermore, concurrent knockdown of HDAC1, -2, and -3 resulted in a significant increase in expression from the HIV-1 promoter. Using small molecule HDAC inhibitors of differing selectivity to ablate the residual HDAC activity that remained after (sh)RNA depletion, the effect of depletion of HDAC3 was further enhanced. Enzymatic inhibition of HDAC3 with the selective small-molecule inhibitor BRD3308 activated HIV-1 transcription in the 2D10 cell line. Furthermore, ex vivo exposure to BRD3308 induced outgrowth of HIV-1 from resting CD4+ T cells isolated from antiretroviral-treated, aviremic HIV+ patients. Taken together these findings suggest that HDAC3 is an essential target to disrupt HIV-1 latency, and inhibition of HDAC2 may also contribute to the effort to purge and eradicate latent HIV-1 infection.


Asunto(s)
Fármacos Anti-VIH/farmacología , Linfocitos T CD4-Positivos/efectos de los fármacos , VIH-1/fisiología , Histona Desacetilasa 1/genética , Histona Desacetilasa 2/genética , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/genética , Linfocitos T CD4-Positivos/enzimología , Linfocitos T CD4-Positivos/virología , Línea Celular Transformada , Eliminación de Gen , Regulación de la Expresión Génica , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/enzimología , Infecciones por VIH/genética , Infecciones por VIH/virología , VIH-1/efectos de los fármacos , VIH-1/patogenicidad , Histona Desacetilasa 1/antagonistas & inhibidores , Histona Desacetilasa 1/deficiencia , Histona Desacetilasa 2/antagonistas & inhibidores , Histona Desacetilasa 2/deficiencia , Histona Desacetilasas/deficiencia , Interacciones Huésped-Patógeno , Humanos , Regiones Promotoras Genéticas , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Latencia del Virus
11.
Haematologica ; 99(8): 1292-303, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24763403

RESUMEN

Class I histone deacetylases are critical regulators of gene transcription by erasing lysine acetylation. Targeting histone deacetylases using relative non-specific small molecule inhibitors is of major interest in the treatment of cancer, neurological disorders and acquired immune deficiency syndrome. Harnessing the therapeutic potential of histone deacetylase inhibitors requires full knowledge of individual histone deacetylases in vivo. As hematologic malignancies show increased sensitivity towards histone deacetylase inhibitors we targeted deletion of class I Hdac1 and Hdac2 to hematopoietic cell lineages. Here, we show that Hdac1 and Hdac2 together control hematopoietic stem cell homeostasis, in a cell-autonomous fashion. Simultaneous loss of Hdac1 and Hdac2 resulted in loss of hematopoietic stem cells and consequently bone marrow failure. Bone-marrow-specific deletion of Sin3a, a major Hdac1/2 co-repressor, phenocopied loss of Hdac1 and Hdac2 indicating that Sin3a-associated HDAC1/2-activity is essential for hematopoietic stem cell homeostasis. Although Hdac1 and Hdac2 show compensatory and overlapping functions in hematopoiesis, mice expressing mono-allelic Hdac1 or Hdac2 revealed that Hdac1 and Hdac2 contribute differently to the development of specific hematopoietic lineages.


Asunto(s)
Hematopoyesis/fisiología , Células Madre Hematopoyéticas/metabolismo , Histona Desacetilasa 1/deficiencia , Histona Desacetilasa 2/deficiencia , Homeostasis/fisiología , Proteínas Represoras/deficiencia , Animales , Células de la Médula Ósea/fisiología , Linaje de la Célula/fisiología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Complejo Correpresor Histona Desacetilasa y Sin3
12.
Am J Physiol Gastrointest Liver Physiol ; 306(7): G594-605, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24525021

RESUMEN

Histone deacetylases (Hdac) remove acetyl groups from proteins, influencing global and specific gene expression. Hdacs control inflammation, as shown by Hdac inhibitor-dependent protection from dextran sulfate sodium (DSS)-induced murine colitis. Although tissue-specific Hdac knockouts show redundant and specific functions, little is known of their intestinal epithelial cell (IEC) role. We have shown previously that dual Hdac1/Hdac2 IEC-specific loss disrupts cell proliferation and determination, with decreased secretory cell numbers and altered barrier function. We thus investigated how compound Hdac1/Hdac2 or Hdac2 IEC-specific deficiency alters the inflammatory response. Floxed Hdac1 and Hdac2 and villin-Cre mice were interbred. Compound Hdac1/Hdac2 IEC-deficient mice showed chronic basal inflammation, with increased basal disease activity index (DAI) and deregulated Reg gene colonic expression. DSS-treated dual Hdac1/Hdac2 IEC-deficient mice displayed increased DAI, histological score, intestinal permeability, and inflammatory gene expression. In contrast to double knockouts, Hdac2 IEC-specific loss did not affect IEC determination and growth, nor result in chronic inflammation. However, Hdac2 disruption protected against DSS colitis, as shown by decreased DAI, intestinal permeability and caspase-3 cleavage. Hdac2 IEC-specific deficient mice displayed increased expression of IEC gene subsets, such as colonic antimicrobial Reg3b and Reg3g mRNAs, and decreased expression of immune cell function-related genes. Our data show that Hdac1 and Hdac2 are essential IEC homeostasis regulators. IEC-specific Hdac1 and Hdac2 may act as epigenetic sensors and transmitters of environmental cues and regulate IEC-mediated mucosal homeostatic and inflammatory responses. Different levels of IEC Hdac activity may lead to positive or negative outcomes on intestinal homeostasis during inflammation.


Asunto(s)
Colitis/enzimología , Colon/enzimología , Células Epiteliales/enzimología , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/metabolismo , Mucosa Intestinal/enzimología , Animales , Colitis/genética , Colitis/inmunología , Colitis/patología , Colon/inmunología , Colon/patología , Sulfato de Dextran , Modelos Animales de Enfermedad , Epigénesis Genética , Células Epiteliales/inmunología , Células Epiteliales/patología , Regulación de la Expresión Génica , Genotipo , Histona Desacetilasa 1/deficiencia , Histona Desacetilasa 1/genética , Histona Desacetilasa 2/deficiencia , Histona Desacetilasa 2/genética , Homeostasis , Inmunidad Mucosa , Mediadores de Inflamación/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Ratones , Ratones Noqueados , Permeabilidad , Fenotipo , Factores de Tiempo
13.
Hepatology ; 58(6): 2089-98, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23744762

RESUMEN

UNLABELLED: Histone deacetylases 1 and 2 (HDAC1 and HDAC2) are ubiquitously expressed in tissues, including the liver, and play critical roles in numerous physiopathological processes. Little is known regarding the role of HDAC1 and HDAC2 in liver regeneration. In this study we generated mice in which Hdac1, Hdac2 or both genes were selectively knocked out in hepatocytes to investigate the role of these genes in liver regeneration following hepatic injury induced by partial hepatectomy or carbon tetrachloride administration. The loss of HDAC1 and/or HDAC2 (HDAC1/2) protein resulted in impaired liver regeneration. HDAC1/2 inactivation did not decrease hepatocytic 5-bromo-2-deoxyuridine uptake or the expression of proliferating cell nuclear antigen, cyclins, or cyclin-dependent kinases. However, the levels of Ki67, a mitotic marker that is expressed from the mid-G1 phase to the end of mitosis and is closely involved in the regulation of mitotic progression, were greatly decreased, and abnormal mitosis lacking Ki67 expression was frequently observed in HDAC1/2-deficient livers. The down-regulation of either HDAC1/2 or Ki67 in the mouse liver cancer cell line Hepa1-6 resulted in similar mitotic defects. Finally, both HDAC1 and HDAC2 proteins were associated with the Ki67 gene mediated by CCAAT/enhancer-binding protein ß. CONCLUSION: Both HDAC1 and HDAC2 play crucial roles in the regulation of liver regeneration. The loss of HDAC1/2 inhibits Ki67 expression and results in defective hepatocyte mitosis and impaired liver regeneration.


Asunto(s)
Histona Desacetilasa 1/deficiencia , Histona Desacetilasa 2/deficiencia , Antígeno Ki-67/biosíntesis , Regeneración Hepática/fisiología , Animales , Apoptosis , Intoxicación por Tetracloruro de Carbono , Línea Celular Tumoral , Regulación hacia Abajo , Hepatectomía , Masculino , Ratones , Ratones Noqueados , Mitosis
14.
Transplant Proc ; 45(2): 463-73, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23498780

RESUMEN

BACKGROUND: Hepatocytes undergo DNA synthesis shortly after liver damage triggered by partial hepatectomy (PH). This study revealed that the rate of liver damage recovery is gender-dependent. Furthermore, histone deacetylase (HDAC) epigenetic factors were discovered, at least in part, to govern the different liver recovery rates that were observed for male and female mice. MATERIALS AND METHODS: Experimental mice were divided into the following three groups: wild-type male mice, wild-type female mice, and HDAC1(flox/flox) HDAC2(flox/flox) Alb-Cre male mice. The different groups underwent a 2/3 PH surgery and were sacrificed after the PH. RESULTS: Immunohistochemical analysis showed that the peak of 5-bromo-2'-deoxyuridine and the number of proliferating cell nuclear antigen-positive cells is delayed in female livers relative to male livers. Consistent with these results, the expression of cyclin D1, cyclin-dependent kinase 2 (CDK2) and cyclin-dependent kinase 4 (CDK4) in females is lower than that in males. Western blot analysis examining HDAC1 and HDAC2 expression in the male and female liver showed the same trend as the cyclin products listed above or decreased protein expression in females relative to males. The results of immunohistochemistry and Western blot analysis of the HDAC1(flox/flox) HDAC2(flox/flox) Alb-Cre liver are consistent with the interesting phenomenon observed in the female mouse liver. Additionally, the hepatocyte proliferation inhibitor B-myc was evaluated as an HDAC1 and HDAC2 target gene. The mRNA levels of B-myc were increased in the female liver compared with the male liver. A chromatin immunoprecipitation assay showed the HDACs directly occupied the B-myc promoter. CONCLUSIONS: The processes of hepatocyte replication and liver mass reconstruction differed in male and female mice. Female subjects show a significantly delayed or decreased rate in these processes, which could be explained by differences in HDAC regulation.


Asunto(s)
Proliferación Celular , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/metabolismo , Regeneración Hepática , Hígado/enzimología , Animales , Sitios de Unión , Biomarcadores/metabolismo , Western Blotting , Inmunoprecipitación de Cromatina , Ciclina D1/metabolismo , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasa 4 Dependiente de la Ciclina/metabolismo , Femenino , Regulación de la Expresión Génica , Genotipo , Hepatectomía , Histona Desacetilasa 1/deficiencia , Histona Desacetilasa 1/genética , Histona Desacetilasa 2/deficiencia , Histona Desacetilasa 2/genética , Inmunohistoquímica , Hígado/patología , Hígado/cirugía , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Fenotipo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Mensajero/metabolismo , Factores Sexuales , Factores de Tiempo
15.
BMC Biotechnol ; 12: 71, 2012 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-23046873

RESUMEN

BACKGROUND: Gene targeting is a powerful method that can be used for examining the functions of genes. Traditionally, the construction of knockout (KO) vectors requires an amplification step to obtain two homologous, large fragments of genomic DNA. Restriction enzymes that cut at unique recognitions sites and numerous cloning steps are then carried out; this is often a time-consuming and frustrating process. RESULTS: We have developed a one-step cloning method for the insertion of two arms into a KO vector using exonuclease III. We modified an adeno-associated virus KO shuttle vector (pTK-LoxP-NEO-AAV) to yield pAAV-LIC, which contained two cassettes at the two multiple-cloning sites. The vector was digested with EcoRV to give two fragments. The two homologous arms, which had an overlap of 16 bases with the ends of the vector fragments, were amplified by polymerase chain reaction. After purification, the four fragments were mixed and treated with exonuclease III, then transformed into Escherichia coli to obtain the desired clones. Using this method, we constructed SirT1 and HDAC2 KO vectors, which were used to establish SirT1 KO cells from the colorectal cancer cell line (HCT116) and HDAC2 KO cells from the colorectal cancer cell line (DLD1). CONCLUSIONS: Our method is a fast, simple, and efficient technique for cloning, and has great potential for high-throughput construction of KO vectors.


Asunto(s)
Clonación Molecular/métodos , Marcación de Gen , Vectores Genéticos/genética , Dependovirus/genética , Escherichia coli/metabolismo , Exodesoxirribonucleasas/genética , Exodesoxirribonucleasas/metabolismo , Técnicas de Inactivación de Genes , Vectores Genéticos/metabolismo , Células HCT116 , Células HEK293 , Histona Desacetilasa 2/deficiencia , Histona Desacetilasa 2/genética , Histona Desacetilasa 2/metabolismo , Humanos , Sirtuina 1/deficiencia , Sirtuina 1/genética , Sirtuina 1/metabolismo
16.
Nature ; 483(7388): 222-6, 2012 Feb 29.
Artículo en Inglés | MEDLINE | ID: mdl-22388814

RESUMEN

Cognitive decline is a debilitating feature of most neurodegenerative diseases of the central nervous system, including Alzheimer's disease. The causes leading to such impairment are only poorly understood and effective treatments are slow to emerge. Here we show that cognitive capacities in the neurodegenerating brain are constrained by an epigenetic blockade of gene transcription that is potentially reversible. This blockade is mediated by histone deacetylase 2, which is increased by Alzheimer's-disease-related neurotoxic insults in vitro, in two mouse models of neurodegeneration and in patients with Alzheimer's disease. Histone deacetylase 2 associates with and reduces the histone acetylation of genes important for learning and memory, which show a concomitant decrease in expression. Importantly, reversing the build-up of histone deacetylase 2 by short-hairpin-RNA-mediated knockdown unlocks the repression of these genes, reinstates structural and synaptic plasticity, and abolishes neurodegeneration-associated memory impairments. These findings advocate for the development of selective inhibitors of histone deacetylase 2 and suggest that cognitive capacities following neurodegeneration are not entirely lost, but merely impaired by this epigenetic blockade.


Asunto(s)
Encéfalo/fisiopatología , Epigénesis Genética , Histona Desacetilasa 2/genética , Trastornos de la Memoria/genética , Trastornos de la Memoria/fisiopatología , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/fisiopatología , Acetilación/efectos de los fármacos , Enfermedad de Alzheimer/complicaciones , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/fisiopatología , Péptidos beta-Amiloides/toxicidad , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Epigénesis Genética/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Histona Desacetilasa 2/deficiencia , Histona Desacetilasa 2/metabolismo , Histonas/metabolismo , Humanos , Peróxido de Hidrógeno/toxicidad , Trastornos de la Memoria/complicaciones , Ratones , Enfermedades Neurodegenerativas/complicaciones , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/genética , Fragmentos de Péptidos/toxicidad , Fosforilación/efectos de los fármacos , Regiones Promotoras Genéticas/efectos de los fármacos , Regiones Promotoras Genéticas/genética , ARN Polimerasa II/metabolismo , Receptores de Glucocorticoides/metabolismo
18.
Dev Cell ; 19(6): 807-18, 2010 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-21093383

RESUMEN

Epidermal and hair follicle development from surface ectodermal progenitor cells requires coordinated changes in gene expression. Histone deacetylases alter gene expression programs through modification of chromatin and transcription factors. We find that deletion of ectodermal Hdac1 and Hdac2 results in dramatic failure of hair follicle specification and epidermal proliferation and stratification, phenocopying loss of the key ectodermal transcription factor p63. Although expression of p63 and its positively regulated basal cell targets is maintained in Hdac1/2-deficient ectoderm, targets of p63-mediated repression, including p21, 14-3-3σ, and p16/INK4a, are ectopically expressed, and HDACs bind and are active at their promoter regions in normal undifferentiated keratinocytes. Mutant embryos display increased levels of acetylated p53, which opposes p63 functions, and p53 is required for HDAC inhibitor-mediated p21 expression in keratinocytes. Our data identify critical requirements for HDAC1/2 in epidermal development and indicate that HDAC1/2 directly mediate repressive functions of p63 and suppress p53 activity.


Asunto(s)
Epidermis/embriología , Epidermis/metabolismo , Histona Desacetilasa 1/fisiología , Histona Desacetilasa 2/fisiología , Fosfoproteínas/fisiología , Transactivadores/fisiología , Proteína p53 Supresora de Tumor/fisiología , Acetilación , Animales , Apoptosis/genética , Apoptosis/fisiología , Secuencia de Bases , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/deficiencia , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/fisiología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Cartilla de ADN/genética , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Epidermis/anomalías , Femenino , Regulación del Desarrollo de la Expresión Génica , Folículo Piloso/anomalías , Folículo Piloso/embriología , Folículo Piloso/metabolismo , Histona Desacetilasa 1/deficiencia , Histona Desacetilasa 1/genética , Histona Desacetilasa 2/deficiencia , Histona Desacetilasa 2/genética , Humanos , Queratinocitos/citología , Queratinocitos/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Modelos Biológicos , Fosfoproteínas/genética , Embarazo , Distribución Tisular , Transactivadores/genética , Proteína p53 Supresora de Tumor/genética , Regulación hacia Arriba
19.
EMBO J ; 29(15): 2586-97, 2010 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-20571512

RESUMEN

Histone deacetylases (HDACs) counterbalance acetylation of lysine residues, a protein modification involved in numerous biological processes. Here, Hdac1 and Hdac2 conditional knock-out alleles were used to study the function of class I Hdac1 and Hdac2 in cell cycle progression and haematopoietic differentiation. Combined deletion of Hdac1 and Hdac2, or inactivation of their deacetylase activity in primary or oncogenic-transformed fibroblasts, results in a senescence-like G(1) cell cycle arrest, accompanied by up-regulation of the cyclin-dependent kinase inhibitor p21(Cip). Notably, concomitant genetic inactivation of p53 or p21(Cip) indicates that Hdac1 and Hdac2 regulate p53-p21(Cip)-independent pathways critical for maintaining cell cycle progression. In vivo, we show that Hdac1 and Hdac2 are not essential for liver homeostasis. In contrast, total levels of Hdac1 and Hdac2 in the haematopoietic system are critical for erythrocyte-megakaryocyte differentiation. Dual inactivation of Hdac1 and Hdac2 results in apoptosis of megakaryocytes and thrombocytopenia. Together, these data indicate that Hdac1 and Hdac2 have overlapping functions in cell cycle regulation and haematopoiesis. In addition, this work provides insights into mechanism-based toxicities observed in patients treated with HDAC inhibitors.


Asunto(s)
Ciclo Celular , Hematopoyesis , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/metabolismo , Anemia/enzimología , Animales , Apoptosis , Biocatálisis , Células Cultivadas , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/deficiencia , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Histona Desacetilasa 1/deficiencia , Histona Desacetilasa 2/deficiencia , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Trombocitopenia/enzimología , Trombocitopenia/patología , Proteína p53 Supresora de Tumor/metabolismo
20.
Proc Natl Acad Sci U S A ; 107(18): 8242-7, 2010 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-20404188

RESUMEN

Histone deacetylases (HDAC) 1 and 2 are highly similar enzymes that help regulate chromatin structure as the core catalytic components of corepressor complexes. Although tissue-specific deletion of HDAC1 and HDAC2 has demonstrated functional redundancy, germ-line deletion of HDAC1 in the mouse causes early embryonic lethality, whereas HDAC2 does not. To address the unique requirement for HDAC1 in early embryogenesis we have generated conditional knockout embryonic stem (ES) cells in which HDAC1 or HDAC2 genes can be inactivated. Deletion of HDAC1, but not HDAC2, causes a significant reduction in the HDAC activity of Sin3A, NuRD, and CoREST corepressor complexes. This reduced corepressor activity results in a specific 1.6-fold increase in histone H3 K56 acetylation (H3K56Ac), thus providing genetic evidence that H3K56Ac is a substrate of HDAC1. In culture, ES cell proliferation was unaffected by loss of either HDAC1 or HDAC2. Rather, we find that loss of HDAC1 affects ES cell differentiation. ES cells lacking either HDAC1 or HDAC2 were capable of forming embryoid bodies (EBs), which stimulates differentiation into the three primary germ layers. However, HDAC1-deficient EBs were significantly smaller, showed spontaneous rhythmic contraction, and increased expression of both cardiomyocyte and neuronal markers. In summary, our genetic study of HDAC1 and HDAC2 in ES cells, which mimic the embryonic epiblast, has identified a unique requirement for HDAC1 in the optimal activity of HDAC1/2 corepressor complexes and cell fate determination during differentiation.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/citología , Células Madre Embrionarias/enzimología , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/metabolismo , Animales , Biomarcadores , Línea Celular , Proliferación Celular , Regulación Enzimológica de la Expresión Génica , Histona Desacetilasa 1/deficiencia , Histona Desacetilasa 2/deficiencia , Ratones , Unión Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA