Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 67
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Mediators Inflamm ; 2021: 6359652, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34924813

RESUMEN

Ellagic acid (EA) was reported to play protective roles in rheumatoid arthritis (RA). It was found that the level of metastasis-associated gene 1 (MTA1)/histone deacetylase 1 (HDAC1) protein complex was downregulated by polyphenols in several human disorders. Notably, inhibition of MTA1 or HDAC1 has anti-inflammatory effects on RA. Therefore, our study is aimed at investigating whether EA prevents RA progression through regulating the MTA1/HDAC1 complex. Herein, the human fibroblast-like synoviocyte (FLS) cell line MH7A was treated with TNF-α to induce an inflammation model in vitro and then incubated with different concentrations of EA. Western blot analysis showed that EA reduced MTA1 expression in a dose-dependent manner in MH7A cells. Then, TNF-α-treated MH7A cells were incubated with EA alone or together with MTA1 overexpression plasmid (pcDNA-MTA1), and we found that EA inhibited proliferation, inflammation cytokine levels, and oxidative stress marker protein levels and promoted apoptosis in MH7A cells, while MTA1 overexpression abolished these effects. Moreover, coimmunoprecipitation assay verified the interaction between MTA1 and HDAC1. EA downregulated the MTA1/HDAC1 complex in MH7A cells. MTA1 knockdown inhibited proliferation, inflammation, and oxidative stress and promoted apoptosis in MH7A cells, while HDAC1 overexpression reversed these effects. Moreover, chromatin immunoprecipitation assay indicated that EA inhibited HDAC1-mediated Nur77 deacetylation. Rescue experiments demonstrated that Nur77 knockdown reversed the effects of EA on MH7A cell biological behaviors. Additionally, EA treatment attenuated arthritis index, paw swelling, synovial hyperplasia, and inflammation in collagen-induced arthritis (CIA) rats. In conclusion, EA inhibited proliferation, inflammation, and oxidative stress and promoted apoptosis in MH7A cells and alleviated the severity of RA in CIA rats though downregulating MTA1/HDAC1 complex and promoting HDAC1 deacetylation-mediated Nur77 expression.


Asunto(s)
Artritis Reumatoide/tratamiento farmacológico , Ácido Elágico/farmacología , Histona Desacetilasa 1/antagonistas & inhibidores , Proteínas Represoras/antagonistas & inhibidores , Transactivadores/antagonistas & inhibidores , Acetilación , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Histona Desacetilasa 1/fisiología , Humanos , Masculino , Miembro 1 del Grupo A de la Subfamilia 4 de Receptores Nucleares/fisiología , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Wistar , Proteínas Represoras/fisiología , Transactivadores/fisiología
2.
Nucleic Acids Res ; 49(17): 9783-9798, 2021 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-34450641

RESUMEN

The activity of hematopoietic factor GATA-1 is modulated through p300/CBP-mediated acetylation and FOG-1 mediated indirect interaction with HDAC1/2 containing NuRD complex. Although GATA-1 acetylation is implicated in GATA-1 activation, the role of deacetylation is not studied. Here, we found that the FOG-1/NuRD does not deacetylate GATA-1. However, HDAC1/2 can directly bind and deacetylate GATA-1. Two arginine residues within the GATA-1 linker region mediates direct interaction with HDAC1. The arginine to alanine mutation (2RA) blocks GATA-1 deacetylation and fails to induce erythroid differentiation. Gene expression profiling and ChIP-seq analysis further demonstrate the importance of GATA-1 deacetylation for gene activation and chromatin recruitment. GATA-12RA knock-in (KI) mice suffer mild anemia and thrombocytopenia with accumulation of immature erythrocytes and megakaryocytes in bone marrow and spleen. Single cell RNA-seq analysis of Lin- cKit+ (LK) cells further reveal a profound change in cell subpopulations and signature gene expression patterns in HSC, myeloid progenitors, and erythroid/megakaryocyte clusters in KI mice. Thus, GATA-1 deacetylation and its interaction with HDAC1 modulates GATA-1 chromatin binding and transcriptional activity that control erythroid/megakaryocyte commitment and differentiation.


Asunto(s)
Cromatina/metabolismo , Factor de Transcripción GATA1/metabolismo , Hematopoyesis/genética , Histona Desacetilasa 1/metabolismo , Transcripción Genética , Anemia/genética , Animales , Sitios de Unión , Células Eritroides/citología , Células Eritroides/metabolismo , Factor de Transcripción GATA1/genética , Factor de Transcripción GATA1/fisiología , Regulación de la Expresión Génica , Técnicas de Sustitución del Gen , Histona Desacetilasa 1/fisiología , Megacariocitos/citología , Megacariocitos/metabolismo , Ratones , Trombocitopenia/genética
3.
IUBMB Life ; 73(5): 784-799, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33715270

RESUMEN

The epigenetic abnormality is believed as a major driver for cancer initiation. Histone modification plays a vital role in tumor formation and progression. Particularly, alteration in histone acetylation has been highly associated with gene expression, cell cycle, as well as carcinogenesis. By analyzing glioblastoma (GBM)-related microarray from the GEO database and conducting chromatin immunoprecipitation-sequencing (ChIP-seq), we discovered that solute carrier family 30 member 3 (SLC30A3), a super enhancer (SE)-regulated factor, was significantly reduced in GBM tissues. Furthermore, histone deacetylase 1 (HDAC1), overexpressed in GBM tissues, could inhibit SLC30A3 expression by promoting histone H3K27ac deacetylation modification of the SE region of SLC30A3. Our functional validation revealed that SLC30A3 can inhibit the growth and metastatic spread of GBM cells in vitro and in vivo, and can activate the MAPK signaling pathway to promote apoptosis of GBM cells. Moreover, overexpression of HDAC1 resulted in a significant increase in DNA replication activity, a significant decline in apoptosis and cell cycle arrest in GBM cells. In a word, these findings indicate that combined epigenetic targeting of SLC30A3 by HDAC1 and SE is potentially therapeutically feasible in GBM.


Asunto(s)
Neoplasias Encefálicas/patología , Proteínas de Transporte de Catión/genética , Glioblastoma/patología , Código de Histonas , Histona Desacetilasa 1/fisiología , Proteínas de Neoplasias/fisiología , Acetilación , Adulto , Anciano , Animales , Apoptosis , Neoplasias Encefálicas/genética , Puntos de Control del Ciclo Celular , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , ADN de Neoplasias/genética , Transición Epitelial-Mesenquimal , Femenino , Glioblastoma/genética , Xenoinjertos , Secuenciación de Nucleótidos de Alto Rendimiento , Histona Desacetilasa 1/genética , Humanos , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Persona de Mediana Edad , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , Fenotipo , Procesamiento Proteico-Postraduccional , Adulto Joven
4.
Cell Death Dis ; 12(1): 5, 2021 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-33414424

RESUMEN

Acute pancreatitis (AP), an acute inflammatory process, can be difficult to diagnose. Activating transcription factor 4 (ATF4) has been reported to participate in the pathogenesis of AP. Additionally, histone deacetylases (HDACs) are shown to be closely related to the development of a variety of diseases, including inflammation disease. In our study, we tried to highlight the role of ATF4 in AP through regulation of HDAC1. Firstly, we validated the effect of ATF4 on pancreatic acinar cell proliferation, apoptosis, and inflammation through in vitro experiments on cellular models of caerulein-induced AP. Next, we examined the correlation between ATF4 and HDAC1, and between HDAC1 with neutral endopeptidase (NEP) and kruppel-like factor 4 (KLF4). Finally, the regulatory role of ATF4 in AP was further assessed by determination of pathological conditions, biochemical indicators and inflammation through in vivo experiments on caerulein-induced AP mouse models. After AP induction, highly expressed ATF4 was observed, and silencing ATF4 could promote pancreatic acinar cell proliferation and inhibit apoptosis. ATF4 could bind to the HDAC1 promoter and upregulate its expression in AP. Moreover, HDAC1 could increase KLF4 expression by inhibiting NEP expression. Functionally, silencing ATF4 could suppress AP through regulation of NEP-mediated KLF4 via downregulation of HDAC1. Above all, our study uncovered the promotive role of ATF4 in AP through upregulation of HDAC1.


Asunto(s)
Células Acinares , Factor de Transcripción Activador 4/metabolismo , Histona Desacetilasa 1/fisiología , Páncreas , Pancreatitis/metabolismo , Células Acinares/metabolismo , Células Acinares/patología , Animales , Apoptosis , Proliferación Celular , Células Cultivadas , Inflamación , Factor 4 Similar a Kruppel , Ratones , Ratones Endogámicos C57BL , Páncreas/metabolismo , Páncreas/patología
5.
Life Sci ; 265: 118760, 2021 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-33212149

RESUMEN

AIMS: Epigenetic regulation plays an important role in the progression of Alzheimer's disease (AD). Here, we identified differential methylation probes (DMP) and investigated their potential mechanistic roles in AD. MAIN METHODS: DMPs were identified via bioinformatic analysis of GSE66351, which was made up with 106 AD samples and 84 control samples derived from three separate brain regions. Differentially expressed genes (DEGs) were analyzed based on GSE5281 comprising 45 control samples and 58 AD samples. Gene ontology (GO), gene set enrichment analysis (GSEA), and protein-protein interaction (PPI) were used to identify pathways and hub genes. KEY FINDINGS: We found 9007 DMPs in Occipital Cortex glia, 1527 in OC neurons, 100 in Temporal Cortex, and 194 in Frontal Cortex. 74 DEGs were identified in Primary Visual Cortex, 67 of which were downregulated while seven upregulated. 482 were upregulated and 697 downregulated in medial temporal gyrus. In superior frontal gyrus, 687 were upregulated and 85 downregulated. GO and PPI revealed that pathways involving epithelial-cell differentiation, cellular responses to lipids, transcription corepressor activities, apoptotic and organ growth were modulated by histone deacetylase 1 (HDAC1) and associated with AD. Additionally, GSEA illustrated that the transforming growth factor beta signaling pathway was significantly enriched in some brain regions and HDAC1 played an important role in this pathway. SIGNIFICANCE: We found the glial-specific 3'UTR of HDAC1 was hypermethylated and HDAC1 was overexpressed in AD patients. Moreover, we also speculate that HDAC1 triggered signaling pathways linked to many different biological processes and functions via the regulation of histone deacetylation.


Asunto(s)
Regiones no Traducidas 3' , Enfermedad de Alzheimer/metabolismo , Histona Desacetilasa 1/metabolismo , Neuroglía/metabolismo , Transducción de Señal , Enfermedad de Alzheimer/enzimología , Metilación de ADN , Regulación de la Expresión Génica , Histona Desacetilasa 1/fisiología , Humanos , Neuroglía/enzimología , Mapas de Interacción de Proteínas
6.
J Cell Mol Med ; 24(18): 10876-10888, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32743904

RESUMEN

Osteosarcoma (OS) is a malignant bone cancer lacking of effective treatment target when the metastasis occurred. This study investigated the implication of MicroRNA-326 in OS proliferation and metastasis to provide the clue for the treatment of metastatic OS. This study knocked down SP1 in MG63 and 143B cells and then performed Microarray assay to find the expression of miRNAs that were influenced by SP1. MTT, EdU, wound-healing and cell invasion assays were performed to evaluated cell proliferation and invasion. OS metastasis to lung was detected in a nude mice model. ChIP assay and DAPA were applied to determine the regulatory effect of SP1 and histone deacetylase 1 (HDAC) complex on miR-326 expression. Human OS tissues showed lowly expressed miR-326 but highly expressed Sp1 and HDAC. Sp1 recruited HDAC1 to miR-326 gene promoter, which caused the histone deacetylation and subsequent transcriptional inhibition of miR-326 gene. miR-326 deficiency induced the stimulation of SMO/Hedgehog pathway and promoted the proliferation and invasion of 143B and MG63 cells as well as the growth and metastasis in nude mice. SP1/HDAC1 caused the transcriptional inhibition of miR-326 gene by promoting histone deacetylation; miR-326 deficiency conversely stimulated SMO/Hedgehog pathway that was responsible for the proliferation and metastasis of OS.


Asunto(s)
Neoplasias Óseas/patología , Histona Desacetilasa 1/fisiología , MicroARNs/antagonistas & inhibidores , Metástasis de la Neoplasia/fisiopatología , Proteínas de Neoplasias/fisiología , Osteosarcoma/patología , ARN Neoplásico/antagonistas & inhibidores , Receptor Smoothened/biosíntesis , Factor de Transcripción Sp1/fisiología , Adolescente , Adulto , Animales , Neoplasias Óseas/genética , División Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Femenino , Técnicas de Silenciamiento del Gen , Histona Desacetilasa 1/antagonistas & inhibidores , Histona Desacetilasa 1/genética , Humanos , Masculino , Metaloproteinasa 9 de la Matriz/fisiología , Ratones , Ratones Desnudos , MicroARNs/biosíntesis , MicroARNs/genética , Metástasis de la Neoplasia/genética , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Osteosarcoma/genética , Osteosarcoma/secundario , Interferencia de ARN , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Neoplásico/biosíntesis , ARN Neoplásico/genética , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Receptor Smoothened/genética , Factor de Transcripción Sp1/antagonistas & inhibidores , Factor de Transcripción Sp1/genética , Ensayos Antitumor por Modelo de Xenoinjerto , Adulto Joven , Proteína con Dedos de Zinc GLI1/fisiología
7.
JCI Insight ; 5(4)2020 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-32102981

RESUMEN

Some effector CD4+ T cell subsets display cytotoxic activity, thus breaking the functional dichotomy of CD4+ helper and CD8+ cytotoxic T lymphocytes. However, molecular mechanisms regulating CD4+ cytotoxic T lymphocyte (CD4+ CTL) differentiation are poorly understood. Here we show that levels of histone deacetylases 1 and 2 (HDAC1-HDAC2) are key determinants of CD4+ CTL differentiation. Deletions of both Hdac1 and 1 Hdac2 alleles (HDAC1cKO-HDAC2HET) in CD4+ T cells induced a T helper cytotoxic program that was controlled by IFN-γ-JAK1/2-STAT1 signaling. In vitro, activated HDAC1cKO-HDAC2HET CD4+ T cells acquired cytolytic activity and displayed enrichment of gene signatures characteristic of effector CD8+ T cells and human CD4+ CTLs. In vivo, murine cytomegalovirus-infected HDAC1cKO-HDAC2HET mice displayed a stronger induction of CD4+ CTL features compared with infected WT mice. Finally, murine and human CD4+ T cells treated with short-chain fatty acids, which are commensal-produced metabolites acting as HDAC inhibitors, upregulated CTL genes. Our data demonstrate that HDAC1-HDAC2 restrain CD4+ CTL differentiation. Thus, HDAC1-HDAC2 might be targets for the therapeutic induction of CD4+ CTLs.


Asunto(s)
Linfocitos T CD4-Positivos/citología , Diferenciación Celular/fisiología , Histona Desacetilasa 1/fisiología , Histona Desacetilasa 2/fisiología , Linfocitos T Citotóxicos/fisiología , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Ácidos Grasos/farmacología , Histona Desacetilasa 1/genética , Histona Desacetilasa 2/genética , Humanos , Ratones , Ratones Noqueados , Transducción de Señal/fisiología , Linfocitos T Citotóxicos/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología
8.
J Clin Invest ; 129(12): 5381-5399, 2019 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-31487270

RESUMEN

Oral squamous cell carcinoma (OSCC) frequently invades the maxillary or mandibular bone, and this bone invasion is closely associated with poor prognosis and survival. Here, we show that CCL28 functions as a negative regulator of OSCC bone invasion. CCL28 inhibited invasion and epithelial-mesenchymal transition (EMT), and its inhibition of EMT was characterized by induced E-cadherin expression and reduced nuclear localization of ß-catenin in OSCC cells with detectable RUNX3 expression levels. CCL28 signaling via CCR10 increased retinoic acid receptor-ß (RARß) expression by reducing the interaction between RARα and HDAC1. In addition, CCL28 reduced RANKL production in OSCC and osteoblastic cells and blocked RANKL-induced osteoclastogenesis in osteoclast precursors. Intraperitoneally administered CCL28 inhibited tumor growth and osteolysis in mouse calvaria and tibia inoculated with OSCC cells. RARß expression was also increased in tumor tissues. In patients with OSCC, low CCL28, CCR10, and RARß expression levels were highly correlated with bone invasion. Patients with OSCC who had higher expression of CCL28, CCR10, or RARß had significantly better overall survival. These findings suggest that CCL28, CCR10, and RARß are useful markers for the prediction and treatment of OSCC bone invasion. Furthermore, CCL28 upregulation in OSCC cells or CCL28 treatment can be a therapeutic strategy for OSCC bone invasion.


Asunto(s)
Huesos/patología , Quimiocinas CC/farmacología , Neoplasias de la Boca/patología , Receptores de Ácido Retinoico/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Animales , Línea Celular Tumoral , Transición Epitelial-Mesenquimal , Histona Desacetilasa 1/fisiología , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos ICR , Invasividad Neoplásica , Osteoclastos/citología , Ligando RANK/fisiología , Receptores CCR10/fisiología , Receptor alfa de Ácido Retinoico/fisiología
9.
FASEB J ; 33(7): 8008-8021, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30913399

RESUMEN

Schwann cells are the main supportive cells of the peripheral nerves. Schwann cells suffer inhibition of autophagy under hyperglycemia treatment in diabetic peripheral neuropathy (DPN). However, the exact mechanism is still not fully elucidated. We first observed the decrease of autophagy markers (LC3-II/LC3-I, P62) in the sciatic nerves of diabetic mice vs. normal mice, accompanied with the loss of myelinated nerve fibers and abnormal myelin sheath. In line with this, LC3-II/LC3-I and P62 were also significantly reduced in high glucose-treated rat Schwann cell 96 (RSC96) cells compared with normal glucose-treated cells. Furthermore, we found that trichostatin A [an inhibitor of histone deacetylase (HDAC)] evidently improved LC3-II/LC3-I in high glucose-treated RSC96 cells, without an effect on P62 expression. Again, HDAC1 and HDAC5 were revealed to be increased in RSC96 cells stimulated with high glucose. Inhibition of HDAC1 but not HDAC5 by small hairpin RNA vector enhanced LC3-II/LC3-I in high glucose-cultured RSC96 cells. In addition, LC3-II conversion regulators [autophagy-related protein (Atg)3, Atg5, and Atg7] were detected in high glucose-treated and HDAC1-knockdown RSC96 cells, and Atg3 was proven to be the key target of HDAC1. The presuppression of Atg3 offset the improvement of LC3-II/LC3-I resulting from HDAC1 inhibition in high glucose-treated RSC96 cells. The Janus kinase (JAK)-signal transducer and activator of transcription (STAT) signaling pathway was activated in RSC96 cells treated with high glucose, which was indicated by increased STAT3 phosphorylation. Blocking STAT3 phosphorylation by chemical inhibitor AG490 induced HDAC1 down-regulation followed by increases in Atg3 and LC3-II/LC3-I. Interestingly, we also found that AG490 treatment enhanced P62 expression in high glucose-stimulated RSC96 cells. Taken together, our findings demonstrate that hyperglycemia inhibits LC3-II/LC3-I in an HDAC1-Atg3-dependent manner and decreases P62 expression in an HDAC-independent manner via the JAK-STAT3 signaling pathway in the Schwann cells of DPN.-Du, W., Wang, N., Li, F. Jia, K., An, J., Liu, Y., Wang, Y., Zhu, L., Zhao, S. Hao, J. STAT3 phosphorylation mediates high glucose-impaired cell autophagy in an HDAC1-dependent and -independent manner in Schwann cells of diabetic peripheral neuropathy.


Asunto(s)
Autofagia/efectos de los fármacos , Neuropatías Diabéticas/metabolismo , Glucosa/farmacología , Histona Desacetilasa 1/fisiología , Procesamiento Proteico-Postraduccional , Factor de Transcripción STAT3/metabolismo , Células de Schwann/efectos de los fármacos , Animales , Proteínas Relacionadas con la Autofagia/antagonistas & inhibidores , Proteínas Relacionadas con la Autofagia/biosíntesis , Proteínas Relacionadas con la Autofagia/genética , Biomarcadores , Línea Celular , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Neuropatías Diabéticas/patología , Técnicas de Silenciamiento del Gen , Histona Desacetilasa 1/antagonistas & inhibidores , Histona Desacetilasa 1/genética , Histona Desacetilasas/genética , Histona Desacetilasas/fisiología , Ácidos Hidroxámicos/farmacología , Ratones , Proteínas Asociadas a Microtúbulos/biosíntesis , Proteínas Asociadas a Microtúbulos/genética , Vaina de Mielina/patología , Fibras Nerviosas Mielínicas/patología , Péptido Sintasas/antagonistas & inhibidores , Péptido Sintasas/biosíntesis , Péptido Sintasas/genética , Fosforilación , Interferencia de ARN , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/farmacología , Ratas , Células de Schwann/metabolismo , Nervio Ciático/metabolismo , Nervio Ciático/patología , Tirfostinos/farmacología , Regulación hacia Arriba
10.
Cell Mol Life Sci ; 76(5): 1005-1025, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30599067

RESUMEN

BACKGROUND: The ADAM10-mediated cleavage of transmembrane proteins regulates cellular processes such as proliferation or migration. Substrate cleavage by ADAM10 has also been implicated in pathological situations such as cancer or Morbus Alzheimer. Therefore, identifying endogenous molecules, which modulate the amount and consequently the activity of ADAM10, might contribute to a deeper understanding of the enzyme's role in both, physiology and pathology. METHOD: To elucidate the underlying cellular mechanism of the TBX2-mediated repression of ADAM10 gene expression, we performed overexpression, RNAi-mediated knockdown and pharmacological inhibition studies in the human neuroblastoma cell line SH-SY5Y. Expression analysis was conducted by e.g. real-time RT-PCR or western blot techniques. To identify the binding region of TBX2 within the ADAM10 promoter, we used luciferase reporter assay on deletion constructs and EMSA/WEMSA experiments. In addition, we analyzed a TBX2 loss-of-function Drosophila model regarding the expression of ADAM10 orthologs by qPCR. Furthermore, we quantified the mRNA level of TBX2 in post-mortem brain tissue of AD patients. RESULTS: Here, we report TBX2 as a transcriptional repressor of ADAM10 gene expression: both, the DNA-binding domain and the repression domain of TBX2 were necessary to effect transcriptional repression of ADAM10 in neuronal SH-SY5Y cells. This regulatory mechanism required HDAC1 as a co-factor of TBX2. Transcriptional repression was mediated by two functional TBX2 binding sites within the core promoter sequence (- 315 to - 286 bp). Analysis of a TBX2 loss-of-function Drosophila model revealed that kuzbanian and kuzbanian-like, orthologs of ADAM10, were derepressed compared to wild type. Vice versa, analysis of cortical brain samples of AD-patients, which showed reduced ADAM10 mRNA levels, revealed a 2.5-fold elevation of TBX2, while TBX3 and TBX21 levels were not affected. CONCLUSION: Our results characterize TBX2 as a repressor of ADAM10 gene expression and suggest that this regulatory interaction is conserved across tissues and species.


Asunto(s)
Proteína ADAM10/genética , Enfermedad de Alzheimer/etiología , Regulación de la Expresión Génica , Proteínas de Dominio T Box/fisiología , Secretasas de la Proteína Precursora del Amiloide/genética , Animales , Ácido Aspártico Endopeptidasas/genética , Sitios de Unión , Encéfalo/metabolismo , Células Cultivadas , Desintegrinas/genética , Drosophila , Proteínas de Drosophila/genética , Histona Desacetilasa 1/fisiología , Humanos , Metaloendopeptidasas/genética , Ratones , Ratones Endogámicos C57BL , Neuronas/metabolismo , Regiones Promotoras Genéticas , Proteínas de Dominio T Box/química , Transcripción Genética
11.
PLoS Genet ; 14(8): e1007578, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30110327

RESUMEN

SMYD4 belongs to a family of lysine methyltransferases. We analyzed the role of smyd4 in zebrafish development by generating a smyd4 mutant zebrafish line (smyd4L544Efs*1) using the CRISPR/Cas9 technology. The maternal and zygotic smyd4L544Efs*1 mutants demonstrated severe cardiac malformations, including defects in left-right patterning and looping and hypoplastic ventricles, suggesting that smyd4 was critical for heart development. Importantly, we identified two rare SMYD4 genetic variants in a 208-patient cohort with congenital heart defects. Both biochemical and functional analyses indicated that SMYD4(G345D) was pathogenic. Our data suggested that smyd4 functions as a histone methyltransferase and, by interacting with HDAC1, also serves as a potential modulator for histone acetylation. Transcriptome and bioinformatics analyses of smyd4L544Efs*1 and wild-type developing hearts suggested that smyd4 is a key epigenetic regulator involved in regulating endoplasmic reticulum-mediated protein processing and several important metabolic pathways in developing zebrafish hearts.


Asunto(s)
Epigénesis Genética , Histona Metiltransferasas/fisiología , N-Metiltransferasa de Histona-Lisina/fisiología , Proteínas de Pez Cebra/fisiología , Pez Cebra/genética , Adolescente , Animales , Sistemas CRISPR-Cas , Niño , Preescolar , Estudios de Cohortes , Modelos Animales de Enfermedad , Desarrollo Embrionario/efectos de los fármacos , Femenino , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Corazón/efectos de los fármacos , Corazón/embriología , Cardiopatías Congénitas/genética , Histona Desacetilasa 1/genética , Histona Desacetilasa 1/fisiología , Histona Metiltransferasas/genética , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Lactante , Masculino , Mutación Missense , Conformación Proteica , Análisis de Secuencia de ARN , Transcriptoma , Secuenciación del Exoma , Pez Cebra/embriología , Proteínas de Pez Cebra/genética
12.
Nat Commun ; 8(1): 728, 2017 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-28959017

RESUMEN

Bilateral symmetry is a striking feature of the vertebrate body plan organization. Vertebral precursors, called somites, provide one of the best illustrations of embryonic symmetry. Maintenance of somitogenesis symmetry requires retinoic acid (RA) and its coactivator Rere/Atrophin2. Here, using a proteomic approach we identify a protein complex, containing Wdr5, Hdac1, Hdac2 and Rere (named WHHERE), which regulates RA signaling and controls embryonic symmetry. We demonstrate that Wdr5, Hdac1, and Hdac2 are required for RA signaling in vitro and in vivo. Mouse mutants for Wdr5 and Hdac1 exhibit asymmetrical somite formation characteristic of RA-deficiency. We also identify the Rere-binding histone methyltransferase Ehmt2/G9a, as a RA coactivator controlling somite symmetry. Upon RA treatment, WHHERE and Ehmt2 become enriched at RA target genes to promote RNA polymerase II recruitment. Our work identifies a protein complex linking key epigenetic regulators acting in the molecular control of embryonic bilateral symmetry.Retinoic acid (RA) regulates the maintenance of somitogenesis symmetry. Here, the authors use a proteomic approach to identify a protein complex of Wdr5, Hdac1, Hdac2 that act together with RA and coactivator Rere/Atrophin2 and a histone methyltransferase Ehmt2 to regulate embryonic symmetry.


Asunto(s)
Embrión de Mamíferos/metabolismo , Desarrollo Embrionario , Tretinoina/fisiología , Animales , Proteína p300 Asociada a E1A/genética , Proteína p300 Asociada a E1A/metabolismo , Proteína p300 Asociada a E1A/fisiología , Embrión de Mamíferos/citología , Epigénesis Genética , Histona Desacetilasa 1/genética , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 1/fisiología , Histona Desacetilasa 2/genética , Histona Desacetilasa 2/metabolismo , Histona Desacetilasa 2/fisiología , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , N-Metiltransferasa de Histona-Lisina/fisiología , Histonas/química , Histonas/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/fisiología , Proteínas/genética , Proteínas/metabolismo , Proteínas/fisiología , Proteómica , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Proteínas Represoras/fisiología , Transducción de Señal , Somitos/crecimiento & desarrollo , Somitos/metabolismo , Somitos/ultraestructura , Tretinoina/metabolismo
13.
Br J Haematol ; 178(5): 728-738, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28480959

RESUMEN

PTPN6, a tyrosine phosphatase protein, plays a negative role in cell signal transduction and is negatively correlated with tumour formation and growth. However, epigenetic regulation mechanism of the PTPN6 gene in advanced chronic myeloid leukaemia (CML) remains unclear. This study investigated bone marrow or blood samples from 44 CML patients and 10 healthy volunteers. KCL22 and K562 cells were cultured and treated with demethylation drugs and histone deacetylase inhibitors. Real time quantitative polymerase chain reaction (qPCR), methylation-specific PCR, bisulfite sequencing PCR, Western blotting, co-immunoprecipitation and chromatin immunoprecipitation (ChIP) was performed. PTPN6 was down-regulated in cell lines and patients with advanced phase CML, whereas DNMT1, DNMT3A, MECP2, MBD2 and HDAC1 were up-regulated. Treatment with 5-azacytidine, decitabine, sodium valproate and LBH589 increased PTPN6 expression, but decreased that of DNMT1, DNMT3A, MECP2, MBD2 and HDAC1. Immunoprecipitation and mass spectrometry showed that HDAC1 combined directly with PTPN6. ChIP-seq showed that HDAC1 did not combine with the promoter region of PTPN6, while MAPK, AKT, STAT5, JAK2 and MYC promoter regions all combined with HDAC1. PTPN6 is associated with progression of CML. Low expression level of PTPN6 was associated with DNA methylation and regulated by histone acetylation. HDAC1 participates in the regulation of PTPN6.


Asunto(s)
Epigénesis Genética , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 6/genética , Adolescente , Adulto , Anciano , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Islas de CpG/genética , Metilación de ADN , Relación Dosis-Respuesta a Droga , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 1/fisiología , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Regiones Promotoras Genéticas , Proteína Tirosina Fosfatasa no Receptora Tipo 6/biosíntesis , ARN Mensajero/genética , ARN Neoplásico/genética , Células Tumorales Cultivadas/efectos de los fármacos , Adulto Joven
14.
Cancer Res ; 77(4): 886-896, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-27923833

RESUMEN

Maspin (SerpinB5) is an epithelial-specific tumor suppressor gene product that displays context-dependent cellular functions. Maspin-deficient mouse models created to date have not definitively established maspin functions critical for cancer suppression. In this study, we generated a mouse strain in which exon 4 of the Maspin gene was deleted, confirming its essential role in development but also enabling a breeding scheme to bypass embryonic lethality. Phenotypic characterization of this viable strain established that maspin deficiency was associated with a reduction in maximum body weight and a variety of context-dependent epithelial abnormalities. Specifically, maspin-deficient mice exhibited pulmonary adenocarcinoma, myoepithelial hyperplasia of the mammary gland, hyperplasia of luminal cells of dorsolateral and anterior prostate, and atrophy of luminal cells of ventral prostate and stratum spinosum of epidermis. These cancer phenotypes were accompanied by increased inflammatory stroma. These mice also displayed the autoimmune disorder alopecia aerate. Overall, our findings defined context-specific tumor suppressor roles for maspin in a clinically relevant model to study maspin functions in cancer and other pathologies. Cancer Res; 77(4); 886-96. ©2017 AACR.


Asunto(s)
Desarrollo Embrionario , Serpinas/fisiología , Proteínas Supresoras de Tumor/fisiología , Alopecia Areata/etiología , Animales , Femenino , Histona Desacetilasa 1/fisiología , Masculino , Glándulas Mamarias Animales/patología , Ratones , Ratones Endogámicos C57BL , Especificidad de Órganos , Próstata/patología , Serpinas/genética
15.
Mol Biol Cell ; 28(2): 346-355, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-27903773

RESUMEN

The Wnt signaling pathway is essential in regulating various cellular processes. Different mechanisms of inhibition for Wnt signaling have been proposed. Besides ß-catenin degradation through the proteasome, nemo-like kinase (NLK) is another molecule that is known to negatively regulate Wnt signaling. However, the mechanism by which NLK mediates the inhibition of Wnt signaling was not known. In the present study, we used primary embryonic fibroblast cells isolated from NLK-deficient mice and showed that these cells proliferate faster and have a shorter cell cycle than wild-type cells. In NLK-knockout cells, we observed sustained interaction between Lef1 and ß-catenin, leading to elevated luciferase reporter of ß-catenin/Lef1-mediated transcriptional activation. The mechanism for the reduced ß-catenin/Lef1 promoter activation was explained by phosphorylation of HDAC1 at serine 421 via NLK. The phosphorylation of HDAC1 was achieved only in the presence of wild-type NLK because a catalytically inactive mutant of NLK was unable to phosphorylate HDAC1 and reduced the luciferase reporter of ß-catenin/Lef1-mediated transcriptional activation. This result suggests that NLK and HDAC1 together negatively regulate Wnt signaling, which is vital in preventing aberrant proliferation of nontransformed primary fibroblast cells.


Asunto(s)
Histona Desacetilasa 1/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Animales , Ciclo Celular , Fibroblastos , Histona Desacetilasa 1/fisiología , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Quinasas Quinasa Quinasa PAM/metabolismo , Ratones Noqueados , Proteínas Quinasas Activadas por Mitógenos/fisiología , Fosforilación , Cultivo Primario de Células , Proteínas Serina-Treonina Quinasas , Transducción de Señal/fisiología , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Activación Transcripcional , Proteínas Wnt/metabolismo , Vía de Señalización Wnt/genética , Vía de Señalización Wnt/fisiología , beta Catenina/metabolismo
16.
Anticancer Res ; 36(6): 2819-26, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27272793

RESUMEN

BACKGROUND: Loss of FAS expression in ovarian cancer cells has recently been associated with resistance to chemotherapeutic drugs. However, the mechanism for suppression of FAS expression is unknown. MATERIALS AND METHODS: The cell surface and transcript expressions of death receptors in parental chemosensitive (A2780) and their derivative chemoresistant (A2780-AD) ovarian cancer cells were determined by flow cytometry and quantitative real-time polymerase chain reaction, respectively. The epigenetic regulation of FAS promoters in both A2780 and A2780-AD ovarian epithelial cells were determined by chromatin immunoprecipitation assays. CONCLUSION: This study demonstrated that expression of FAS is suppressed in A2780-AD cells compared to parental A2780 ovarian cells. No difference in DNA methylation was observed at FAS promoters between A2780-AD cells compared to parental cells. However, the level of acetylated histone H3 associated with FAS promoter in A2780-AD cells was significantly lower compared to parental cells, and there was a corresponding increase in histone deacetylase 1 (HDAC1) enzyme associated with the FAS promoter in resistant cells. Knockdown of HDAC1 expression, and pharmacological inhibition of HDAC enzymatic activity significantly increased FAS expression in resistant A2780-AD cells. These results suggest that epigenetic changes in histone modifications may contribute to the loss of FAS expression in chemoresistant ovarian cancer cells and that enhancement of FAS expression could increase tumor cell sensitivity to immune cells.


Asunto(s)
Histona Desacetilasa 1/fisiología , Neoplasias Ováricas/tratamiento farmacológico , Receptor fas/genética , Acetilación , Cisplatino/farmacología , Metilación de ADN , Resistencia a Antineoplásicos , Femenino , Humanos , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Regiones Promotoras Genéticas
17.
Exp Gerontol ; 86: 124-128, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-26927903

RESUMEN

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.


Asunto(s)
Proteínas de Drosophila/fisiología , Histona Desacetilasa 1/fisiología , Longevidad/fisiología , Envejecimiento/fisiología , Animales , Dieta , Drosophila/genética , Drosophila/metabolismo , Drosophila/fisiología , Proteínas de Drosophila/deficiencia , Proteínas de Drosophila/genética , Epigénesis Genética/fisiología , Histona Desacetilasa 1/deficiencia , Histona Desacetilasa 1/genética , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/fisiología , Sirtuinas/fisiología
19.
Neurobiol Aging ; 37: 103-116, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26545632

RESUMEN

With increased histone deacetylase (HDAC) activity and histone hypoacetylation being implicated in neurodegeneration, HDAC inhibitors have been reported to have considerable therapeutic potential. Yet, existing inhibitors lack specificity and may show substantial adverse effect. In this study, we identified a novel HDAC1/2 isoform-specific inhibitor, K560, with protective effects against 1-methyl-4-phenylpyridinium (MPP(+))- and/or 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced neuronal death in both in vitro and in vivo Parkinson's disease model. K560 attenuated cell death induced by MPP(+) in differentiated SH-SY5Y cells through the sustained expression of an antiapoptotic protein, X-linked inhibitor of apoptosis (XIAP). Inhibition of XIAP expression by locked nucleic acid antisense oligonucleotides abolished the protective effect of K560. Inactivation of mitogen-activated protein kinase cascades, reduced p53 phosphorylation, and down-regulation of p53-upregulated modulator of apoptosis on K560 treatment were also observed. Furthermore, pre- and post-oral administration of K560 to mice prevented MPTP-induced loss of dopaminergic neurons in substantia nigra, suggesting that selective inhibition of HDAC1 and HDAC2 by K560 may pave the way to new strategies for Parkinson's disease treatment.


Asunto(s)
Benzamidas/uso terapéutico , Dicetopiperazinas/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Histona Desacetilasa 1/antagonistas & inhibidores , Histona Desacetilasa 2/antagonistas & inhibidores , Terapia Molecular Dirigida , Fármacos Neuroprotectores/uso terapéutico , Enfermedad de Parkinson/tratamiento farmacológico , Acetilación , Administración Oral , Animales , Benzamidas/administración & dosificación , Benzamidas/farmacología , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Línea Celular Tumoral , Dicetopiperazinas/administración & dosificación , Dicetopiperazinas/farmacología , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/farmacología , Expresión Génica/efectos de los fármacos , Histona Desacetilasa 1/fisiología , Histona Desacetilasa 2/fisiología , Histonas/metabolismo , Humanos , Isoenzimas , Ratones , Fármacos Neuroprotectores/farmacología , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/patología , Proteína Inhibidora de la Apoptosis Ligada a X/genética , Proteína Inhibidora de la Apoptosis Ligada a X/metabolismo
20.
EMBO Rep ; 16(10): 1288-98, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26303947

RESUMEN

The histone H3K27 demethylase, UTX, is a known component of the H3K4 methyltransferase MLL complex, but its functional association with H3K4 methylation in human cancers remains largely unknown. Here we demonstrate that UTX loss induces epithelial-mesenchymal transition (EMT)-mediated breast cancer stem cell (CSC) properties by increasing the expression of the SNAIL, ZEB1 and ZEB2 EMT transcription factors (EMT-TFs) and of the transcriptional repressor CDH1. UTX facilitates the epigenetic silencing of EMT-TFs by inducing competition between MLL4 and the H3K4 demethylase LSD1. EMT-TF promoters are occupied by c-Myc and MLL4, and UTX recognizes these proteins, interrupting their transcriptional activation function. UTX decreases H3K4me2 and H3 acetylation at these promoters by forming a transcriptional repressive complex with LSD1, HDAC1 and DNMT1. Taken together, our findings indicate that UTX is a prominent tumour suppressor that functions as a negative regulator of EMT-induced CSC-like properties by epigenetically repressing EMT-TFs.


Asunto(s)
Represión Epigenética , Transición Epitelial-Mesenquimal , Histona Demetilasas/genética , Células Madre Neoplásicas/fisiología , Proteínas Nucleares/genética , Proteínas Represoras/genética , Neoplasias de la Mama , Línea Celular Tumoral , Transición Epitelial-Mesenquimal/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Histona Desacetilasa 1/genética , Histona Desacetilasa 1/fisiología , Histona Demetilasas/fisiología , Humanos , Regiones Promotoras Genéticas , Procesamiento Proteico-Postraduccional , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA