Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cells ; 10(2)2021 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-33498747

RESUMO

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.


Assuntos
Células Epiteliais/metabolismo , Histona Desacetilase 1/deficiência , Histona Desacetilase 2/deficiência , Homeostase , Intestinos/citologia , Janus Quinases/metabolismo , Fatores de Transcrição STAT/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Deleção de Genes , Histona Desacetilase 1/genética , Histona Desacetilase 1/metabolismo , Histona Desacetilase 2/genética , Histona Desacetilase 2/metabolismo , Homeostase/efeitos dos fármacos , Contagem de Linfócitos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Organoides/efeitos dos fármacos , Organoides/crescimento & desenvolvimento , Celulas de Paneth/efeitos dos fármacos , Celulas de Paneth/metabolismo , Piperidinas/farmacologia , Pirimidinas/farmacologia , Linfócitos T/efeitos dos fármacos
2.
J Pharmacol Exp Ther ; 370(3): 490-503, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31308194

RESUMO

Valproic acid (VPA) has been shown to regulate the levels of brain-derived neurotrophic factor (BDNF), but it is not known whether this drug can affect the neuronal responses to BDNF. In the present study, we show that in retinoic acid-differentiated SH-SY5Y human neuroblastoma cells, prolonged exposure to VPA reduces the expression of the BDNF receptor TrkB at the protein and mRNA levels and inhibits the intracellular signaling, neurotrophic activity, and prosurvival function of BDNF. VPA downregulates TrkB and curtails BDNF-induced signaling also in differentiated Kelly and LAN-1 neuroblastoma cells and primary mouse cortical neurons. The VPA effect is mimicked by several histone deacetylase (HDAC) inhibitors, including the class I HDAC inhibitors entinostat and romidepsin. Conversely, the class II HDAC inhibitor MC1568, the HDAC6 inhibitor tubacin, the HDAC8 inhibitor PCI-34051, and the VPA derivative valpromide have no effect. In neuroblastoma cells and primary neurons both VPA and entinostat increase the cellular levels of the transcription factor RUNX3, which negatively regulates TrkB gene expression. Treatment with RUNX3 siRNA attenuates VPA-induced RUNX3 elevation and TrkB downregulation. VPA, entinostat, HDAC1 depletion by siRNA, and 3-deazaneplanocin A (DZNep), an inhibitor of the polycomb repressor complex 2 (PRC2), decrease the PRC2 core component EZH2, a RUNX3 suppressor. Like VPA, HDAC1 depletion and DZNep increase RUNX3 and decrease TrkB expression. These results indicate that VPA downregulates TrkB through epigenetic mechanisms involving the EZH2/RUNX3 axis and provide evidence that this effect implicates relevant consequences with regard to BDNF efficacy in stimulating intracellular signaling and functional responses. SIGNIFICANCE STATEMENT: The tropomyosin-related kinase receptor B (TrkB) mediates the stimulatory effects of brain-derived neurotrophic factor (BDNF) on neuronal growth, differentiation, and survival and is highly expressed in aggressive neuroblastoma and other tumors. Here we show that exposure to valproic acid (VPA) downregulates TrkB expression and functional activity in retinoic acid-differentiated human neuroblastoma cell lines and primary mouse cortical neurons. The effects of VPA are mimicked by other histone deacetylase (HDAC) inhibitors and HDAC1 knockdown and appear to be mediated by an epigenetic mechanism involving the upregulation of RUNX3, a suppressor of TrkB gene expression. TrkB downregulation may have relevance for the use of VPA as a potential therapeutic agent in neuroblastoma and other pathologies characterized by an excessive BDNF/TrkB signaling.


Assuntos
Regulação para Baixo/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Receptor trkB/genética , Receptor trkB/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ácido Valproico/farmacologia , Animais , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Subunidade alfa 3 de Fator de Ligação ao Core/genética , Ativação Enzimática/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Histona Desacetilase 1/deficiência , Histona Desacetilase 1/genética , Humanos , Camundongos , Neurônios/efeitos dos fármacos , Neurônios/patologia
3.
Cell Commun Signal ; 17(1): 86, 2019 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-31358016

RESUMO

OBJECTIVE: This study aimed to investigate the function and mechanism of neddylation of HDAC1 underlying drug resistance of AML cells. METHODS: Evaluation experiments of effects of HDAC1 on drug resistance of AML cells were performed with AML cell transfected with constructs overexpressing HDAC1 or multi-drug resistance AML cells transfected with siRNA for HDAC1 through observing cell viability, percentage of apoptotic cell, doxorubicin-releasing index and multidrug resistance associated protein 1 (MRP1) expression. Neddylation or ubiquitination of HDAC1 was determined by immunoprecipitation or Ni2+ pull down assay followed by western blot. The role of HDAC1 was in vivo confirmed by xenograft in mice. RESULTS: HDAC1 was significantly upregulated in refractory AML patients, and in drug-resistant AML cells (HL-60/ADM and K562/A02). Intracellular HDAC1 expression promoted doxorubicin resistance of HL-60, K562, and primary bone marrow cells (BMCs) of remission AML patients as shown by increasing cell viability and doxorubicin-releasing index, inhibiting cell apoptosis. Moreover, HDAC1 protein level in AML cells was regulated by the Nedd8-mediated neddylation and ubiquitination, which further promoted HDAC1 degradation. In vivo, HDAC1 overexpression significantly increased doxorubicin resistance; while HDACs inhibitor Panobinostat markedly improved the inhibitory effect of doxorubicin on tumor growth. Furthermore, HDAC1 silencing by Panobinostat and/or lentivirus mediated RNA interference against HDAC1 effectively reduced doxorubicin resistance, resulting in the inhibition of tumor growth in AML bearing mice. CONCLUSION: Our findings suggested that HDAC1 contributed to the multidrug resistance of AML and its function turnover was regulated, at least in part, by post-translational modifications, including neddylation and ubiquitination.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Histona Desacetilase 1/biossíntese , Histona Desacetilase 1/metabolismo , Leucemia Mieloide Aguda/patologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Adulto , Idoso , Animais , Linhagem Celular Tumoral , Doxorrubicina/farmacologia , Indução Enzimática/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Histona Desacetilase 1/deficiência , Histona Desacetilase 1/genética , Humanos , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Leucemia Mieloide Aguda/tratamento farmacológico , Masculino , Camundongos , Pessoa de Meia-Idade , Proteína NEDD8/metabolismo , Ubiquitinação/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto , Adulto Jovem
4.
Stem Cell Reports ; 10(4): 1369-1383, 2018 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-29641990

RESUMO

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.


Assuntos
Células Endoteliais/citologia , Células Endoteliais/metabolismo , Hematopoese , Histona Desacetilase 1/metabolismo , Histona Desacetilase 2/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Animais , Benzamidas/farmacologia , Diferenciação Celular/efeitos dos fármacos , Dioxóis/farmacologia , Células Endoteliais/efeitos dos fármacos , Deleção de Genes , Hemangioblastos/citologia , Hematopoese/efeitos dos fármacos , Histona Desacetilase 1/deficiência , Histona Desacetilase 2/deficiência , Inibidores de Histona Desacetilases/farmacologia , Camundongos , Transdução de Sinais/efeitos dos fármacos
5.
Biol Chem ; 399(6): 603-610, 2018 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-29537214

RESUMO

Non-small cell lung cancer (NSCLC) is a common malignant tumor. Although the abnormal expression and potential clinical prognostic value of histone deacetylase 1 (HDAC1) were recently discovered in many kinds of cancer, the roles and molecular mechanisms of HDAC1 in NSCLC is still limited. The CCK-8 assay is used to evaluate the viability of NSCLC cells. Downregulation of HDAC1 by shRNA. The TUNEL assay was used to evaluate the role of HDAC1 in NSCLC apoptosis. To evaluate the role of HDAC1 in NSCLC cells migration, the Boyden chamber transwell assay and wound healing assay were used. To evaluate the cells invasion, the matrigel precoated Transwell assay was used. Enzyme-linked immunosorbent assays (ELISAs) were used to detect the level of vascular endothelial growth factor (VEGF) and IL-8 in NSCLC. To investigate the role of HDAC1 in angiogenesis, the tube formation assay was investigated. In this study, we showed that HDAC1 expression was elevated in NSCLC lines compared to that in normal liver cells LO2. Furthermore, downregulation of HDAC1 inhibited cell proliferation, prevented cell migration, decreased cell invasion, reduced tumor angiogenesis and induced cell apoptosis. In summary, HDAC1 may be regarded as a potential indicator for NSCLC patient treatment.


Assuntos
Apoptose/genética , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Histona Desacetilase 1/deficiência , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Invasividade Neoplásica/genética , Sobrevivência Celular , Regulação para Baixo , Técnicas de Silenciamento de Genes , Humanos , Células Tumorais Cultivadas , Cicatrização
6.
Antioxid Redox Signal ; 28(13): 1224-1237, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29113455

RESUMO

AIMS: Iron-overload disorders are common and could lead to significant morbidity and mortality worldwide. Due to limited treatment options, there is a great need to develop novel strategies to remove the excess body iron. To discover potential epigenetic modulator in hepcidin upregulation and subsequently decreasing iron burden, we performed an epigenetic screen. The in vivo effects of the identified compounds were further tested in iron-overload mouse models, including Hfe-/-, Hjv-/-, and hepatocyte-specific Smad4 knockout (Smad4fl/fl;Alb-Cre+) mice. RESULTS: Entinostat (MS-275), the clinical used histone deacetylase 1 (HDAC1) inhibitor, was identified the most potent hepcidin agonist. Consistently, Hdac1-deficient mice also presented higher hepcidin levels than wild-type controls. Notably, the long-term treatment with entinostat in Hfe-/- mice significantly alleviated iron overload through upregulating hepcidin transcription. In contrast, entinostat showed no effect on hepcidin expression and iron levels in Smad4fl/fl;Alb-Cre+ mice. Further mechanistic studies revealed that HDAC1 suppressed expression of hepcidin through interacting with SMAD4 rather than deacetylation of SMAD4 or histone-H3 on the hepcidin promoter. INNOVATION: The findings uncovered HDAC1 as a novel hepcidin suppressor through complexing with SMAD4 but not deacetylation of either histone 3 or SMAD4. In addition, our study suggested a novel implication of entinostat in treating iron-overload disorders. CONCLUSIONS: Based on our results, we conclude that entinostat strongly activated hepcidin in vivo and in vitro. HDAC1 could serve as a novel hepcidin suppressor by binding to SMAD4, effect of which is independent of BMP/SMAD1/5/8 signaling. Antioxid. Redox Signal. 28, 1224-1237.


Assuntos
Modelos Animais de Doenças , Histona Desacetilase 1/metabolismo , Histonas/metabolismo , Homeostase , Sobrecarga de Ferro/metabolismo , Ferro/metabolismo , Acetilação , Animais , Benzamidas/farmacologia , Relação Dose-Resposta a Droga , Histona Desacetilase 1/antagonistas & inibidores , Histona Desacetilase 1/deficiência , Inibidores de Histona Desacetilases/farmacologia , Homeostase/efeitos dos fármacos , Ferro/efeitos adversos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Piridinas/farmacologia , Relação Estrutura-Atividade
7.
Biol Chem ; 398(12): 1347-1356, 2017 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-28779562

RESUMO

Targeted inhibition of histone deacetylase (HDAC) is one of the potent anticancer therapy approaches. Our data showed that mRNA and protein levels of HDAC1 in breast cancer cells were greater than that in normal fibroblast 3T3 cells and normal epithelial breast MCF10A cells. The mRNA levels of HDAC1 in 75% of breast cancer tissues (18/24) were greater than that in their corresponding adjacent normal tissues. Knockdown of HDAC1 by specific siRNAs can suppress the proliferation and migration of breast cancer cells and inhibit the expression of interleukin-8 (IL-8), while not IL-6. While recombinant IL-8 (rIL-8) can attenuate the suppression effects of si-HDAC1 on the proliferation and migration of breast cancer cells. HDAC1 can positively regulate the transcription and promoter activities of IL-8. While NF-κB and MAPK, two important signals responsible for the transcription of IL-8, did not mediate HDAC1 regulated IL-8 expression. The expression and nuclear translocation of Snail were increased in HDAC1 over expressed breast cancer cells. Targeted inhibition of Snail can attenuate HDAC1 over expression induced cell proliferation and migration. Collectively, our data showed that HDAC1 can trigger the proliferation and migration of breast cancer cells via activation of Snail/IL-8 signals.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Movimento Celular , Histona Desacetilase 1/metabolismo , Interleucina-8/metabolismo , Regulação para Cima , Células 3T3 , Animais , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Histona Desacetilase 1/deficiência , Histona Desacetilase 1/genética , Humanos , Camundongos , RNA Interferente Pequeno/genética , Transdução de Sinais/genética
8.
J Pharmacol Exp Ther ; 363(1): 1-11, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28798030

RESUMO

γ-Aminobutyric acid A receptors (GABAA-Rs) mediate the majority of inhibitory neurotransmission in the adult brain. The α1-containing GABAA-Rs are the most prominent subtype in the adult brain and are important in both homeostatic function and several disease pathologies including alcohol dependence, epilepsy, and stress. Ethanol exposure causes a decrease of α1 transcription and peptide expression both in vivo and in vitro, but the mechanism that controls the transcriptional regulation is unknown. Because ethanol is known to activate epigenetic regulation of gene expression, we tested the hypothesis that ethanol regulates α1 expression through histone modifications in cerebral cortical cultured neurons. We found that class I histone deacetylases (HDACs) regulate ethanol-induced changes in α1 gene and protein expression as pharmacologic inhibition or knockdown of HDAC1-3 prevents the effects of ethanol exposure. Targeted histone acetylation associated with the Gabra1 promoter using CRISPR (clustered regularly interspaced palindromic repeat) dCas9-P300 (a nuclease-null Cas9 fused with a histone acetyltransferase) increases histone acetylation and prevents the decrease of Gabra1 expression. In contrast, there was no effect of a mutant histone acetyltransferase or generic transcriptional activator or targeting P300 to a distant exon. Conversely, using a dCas9-KRAB construct that increases repressive methylation (H3K9me3) does not interfere with ethanol-induced histone deacetylation. Overall our results indicate that ethanol deacetylates histones associated with the Gabra1 promoter through class I HDACs and that pharmacologic, genetic, or epigenetic intervention prevents decreases in α1 expression in cultured cortical neurons.


Assuntos
Córtex Cerebral/citologia , Etanol/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Histonas/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Regiões Promotoras Genéticas/genética , Receptores de GABA-A/genética , Acetilação/efeitos dos fármacos , Animais , Feminino , Técnicas de Silenciamento de Genes , Histona Desacetilase 1/antagonistas & inibidores , Histona Desacetilase 1/deficiência , Histona Desacetilase 1/genética , Inibidores de Histona Desacetilases/farmacologia , Masculino , Ratos , Ratos Sprague-Dawley
9.
J Am Heart Assoc ; 6(7)2017 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-28679560

RESUMO

BACKGROUND: Cardiac mesenchymal cell (CMC) administration improves cardiac function in animal models of heart failure. Although the precise mechanisms remain unclear, transdifferentiation and paracrine signaling are suggested to underlie their cardiac reparative effects. We have shown that histone deacetylase 1 (HDAC1) inhibition enhances CMC cardiomyogenic lineage commitment. Here, we investigated the impact of HDAC1 on CMC cytokine secretion and associated paracrine-mediated activities on endothelial cell function. METHODS AND RESULTS: CMCs were transduced with shRNA constructs targeting HDAC1 (shHDAC1) or nontarget (shNT) control. Cytokine arrays were used to assess the expression of secreted proteins in conditioned medium (CM) from shHDAC1 or shNT-transduced CMCs. In vitro functional assays for cell proliferation, protection from oxidative stress, cell migration, and tube formation were performed on human endothelial cells incubated with CM from the various treatment conditions. CM from shHDAC1-transduced CMCs contained more cytokines involved in cell growth/differentiation and more efficiently promoted endothelial cell proliferation and tube formation compared with CM from shNT. After evaluating key cytokines previously implicated in cell-therapy-mediated cardiac repair, we found that basic fibroblast growth factor was significantly upregulated in shHDAC1-transduced CMCs. Furthermore, shRNA-mediated knockdown of basic fibroblast growth factor in HDAC1-depleted CMCs inhibited the effects of shHDAC1 CM in promoting endothelial proliferation and tube formation-indicating that HDAC1 depletion activates CMC proangiogenic paracrine signaling in a basic fibroblast growth factor-dependent manner. CONCLUSIONS: These results reveal a hitherto unknown role for HDAC1 in the modulation of CMC cytokine secretion and implicate the targeted inhibition of HDAC1 in CMCs as a means to enhance paracrine-mediated neovascularization in cardiac cell therapy applications.


Assuntos
Proteínas Angiogênicas/biossíntese , Fator 2 de Crescimento de Fibroblastos/biossíntese , Coração , Histona Desacetilase 1/deficiência , Células Endoteliais da Veia Umbilical Humana/metabolismo , Células-Tronco Mesenquimais/enzimologia , Miócitos Cardíacos/enzimologia , Neovascularização Fisiológica , Comunicação Parácrina , Proteínas Angiogênicas/metabolismo , Diferenciação Celular , Linhagem da Célula , Movimento Celular , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Meios de Cultivo Condicionados/metabolismo , Citocinas/metabolismo , Repressão Enzimática , Fator 2 de Crescimento de Fibroblastos/metabolismo , Coração/metabolismo , Histona Desacetilase 1/genética , Humanos , Células-Tronco Mesenquimais/metabolismo , Miócitos Cardíacos/metabolismo , Estresse Oxidativo , Transdução de Sinais , Fatores de Tempo , Transdução Genética , Transfecção
10.
Blood ; 130(2): 146-155, 2017 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-28550044

RESUMO

Histone acetylation and the families of enzymes responsible for controlling these epigenetic marks have been implicated in regulating T-cell maturation and phenotype. Here, we demonstrate a previously undefined role of histone deacetylase 11 (HDAC11) in regulating T-cell effector functions. Using EGFP-HDAC11 transgenic reporter mice, we found that HDAC11 expression was lower in effector relative to naive and central memory T-cell populations, and activation of resting T cells resulted in its decreased expression. Experiments using HDAC11 knockout (KO) mice revealed that T cells from these mice displayed enhanced proliferation, proinflammatory cytokine production, and effector molecule expression. In addition, HDAC11KO T cells had increased expression of Eomesodermin (Eomes) and TBX21 (Tbet), transcription factors previously shown to regulate inflammatory cytokine and effector molecule production. Conversely, overexpression of HDAC11 resulted in decreased expression of these genes. Chromatin immunoprecipitation showed the presence of HDAC11 at the Eomes and Tbet gene promoters in resting T cells, where it rapidly disassociated following T-cell activation. In vivo, HDAC11KO T cells were refractory to tolerance induction. HDAC11KO T cells also mediated accelerated onset of acute graft-versus-host disease (GVHD) in a murine model, characterized by increased proliferation of T cells and expression of interferon-γ, tumor necrosis factor, and EOMES. In addition, adoptive transfer of HDAC11KO T cells resulted in significantly reduced tumor burden in a murine B-cell lymphoma model. Taken together, these data demonstrate a previously unknown role of HDAC11 as a negative epigenetic regulator of T-cell effector phenotype and function.


Assuntos
Regulação Neoplásica da Expressão Gênica , Doença Enxerto-Hospedeiro/imunologia , Histona Desacetilase 1/genética , Linfoma de Células B/imunologia , Proteínas com Domínio T/genética , Linfócitos T/imunologia , Transferência Adotiva , Animais , Linfócitos B/imunologia , Linfócitos B/patologia , Cromatina/química , Cromatina/metabolismo , Imunoprecipitação da Cromatina , Modelos Animais de Doenças , Doença Enxerto-Hospedeiro/genética , Doença Enxerto-Hospedeiro/patologia , Histona Desacetilase 1/deficiência , Histona Desacetilase 1/imunologia , Interferon gama/genética , Interferon gama/imunologia , Ativação Linfocitária , Linfoma de Células B/genética , Linfoma de Células B/patologia , Camundongos , Camundongos Knockout , Regiões Promotoras Genéticas , Transdução de Sinais , Proteínas com Domínio T/imunologia , Linfócitos T/patologia , Linfócitos T/transplante , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/imunologia
11.
Nat Commun ; 8: 14272, 2017 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-28139683

RESUMO

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.


Assuntos
Benzamidas/farmacologia , Histona Desacetilase 1/genética , Histona Desacetilase 2/genética , Inibidores de Histona Desacetilases/farmacologia , Regeneração Nervosa/efeitos dos fármacos , Traumatismos dos Nervos Periféricos/tratamento farmacológico , Pirimidinas/farmacologia , Células de Schwann/efeitos dos fármacos , Animais , Axônios/efeitos dos fármacos , Axônios/metabolismo , Proteína 2 de Resposta de Crescimento Precoce/genética , Proteína 2 de Resposta de Crescimento Precoce/metabolismo , Regulação da Expressão Gênica , Genes Reporter , Histona Desacetilase 1/antagonistas & inibidores , Histona Desacetilase 1/deficiência , Histona Desacetilase 2/antagonistas & inibidores , Histona Desacetilase 2/deficiência , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Luciferases/genética , Luciferases/metabolismo , Camundongos , Camundongos Knockout , Regeneração Nervosa/genética , Fator de Transcrição PAX3/genética , Fator de Transcrição PAX3/metabolismo , Traumatismos dos Nervos Periféricos/genética , Traumatismos dos Nervos Periféricos/metabolismo , Traumatismos dos Nervos Periféricos/patologia , Recuperação de Função Fisiológica/efeitos dos fármacos , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXB1/metabolismo , Células de Schwann/metabolismo , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
12.
Methods Mol Biol ; 1510: 169-192, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27761821

RESUMO

Histone deacetylases (HDACs) play crucial roles during mammalian development and for cellular homeostasis. In addition, these enzymes are promising targets for small molecule inhibitors in the treatment of cancer and neurological diseases. Conditional HDAC knock-out mice are excellent tools for defining the functions of individual HDACs in vivo and for identifying the molecular targets of HDAC inhibitors in disease. Here, we describe the generation of tissue-specific HDAC knock-out mice and delineate a strategy for the generation of conditional HDAC knock-in mice.


Assuntos
Blastocisto/enzimologia , Cromatina/metabolismo , Epigênese Genética , Vetores Genéticos/metabolismo , Histona Desacetilase 1/genética , Células-Tronco Embrionárias Murinas/enzimologia , Animais , Blastocisto/citologia , Southern Blotting , Sistemas CRISPR-Cas , Cromatina/química , Cromossomos Artificiais Bacterianos/química , Cromossomos Artificiais Bacterianos/metabolismo , Cruzamentos Genéticos , Feminino , Técnicas de Introdução de Genes , Vetores Genéticos/química , Histona Desacetilase 1/deficiência , Recombinação Homóloga , Integrases/genética , Integrases/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células-Tronco Embrionárias Murinas/citologia , Especificidade de Órgãos , RNA Guia de Cinetoplastídeos/genética , RNA Guia de Cinetoplastídeos/metabolismo
13.
Methods Mol Biol ; 1510: 193-209, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27761822

RESUMO

The protein sequences of class I HDACs in mice and humans are 96-99 % identical. These highly conserved proteins have crucial roles in biological processes, such as proliferation and development, which is reflected in the lethality that occurs in conventional whole body knockout mice. Therefore, conditional knockouts are inevitable to investigate the functions of class I HDACs in mice. Here, we describe the generation of conditional class I Hdac knockout mice, using Hdac1 as an example. We explain a relatively quick procedure to generate the necessary target vectors by recombination-mediated genetic engineering and gateway techniques. Furthermore, we show how to culture, target, and screen for positively recombined ES cells. Additionally, we present a dual recombination system, which allows the deletion of class I Hdacs at any time by a tamoxifen inducible Cre.


Assuntos
Blastocisto/enzimologia , Epigênese Genética , Vetores Genéticos/metabolismo , Histona Desacetilase 1/genética , Recombinação Homóloga , Células-Tronco Embrionárias Murinas/enzimologia , Animais , Blastocisto/citologia , Southern Blotting , Cruzamentos Genéticos , Feminino , Vetores Genéticos/química , Proteínas de Choque Térmico HSP90/genética , Proteínas de Choque Térmico HSP90/metabolismo , Histona Desacetilase 1/deficiência , Integrases/genética , Integrases/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células-Tronco Embrionárias Murinas/citologia , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Tamoxifeno/farmacologia
14.
PLoS One ; 11(12): e0167554, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27907135

RESUMO

Epigenetic regulation in starvation is important but not fully understood yet. Here we identified the Rpd3 gene, a Drosophila homolog of histone deacetylase 1, as a critical epigenetic regulator for acquiring starvation stress resistance. Immunostaining analyses of Drosophila fat body revealed that the subcellular localization and levels of Rpd3 dynamically changed responding to starvation stress. In response to starvation stress, the level of Rpd3 rapidly increased, and it accumulated in the nucleolus in what appeared to be foci. These observations suggest that Rpd3 plays a role in regulation of rRNA synthesis in the nucleolus. The RT-qPCR and ChIP-qPCR analyses clarified that Rpd3 binds to the genomic region containing the rRNA promoters and activates rRNA synthesis in response to starvation stress. Polysome analyses revealed that the amount of polysomes was decreased in Rpd3 knockdown flies under starvation stress compared with the control flies. Since the autophagy-related proteins are known to be starvation stress tolerance proteins, we examined autophagy activity, and it was reduced in Rpd3 knockdown flies. Taken together, we conclude that Rpd3 accumulates in the nucleolus in the early stage of starvation, upregulates rRNA synthesis, maintains the polysome amount for translation, and finally increases stress tolerance proteins, such as autophagy-related proteins, to acquire starvation stress resistance.


Assuntos
Adaptação Fisiológica/genética , Proteínas Relacionadas à Autofagia/genética , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Epigênese Genética , Histona Desacetilase 1/genética , Inanição/genética , Animais , Autofagia , Proteínas Relacionadas à Autofagia/metabolismo , Nucléolo Celular/metabolismo , Nucléolo Celular/ultraestrutura , Proteínas de Drosophila/deficiência , Drosophila melanogaster/metabolismo , Corpo Adiposo/metabolismo , Corpo Adiposo/ultraestrutura , Técnicas de Silenciamento de Genes , Histona Desacetilase 1/deficiência , Histonas/genética , Histonas/metabolismo , Polirribossomos/química , Polirribossomos/metabolismo , Regiões Promotoras Genéticas , RNA Ribossômico/biossíntese , RNA Ribossômico/genética , Estresse Fisiológico/genética , Análise de Sobrevida
15.
J Neurochem ; 139(3): 369-380, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27534825

RESUMO

During early postnatal development, neuronal circuits are sculpted by sensory experience provided by the external environment. This experience-dependent regulation of circuitry development consolidates the balance of excitatory-inhibitory (E/I) neurons in the brain. The cortical barrel-column that innervates a single principal whisker is used to provide a clear reference frame for studying the consolidation of E/I circuitry. Sensory deprivation of S1 at birth disrupts the consolidation of excitatory-inhibitory balance by decreasing inhibitory transmission of parvalbumin interneurons. The molecular mechanisms underlying this decrease in inhibition are not completely understood. Our findings show that epigenetic mechanisms, in particular histone deacetylation by histone deacetylases, negatively regulate the expression of brain-derived neurotrophic factor (Bdnf) and parvalbumin (Pvalb) genes during development, which are required for the maturation of parvalbumin interneurons. After whisker deprivation, increased histone deacetylase 1 expression and activity led to increased histone deacetylase 1 binding and decreased histone acetylation at Bdnf promoters I-IV and Pvalb promoter, resulting in the repression of Bdnf and Pvalb gene transcription. The decrease in Bdnf expression further affected parvalbumin interneuron maturation at layer II/III in S1, demonstrated by decreased parvalbumin expression, a marker for parvalbumin interneuron maturation. Knockdown of HDAC1 recovered Bdnf and Pvalb gene transcription and also prevented the decrease of inhibitory synapses accompanying whisker deprivation.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Histona Desacetilase 1/metabolismo , Interneurônios/metabolismo , Parvalbuminas/metabolismo , Parvalbuminas/fisiologia , Animais , Animais Recém-Nascidos , Fator Neurotrófico Derivado do Encéfalo/genética , Epigênese Genética/fisiologia , Histona Desacetilase 1/deficiência , Histona Desacetilase 1/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Vias Neurais/crescimento & desenvolvimento , Parvalbuminas/genética , Células Piramidais/metabolismo , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Sensação , Privação Sensorial , Córtex Somatossensorial/crescimento & desenvolvimento , Córtex Somatossensorial/fisiologia , Vibrissas/inervação , Vibrissas/fisiologia
16.
Med Sci Monit ; 22: 1291-6, 2016 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-27086779

RESUMO

BACKGROUND HDAC1 has been shown to be closely associated with the occurrence of tumors. We aimed to investigate the effects of siRNA-mediated HDAC1 knockdown on the biological behavior of esophageal carcinoma cell lines. MATERIAL AND METHODS HDAC1 expression in esophageal cancer cell lines TE-1, Eca109, and EC9706 was compared by Western blot analysis. These cells were transfected with siRNA-HDAC1 and cell proliferation was evaluated by MTT assay to select the optimum cell line for subsequent experiments. The effects of siRNA-HDAC1 on the migration and invasion of the selected cell line were assessed by transwell assay. The expression of cell cycle-related proteins cyclinD1, p21 and p27, and epithelial-mesenchymal transition (EMT)-related protein zonula occludens-1 (ZO-1), E-cadherin and vimentin was determined by Western blot analysis. RESULTS HDAC1 expression in TE-1, Eca109 and EC9706 cells was significantly higher compared with normal esophageal cell line HEEC (P<0.01). MTT assay, Western blot and RT-PCR analyses demonstrated that the inhibitory effects of siRNA on HDAC1 expression and cell viability in TE-1 cells were the highest among all cell lines, which was therefore used in subsequent experiments. After TE-1 cells were transfected with siRNA-HDAC1, their migration and invasion were significantly lower compared with the controls (P<0.01). CyclinD1 and vimentin expression was significantly lower compared with the controls (P<0.01), whereas the expression of p21, p27, ZO-1 and E-cadherin was significantly higher (P<0.01). CONCLUSIONS The siRNA-mediated HDAC1 knockdown significantly inhibited the proliferation, migration and invasion of TE-1 cells probably by regulating the expression of cell cycle- and EMT-related proteins.


Assuntos
Carcinoma de Células Escamosas/enzimologia , Neoplasias Esofágicas/enzimologia , Neoplasias Esofágicas/patologia , Histona Desacetilase 1/deficiência , Histona Desacetilase 1/genética , Apoptose/fisiologia , Western Blotting , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Regulação para Baixo , Neoplasias Esofágicas/genética , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Histona Desacetilase 1/metabolismo , Humanos , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Transfecção
17.
Exp Gerontol ; 86: 124-128, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-26927903

RESUMO

The epigenetic regulation of DNA structure and function is essential for changes in gene expression involved in development, growth, and maintenance of cellular function. Epigenetic changes include histone modifications such as methylation, acetylation, ubiquitination, and phosphorylation. Histone deacetylase (HDAC) proteins have a major role in epigenetic regulation of chromatin structure. HDACs are enzymes that catalyze the removal of acetyl groups from lysine residues within histones, as well as a range of other proteins including transcriptional factors. HDACs are highly conserved proteins divided into two families and based on sequence similarity in four classes. Here we will discuss the roles of Rpd3 in physiology and longevity with emphasis on its role in flies. Rpd3, the Drosophila HDAC1 homolog, is a class I lysine deacetylase and a member of a large family of HDAC proteins. Rpd3 has multiple functions including control of proliferation, development, metabolism, and aging. Pharmacological and dietary HDAC inhibitors have been used as therapeutics in psychiatry, cancer, and neurology.


Assuntos
Proteínas de Drosophila/fisiologia , Histona Desacetilase 1/fisiologia , Longevidade/fisiologia , Envelhecimento/fisiologia , Animais , Dieta , Drosophila/genética , Drosophila/metabolismo , Drosophila/fisiologia , Proteínas de Drosophila/deficiência , Proteínas de Drosophila/genética , Epigênese Genética/fisiologia , Histona Desacetilase 1/deficiência , Histona Desacetilase 1/genética , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/fisiologia , Sirtuínas/fisiologia
18.
J Cell Physiol ; 231(2): 436-48, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26174178

RESUMO

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.


Assuntos
Histona Desacetilase 1/metabolismo , Histona Desacetilase 2/metabolismo , Mucosa Intestinal/enzimologia , Animais , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Proliferação de Células/genética , Proliferação de Células/fisiologia , Colite/enzimologia , Colite/genética , Colite/patologia , Dano ao DNA , Modelos Animais de Doenças , Células Epiteliais/enzimologia , Células Caliciformes/citologia , Células Caliciformes/enzimologia , Histona Desacetilase 1/deficiência , Histona Desacetilase 1/genética , Histona Desacetilase 2/deficiência , Histona Desacetilase 2/genética , Homeostase , Imunidade nas Mucosas , Mucosa Intestinal/anormalidades , Mucosa Intestinal/citologia , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Notch/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo
19.
FEBS Lett ; 589(19 Pt B): 2776-83, 2015 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-26297832

RESUMO

By using acetyl-CoA as a substrate, acetyltransferases and histone deacetylases regulate protein acetylation by adding or removing an acetyl group on lysines. Nuclear-located Hdac1 is a regulator of intestinal homeostasis. We have previously shown that Hdac1 define specific intestinal epithelial cell basal and inflammatory-dependent gene expression patterns and control cell proliferation. We show here that Hdac1 depletion in cellulo leads to increased histone acetylation after metabolic stresses, and to metabolic disturbances resulting in impaired responses to oxidative stresses, AMPK kinase activation and mitochondrial biogenesis. Thus, nuclear Hdac1 may control intestinal epithelial cell metabolism by regulating the supply of acetyl groups.


Assuntos
Células Epiteliais/metabolismo , Histona Desacetilase 1/deficiência , Intestinos/citologia , Proteínas Quinases Ativadas por AMP/metabolismo , Acetilação , Animais , Linhagem Celular , Proliferação de Células , Ativação Enzimática , Células Epiteliais/citologia , Técnicas de Silenciamento de Genes , Histona Desacetilase 1/genética , Histonas/metabolismo , Biogênese de Organelas , Estresse Oxidativo , RNA Interferente Pequeno/genética , Ratos , Transdução de Sinais
20.
ASN Neuro ; 7(3)2015.
Artigo em Inglês | MEDLINE | ID: mdl-26129908

RESUMO

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.


Assuntos
Sobrevivência Celular/fisiologia , Genes p53 , Histona Desacetilase 1/deficiência , Histona Desacetilase 2/deficiência , Neuroproteção , Traumatismos do Nervo Óptico/enzimologia , Células Ganglionares da Retina/enzimologia , Células Ganglionares da Retina/fisiologia , Acetilação , Doença Aguda , Animais , Apoptose/genética , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/metabolismo , Axotomia , Modelos Animais de Doenças , Regulação da Expressão Gênica , Histona Desacetilase 1/genética , Histona Desacetilase 2/genética , Sistema de Sinalização das MAP Quinases , Camundongos Knockout , Traumatismos do Nervo Óptico/patologia , Células Ganglionares da Retina/patologia , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...