Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mol Neurobiol ; 57(1): 393-407, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31364026

RESUMO

Ten to 20% of western countries population suffers from major depression disorder (MDD). Stressful life events represent the main environmental risk factor contributing to the onset of MDD and other stress-related neuropsychiatric disorders. In this regard, investigating brain physiology of stress response underlying the remarkable individual variability in terms of behavioral outcome may uncover stress-vulnerability pathways as a source of candidate targets for conceptually new antidepressant treatments. Serum response factor (SRF) has been addressed as a stress transducer via promoting inherent experience-induced Immediate Early Genes (IEGs) expression in neurons. However, in resting conditions, SRF also represents a transcriptional repressor able to assemble the core LSD1/CoREST/HDAC2 corepressor complex, including demethylase and deacetylase activities. We here show that dominant negative SRF splicing isoform lacking most part of the transactivation domain, namely SRFΔ5, owes its transcriptional repressive behavior to the ability of assembling LSD1/CoREST/HDAC2 corepressor complex meanwhile losing its affinity for transcription-permissive cofactor ELK1. SRFΔ5 is highly expressed in the brain and developmentally regulated. In the light of its activity as negative modulator of dendritic spine density, SRFΔ5 increase along with brain maturation suggests a role in synaptic pruning. Upon acute psychosocial stress, SRFΔ5 isoform transiently increases its levels. Remarkably, when stress is chronically repeated, a different picture occurs where SRF protein becomes stably upregulated in vulnerable mice but not in resilient animals. These data suggest a role for SRFΔ5 that is restricted to acute stress response, while positive modulation of SRF during chronic stress matches the criteria for stress-vulnerability hallmark.


Assuntos
Processamento Alternativo/genética , Proteínas Correpressoras/metabolismo , Histona Desmetilases/metabolismo , Plasticidade Neuronal , Fator de Resposta Sérica/genética , Estresse Fisiológico , Animais , Forma Celular , Espinhas Dendríticas/metabolismo , Células HeLa , Hipocampo/metabolismo , Histona Desacetilase 2/metabolismo , Humanos , Camundongos Endogâmicos C57BL , Ligação Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Fator de Resposta Sérica/metabolismo , Estresse Psicológico/patologia
2.
World J Biol Psychiatry ; 20(7): 555-566, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-30058429

RESUMO

Objectives: Although stress is considered a primary risk factor for neuropsychiatric disorders, a majority of individuals are resilient to the effects of stress exposure and successfully adapt to adverse life events, while others, the so-called susceptible individuals, may have problems to properly adapt to environmental changes. However, the mechanisms underlying these different responses to stress exposure are poorly understood.Methods: Adult male C57BL/6J mice were exposed to chronic social defeat stress protocol and levels of brain derived neurotrophic factor (BDNF) transcripts and epigenetic modifying enzymes were analysed by real-time PCR in the hippocampus (HPC) and prefrontal cortex (PFC) of susceptible and resilient mice.Results: We found a selective reduction of BDNF-6 transcript in the HPC and an increase of BDNF-4 transcript in the PFC of susceptible mice. Moreover, susceptible mice showed a selective reduction of the g9a mRNA levels in the HPC, while HDAC-5 and DNMT3a mRNA levels were specifically reduced in the PFC.Conclusions: Overall, our results, showing a different expression of BDNF transcripts and epigenetic modifying enzymes in susceptible and resilient mice, suggest that stress resilience is not simply a lack of activation of stress-related pathways, but is related to the activation of additional different specific mechanisms.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Hipocampo/enzimologia , Córtex Pré-Frontal/enzimologia , Estresse Psicológico/enzimologia , Adaptação Psicológica , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , DNA (Citosina-5-)-Metiltransferases/genética , DNA Metiltransferase 3A , Suscetibilidade a Doenças , Epigênese Genética , Histona Desacetilases/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estresse Psicológico/genética
3.
Hippocampus ; 28(11): 783-795, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30067287

RESUMO

Brain-derived neurotrophic factor (BDNF), a neurotrophin highly expressed in the hippocampus, plays crucial roles in cognition, neuroplasticity, synaptic function, and dendritic remodeling. The common human Val66Met polymorphism of BDNF has been implicated in the pathophysiology of neuropsychiatric and neurodegenerative disorders, and in the outcome of pro-adaptive and therapeutic treatments. Altered gene-expression profile has been previously shown in BDNF Val66Met knock-in mice, which recapitulate the phenotypic hallmarks of individuals carrying the BDNF Met allele. The aim of this study was to investigate the impact of the BDNF Val66Met polymorphism in the knock-in mouse model on two hippocampal epigenetic marks for transcriptional repression and activation, respectively: trimethylation of lysine 27 on histone H3 (H3K27me3) and acetylation of histone H3 (AcH3), using a genome-wide approach. Chromatin immunoprecipitation followed by deep sequencing of immunoprecipitated DNA (ChIP-Seq) was carried out with specific antibodies for H3K27me3 and AcH3. Our results revealed broad alteration of H3K27me3 and AcH3 marks association profiles in BDNFMet/Met , compared to BDNFVal/Val mice. Bioinformatics analysis showed changes in several biological functions and related pathways, affected by the presence of the polymorphism. In particular, a number of networks of functional interaction contained BDNF as central node. Quantitative PCR analysis confirmed epigenetically related significant changes in the expression of five genes: Dvl1, Nos3, Reln, Lypd6, and Sh3gl2. The first three are involved in dendrite and spine remodeling, morphological features altered in BDNFMet/Met mice. This work in homozygous knock-in mice shows that the human BDNF Val66Met polymorphism induces an array of histone H3 epigenetic modifications, in turn altering the expression of select genes crucial for structural and functional neuronal features.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Dendritos/metabolismo , Epigênese Genética , Hipocampo/metabolismo , Polimorfismo Genético , Animais , Biologia Computacional , Técnicas de Introdução de Genes , Histonas/genética , Histonas/metabolismo , Humanos , Masculino , Camundongos Transgênicos , Proteína Reelina , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
4.
Neuropsychopharmacology ; 41(13): 3070-3079, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27388329

RESUMO

Several studies have shown that exercise improves cognitive functions and emotional behaviors. Positive effects of exercise have been associated with enhanced brain plasticity, adult hippocampal neurogenesis, and increased levels of brain-derived neurotrophic factor (BDNF). However, a substantial variability of individual response to exercise has been described, which may be accounted for by individual genetic variants. Here, we have assessed whether and how the common human BDNF Val66Met polymorphism influences the neurobiological effects modulated by exercise in BDNF Val66Met knock-in male mice. Wild-type (BDNFVal/Val) and homozygous BDNF Val66Met (BDNFMet/Met) male mice were housed in cages equipped with or without running wheels for 4 weeks. Changes in behavioral phenotype, hippocampal adult neurogenesis, and gene expression were evaluated in exercised and sedentary control mice. We found that exercise reduced the latency to feed in the novelty suppressed feeding and the immobility time in the forced swimming test in BDNFVal/Val but not in BDNFMet/Met mice. Hippocampal neurogenesis was reduced in BDNFMet/Met mice compared with BDNFVal/Val mice. BDNFMet/Met mice had lower basal BDNF protein levels in the hippocampus, which was not recovered following exercise. Moreover, exercise-induced expression of total BDNF, BDNF splice variants 1, 2, 4, 6 and fibronectin type III domain-containing protein 5 (FNDC5) mRNA levels were absent or reduced in the dentate gyrus of BDNFMet/Met mice. Exercise failed to enhance PGC-1α and FNDC5 mRNA levels in the BDNFMet/Met muscle. Overall these results indicate that, in adult male mice, the BDNF Val66Met polymorphism impairs the beneficial behavioral and neuroplasticity effects induced by physical exercise.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/genética , Metionina/genética , Plasticidade Neuronal/genética , Condicionamento Físico Animal/fisiologia , Polimorfismo de Nucleotídeo Único/genética , Valina/genética , Animais , Peso Corporal/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Modelos Animais de Doenças , Fibronectinas/genética , Fibronectinas/metabolismo , Regulação da Expressão Gênica/genética , Genótipo , Hipocampo/citologia , Hipocampo/fisiologia , Humanos , Masculino , Transtornos Mentais/genética , Camundongos , Camundongos Transgênicos , Neurogênese/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo , Natação/fisiologia
5.
Proc Natl Acad Sci U S A ; 113(13): 3651-6, 2016 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-26976584

RESUMO

Behavioral changes in response to stressful stimuli can be controlled via adaptive epigenetic changes in neuronal gene expression. Here we indicate a role for the transcriptional corepressor Lysine-Specific Demethylase 1 (LSD1) and its dominant-negative splicing isoform neuroLSD1, in the modulation of emotional behavior. In mouse hippocampus, we show that LSD1 and neuroLSD1 can interact with transcription factor serum response factor (SRF) and set the chromatin state of SRF-targeted genes early growth response 1 (egr1) and c-fos Deletion or reduction of neuro LSD1 in mutant mice translates into decreased levels of activating histone marks at egr1 and c-fos promoters, dampening their psychosocial stress-induced transcription and resulting in low anxiety-like behavior. Administration of suberoylanilide hydroxamine to neuroLSD1(KO)mice reactivates egr1 and c-fos transcription and restores the behavioral phenotype. These findings indicate that LSD1 is a molecular transducer of stressful stimuli as well as a stress-response modifier. Indeed, LSD1 expression itself is increased acutely at both the transcriptional and splicing levels by psychosocial stress, suggesting that LSD1 is involved in the adaptive response to stress.


Assuntos
Emoções/fisiologia , Genes Precoces , Histona Desmetilases/fisiologia , Processamento Alternativo , Animais , Proteína 1 de Resposta de Crescimento Precoce/genética , Epigênese Genética , Genes fos , Histona Desmetilases/deficiência , Histona Desmetilases/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Plasticidade Neuronal , Fenótipo , Fator de Resposta Sérica/fisiologia , Estresse Psicológico , Transcrição Gênica
6.
Neural Plast ; 2016: 6212983, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26881124

RESUMO

Stress is a major risk factor in the onset of several neuropsychiatric disorders including anxiety and depression. Although several studies have shown that social isolation stress during postweaning period induces behavioral and brain molecular changes, the effects of social isolation on behavior during adulthood have been less characterized. Aim of this work was to investigate the relationship between the behavioral alterations and brain molecular changes induced by chronic social isolation stress in adult male mice. Plasma corticosterone levels and adrenal glands weight were also analyzed. Socially isolated (SI) mice showed higher locomotor activity, spent less time in the open field center, and displayed higher immobility time in the tail suspension test compared to group-housed (GH) mice. SI mice exhibited reduced plasma corticosterone levels and reduced difference between right and left adrenal glands. SI showed lower mRNA levels of the BDNF-7 splice variant, c-Fos, Arc, and Egr-1 in both hippocampus and prefrontal cortex compared to GH mice. Finally, SI mice exhibited selectively reduced mGluR1 and mGluR2 levels in the prefrontal cortex. Altogether, these results suggest that anxious- and depressive-like behavior induced by social isolation stress correlates with reduction of several neuroplasticity-related genes in the hippocampus and prefrontal cortex of adult male mice.


Assuntos
Ansiedade/genética , Depressão/genética , Plasticidade Neuronal/genética , Isolamento Social , Estresse Psicológico/complicações , Estresse Psicológico/genética , Glândulas Suprarrenais/patologia , Animais , Ansiedade/etiologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Corticosterona/sangue , Proteínas do Citoesqueleto/metabolismo , Depressão/etiologia , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Hipocampo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/metabolismo , Tamanho do Órgão , Córtex Pré-Frontal/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Estresse Psicológico/sangue
7.
Int J Neuropsychopharmacol ; 18(12)2015 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-26108221

RESUMO

BACKGROUND: The human Val66Met polymorphism in brain-derived neurotrophic factor (BDNF), a key factor in neuroplasticity, synaptic function, and cognition, has been implicated in the pathophysiology of neuropsychiatric and neurodegenerative disorders. BDNF is encoded by multiple transcripts with distinct regulation and localization, but the impact of the Val66Met polymorphism on BDNF regulation remains unclear. METHODS: In BDNF Val66Met knock-in mice, which recapitulate the phenotypic hallmarks of individuals carrying the BDNF(Met) allele, we measured expression levels, epigenetic changes at promoters, and dendritic trafficking of distinct BDNF transcripts using quantitative PCR, chromatin immunoprecipitation (ChIP), and in situ hybridization. RESULTS: BDNF-4 and BDNF-6 transcripts were reduced in BDNF(Met/Met) mice, compared with BDNF(Val/Val) mice. ChIP for acetyl-histone H3, a marker of active gene transcription, and trimethyl-histone-H3-Lys27 (H3K27me3), a marker of gene repression, showed higher H3K27me3 binding to exon 5, 6, and 8 promoters in BDNF(Met/Met). The H3K27 methyltransferase enhancer of zeste homolog 2 (EZH2) is involved in epigenetic regulation of BDNF expression, because in neuroblastoma cells BDNF expression was increased both by short interference RNA for EZH2 and incubation with 3-deazaneplanocin A, an inhibitor of EZH2. In situ hybridization for BDNF-2, BDNF-4, and BDNF-6 after pilocarpine treatment showed that BDNF-6 transcript was virtually absent from distal dendrites of the CA1 and CA3 regions in BDNF(Met/Met) mice, while no changes were found for BDNF-2 and BDNF-4. CONCLUSIONS: Impaired BDNF expression and dendritic targeting in BDNF(Met/Met) mice may contribute to reduced regulated secretion of BDNF at synapses, and may be a specific correlate of pathology in individuals carrying the Met allele.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Dendritos/metabolismo , Polimorfismo Genético , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Imunoprecipitação da Cromatina , Dendritos/efeitos dos fármacos , Proteína Potenciadora do Homólogo 2 de Zeste , Epigênese Genética , Perfilação da Expressão Gênica , Técnicas de Introdução de Genes , Humanos , Hibridização In Situ , Histona Desmetilases com o Domínio Jumonji/metabolismo , Masculino , Camundongos Transgênicos , Agonistas Muscarínicos/farmacologia , Pilocarpina/farmacologia , Complexo Repressor Polycomb 2/metabolismo , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas , Isoformas de Proteínas , Transporte Proteico/efeitos dos fármacos
8.
Hippocampus ; 25(11): 1380-92, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25820928

RESUMO

Physical exercise and stressful experiences have been shown to exert opposite effects on behavioral functions and brain plasticity, partly by involving the action of brain-derived neurotrophic factor (BDNF). Although epigenetic modifications are known to play a pivotal role in the regulation of the different BDNF transcripts, it is poorly understood whether epigenetic mechanisms are also implied in the BDNF modulation induced by physical exercise and stress. Here, we show that total BDNF mRNA levels and BDNF transcripts 1, 2, 3, 4, 6, and 7 were reduced immediately after acute restraint stress (RS) in the hippocampus of mice, and returned to control levels 24 h after the stress session. On the contrary, exercise increased BDNF mRNA expression and counteracted the stress-induced decrease of BDNF transcripts. Physical exercise-induced up-regulation of BDNF transcripts was accounted for by increase in histone H3 acetylated levels at specific BDNF promoters, whereas the histone H3 trimethylated lysine 27 and dimethylated lysine 9 levels were unaffected. Acute RS did not change the levels of acetylated and methylated histone H3 at the BDNF promoters. Furthermore, we found that physical exercise and RS were able to differentially modulate the histone deacetylases mRNA levels. Finally, we report that a single treatment with histone deacetylase inhibitors, prior to acute stress exposure, prevented the down-regulation of total BDNF and BDNF transcripts 1, 2, 3, and 6, partially reproducing the effect of physical exercise. Overall, these results suggest that physical exercise and stress are able to differentially modulate the expression of BDNF transcripts by possible different epigenetic mechanisms.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Epigênese Genética/fisiologia , Hipocampo/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Atividade Motora/fisiologia , Restrição Física/fisiologia , Estresse Psicológico/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/efeitos dos fármacos , Epigênese Genética/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Atividade Motora/efeitos dos fármacos , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/metabolismo , Estresse Psicológico/prevenção & controle
9.
Int J Neuropsychopharmacol ; 18(3)2014 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-25522407

RESUMO

BACKGROUND: Major depression is a severe mental illness that causes heavy social and economic burdens worldwide. A number of studies have shown that interaction between individual genetic vulnerability and environmental risk factors, such as stress, is crucial in psychiatric pathophysiology. In particular, the experience of stressful events in childhood, such as neglect, abuse, or parental loss, was found to increase the risk for development of depression in adult life. Here, to reproduce the gene x environment interaction, we employed an animal model that combines genetic vulnerability with early-life stress. METHODS: The Flinders Sensitive Line rats (FSL), a validated genetic animal model of depression, and the Flinders Resistant Line (FRL) rats, their controls, were subjected to a standard protocol of maternal separation (MS) from postnatal days 2 to 14. A basal comparison between the two lines for the outcome of the environmental manipulation was performed at postnatal day 73, when the rats were into adulthood. We carried out a global proteomic analysis of purified synaptic terminals (synaptosomes), in order to study a subcellular compartment enriched in proteins involved in synaptic function. Two-dimensional gel electrophoresis (2-DE), mass spectrometry, and bioinformatic analysis were used to analyze proteins and related functional networks that were modulated by genetic susceptibility (FSL vs. FRL) or by exposure to early-life stress (FRL + MS vs. FRL and FSL + MS vs. FSL) RESULTS: We found that, at a synaptic level, mainly proteins and molecular pathways related to energy metabolism and cellular remodeling were dysregulated. CONCLUSIONS: The present results, in line with previous works, suggest that dysfunction of energy metabolism and cytoskeleton dynamics at a synaptic level could be features of stress-related pathologies, in particular major depression.


Assuntos
Depressão/etiologia , Depressão/genética , Metabolismo Energético/genética , Interação Gene-Ambiente , Sinaptofisina/metabolismo , Sinaptossomos/metabolismo , Fatores Etários , Animais , Animais Recém-Nascidos , Biologia Computacional , Modelos Animais de Doenças , Eletroforese em Gel Bidimensional , Espectrometria de Massas , Privação Materna , Proteômica/métodos , Ratos
10.
BMC Neurosci ; 15: 119, 2014 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-25332063

RESUMO

BACKGROUND: The novel antidepressant agomelatine, a melatonergic MT1/MT2 agonist combined with 5-HT2c serotonin antagonist properties, showed antidepressant action in preclinical and clinical studies. There is a general agreement that the therapeutic action of antidepressants needs the activation of slow-onset adaptations in downstream signalling pathways finally regulating neuroplasticity. In the last several years, particular attention was given to cAMP-responsive element binding protein (CREB)-related pathways, since it was shown that chronic antidepressants increase CREB phosphorylation and transcriptional activity, through the activation of calcium/calmodulin-dependent (CaM) and mitogen activated protein kinase cascades (MAPK/Erk1/2). Aim of this work was to analyse possible effects of chronic agomelatine on time-dependent changes of different intracellular signalling pathways in hippocampus and prefrontal/frontal cortex of male rats. To this end, measurements were performed 1 h or 16 h after the last agomelatine or vehicle injection. RESULTS: We have found that in naïve rats chronic agomelatine, contrary to traditional antidepressants, did not increase CREB phosphorylation, but modulates the time-dependent regulation of MAPK/Erk1/2 and Akt/glycogen synthase kinase-3 (GSK-3) pathways. CONCLUSION: Our results suggest that the intracellular molecular mechanisms modulated by chronic agomelatine may be partly different from those of traditional antidepressants and involve the time-dependent regulation of MAPK/Erk1/2 and Akt/GSK-3 signalling pathways. This could exert a role in the antidepressant efficacy of the drug.


Assuntos
Acetamidas/farmacologia , Antidepressivos/farmacologia , Lobo Frontal/efeitos dos fármacos , Quinase 3 da Glicogênio Sintase/metabolismo , Hipocampo/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Animais , Western Blotting , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Lobo Frontal/enzimologia , Hipocampo/enzimologia , Masculino , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Distribuição Aleatória , Ratos Sprague-Dawley , Fatores de Tempo
11.
BMC Neurosci ; 14: 75, 2013 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-23895555

RESUMO

BACKGROUND: Growing compelling evidence from clinical and preclinical studies has demonstrated the primary role of alterations of glutamatergic transmission in cortical and limbic areas in the pathophysiology of mood disorders. Chronic antidepressants have been shown to dampen endogenous glutamate release from rat hippocampal synaptic terminals and to prevent the marked increase of glutamate overflow induced by acute behavioral stress in frontal/prefrontal cortex. Agomelatine, a new antidepressant endowed with MT1/MT2 agonist and 5-HT2C serotonergic antagonist properties, has shown efficacy at both preclinical and clinical levels. RESULTS: Chronic treatment with agomelatine, or with the reference drug venlafaxine, induced a marked decrease of depolarization-evoked endogenous glutamate release from purified hippocampal synaptic terminals in superfusion. No changes were observed in GABA release. This effect was accompanied by reduced accumulation of SNARE protein complexes, the key molecular effector of vesicle docking, priming and fusion at presynaptic membranes. CONCLUSIONS: Our data suggest that the novel antidepressant agomelatine share with other classes of antidepressants the ability to modulate glutamatergic transmission in hippocampus. Its action seems to be mediated by molecular mechanisms located on the presynaptic membrane and related with the size of the vesicle pool ready for release.


Assuntos
Acetamidas/farmacologia , Antidepressivos/farmacologia , Cicloexanóis/farmacologia , Ácido Glutâmico/metabolismo , Hipocampo/citologia , Sinaptossomos/efeitos dos fármacos , Análise de Variância , Animais , Ionóforos de Cálcio/farmacologia , Ionomicina/farmacologia , Masculino , Cloreto de Potássio/farmacologia , Ratos , Ratos Sprague-Dawley , Proteínas SNARE/metabolismo , Sintaxina 1/metabolismo , Cloridrato de Venlafaxina , Ácido gama-Aminobutírico/metabolismo
12.
Biol Psychiatry ; 73(12): 1180-8, 2013 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-23273725

RESUMO

Recent compelling evidence has suggested that the glutamate system is a primary mediator of psychiatric pathology and also a target for rapid-acting antidepressants. Clinical research in mood and anxiety disorders has shown alterations in levels, clearance, and metabolism of glutamate and consistent volumetric changes in brain areas where glutamate neurons predominate. In parallel, preclinical studies with rodent stress and depression models have found dendritic remodeling and synaptic spines reduction in corresponding areas, suggesting these as major factors in psychopathology. Enhancement of glutamate release/transmission, in turn induced by stress/glucocorticoids, seems crucial for structural/functional changes. Understanding mechanisms of maladaptive plasticity may allow identification of new targets for drugs and therapies. Interestingly, traditional monoaminergic-based antidepressants have been repeatedly shown to interfere with glutamate system function, starting with modulation of N-methyl-D-aspartate (NMDA) receptors. Subsequently, it has been shown that antidepressants reduce glutamate release and synaptic transmission; in particular, it was found antidepressants prevent the acute stress-induced enhancement of glutamate release. Additional studies have shown that antidepressants may partly reverse the maladaptive changes in synapses/circuitry in stress and depression models. Finally, a number of studies over the years have shown that these drugs regulate glutamate receptors, reducing the function of NMDA receptors, potentiating the function of α-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid receptors, and, more recently, exerting variable effects on different subtypes of metabotropic glutamate receptors. The development of NMDA receptor antagonists has opened new avenues for glutamatergic, rapid acting, antidepressants, while additional targets in the glutamate synapse await development of new compounds for better, faster antidepressant action.


Assuntos
Antidepressivos/farmacologia , Ácido Glutâmico/metabolismo , Receptores de Glutamato/metabolismo , Animais , Antidepressivos/uso terapêutico , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Desenho de Fármacos , Humanos
13.
Expert Opin Investig Drugs ; 22(2): 217-33, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23253113

RESUMO

INTRODUCTION: Mood and anxiety disorders are among the major causes of disability worldwide. Despite clear need for better therapies, efforts to develop novel drugs have been relatively unsuccessful. One major reason is lack of translation into neuropsychopharmacology of the impressive recent array of knowledge accrued by clinical and preclinical researches on the brain. Here focus is on epigenetics mechanisms, including microRNAs, which seem particularly promising for the identification of new targets for alternative pharmacological approaches. AREAS COVERED: First, the current knowledge about epigenetic mechanisms, including DNA methylation, posttranslational modification of histone proteins, focusing on histone methylation and acetylation, and posttranscriptional modulation of gene expression by microRNAs is described. Then evidence showing involvement of epigenetics and microRNAs in the pathophysiology of mood and anxiety disorders as well as evidence showing that some of the currently employed antidepressants and mood stabilizers also affect epigenetic and microRNA mechanisms are reviewed. Finally current evidence and novel approaches in favor of drugs regulating epigenetic and microRNA mechanisms as potential therapeutics for these disorders are discussed. EXPERT OPINION: Although still in its infancy, research investigating the effects of pharmacological modulation of epigenetic and microRNA mechanisms in neuropsychiatric disorders continues to provide encouraging findings, suggesting new avenues for treatment of mood and anxiety disorders.


Assuntos
Transtornos de Ansiedade/tratamento farmacológico , Epigênese Genética/efeitos dos fármacos , MicroRNAs/genética , Transtornos do Humor/tratamento farmacológico , Neuropsicologia , Psicotrópicos/uso terapêutico , Animais , Transtornos de Ansiedade/genética , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Metilação de DNA/efeitos dos fármacos , Metilação de DNA/genética , Histonas/genética , Histonas/metabolismo , Humanos , Transtornos do Humor/genética , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Psicotrópicos/administração & dosagem , Psicotrópicos/efeitos adversos , Psicotrópicos/farmacologia
14.
BMC Psychiatry ; 12: 145, 2012 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-22989054

RESUMO

BACKGROUND: In recent years, the identification of peripheral biomarkers that are associated with psychiatric diseases, such as Major Depressive Disorder (MDD), has become relevant because these biomarkers may improve the efficiency of the differential diagnosis process and indicate targets for new antidepressant drugs. Two recent candidate genes, ErbB3 and Fgfr1, are growth factors whose mRNA levels have been found to be altered in the leukocytes of patients that are affected by bipolar disorder in a depressive state. On this basis, the aim of the study was to determine if ErbB3 and Fgfr1 mRNA levels could be a biomarkers of MDD. METHODS: We measured by Real Time PCR ErbB3 and Fgfr1 mRNA expression levels in leukocytes of MDD patients compared with controls. Successively, to assess whether ErbB3 mRNA levels were influenced by previous antidepressant treatment we stratified our patients sample in two cohorts, comparing drug-naive versus drug-free patients. Moreover, we evaluated the levels of the transcript in MDD patients after 12 weeks of antidepressant treatment, and in prefrontal cortex of rats stressed and treated with an antidepressant drug of the same class. RESULTS: These results showed that ErbB3 but not Fgfr1 mRNA levels were reduced in leukocytes of MDD patients compared to healthy subjects. Furthermore, ErbB3 levels were not affected by antidepressant treatment in either human or animal models CONCLUSIONS: Our data suggest that ErbB3 might be considered as a biomarker for MDD and that its deficit may underlie the pathophysiology of the disease and is not a consequence of treatment. Moreover the study supports the usefulness of leukocytes as a peripheral system for identifying biomarkers in psychiatric diseases.


Assuntos
Transtorno Depressivo Maior/genética , Leucócitos/metabolismo , RNA Mensageiro/genética , Receptor ErbB-3/genética , Adulto , Animais , Antidepressivos/administração & dosagem , Antidepressivos/efeitos adversos , Antidepressivos/farmacologia , Biomarcadores/sangue , Estudos de Coortes , Transtorno Depressivo Maior/diagnóstico , Transtorno Depressivo Maior/tratamento farmacológico , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/patologia , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética
15.
Neuropsychopharmacology ; 37(7): 1600-11, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22318196

RESUMO

Brain-derived neurotrophic factor (BDNF) is encoded by multiple BDNF transcripts, whose function is unclear. We recently showed that a subset of BDNF transcripts can traffic into distal dendrites in response to electrical activity, while others are segregated into the somatoproximal domains. Physical exercise and antidepressant treatments exert their beneficial effects through upregulation of BDNF, which is required to support survival and differentiation of newborn dentate gyrus (DG) neurons. While these DG processes are required for the antidepressant effect, a role for CA1 in antidepressant action has been excluded, and the effect on CA3 neurons remains unclear. Here, we show for the first time that physical exercise and antidepressants induce local increase of BDNF in CA3. Voluntary physical exercise for 28 consecutive days, or 2-week treatment with 10 mg/kg per day fluoxetine or reboxetine, produced a global increase of BDNF mRNA and protein in the neuronal somata of the whole hippocampus and a specific increase of BDNF in dendrites of CA3 neurons. This increase was accounted for by BDNF exon 6 variant. In cultured hippocampal neurons, application of serotonin or norepinephrine (10-50 µM) induced increase in synaptic transmission and targeting of BDNF mRNA in dendrites. The increased expression of BDNF in CA3 dendrites following antidepressants or exercise further supports the neurotrophin hypothesis of antidepressants action and confirms that the differential subcellular localization of BDNF mRNA splice variants provides a spatial code for a selective expression of BDNF in specific subcellular districts. This selective expression may be exploited to design more specific antidepressants.


Assuntos
Antidepressivos/farmacologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Região CA3 Hipocampal/metabolismo , Dendritos/metabolismo , Condicionamento Físico Animal/fisiologia , Animais , Região CA3 Hipocampal/efeitos dos fármacos , Células Cultivadas , Dendritos/efeitos dos fármacos , Fluoxetina/farmacologia , Masculino , Camundongos , Morfolinas/farmacologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Norepinefrina/farmacologia , Isoformas de Proteínas/metabolismo , Ratos , Ratos Sprague-Dawley , Reboxetina , Serotonina/farmacologia , Transmissão Sináptica/efeitos dos fármacos , Transmissão Sináptica/fisiologia , Regulação para Cima/efeitos dos fármacos
16.
Neurochem Int ; 59(6): 896-905, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21839792

RESUMO

Growing evidence suggests a pivotal role for glutamatergic neurotransmission in the pathophysiology of major depressive disorder and in the action of antidepressants. The main aim of this study was to elucidate the temporal profile of α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptors expression and their functional regulation in prefrontal/frontal cortex (P/FC) and hippocampus (HC) of rats chronically treated with two different antidepressants: fluoxetine (FLX) and reboxetine (RBX). Rat groups were treated for 1, 2 or 3 weeks with the two drugs and, in additional groups, the treatments were followed by 1 week of drug washout (3+1). We found that both drugs induced strong increases in AMPAR subunit protein expression that were time dependent and subunit specific. Especially in P/FC, FLX had the main effect on GluA2 and GluA4 subunits, reaching a 5-fold increase after the drug washout; RBX mostly affected GluA1 and GluA3, reaching a 4-fold increase at the end of the treatment. Furthermore, in HC, the two drugs induced a time specific increase in subunit protein levels, with GluA3 and GluA4 presenting the main changes, albeit with different kinetics. In addition, our data indicate that antidepressants might alter, though by small changes, the R/G editing levels for GluA2, mostly in P/FC, and in turn may induce fine-tuning of glutamate neurotransmission. Overall, we showed that antidepressant treatments induced marked changes in AMPA receptor subunits expression, with time-dependent effects that are consistent with the onset of therapeutic effect of these drugs. These data confirm the involvement of glutamate neurotransmission in the effects of these drugs and further suggest the targeting of AMPA receptors as a therapeutic approach for the treatment of depression.


Assuntos
Antidepressivos/administração & dosagem , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Transtorno Depressivo/tratamento farmacológico , Subunidades Proteicas/biossíntese , Receptores de AMPA/biossíntese , Regulação para Cima/efeitos dos fármacos , Animais , Antidepressivos/farmacologia , Esquema de Medicação , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Masculino , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Regulação para Cima/fisiologia
17.
Neuropharmacology ; 60(7-8): 1243-53, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21195720

RESUMO

Although depression is a severe and life-threatening psychiatric illness, its pathogenesis still is essentially unknown. Recent studies highlighted the influence of environmental stress factors on an individual's genetic predisposition to develop mood disorders. In the present study, we employed a well-validated stress-induced animal model of depression, Learned Helplessness paradigm, in rats. Learned helpless (LH) and non-learned helpless (NLH) rats were treated with nortriptyline, a tricyclic antidepressant. The resulting 4 groups (LH vs. NLH, treated vs. non-treated), were subjected to global analysis of protein expression, a powerful approach to gain insight into the molecular mechanisms underlying vulnerability to psychiatric disorders and the long-term action of drug treatments. Many of the biological targets of antidepressant drugs are localized at synapses. Thus, to reduce the complexity of the proteome analyzed and to enrich for less abundant synaptic proteins, purified nerve terminals (synaptosomes) from prefrontal/frontal cortex (P/FC) and hippocampus (HPC) of LH-NLH rats were used. Synaptosomes were purified by differential centrifugation on Percoll gradients and analyzed by two-dimensional polyacrylamide gel electrophoresis (2-DE). Protein spots differently regulated in the various comparisons were excised from gels and identified by mass spectrometry. Proteins involved in energy metabolism and cellular remodeling were primarily dysregulated, when LH and NLH rats were compared. Moreover, several proteins (aconitate hydratase, pyruvate dehydrogenase E1, dihydropyrimidinase-related protein-2 and stathmin) were found to be regulated in opposite directions by stress and drug treatment. These proteins could represent new molecular correlates of both vulnerability to stress and response to drugs, and putative targets for the development of novel drugs with antidepressant action. This article is part of a Special Issue entitled 'Trends in neuropharmacology: in memory of Erminio Costa'.


Assuntos
Antidepressivos Tricíclicos/uso terapêutico , Transtorno Depressivo/metabolismo , Hipocampo/patologia , Proteoma/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sinapses/fisiologia , Animais , Antidepressivos Tricíclicos/farmacologia , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/fisiologia , Simulação por Computador , Depressão/tratamento farmacológico , Transtorno Depressivo/tratamento farmacológico , Transtorno Depressivo/patologia , Modelos Animais de Doenças , Regulação para Baixo , Metabolismo Energético , Desamparo Aprendido , Hipocampo/metabolismo , Masculino , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/fisiologia , Proteínas/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/fisiologia , Software , Sinapses/metabolismo , Resultado do Tratamento , Regulação para Cima
18.
Neuropsychobiology ; 63(3): 160-8, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21228608

RESUMO

BACKGROUND/AIMS: Compelling evidence would suggest the involvement of the serotonin 2C receptor in the pathophysiology of affective disorders and in the action of antidepressants. We analyzed the time course of 5-HT2C receptor (5-HTR2C) mRNA expression during antidepressant treatment in the prefrontal/frontal cortex (P/FC) and in the hippocampus (HC) of rats chronically treated with fluoxetine (a selective serotonin reuptake inhibitor) and reboxetine (a selective noradrenaline reuptake inhibitor). We also analyzed the 5-HTR2C RNA-editing levels at the sites called A, B, C, C' and D, which are known to modulate 5-HTR2C receptor function. RESULTS: The expression profile of 5-HTR2C mRNA was modified during treatment with both antidepressants. In particular, we found a general down-regulation of 5-HTR2C mRNA expression in P/FC, which became significant after 3 weeks of treatment with both antidepressants and persisted after a fourth week of drug withdrawal (-46% with fluoxetine, -41% with reboxetine, p < 0.05). In HC, however, reboxetine induced significant down-regulation (-56%, p < 0.05) of 5-HTR2C mRNA after 3 weeks, while fluoxetine induced threefold up-regulation (p < 0.01) by the 2nd and 3rd week, returning to the base level after drug withdrawal of both antidepressants. Moreover, the frequency of 5-HTR2C-edited isoforms showed no significant alterations, although analysis of the RNA-editing level at the single editing sites showed small decreases in the C' and D sites induced by reboxetine in P/FC. CONCLUSION: Our results suggest that chronic administration of antidepressants in rats slightly modifies the editing levels of 5-HT2C receptor but has considerable influence on its mRNA expression patterns in a way that is area- and time-specific.


Assuntos
Fluoxetina/farmacologia , Lobo Frontal/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Morfolinas/farmacologia , Córtex Pré-Frontal/efeitos dos fármacos , Receptor 5-HT2C de Serotonina/genética , Receptor 5-HT2C de Serotonina/metabolismo , Inibidores da Captação Adrenérgica/farmacologia , Animais , Regulação para Baixo/efeitos dos fármacos , Lobo Frontal/metabolismo , Hipocampo/metabolismo , Masculino , Córtex Pré-Frontal/metabolismo , Edição de RNA/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Reboxetina , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Fatores de Tempo , Regulação para Cima/efeitos dos fármacos
19.
PLoS One ; 5(9): e12596, 2010 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-20830301

RESUMO

BACKGROUND: The Flinders model is a validated genetic rat model of depression that exhibits a number of behavioural, neurochemical and pharmacological features consistent with those observed in human depression. PRINCIPAL FINDINGS: In this study we have used genome-wide microarray expression profiling of the hippocampus and prefrontal/frontal cortex of Flinders Depression Sensitive (FSL) and control Flinders Depression Resistant (FRL) lines to understand molecular basis for the differences between the two lines. We profiled two independent cohorts of Flinders animals derived from the same colony six months apart, each cohort statistically powered to allow independent as well as combined analysis. Using this approach, we were able to validate using real-time-PCR a core set of gene expression differences that showed statistical significance in each of the temporally distinct cohorts, representing consistently maintained features of the model. Small but statistically significant increases were confirmed for cholinergic (chrm2, chrna7) and serotonergic receptors (Htr1a, Htr2a) in FSL rats consistent with known neurochemical changes in the model. Much larger gene changes were validated in a number of novel genes as exemplified by TMEM176A, which showed 35-fold enrichment in the cortex and 30-fold enrichment in hippocampus of FRL animals relative to FSL. CONCLUSIONS: These data provide significant insights into the molecular differences underlying the Flinders model, and have potential relevance to broader depression research.


Assuntos
Depressão/genética , Depressão/psicologia , Perfilação da Expressão Gênica , Animais , Comportamento Animal , Depressão/metabolismo , Modelos Animais de Doenças , Humanos , Masculino , Ratos
20.
Prog Neuropsychopharmacol Biol Psychiatry ; 34(6): 1037-48, 2010 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-20580919

RESUMO

BACKGROUND: Availability of peripheral biomarkers for depression could aid diagnosis and help to predict treatment response. The objective of this work was to analyse the peripheral biomarker response in a gene-environment interaction model of depression. Genetically selected Flinders Sensitive Line (FSL) rats were subjected to maternal separation (MS), since early-life trauma is an important antecedent of depression. An open-ended approach based on a proteomic analysis of serum was combined with the evaluation of depression-associated proteins. METHODS: Rats experienced MS and chronically received escitalopram (ESC) or nortryptiline (NOR). Serum proteins were compared by two-dimensional gel electrophoresis. Corticosterone, cytokines, BDNF and C-reactive protein (CRP) were measured by immunoassays. RESULTS: Comparing FSL with the control Flinders Resistant Line (FRL), Apo-AI and Apo-AIV, alpha1-macroglobulin, glutathione peroxidase and complement-C3 were significantly modulated. Significant increases were detected in leptin, interleukin (IL) 1alpha and BDNF. CRP levels were significantly reduced. The impact of early-life stress was assessed by comparing FSL+MS versus FSL. Apo-E, alpha1-macroglobulin, complement-C3, transferrin and hemopexin were significantly modulated. The effect of stress in antidepressant response was then evaluated. In the comparison FSL+ESC+MS versus FSL+ESC, albumin, alpha1-macroglobulin, glutathione peroxidase and complement-C3 were modulated and significant reductions were detected in IL4, IL6, IL10, CRP and BDNF. By comparing FSL+NOR+MS versus FSL+NOR proteins like Apo-AIV, pyruvate dehydrogenase, alpha1-macroglobulin, transferrin and complement-C3 showed different levels. CONCLUSIONS: Lipid metabolism and immunity proteins were differently expressed in FSL in comparison with FRL. Exposure to MS induced changes in inflammation and transport proteins which became apparent in response to antidepressant treatments. Modulated proteins could suggest biomarker studies in humans.


Assuntos
Citalopram/farmacologia , Transtorno Depressivo/metabolismo , Privação Materna , Nortriptilina/farmacologia , Estresse Psicológico/metabolismo , Animais , Biomarcadores/metabolismo , Fator Neurotrófico Derivado do Encéfalo/sangue , Proteína C-Reativa/metabolismo , Corticosterona/sangue , Citocinas/sangue , Transtorno Depressivo/tratamento farmacológico , Transtorno Depressivo/psicologia , Modelos Animais de Doenças , Eletroforese em Gel Bidimensional , Ensaio de Imunoadsorção Enzimática , Imunoensaio , Proteômica , Ratos , Estresse Psicológico/psicologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...