Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
J Neurosci ; 41(17): 3799-3807, 2021 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-33741723

RESUMEN

The release of small extracellular vesicles (sEVs) has recently been reported, but knowledge of their function in neuron development remains limited. Using LC-MS/MS, we found that sEVs released from developing cortical neurons in vitro obtained from mice of both sexes were enriched in cytoplasm, exosome, and protein-binding and DNA/RNA-binding pathways. The latter included HDAC2, which was of particular interest, because HDAC2 regulates spine development, and populations of neurons expressing different levels of HDAC2 co-exist in vivo during the period of spine growth. Here, we found that HDAC2 levels decrease in neurons as they acquire synapses and that sEVs from HDAC2-rich neurons regulate HDAC2 signaling in HDAC2-low neurons possibly through HDAC2 transfer. This regulation led to a transcriptional decrease in HDAC2 synaptic targets and the density of excitatory synapses. These data suggest that sEVs provide inductive cell-cell signaling that coordinates the development of dendritic spines via the activation of HDAC2-dependent transcriptional programs.SIGNIFICANCE STATEMENT A role of small extracellular vesicles (sEVs; also called exosomes) in neuronal development is of particular interest, because sEVs could provide a major signaling modality between developing neurons when synapses are not fully functional or immature. However, knowledge of sEVs on neuron, and more precisely spine development, is limited. We provide several lines of evidence that sEVs released from developing cortical neurons regulate the development of dendritic spines via the regulation of HDAC2 signaling. This paracrine communication is temporally restricted during development because of the age-dependent decrease in sEV release as neurons mature and acquire spines.


Asunto(s)
Espinas Dendríticas/fisiología , Espacio Extracelular/fisiología , Histona Desacetilasa 2/genética , Histona Desacetilasa 2/fisiología , Transducción de Señal/genética , Transducción de Señal/fisiología , Animales , Citoplasma/metabolismo , Exosomas/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Masculino , Ratones , Cultivo Primario de Células , Proteómica , Sinapsis/fisiología
2.
Mol Cell Neurosci ; 109: 103570, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33160016

RESUMEN

Alzheimer's disease (AD) is an age-related neurodegenerative disorder hallmarked by amyloid-ß (Aß) plaque accumulation, neuronal cell death, and cognitive deficits that worsen during disease progression. Histone acetylation dysregulation, caused by an imbalance between reduced histone acetyltransferases (HAT) Tip60 and increased histone deacetylase 2 (HDAC2) levels, can directly contribute to AD pathology. However, whether such AD-associated neuroepigenetic alterations occur in response to Aß peptide production and can be protected against by increasing Tip60 levels over the course of neurodegenerative progression remains unknown. Here we profile Tip60 HAT/HDAC2 dynamics and transcriptome-wide changes across early and late stage AD pathology in the Drosophila brain produced solely by human amyloid-ß42. We show that early Aß42 induction leads to disruption of Tip60 HAT/HDAC2 balance during early neurodegenerative stages preceding Aß plaque accumulation that persists into late AD stages. Correlative transcriptome-wide studies reveal alterations in biological processes we classified as transient (early-stage only), late-onset (late-stage only), and constant (both). Increasing Tip60 HAT levels in the Aß42 fly brain protects against AD functional pathologies that include Aß plaque accumulation, neural cell death, cognitive deficits, and shorter life-span. Strikingly, Tip60 protects against Aß42-induced transcriptomic alterations via distinct mechanisms during early and late stages of neurodegeneration. Our findings reveal distinct modes of neuroepigenetic gene changes and Tip60 neuroprotection in early versus late stages in AD that can serve as early biomarkers for AD, and support the therapeutic potential of Tip60 over the course of AD progression.


Asunto(s)
Péptidos beta-Amiloides/toxicidad , Proteínas de Drosophila/fisiología , Drosophila melanogaster/genética , Histona Acetiltransferasas/fisiología , Degeneración Nerviosa/genética , Fragmentos de Péptidos/toxicidad , Transcriptoma , Acetilación , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Animales , Apoptosis , Aprendizaje por Asociación/fisiología , Modelos Animales de Enfermedad , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/metabolismo , Epigénesis Genética , Regulación de la Expresión Génica , Código de Histonas , Histona Desacetilasa 2/fisiología , Larva , Locomoción , Longevidad , Aprendizaje por Laberinto , Odorantes , Procesamiento Proteico-Postraduccional , Olfato/fisiología
3.
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
4.
J Am Soc Nephrol ; 31(2): 337-349, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31843983

RESUMEN

BACKGROUND: The progression rate of CKD varies substantially among patients. The genetic and epigenetic contributions that modify how individual patients respond to kidney injury are largely unknown. Emerging evidence has suggested that histone H3 K79 methyltransferase Dot1l has an antifibrotic effect by repressing Edn1, which encodes endothelin 1 in the connecting tubule/collecting duct. METHODS: To determine if deletion of the Dot1l gene is a genetic and epigenetic risk factor through regulating Edn1, we studied four groups of mice: wild-type mice, connecting tubule/collecting duct-specific Dot1l conditional knockout mice (Dot1lAC ), Dot1l and Edn1 double-knockout mice (DEAC ), and Edn1 connecting tubule/collecting duct-specific conditional knockout mice (Edn1AC ), under three experimental conditions (streptozotocin-induced diabetes, during normal aging, and after unilateral ureteral obstruction). We used several approaches (colocalization, glutathione S-transferase pulldown, coimmunoprecipitation, yeast two-hybrid, gel shift, and chromatin immunoprecipitation assays) to identify and confirm interaction of Dot1a (the major Dot1l splicing variant in the mouse kidney) with histone deacetylase 2 (HDAC2), as well as the function of the Dot1a-HDAC2 complex in regulating Edn1 transcription. RESULTS: In each case, Dot1lAC mice developed more pronounced kidney fibrosis and kidney malfunction compared with wild-type mice. These Dot1lAC phenotypes were ameliorated in the double-knockout DEAC mice. The interaction between Dot1a and HDAC2 prevents the Dot1a-HDAC2 complex from association with DNA, providing a counterbalancing mechanism governing Edn1 transcription by modulating H3 K79 dimethylation and H3 acetylation at the Edn1 promoter. CONCLUSIONS: Our study confirms Dot1l to be a genetic and epigenetic modifier of kidney fibrosis, reveals a new mechanism regulating Edn1 transcription by Dot1a and HDAC2, and reinforces endothelin 1 as a therapeutic target of kidney fibrosis.


Asunto(s)
Endotelina-1/genética , Histona Desacetilasa 2/fisiología , N-Metiltransferasa de Histona-Lisina/fisiología , Riñón/patología , Factores de Edad , Animales , Núcleo Celular/metabolismo , Células Cultivadas , ADN/metabolismo , Fibrosis , Histonas/metabolismo , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas , Regulación hacia Arriba
5.
Int J Mol Sci ; 20(20)2019 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-31627491

RESUMEN

The current study was undertaken to investigate whether histone deacetylases (HDACs) can modulate the viability of retinal ganglion cells (RGCs) and the activity of glial cells in a mouse model of retinal ischemia-reperfusion (IR) injury. C57BL/6J mice were subjected to constant elevation of intraocular pressure for 60 min to induce retinal IR injury. Expression of macroglial and microglial cell markers (GFAP and Iba1), hypoxia inducing factor (HIF)-1α, and histone acetylation was analyzed after IR injury. To investigate the role of HDACs in the activation of glial cells, overexpression of HDAC1 and HDAC2 isoforms was performed. To determine the effect of HDAC inhibition on RGC survival, trichostatin-A (TSA, 2.5 mg/kg) was injected intraperitoneally. After IR injury, retinal GFAP, Iba1, and HIF-1α were upregulated. Conversely, retinal histone acetylation was downregulated. Notably, adenoviral-induced overexpression of HDAC2 enhanced glial activation following IR injury, whereas overexpression of HDAC1 did not significantly affect glial activation. TSA treatment significantly increased RGC survival after IR injury. Our results suggest that increased activity of HDAC2 is closely related to glial activation in a mouse model of retinal IR injury and inhibition of HDACs by TSA showed neuroprotective potential in retinas with IR injuries.


Asunto(s)
Histona Desacetilasa 2/fisiología , Neuroglía/fisiología , Daño por Reperfusión/patología , Retina/patología , Células Ganglionares de la Retina/patología , Acetilación , Animales , Regulación hacia Abajo , Proteína Ácida Fibrilar de la Glía/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Histona Desacetilasa 2/genética , Histona Desacetilasa 2/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Histonas/metabolismo , Ácidos Hidroxámicos/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Neuroglía/metabolismo , Retina/metabolismo , Regulación hacia Arriba
6.
J Pharmacol Sci ; 140(2): 137-143, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31255517

RESUMEN

Stroke is one of the leading causes of long-term disabilities worldwide. Although exposure to an enriched environment (EE) initiated in the acute phase after stroke has neuroprotective effects and improves stroke outcome, it remains unclear whether EE has positive effects when started in a delayed time frame. Here we show that exposure to EE in the delayed phase notably ameliorates the ischemia-induced impairments in neurological functions and spatial learning and memory. In addition, delayed EE exposure after stroke significantly promotes the survival and neuronal fate choice of hippocampal newborn cells, increases synaptic density of hippocampal mature neurons, and enhances the migration of subventricular zone (SVZ)-derived cells towards the ischemic striatum. Histone deacetylase 2 (HDAC2), synapse-associated proteins and brain-derived neurotrophic factor (BDNF) may respectively mediate these roles of delayed EE. Our findings provide the suggestion that exposure to EE initiated in the delayed phase after stroke promotes plastic changes via affecting neurogenesis, synaptogenesis and neuronal migration, and thus improves stroke outcome. Because EE initiated earlier than 24 h is clinically feasible, our work could be introduced into clinical studies of stroke directly and may provide stroke survivors with a new strategy for their functional recovery.


Asunto(s)
Ambiente , Memoria , Recuperación de la Función , Aprendizaje Espacial , Rehabilitación de Accidente Cerebrovascular/métodos , Accidente Cerebrovascular/fisiopatología , Accidente Cerebrovascular/psicología , Animales , Factor Neurotrófico Derivado del Encéfalo/fisiología , Movimiento Celular , Hipocampo/citología , Histona Desacetilasa 2/fisiología , Masculino , Neurogénesis , Ratas Sprague-Dawley , Factores de Tiempo
7.
Brain Pathol ; 29(2): 164-175, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30028551

RESUMEN

Histone deacetylases (HDACs) catalyze acetyl group removal from histone proteins, leading to altered chromatin structure and gene expression. HDAC2 is highly expressed in adult brain, and HDAC2 levels are elevated in Alzheimer's disease (AD) brain. We previously reported that neuron-specific splice isoforms of Endophilin-B1 (Endo-B1) promote neuronal survival, but are reduced in human AD brain and mouse models of AD and stroke. Here, we demonstrate that HDAC2 suppresses Endo-B1 expression. HDAC2 knockdown or knockout enhances expression of Endo-B1. Conversely, HDAC2 overexpression decreases Endo-B1 expression. We also demonstrate that neurons exposed to beta-amyloid increase HDAC2 and reduce histone H3 acetylation while HDAC2 knockdown prevents Aß induced loss of histone H3 acetylation, mitochondrial dysfunction, caspase-3 activation, and neuronal death. The protective effect of HDAC2 knockdown was abrogated by Endo-B1 shRNA and in Endo-B1-null neurons, suggesting that HDAC2-induced neurotoxicity is mediated through suppression of Endo-B1. HDAC2 overexpression also modulates neuronal expression of mitofusin2 (Mfn2) and mitochondrial fission factor (MFF), recapitulating the pattern of change observed in AD. HDAC2 knockout mice demonstrate reduced injury in the middle cerebral artery occlusion with reperfusion (MCAO/R) model of cerebral ischemia demonstrating enhanced neuronal survival, minimized loss of Endo-B1, and normalized expression of Mfn2. These findings support the hypothesis that HDAC2 represses Endo-B1, sensitizing neurons to mitochondrial dysfunction and cell death in stroke and AD.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Histona Desacetilasa 2/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/genética , Péptidos beta-Amiloides/metabolismo , Péptidos beta-Amiloides/toxicidad , Animales , Encéfalo/metabolismo , Isquemia Encefálica/metabolismo , Modelos Animales de Enfermedad , GTP Fosfohidrolasas/metabolismo , Regulación de la Expresión Génica/genética , Histona Desacetilasa 2/genética , Histona Desacetilasa 2/metabolismo , Histona Desacetilasas/genética , Histonas/genética , Isquemia/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas/fisiología , Accidente Cerebrovascular/fisiopatología
8.
Neurobiol Aging ; 68: 134-141, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29803514

RESUMEN

Histone deacetylase (HDAC) inhibitors have been widely reported to have considerable therapeutic potential in a host of neurodegenerative diseases. However, HDAC inhibitor selectivity and specificity in specific cell classes have been a source of much debate. To address the role of HDAC2 in specific cell classes, and in disease, we examined glial protein and mRNA levels in the substantia nigra (SN) of Parkinson's disease (PD) and normal controls (NCs) by immunohistochemistry and laser captured microdissection followed by quantitative real time polymerase chain reaction. Differential expression analysis in immunohistochemically defined laser capture microglia revealed significant upregulation of HDAC2 in the PD SN compared to NC subjects. Complementary in vivo evidence reveals significant upregulation of HDAC2 protein levels in PD SN microglia compared to NC subjects. Correspondingly, human immortalized telencephalic/mesencephalic microglial cells reveal significant upregulation of HDAC2 in the presence of the potent microglial activator lipopolysaccharide. These data provide evidence that selective inhibition of HDAC2 in PD SN microglia could be a promising approach to treat microglial-initiated nigral dopaminergic neuronal cell loss in PD.


Asunto(s)
Histona Desacetilasa 2/antagonistas & inhibidores , Histona Desacetilasa 2/metabolismo , Microglía/enzimología , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/genética , Sustancia Negra/citología , Sustancia Negra/enzimología , Anciano , Anciano de 80 o más Años , Células Cultivadas , Neuronas Dopaminérgicas/patología , Femenino , Histona Desacetilasa 2/fisiología , Humanos , Inmunohistoquímica , Captura por Microdisección con Láser , Masculino , Terapia Molecular Dirigida , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/patología , Reacción en Cadena de la Polimerasa , Regulación hacia Arriba
9.
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
10.
Pharmacology ; 99(1-2): 27-39, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27673327

RESUMEN

Recent studies indicate that histone deacetylases (HDACs) activity is associated with the development and progression of cardiac hypertrophy. In this study, we investigated the effects of a HDACs inhibitor, valproic acid sodium (VPA), on cardiac remodeling and the differential expression of HDACs in left ventricles (LVs) of renovascular hypertensive rats. Renovascular hypertension was induced in rats by the two-kidney two-clip (2K2C) method. Cardiac remodeling, heart function and the differential expression of HDACs were examined at different weeks after 2K2C operation. The effects of VPA on cardiac remodeling, the expressions of HDACs, transforming growth factor-beta 1 (TGF-ß1) and connective tissue growth factor (CTGF) in LV were investigated. The expressions of atrial natriuretic factor, ß-myosin heavy chain, HDAC2 and HDAC8 increased in LV of 2K2C rats at 4, 8, 12 weeks after operation. Cardiac dysfunction, cardiac hypertrophy and fibrosis were markedly attenuated by VPA treatment in 2K2C rats. Further studies revealed that VPA inhibited the expressions of HDAC2, HDAC8, TGF-ß1 and CTGF in LV of 2K2C rats. In summary, these data indicate that HDAC2 and HDAC8 play a key role in cardiac remodeling in renovascular hypertensive rats and that VPA attenuates hypertension and cardiac remodeling. The effect of VPA is possibly exerted via decreasing HDAC2, HDAC8, TGF-ß1 and CTGF expressions in LV of 2K2C rats.


Asunto(s)
Histona Desacetilasa 2/fisiología , Histona Desacetilasas/fisiología , Hipertensión Renovascular/tratamiento farmacológico , Hipertensión Renovascular/enzimología , Ácido Valproico/uso terapéutico , Remodelación Ventricular/efectos de los fármacos , Animales , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Histona Desacetilasa 2/antagonistas & inhibidores , Hipertensión Renovascular/patología , Masculino , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Resultado del Tratamiento , Ácido Valproico/farmacología , Remodelación Ventricular/fisiología
11.
Cancer Chemother Pharmacol ; 78(6): 1289-1296, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27832326

RESUMEN

Histone deacetylases (HDACs) can regulate the progression of various cancers, while their roles in glioblastoma multiforme (GBM) are not well known. Our present study investigated the expression of class I HDACs (HDAC1, 2, 3, 8) in GBM U87, A172, U251, and LN229 cells and compared their levels with that in primary normal human astrocytes (NHA) cells. It showed that HDAC2 expression is significantly up-regulated in GBM cells. Silencing of HDAC2 via its specific siRNAs can suppress the in vitro proliferation, migration, and invasion of GBM U87 and A172 cells. Furthermore, silencing of HDAC2 can increase the sensitivity of GBM cells to temozolomide (TMZ), a standard-of-care during clinical GBM treatment. This might be due to that si-HDAC can significantly down-regulate the mRNA and protein expression of MRP1, while has no effect on ABCB1 and ABCG2. Schisandrin B (Sch B), a specific inhibitor of MRP1, can further increase the TMZ sensitivity in HDAC2-knocked down GBM cells. Collectively, our data revealed that targeted HDAC2 can suppress the malignancy of GBM cells and increase their sensitivity of TMZ via down-regulation of MRP1. It suggested that HDAC2 might be a potential target for GBM therapy and improvement in TMZ therapy efficiency.


Asunto(s)
Antineoplásicos Alquilantes/farmacología , Neoplasias Encefálicas/patología , Movimiento Celular , Proliferación Celular , Dacarbazina/análogos & derivados , Glioblastoma/patología , Histona Desacetilasa 2/fisiología , Neoplasias Encefálicas/enzimología , Neoplasias Encefálicas/terapia , Línea Celular Tumoral , Dacarbazina/farmacología , Glioblastoma/enzimología , Glioblastoma/terapia , Histona Desacetilasa 2/antagonistas & inhibidores , Humanos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/análisis , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/fisiología , Invasividad Neoplásica , ARN Interferente Pequeño/genética , Temozolomida
12.
Neurotox Res ; 30(1): 32-40, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26721795

RESUMEN

Methamphetamine (METH) administration alters gene expression in the nucleus accumbens (NAc). We recently demonstrated that an acute METH injection produced prolonged increases in the expression of immediate early genes in the NAc of HDAC2-deficient mice, suggesting that HDAC2 might be an important regulator of gene expression in the rodent brain. Here, we tested the possibility that HDAC2 deletion might also impact METH-induced changes in the expression of various HDAC classes in the NAc. Wild-type (WT) and HDAC2 knockout (KO) mice were given a METH (20 mg/kg) injection, and NAc tissue was collected at 1, 2, and 8 h post treatment. We found that METH decreased HDAC3, HDAC4, HDAC7, HDAC8, and HDAC11 mRNA expression but increased HDAC6 mRNA levels in the NAc of WT mice. In contrast, the METH injection increased HDAC3, HDAC4, HDAC7, HDAC8, and HDAC11 mRNA levels in HDAC2KO mice. These observations suggest that METH may induce large-scale transcriptional changes in the NAc by regulating the expression of several HDACs, in part, via HDAC2-dependent mechanisms since some of the HDACs showed differential responses between the two genotypes. Our findings further implicate HDACs as potential novel therapeutic targets for neurotoxic complications associated with the abuse of certain psychostimulants.


Asunto(s)
Regulación hacia Abajo/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Histona Desacetilasa 2/fisiología , Metanfetamina/administración & dosificación , Metanfetamina/farmacología , Núcleo Accumbens/efectos de los fármacos , Núcleo Accumbens/metabolismo , Animales , Regulación Enzimológica de la Expresión Génica/genética , Histona Desacetilasa 2/biosíntesis , Histona Desacetilasa 2/genética , Masculino , Ratones , Ratones Noqueados , Regulación hacia Arriba/efectos de los fármacos
13.
Science ; 351(6268): aac6633, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26722000

RESUMEN

Eusocial insects organize themselves into behavioral castes whose regulation has been proposed to involve epigenetic processes, including histone modification. In the carpenter ant Camponotus floridanus, morphologically distinct worker castes called minors and majors exhibit pronounced differences in foraging and scouting behaviors. We found that these behaviors are regulated by histone acetylation likely catalyzed by the conserved acetyltransferase CBP. Transcriptome and chromatin analysis in brains of scouting minors fed pharmacological inhibitors of CBP and histone deacetylases (HDACs) revealed hundreds of genes linked to hyperacetylated regions targeted by CBP. Majors rarely forage, but injection of a HDAC inhibitor or small interfering RNAs against the HDAC Rpd3 into young major brains induced and sustained foraging in a CBP-dependent manner. Our results suggest that behavioral plasticity in animals may be regulated in an epigenetic manner via histone modification.


Asunto(s)
Hormigas , Conducta Animal , Epigénesis Genética , Histona Desacetilasa 2 , Conducta Social , Animales , Acetilación , Hormigas/efectos de los fármacos , Hormigas/genética , Hormigas/fisiología , Cromatina/metabolismo , Histona Desacetilasa 2/antagonistas & inhibidores , Histona Desacetilasa 2/genética , Histona Desacetilasa 2/fisiología , Inhibidores de Histona Desacetilasas/farmacología , Procesamiento Proteico-Postraduccional , Transcriptoma
14.
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
15.
Am J Respir Crit Care Med ; 193(2): 143-53, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26426522

RESUMEN

RATIONALE: Corticosteroid resistance is a major barrier to the effective treatment of chronic obstructive pulmonary disease (COPD). Several molecular mechanisms have been proposed, such as activations of the phosphoinositide-3-kinase/Akt pathway and p38 mitogen-activated protein kinase. However, the mechanism for corticosteroid resistance is still not fully elucidated. OBJECTIVES: To investigate the role of mammalian target of rapamycin (mTOR) in corticosteroid sensitivity in COPD. METHODS: The corticosteroid sensitivity of peripheral blood mononuclear cells collected from patients with COPD, smokers, and nonsmoking control subjects, or of human monocytic U937 cells exposed to cigarette smoke extract (CSE), was quantified as the dexamethasone concentration required to achieve 30% inhibition of tumor necrosis factor-α-induced CXCL8 production in the presence or absence of the mTOR inhibitor rapamycin. mTOR activity was determined as the phosphorylation of p70 S6 kinase, using Western blotting. MEASUREMENTS AND MAIN RESULTS: mTOR activity was increased in peripheral blood mononuclear cells from patients with COPD, and treatment with rapamycin inhibited this as well as restoring corticosteroid sensitivity. In U937 cells, CSE stimulated mTOR activity and c-Jun expression, but pretreatment with rapamycin inhibited both and also reversed CSE-induced corticosteroid insensitivity. CONCLUSIONS: mTOR inhibition by rapamycin restores corticosteroid sensitivity via inhibition of c-Jun expression, and thus mTOR is a potential novel therapeutic target for COPD.


Asunto(s)
Corticoesteroides/farmacología , Resistencia a Medicamentos/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Proteínas Proto-Oncogénicas c-jun/efectos de los fármacos , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/efectos de los fármacos , Corticoesteroides/uso terapéutico , Anciano , Resistencia a Medicamentos/inmunología , Femenino , Histona Desacetilasa 2/efectos de los fármacos , Histona Desacetilasa 2/fisiología , Humanos , Inmunosupresores/inmunología , Inmunosupresores/farmacología , Inmunosupresores/uso terapéutico , Masculino , Persona de Mediana Edad , Estrés Oxidativo/fisiología , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/fisiología , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/fisiología , Proteínas Proto-Oncogénicas c-jun/fisiología , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Sirolimus/inmunología , Sirolimus/uso terapéutico , Fumar/efectos adversos , Fumar/fisiopatología , Serina-Treonina Quinasas TOR/fisiología , Células U937/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/fisiología
16.
FASEB J ; 29(5): 2070-80, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25648995

RESUMEN

Histone deacetylases (HDACs) are posttranslational modifiers that deacetylate proteins. Despite their crucial role in numerous biological processes, the use of broad-range HDAC inhibitors (HDACi), has shown clinical efficacy. However, undesired side effects highlight the necessity to better understand the biology of different HDACs and target the relevant HDACs. Using a novel mouse model, in which HDAC1 and HDAC2 can be simultaneously deleted in the intestine of adult mice, we show that the simultaneous deletion of HDAC1 and HDAC2 leads to a rapid loss of intestinal homeostasis. Importantly, this deletion cannot be sustained, and 8 days after initial ablation, stem cells that have escaped HDAC1 or HDAC2 deletion swiftly repopulate the intestinal lining. In vitro ablation of HDAC1 and HDAC2 using intestinal organoid cultures resulted in a down-regulation of multiple intestinal stem cell markers and functional loss of clonogenic capacity. Importantly, treatment of wild-type organoids with class I-specific HDACi MS-275 also induced a similar loss of stemness, providing a possible rationale for the gastrointestinal side effects often observed in HDACi-treated patients. In conclusion, these data show that HDAC1 and HDAC2 have a redundant function and are essential to maintain intestinal homeostasis.


Asunto(s)
Histona Desacetilasa 1/fisiología , Histona Desacetilasa 2/fisiología , Homeostasis/fisiología , Intestinos/citología , Células Madre/citología , Animales , Benzamidas/farmacología , Biomarcadores/metabolismo , Western Blotting , Diferenciación Celular , Células Cultivadas , Femenino , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Histona Desacetilasa 1/antagonistas & inhibidores , Histona Desacetilasa 2/antagonistas & inhibidores , Inhibidores de Histona Desacetilasas/farmacología , Homeostasis/efectos de los fármacos , Humanos , Técnicas para Inmunoenzimas , Intestinos/efectos de los fármacos , Intestinos/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Técnicas de Cultivo de Órganos , Piridinas/farmacología , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/efectos de los fármacos , Células Madre/enzimología
17.
Nucleic Acids Res ; 43(4): 2008-21, 2015 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-25653159

RESUMEN

During skeletal muscle differentiation, the activation of some tissue-specific genes occurs immediately while others are delayed. The molecular basis controlling temporal gene regulation is poorly understood. We show that the regulatory sequences, but not other regions of genes expressed at late times of myogenesis, are in close physical proximity in differentiating embryonic tissue and in differentiating culture cells, despite these genes being located on different chromosomes. Formation of these inter-chromosomal interactions requires the lineage-determinant MyoD and functional Brg1, the ATPase subunit of SWI/SNF chromatin remodeling enzymes. Ectopic expression of myogenin and a specific Mef2 isoform induced myogenic differentiation without activating endogenous MyoD expression. Under these conditions, the regulatory sequences of late gene loci were not in close proximity, and these genes were prematurely activated. The data indicate that the spatial organization of late genes contributes to temporal regulation of myogenic transcription by restricting late gene expression during the early stages of myogenesis.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Desarrollo de Músculos/genética , Elementos Reguladores de la Transcripción , Animales , Línea Celular , Ensamble y Desensamble de Cromatina , Cromosomas de los Mamíferos , ADN Helicasas/fisiología , Histona Desacetilasa 2/fisiología , Ratones , Músculo Esquelético/metabolismo , Proteína MioD/fisiología , Proteínas Nucleares/fisiología , Regiones Promotoras Genéticas , Proteínas Represoras/metabolismo , Factores de Transcripción/fisiología
18.
Oncogene ; 34(34): 4509-18, 2015 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-25435372

RESUMEN

Liver receptor homolog-1 (LRH-1, NR5A2) is an orphan nuclear receptor that has an essential role in cancer progression, notably in breast cancer. Although its role in promoting cancer cell proliferation and migration is well documented, the molecular basis is not completely established. Here, we report that LRH-1 inhibition affects two- and three-dimensional cell proliferation of different types of breast cancer cells, including estrogen receptor α (ERα)-positive and triple-negative cells. This phenotype is accompanied by the upregulation of the cyclin-dependent kinase inhibitor CDKN1A (aka p21(CIP1/WAF1)) in a p53-independent manner. Chromatin immunoprecipitation analysis shows that LRH-1 cooperates with FOXA1 and binds directly to CDKN1A promoter and a distal regulatory region found at -62 kb from its transcriptional start sites, allowing repression of CDKN1A transcription. LRH-1 or FOXA1 depletion induces CDKN1A upregulation by removing histone deacetylase 2 from the promoter and distal regulatory elements and permitting histone acetylation in these regions. Analysis of breast cancer samples reveals that a high LRH-1 level is inversely correlated with CDKN1A expression in breast cancer patients and is associated with poor prognosis. This study reveals a novel mechanism of control of cell proliferation by LRH-1 regulating CDKN1A transcription in breast cancer cells, independent of ERα and p53 status. Targeting LRH-1 may provide an attractive prospect for treatment of tumors that are resistant to hormonal and targeted therapy.


Asunto(s)
Neoplasias de la Mama/patología , Proliferación Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Regulación Neoplásica de la Expresión Génica , Receptores Citoplasmáticos y Nucleares/fisiología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/mortalidad , Línea Celular Tumoral , Receptor alfa de Estrógeno/análisis , Femenino , Factor Nuclear 3-alfa del Hepatocito/fisiología , Histona Desacetilasa 2/fisiología , Humanos , Proteína p53 Supresora de Tumor/fisiología
19.
Biochim Biophys Acta ; 1846(2): 524-38, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25072962

RESUMEN

Histone deacetylase 2 (HDAC2) regulates biological processes by deacetylation of histones and non-histone proteins. HDAC2 is overexpressed in numerous cancer types, suggesting general cancer-relevant functions of HDAC2. In human tumors the TP53 gene encoding p53 is frequently mutated and wild-type p53 is often disarmed. Molecular pathways inactivating wild-type p53 often remain to be defined and understood. Remarkably, current data link HDAC2 to the regulation of the tumor suppressor p53 by deacetylation and to the maintenance of genomic stability. Here, we summarize recent findings on HDAC2 overexpression in solid and hematopoietic cancers with a focus on mechanisms connecting HDAC2 and p53 in vitro and in vivo. In addition, we present an evidence-based model that integrates molecular pathways and feedback loops by which p53 and further transcription factors govern the expression and the ubiquitin-dependent proteasomal degradation of HDAC2 and of p53 itself. Understanding the interactions between p53 and HDAC2 might aid in the development of new therapeutic approaches against cancer.


Asunto(s)
Carcinogénesis , Histona Desacetilasa 2/fisiología , Neoplasias/enzimología , Proteína p53 Supresora de Tumor/fisiología , Acetilación , Animales , Humanos
20.
Proc Natl Acad Sci U S A ; 111(27): 9840-5, 2014 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-24958871

RESUMEN

Histone deacetylases 1 and 2 (HDAC1/2) form the core catalytic components of corepressor complexes that modulate gene expression. In most cell types, deletion of both Hdac1 and Hdac2 is required to generate a discernible phenotype, suggesting their activity is largely redundant. We have therefore generated an ES cell line in which Hdac1 and Hdac2 can be inactivated simultaneously. Loss of HDAC1/2 resulted in a 60% reduction in total HDAC activity and a loss of cell viability. Cell death is dependent upon cell cycle progression, because differentiated, nonproliferating cells retain their viability. Furthermore, we observe increased mitotic defects, chromatin bridges, and micronuclei, suggesting HDAC1/2 are necessary for accurate chromosome segregation. Consistent with a critical role in the regulation of gene expression, microarray analysis of Hdac1/2-deleted cells reveals 1,708 differentially expressed genes. Significantly for the maintenance of stem cell self-renewal, we detected a reduction in the expression of the pluripotent transcription factors, Oct4, Nanog, Esrrb, and Rex1. HDAC1/2 activity is regulated through binding of an inositol tetraphosphate molecule (IP4) sandwiched between the HDAC and its cognate corepressor. This raises the important question of whether IP4 regulates the activity of the complex in cells. By rescuing the viability of double-knockout cells, we demonstrate for the first time (to our knowledge) that mutations that abolish IP4 binding reduce the activity of HDAC1/2 in vivo. Our data indicate that HDAC1/2 have essential and pleiotropic roles in cellular proliferation and regulate stem cell self-renewal by maintaining expression of key pluripotent transcription factors.


Asunto(s)
División Celular/fisiología , Células Madre Embrionarias/enzimología , Histona Desacetilasa 1/fisiología , Histona Desacetilasa 2/fisiología , Células Madre Pluripotentes/enzimología , Acetilación , Animales , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Regulación de la Expresión Génica , Histonas/metabolismo , Ratones , Ratones Noqueados , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...