Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
Res Sq ; 2024 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-39011107

RESUMO

Prader-Willi Syndrome (PWS) is caused by loss of expression of paternally expressed genes in the human 15q11.2-q13 imprinting domain. A set of imprinted genes that are active on the paternal but silenced on the maternal chromosome are intricately regulated by a bipartite imprinting center (PWS-IC) located in the PWS imprinting domain. In past work, we discovered that euchromatic histone lysine N-methyltransferase-2 (EHMT2/G9a) inhibitors were capable of un-silencing PWS-associated genes by restoring their expression from the maternal chromosome. Here, in mice lacking the Ehmt2 gene, we document unsilencing of the imprinted Snrpn/Snhg14 gene on the maternal chromosome in the late embryonic and postnatal brain. Using PWS and Angelman syndrome patient derived cells with either paternal or maternal deletion of 15q11-q13, we have found that chromatin of maternal PWS-IC is closed and has compact 3D folding confirmation. We further show that a new and distinct noncoding RNA preferentially transcribed from upstream of the PWS-IC interacts with EHMT2 and forms a heterochromatin complex to silence gene expression of SNRPN in CIS on maternal chromosome. Taken together, these findings demonstrate that allele-specific recruitment of EHMT2 is required to maintain the maternal imprints. Our findings provide novel mechanistic insights and support a new model for imprinting maintenance of the PWS imprinted domain.

2.
Mol Ther ; 2024 May 24.
Artigo em Inglês | MEDLINE | ID: mdl-38796700

RESUMO

Prader-Willi syndrome (PWS) is the prototypic genomic disorder resulting from deficiency of paternally expressed genes in the human chromosome 15q11-q13 region. The unique molecular mechanism involving epigenetic modifications renders PWS as the most attractive candidate to explore a proof-of-concept of epigenetic therapy in humans. The premise is that epigenetic modulations could reactivate the repressed PWS candidate genes from the maternal chromosome and offer therapeutic benefit. Our prior study identifies an EHMT2/G9a inhibitor, UNC0642, that reactivates the expression of PWS genes via reduction of H3K9me2. However, low brain permeability and poor oral bioavailability of UNC0642 preclude its advancement into translational studies in humans. In this study, a newly developed inhibitor, MS152, modified from the structure of UNC0642, has better brain penetration and greater potency and selectivity against EHMT2/G9a. MS152 reactivated maternally silenced PWS genes in PWS patient fibroblasts and in brain and liver tissues of PWS mouse models. Importantly, the molecular efficacy of oral administration is comparable with the intraperitoneal route. MS152 treatment in newborns ameliorates the perinatal lethality and poor growth, maintaining reactivation in a PWS mouse model at postnatal 90 days. Our findings provide strong support for MS152 as a first-in-class inhibitor to advance the epigenetic therapy of PWS in humans.

3.
Mol Psychiatry ; 28(8): 3182-3193, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37626134

RESUMO

Genomic imprinting disorders are caused by the disruption of genomic imprinting processes leading to a deficit or increase of an active allele. Their unique molecular mechanisms underlying imprinted genes offer an opportunity to investigate epigenetic-based therapy for reactivation of an inactive allele or reduction of an active allele. Current treatments are based on managing symptoms, not targeting the molecular mechanisms underlying imprinting disorders. Here, we highlight molecular approaches of therapeutic candidates in preclinical and clinical studies for individual imprinting disorders. These include the significant progress of discovery and testing of small molecules, antisense oligonucleotides, and CRISPR mediated genome editing approaches as new therapeutic strategies. We discuss the significant challenges of translating these promising therapies from the preclinical stage to the clinic, especially for genome editing based approaches.


Assuntos
Edição de Genes , Impressão Genômica , Impressão Genômica/genética , Metilação de DNA
4.
Int Immunopharmacol ; 118: 110132, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37023698

RESUMO

OBJECTIVE: Ankylosing spondylitis (AS) exhibits paradoxical bone features typically characterized by new bone formation and systemic bone loss. Although abnormal kynurenine (Kyn), a tryptophan metabolite, has been closely linked to the disease activity of AS, the distinct role of its pathological bone features remains unknown. METHODS: Kynurenine sera level was collected from healthy control (HC; n = 22) and AS (n = 87) patients and measured by ELISA. In the AS group, we analyzed and compared the Kyn level based on the modified stoke ankylosing spondylitis spinal score (mSASSS), MMP13, and OCN. Under osteoblast differentiation, the treatment with Kyn in AS-osteoprogenitors conducted cell proliferation, alkaline phosphatase activity, bone mineralization-related alizarin red s (ARS), von kossa (VON), hydroxyapatite (HA) staining, and mRNA expression markers (ALP, RUNX2, OCN, and OPG) for bone formation. TRAP and F-actin staining was used for osteoclast formation of mouse osteoclast precursors. RESULTS: Kyn sera level was significantly elevated in the AS group compared to the HC. In addition, Kyn sera level was correlated with mSASSS (r = 0.03888, p = 0.067), MMP13 (r = 0.0327, p = 0.093), and OCN (r = 0.0436, p = 0.052). During osteoblast differentiation, treatment with Kyn exhibited no difference in cell proliferation and alkaline phosphate (ALP) activity for bone matrix maturation but promoted ARS, VON, and HA staining for bone mineralization. Interestingly, osteoprotegerin (OPG) and OCN expressions of AS-osteoprogenitors were augmented in the Kyn treatment during differentiation. In growth medium, Kyn treatment of AS-osteoprogenitors resulted in induction of OPG mRNA, protein expression, and Kyn-response genes (AhRR, CYP1b1, and TIPARP). Secreted OPG proteins were observed in the supernatant of AS-osteoprogenitors treated with Kyn. Notably, the supernatant of Kyn-treated AS-osteoprogenitors interrupted the RANKL-mediated osteoclastogenesis of mouse osteoclast precursor such as TRAP-positive osteoclast formation, NFATc1 expression, and osteoclast differentiation markers. CONCLUSION: Our results revealed that elevated Kyn level increased the bone mineralization of osteoblast differentiation in AS and decreased RANKL-mediated osteoclast differentiation by inducing OPG expression. Out study have implication for potential coupling factors linking osteoclast and osteoblast where abnormal Kyn level could be involved in pathological bone features of AS.


Assuntos
Cinurenina , Espondilite Anquilosante , Animais , Camundongos , Cinurenina/metabolismo , Metaloproteinase 13 da Matriz/metabolismo , Osteoblastos/metabolismo , Regulação da Expressão Gênica , Espondilite Anquilosante/metabolismo , Osteoclastos/metabolismo , Osteoprotegerina/genética , Osteoprotegerina/metabolismo , Diferenciação Celular , RNA Mensageiro/metabolismo , Ligante RANK/metabolismo
5.
Small Methods ; 6(7): e2200430, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35616025

RESUMO

Protective surface coatings on Si anodes are promising for improving the electrochemical performance of lithium-ion batteries (LIBs). Nevertheless, most coating materials have severe issues, including low initial coulombic efficiency, structural fracture, morphology control, and complicated synthetic processing. In this study, a multifunctional TiO2- x /TiO1- y Ny (TTN) formed via a facile and scalable synthetic process is applied as a coating material for Si anodes. A thin layer of amorphous TiO2 is uniformly coated onto Si nanoparticles by a simple sol-gel method and then converted into a two phase TiO2- x /TiO1- y Ny via nitridation. The lithiated TiO2-x provides high ionic and electrical conductivity, while TiO1-y Ny can improve mechanical strength that alleviates volume change of Si to address capacity fading issue. Owing to these synergetic advantages, TiO2- x /TiO1- y Ny -coated Si (Si@TTN) exhibits excellent electrochemical properties, including a high charge capacity of 1650 mA h g-1 at 0.1 A g-1 and 84% capacity retention after 100 cycles at 1 A g-1 . Moreover, a significantly enhanced rate performance can be achieved at a high current density. This investigation presents a facile and effective coating material to use as the high-capacity silicon anode in the emerging Si anode technology in LIBs.

6.
Nanomaterials (Basel) ; 13(1)2022 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-36616042

RESUMO

Surface coating approaches for silicon (Si) have demonstrated potential for use as anodes in lithium-ion batteries (LIBs) to address the large volume change and low conductivity of Si. However, the practical application of these approaches remains a challenge because they do not effectively accommodate the pulverization of Si during cycling or require complex processes. Herein, Si-embedded titanium oxynitride (Si-TiON) was proposed and successfully fabricated using a spray-drying process. TiON can be uniformly coated on the Si surface via self-assembly, which can enhance the Si utilization and electrode stability. This is because TiON exhibits high mechanical strength and electrical conductivity, allowing it to act as a rigid and electrically conductive matrix. As a result, the Si-TiON electrodes delivered an initial reversible capacity of 1663 mA h g-1 with remarkably enhanced capacity retention and rate performance.

7.
Neurobiol Stress ; 15: 100373, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34401409

RESUMO

Depression is accompanied by neuronal atrophy and decreased neuroplasticity. Leucine-rich glioma-inactivated protein 1 (LGI1), a metastasis suppressor, plays an important role in the development of CNS synapses. We found that LGI1 expression was reduced in the hippocampi of mice that underwent chronic unpredictable stress (CUS), and could be rescued by the antidepressant, fluoxetine. Recombinant soluble neuritin, an endogenous protein previously implicated in antidepressant-like behaviors, elevated hippocampal LGI1 expression in a manner dependent on histone deacetylase 5 (HDAC5) phosphorylation. Accordingly, Nrn1 flox/flox ;Pomc-cre (Nrn1 cOE) mice, which conditionally overexpress neuritin, displayed increases in hippocampal LGI1 level under CUS and exhibited resilience to CUS that were blocked by hippocampal depletion of LGI1. Interestingly, neuritin-mediated LGI1 expression was inhibited by HNMPA-(AM)3, an insulin receptor inhibitor, as was neuritin-mediated HDAC5 phosphorylation. We thus establish hippocampal LGI1 as an effector of neurite outgrowth and stress resilience, and suggest that HDAC5-LGI1 plays a critical role in ameliorating pathological depression.

8.
Exp Neurobiol ; 28(4): 537-546, 2019 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-31495082

RESUMO

Silent information regulator 2 (Sirtuin2 / SIRT2) is a NAD+-dependent deacetylase that regulates the cellular oxidative stress response. It modulates transcriptional silencing and protein stability through deacetylation of target proteins including histones. Previous studies have shown that SIRT2 plays a role in mood disorders and hippocampus-dependent cognitive function, but the underlying neurobiological mechanism is poorly understood. Here, we report that chronic stress suppresses SIRT2 expression in the hippocampus. Molecular and biochemical analyses indicate that the stress-induced decrease in the SIRT2 expression downregulates synaptic plasticity-related genes in the hippocampus through the increase of euchromatic histone-lysine N-methyltransferase 2 (Ehmt2) (also known as G9a). shRNA-mediated knockdown of SIRT2 in the dentate gyrus alters the expression of synaptic plasticity- related genes in a way similar to those induced by chronic stress, and produces depression-like behaviors. Our results indicate that SIRT2 plays an important role in the response to stress, thereby modulating depression-like behaviors.

9.
Trends Pharmacol Sci ; 40(9): 605-608, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31353046

RESUMO

Prader-Willi syndrome (PWS) is a neurobehavioral and epigenetic disorder caused by the deficiency of paternally expressed genes in the chromosome 15q11-q13. This unique molecular defect renders PWS an exciting opportunity to explore epigenetic therapy. Here, we briefly highlight recent findings from small molecule screening and CRISPR/Cas9-mediated epigenome editing that offer promising therapeutic options along with the challenges that remain in developing a successful epigenetic therapy for PWS in humans.


Assuntos
Síndrome de Prader-Willi/genética , Síndrome de Prader-Willi/terapia , Epigênese Genética , Epigenômica/métodos , Humanos , Síndrome de Prader-Willi/tratamento farmacológico
10.
Transl Res ; 208: 105-118, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30904443

RESUMO

Prader-Willi syndrome (PWS) is a complex and multisystem neurobehavioral disorder. The molecular mechanism of PWS is deficiency of paternally expressed gene gene or genes from the chromosome 15q11-q13. Due to imprinted gene regulation, the same genes in the maternal chromosome 15q11-q13 are structurally intact but transcriptionally repressed by an epigenetic mechanism. The unique molecular defect underlying PWS renders an exciting opportunity to explore epigenetic-based therapy to reactivate the expression of repressed PWS genes from the maternal chromosome. Inactivation of H3K9m3 methyltransferase SETDB1 and zinc finger protein ZNF274 results in reactivation of SNRPN and SNORD116 cluster from the maternal chromosomes in PWS patient iPSCs and iPSC-derived neurons, respectively. High content screening of small molecule libraries using cells derived from transgenic mice carrying the SNRPN-EGFP fusion protein has discovered that inhibitors of EHMT2/G9a, a histone 3 lysine 9 methyltransferase, are capable of reactivating expression of paternally expressed SNRPN and SNORD116 from the maternal chromosome, both in cultured PWS patient-derived fibroblasts and in a PWS mouse model. Treatment with an EMHT2/G9a inhibitor also rescues perinatal lethality and failure to thrive phenotypes in a PWS mouse model. These findings present the first evidence to support a proof-of-principle for epigenetic-based therapy for the PWS in humans.


Assuntos
Epigênese Genética , Síndrome de Prader-Willi/genética , Animais , Cromossomos Humanos Par 15 , Histona-Lisina N-Metiltransferase , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas Metiltransferases/metabolismo , RNA Nucleolar Pequeno/genética
11.
Proc Natl Acad Sci U S A ; 116(5): 1770-1775, 2019 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-30642955

RESUMO

Major depressive disorder (MDD) is a devastating disease that arises in a background of environmental risk factors, such as chronic stress, that produce reactive oxygen species (ROS) in the brain. The chronic stress-induced ROS production involves Ca2+ signals; however, the mechanism is poorly understood. Transient receptor potential melastatin type 2 (TRPM2) is a Ca2+-permeable cation channel that is highly expressed in the brain. Here we show that in animal models of chronic unpredictable stress (CUS), deletion of TRPM2 (Trpm2-/- ) produces antidepressant-like behaviors in mice. This phenotype correlates with reduced ROS, ROS-induced calpain activation, and enhanced phosphorylation of two Cdk5 targets including synapsin 1 and histone deacetylase 5 that are linked to synaptic function and gene expression, respectively. Moreover, TRPM2 mRNA expression is increased in hippocampal tissue samples from patients with MDD. Our findings suggest that TRPM2 is a key agent in stress-induced depression and a possible target for treating depression.


Assuntos
Transtorno Depressivo Maior/metabolismo , Estresse Fisiológico/fisiologia , Canais de Cátion TRPM/metabolismo , Animais , Cálcio/metabolismo , Expressão Gênica/fisiologia , Hipocampo/metabolismo , Masculino , Camundongos , Camundongos Knockout , Fosforilação/fisiologia , Espécies Reativas de Oxigênio/metabolismo
12.
Arthritis Res Ther ; 20(1): 115, 2018 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-29880011

RESUMO

BACKGROUND: IL-17A has recently emerged as a potential target that regulates the extensive inflammation and abnormal bone formation observed in ankylosing spondylitis (AS). Blocking IL-17A is expected to inhibit bony ankylosis. Here, we investigated the effects of anti IL-17A agents in AS. METHODS: TNFα, IL-17A, and IL-12/23 p40 levels in serum and synovial fluid from patients with ankylosing spondylitis (AS), rheumatoid arthritis (RA), osteoarthritis (OA), or healthy controls (HC) were measured by ELISA. Bone tissue samples were obtained at surgery from the facet joints of ten patients with AS and ten control (Ct) patients with noninflammatory spinal disease. The functional relevance of IL-17A, biological blockades, Janus kinase 2 (JAK2), and non-receptor tyrosine kinase was assessed in vitro with primary bone-derived cells (BdCs) and serum from patients with AS. RESULTS: Basal levels of IL-17A and IL-12/23 p40 in body fluids were elevated in patients with AS. JAK2 was also highly expressed in bone tissue and primary BdCs from patients with AS. Furthermore, addition of exogenous IL-17A to primary Ct-BdCs promoted the osteogenic stimulus-induced increase in ALP activity and mineralization. Intriguingly, blocking IL-17A with serum from patients with AS attenuated ALP activity and mineralization in both Ct and AS-BdCs by inhibiting JAK2 phosphorylation and downregulating osteoblast-involved genes. Moreover, JAK2 inhibitors effectively reduced JAK2-driven ALP activity and JAK2-mediated events. CONCLUSIONS: Our findings indicate that IL-17A regulates osteoblast activity and differentiation via JAK2/STAT3 signaling. They shed light on AS pathogenesis and suggest new rational therapies for clinical AS ankylosis.


Assuntos
Diferenciação Celular/fisiologia , Interleucina-17/metabolismo , Janus Quinase 2/biossíntese , Osteoblastos/metabolismo , Fator de Transcrição STAT3/metabolismo , Espondilite Anquilosante/metabolismo , Adulto , Idoso , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Anticorpos Monoclonais Humanizados , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Feminino , Humanos , Interleucina-17/antagonistas & inibidores , Janus Quinase 2/antagonistas & inibidores , Masculino , Pessoa de Meia-Idade , Osteoblastos/efeitos dos fármacos , Espondilite Anquilosante/tratamento farmacológico , Espondilite Anquilosante/patologia
13.
Exp Mol Med ; 50(3): e455, 2018 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-29520110

RESUMO

Transient receptor potential vanilloid 1 (TRPV1) affects mood and neuroplasticity in the brain, where its role is poorly understood. In the present study we investigated whether capsaicin (8-methyl-N-vanillyl-trans-6-nonenamide), an agonist of TRPV1, induced chromatin remodeling and thereby altered gene expression related to synaptic plasticity. We found that capsaicin treatment resulted in upregulation of histone deacetylase 2 (HDAC2) in the mouse hippocampus and HDAC2 was enriched at Psd95, synaptophysin, GLUR1, GLUR2 promoters. Viral-mediated hippocampal knockdown of HDAC2 induced expression of Synapsin I and prevented the detrimental effects of capsaicin on Synapsin I expression in mice, supporting the role of HDAC2 in regulation of capsaicin-induced Synapsin I expression. Taken together, our findings implicate HDAC2 in capsaicin-induced transcriptional regulation of synaptic molecules and support the view that HDAC2 is a molecular link between TRPV1 activity and synaptic plasticity.


Assuntos
Capsaicina/farmacologia , Histona Desacetilase 2/metabolismo , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Canais de Cátion TRPV/metabolismo , Animais , Western Blotting , Células Cultivadas , Imunoprecipitação da Cromatina , Hipocampo/citologia , Histona Desacetilase 2/genética , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurogênese/efeitos dos fármacos , Neurônios/citologia , Canais de Cátion TRPV/genética
14.
Mol Cells ; 40(7): 485-494, 2017 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-28681592

RESUMO

Oleanolic acid (OA) has neurotrophic effects on neurons, although its use as a neurological drug requires further research. In the present study, we investigated the effects of OA and OA derivatives on the neuronal differentiation of rat hippocampal neural progenitor cells. In addition, we investigated whether the class II histone deacetylase (HDAC) 5 mediates the gene expression induced by OA. We found that OA and OA derivatives induced the formation of neurite spines and the expression of synapse-related molecules. OA and OA derivatives stimulated HDAC5 phosphorylation, and concurrently the nuclear export of HDCA5 and the expression of HDAC5 target genes, indicating that OA and OA derivatives induce neural differentiation and synapse formation via a pathway that involves HDAC5 phosphorylation.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Hipocampo/citologia , Histona Desacetilases/metabolismo , Neurônios/citologia , Neurônios/enzimologia , Ácido Oleanólico/farmacologia , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Animais , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Espinhas Dendríticas/efeitos dos fármacos , Espinhas Dendríticas/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Fatores de Transcrição MEF2/metabolismo , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Neuritos/efeitos dos fármacos , Neuritos/metabolismo , Neurônios/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Proteína Quinase C/metabolismo , Ratos Sprague-Dawley , Frações Subcelulares/efeitos dos fármacos , Frações Subcelulares/metabolismo , Sinapses/efeitos dos fármacos , Sinapses/metabolismo
15.
Cell Rep ; 19(2): 401-412, 2017 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-28402861

RESUMO

Stress causes changes in neurotransmission in the brain, thereby influencing stress-induced behaviors. However, it is unclear how neurotransmission systems orchestrate stress responses at the molecular and cellular levels. Transient receptor potential vanilloid 1 (TRPV1), a non-selective cation channel involved mainly in pain sensation, affects mood and neuroplasticity in the brain, where its role is poorly understood. Here, we show that Trpv1-deficient (Trpv1-/-) mice are more stress resilient than control mice after chronic unpredictable stress. We also found that glucocorticoid receptor (GR)-mediated histone deacetylase 2 (HDAC) 2 expression and activity are reduced in the Trpv1-/- mice and that HDAC2-regulated, cell-cycle- and neuroplasticity-related molecules are altered. Hippocampal knockdown of TRPV1 had similar effects, and its behavioral effects were blocked by HDAC2 overexpression. Collectively, our findings indicate that HDAC2 is a molecular link between TRPV1 activity and stress responses.


Assuntos
Histona Desacetilase 2/genética , Plasticidade Neuronal/genética , Estresse Fisiológico/genética , Canais de Cátion TRPV/genética , Animais , Células Cultivadas , Regulação da Expressão Gênica , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Histona Desacetilase 2/biossíntese , Camundongos , Camundongos Knockout , Receptores de Glucocorticoides/genética , Canais de Cátion TRPV/biossíntese
16.
Proc Natl Acad Sci U S A ; 112(51): 15755-60, 2015 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-26647181

RESUMO

Ketamine produces rapid antidepressant-like effects in animal assays for depression, although the molecular mechanisms underlying these behavioral actions remain incomplete. Here, we demonstrate that ketamine rapidly stimulates histone deacetylase 5 (HDAC5) phosphorylation and nuclear export in rat hippocampal neurons through calcium/calmodulin kinase II- and protein kinase D-dependent pathways. Consequently, ketamine enhanced the transcriptional activity of myocyte enhancer factor 2 (MEF2), which leads to regulation of MEF2 target genes. Transfection of a HDAC5 phosphorylation-defective mutant (Ser259/Ser498 replaced by Ala259/Ala498, HDAC5-S/A), resulted in resistance to ketamine-induced nuclear export, suppression of ketamine-mediated MEF2 transcriptional activity, and decreased expression of MEF2 target genes. Behaviorally, viral-mediated hippocampal knockdown of HDAC5 blocked or occluded the antidepressant effects of ketamine both in unstressed and stressed animals. Taken together, our results reveal a novel role of HDAC5 in the actions of ketamine and suggest that HDAC5 could be a potential mechanism contributing to the therapeutic actions of ketamine.


Assuntos
Antidepressivos/farmacologia , Núcleo Celular/metabolismo , Histona Desacetilases/metabolismo , Ketamina/farmacologia , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Animais , Células Cultivadas , Hipocampo/metabolismo , Fatores de Transcrição MEF2/fisiologia , Fosforilação , Ratos , Ratos Sprague-Dawley
17.
PLoS One ; 9(10): e109253, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25340772

RESUMO

Functional consequences to which vertebrate GATA transcription factors contribute in the adult brain remain largely an open question. The present study examines how human GATA-1 and GATA-2 (hGATA-1 and hGATA-2) are linked to neuronal differentiation and depressive behaviors in rats. We investigated the effects of adeno-associated viral expression of hGATA-1 and hGATA-2 (AAV-hGATA1 and AAV-hGATA2) in the dentate gyrus (DG) of the dorsal hippocampus on dendrite branching and spine number. We also examined the influence of AAV-hGATA1 and AAV-hGATA2 infusions into the dorsal hippocampus on rodent behavior in models of depression. Viral expression of hGATA-1 and hGATA-2 cDNA in rat hippocampal neurons impaired dendritic outgrowth and spine formation. Moreover, viral-mediated expression of hGATA-1 and hGATA-2 in the dorsal hippocampus caused depressive-like deficits in the forced swim test and learned helplessness models of depression, and decreased the expression of several synapse-related genes as well as spine number in hippocampal neurons. Conversely, shRNA knockdown of GATA-2 increased synapse-related gene expression, spine number, and dendrite branching. The results demonstrate that hGATA-1 and hGATA-2 expression in hippocampus is sufficient to cause depressive like behaviors that are associated with reduction in spine synapse density and expression of synapse-related genes.


Assuntos
Comportamento Animal , Espinhas Dendríticas/metabolismo , Depressão/metabolismo , Fator de Transcrição GATA1/metabolismo , Fator de Transcrição GATA2/metabolismo , Animais , Células Cultivadas , Dependovirus/metabolismo , Regulação da Expressão Gênica , Proteínas de Fluorescência Verde/metabolismo , Hipocampo/patologia , Humanos , Masculino , RNA Interferente Pequeno/metabolismo , Ratos Sprague-Dawley , Ratos Transgênicos , Sinapses/metabolismo , Transcrição Gênica
18.
Biochem Biophys Res Commun ; 446(1): 79-84, 2014 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-24607903

RESUMO

Both erythropoietin (EPO) and carbamylated EPO (cEPO) have been shown to increase the length of neurites and spine density in neurons. However, the molecular mechanism underlying the EPO- and cEPO-induced neuronal differentiation has yet to be investigated. To address this issue, we investigated epigenetic modifications that regulate gene expression in neurons. Neurons treated with EPO or cEPO display an upregulation of E1A-binding protein (p300) and p300-mediated p53 acetylation, possibly increasing the transactivation activity of p53 on growth-associated protein 43 (GAP43). Treatment of cells with cEPO markedly increases spine formation and potentiates p300-mediated transactivation of PSD95, Shank2 and 3 compared to EPO. These results demonstrate that cEPO controls neuronal differentiation via acetylation of transcription factors and subsequent transactivation of target genes. These findings have important medical implications because cEPO is of interest in the development of therapeutic agents against neuropsychiatric disorders.


Assuntos
Proteína p300 Associada a E1A/metabolismo , Eritropoetina/análogos & derivados , Neuritos/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Animais , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Proteína 4 Homóloga a Disks-Large , Proteína p300 Associada a E1A/genética , Epigênese Genética/efeitos dos fármacos , Eritropoetina/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Sistema de Sinalização das MAP Quinases/genética , Proteínas de Membrana/genética , Proteínas do Tecido Nervoso/genética , Neuritos/metabolismo , Neuritos/ultraestrutura , Neurogênese/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/ultraestrutura , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Fator de Transcrição STAT3/metabolismo , Ativação Transcricional/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...