Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
J Sleep Res ; 24(2): 124-30, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25223586

RESUMO

Brain-derived neurotrophic factor (Bdnf) regulates neuronal plasticity, slow wave activity and sleep homeostasis. Environmental stimuli control Bdnf expression through epigenetic mechanisms, but there are no data on epigenetic regulation of Bdnf by sleep or sleep deprivation. Here we investigated whether 5-methylcytosine (5mC) DNA modification at Bdnf promoters p1, p4 and p9 influences Bdnf1, Bdnf4 and Bdnf9a expression during the normal inactive phase or after sleep deprivation (SD) (3, 6 and 12 h, end-times being ZT3, ZT6 and ZT12) in rats in two brain areas involved in sleep regulation, the basal forebrain and cortex. We found a daytime variation in cortical Bdnf expression: Bdnf1 expression was highest at ZT6 and Bdnf4 lowest at ZT12. Such variation was not observed in the basal forebrain. Also Bdnf p1 and p9 methylation levels differed only in the cortex, while Bdnf p4 methylation did not vary in either area. Factorial analysis revealed that sleep deprivation significantly induced Bdnf1 and Bdnf4 with the similar pattern for Bdnf9a in both basal forebrain and cortex; 12 h of sleep deprivation decreased 5mC levels at the cortical Bdnf p4 and p9. Regression analysis between the 5mC promoter levels and the corresponding Bdnf transcript expression revealed significant negative correlations for the basal forebrain Bdnf1 and cortical Bdnf9a transcripts in only non-deprived rats, while these correlations were lost after sleep deprivation. Our results suggest that Bdnf transcription during the light phase of undisturbed sleep-wake cycle but not after SD is regulated at least partially by brain site-specific DNA methylation.


Assuntos
Prosencéfalo Basal/metabolismo , Fator Neurotrófico Derivado do Encéfalo/genética , Ritmo Circadiano/genética , Metilação de DNA , Lobo Frontal/metabolismo , Privação do Sono/genética , Sono/genética , Vigília/genética , Animais , Epigênese Genética , Masculino , Plasticidade Neuronal , Regiões Promotoras Genéticas/genética , RNA Mensageiro/análise , RNA Mensageiro/genética , Ratos , Privação do Sono/fisiopatologia , Fatores de Tempo
2.
J Affect Disord ; 350: 900-908, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38246279

RESUMO

BACKGROUND: The effects of the selective serotonin reuptake inhibitors (SSRIs), the first-line antidepressant treatment, have been proposed to be affected, at least in part, by the living environment. Since the quality of the environment depends not only on its objective features, but also on the subjective experience, we hypothesized that the latter plays a key role in determining SSRI treatment outcome. METHODS: We chronically administered the SSRI fluoxetine to two groups of adult CD-1 male mice that reportedly show distinct subjective experiences of the environment measured as consistent and significantly different responses to the same emotional and social stimuli. These distinct socioemotional profiles were generated by rearing mice either in standard laboratory conditions (SN) or in a communal nest (CN) where three dams breed together their offspring, sharing caregiving behavior. RESULTS: At adulthood, CN mice displayed higher levels of agonistic and anxiety-like behaviors than SN mice, indicating that they experience the environment as more socially challenging and potentially dangerous. We then administered fluoxetine, which increased offensive and anxious response in SN, while producing opposite effects in CN mice. BDNF regulation was modified by the treatment accordingly. LIMITATIONS: Subjective experience in mice was assessed as behavioral response to the environment. CONCLUSIONS: These results show that the subjective experience of the environment determines fluoxetine outcome. In a translational perspective, our findings suggest considering not only the objective quality, but also the subjective appraisal, of the patient's living environment for developing effective personalized therapeutic approaches in psychiatry.


Assuntos
Antidepressivos , Fluoxetina , Adulto , Camundongos , Masculino , Animais , Humanos , Fluoxetina/farmacologia , Fluoxetina/uso terapêutico , Antidepressivos/farmacologia , Antidepressivos/uso terapêutico , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Inibidores Seletivos de Recaptação de Serotonina/uso terapêutico , Ansiedade , Resultado do Tratamento
3.
Artigo em Inglês | MEDLINE | ID: mdl-36673825

RESUMO

Although the molecular mechanisms underlying methylmercury toxicity are not entirely understood, the observed neurotoxicity in early-life is attributed to the covalent binding of methylmercury to sulfhydryl (thiol) groups of proteins and other molecules being able to affect protein post-translational modifications from numerous molecular pathways, such as glutamate signaling, heat-shock chaperones and the antioxidant glutaredoxin/glutathione system. However, for other organomercurials such as ethylmercury or thimerosal, there is not much information available. Therefore, this review critically discusses current knowledge about organomercurials neurotoxicity-both methylmercury and ethylmercury-following intrauterine and childhood exposure, as well as the prospects and future needs for research in this area. Contrasting with the amount of epidemiological evidence available for methylmercury, there are only a few in vivo studies reporting neurotoxic outcomes and mechanisms of toxicity for ethylmercury or thimerosal. There is also a lack of studies on mechanistic approaches to better investigate the pathways involved in the potential neurotoxicity caused by both organomercurials. More impactful follow-up studies, especially following intrauterine and childhood exposure to ethylmercury, are necessary. Childhood vaccination is critically important for controlling infectious diseases; however, the safety of mercury-containing thimerosal and, notably, its effectiveness as preservative in vaccines are still under debate regarding its potential dose-response effects to the central nervous system.


Assuntos
Mercúrio , Compostos de Metilmercúrio , Síndromes Neurotóxicas , Vacinas , Humanos , Timerosal/toxicidade , Compostos de Metilmercúrio/toxicidade , Conservantes Farmacêuticos , Síndromes Neurotóxicas/etiologia , Compostos de Sulfidrila
4.
Cell Mol Neurobiol ; 30(7): 1117-23, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20614233

RESUMO

Neuronal activity regulates the expression of brain-derived neurotrophic factor (BDNF) in brain. In darkness, reduced neuronal activity in the visual cortex markedly decreases total BDNF transcription level in adult rats. Epigenetic mechanisms are crucially involved in the regulation of gene expression in response to environmental stimuli. In this study, we examined the effect of 1 week of light deprivation (LD) on the activity-dependent changes in BDNF expression from different promoters in the visual cortex and hippocampus. We analyzed the correlation between the chromatin state of Bdnf promoters, exon-specific transcripts levels, and total protein levels in light-deprived rats and in rats reared under normal light-dark cycle. We found that 1 week of LD significantly reduced Bdnf mRNA and protein in the visual cortex but not in the hippocampus. However, epigenetic analysis revealed that LD increased histone-3 methylation and DNA methylation at the Bdnf promoter IV in both the visual cortex and hippocampus. These data highlight the spatial differences in signaling pathways that lead to the BDNF expression in response to diminished ambient light.


Assuntos
Fator Neurotrófico Derivado do Encéfalo , Montagem e Desmontagem da Cromatina , Escuridão , Hipocampo/fisiologia , Córtex Visual/fisiologia , Animais , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Éxons , Humanos , Masculino , Regiões Promotoras Genéticas , Ratos , Ratos Wistar , Receptor trkB/genética , Receptor trkB/metabolismo
5.
J Neurochem ; 106(3): 1378-87, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18485098

RESUMO

Substantial evidence indicates that predisposition to diseases can be acquired during early stages of development and interactions between environmental and genetic factors may be implicated in the onset of many pathological conditions. Data collected over several decades have shown that chemicals are among the relevant factors that can endanger CNS. We previously showed that perinatal exposure to methylmercury (MeHg) causes persistent changes in learning and motivational behavior in mice. In this study, we report that the depression-like behavior in MeHg-exposed male mice is reversed by chronic treatment with the antidepressant fluoxetine. Behavioral alterations are associated with a decrease in brain-derived neurotrophic factor (BDNF) mRNA in the hippocampal dentate gyrus and fluoxetine treatment restores BDNF mRNA expression. We also show that MeHg-exposure induces long-lasting repressive state of the chromatin structure at the BDNF promoter region, in particular DNA hypermethylation, an increase in histone H3-K27 tri-methylation and a decrease in H3 acetylation at the promoter IV. While fluoxetine treatment does not alter hypermethylation of H3-K27, it significantly up-regulates H3 acetylation at the BDNF promoter IV in MeHg-exposed mice. Our study shows that developmental exposure to low levels of MeHg predisposes mice to depression and induces epigenetic suppression of BDNF gene expression in the hippocampus.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/genética , Depressão/metabolismo , Epigênese Genética/efeitos dos fármacos , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Compostos de Metilmercúrio/toxicidade , Efeitos Tardios da Exposição Pré-Natal/metabolismo , Animais , Fator Neurotrófico Derivado do Encéfalo/biossíntese , Depressão/induzido quimicamente , Depressão/tratamento farmacológico , Epigênese Genética/fisiologia , Feminino , Fluoxetina/uso terapêutico , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Imobilização , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Gravidez , Efeitos Tardios da Exposição Pré-Natal/genética
6.
Neuropsychopharmacology ; 43(2): 235-245, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28685757

RESUMO

Escalated or abnormal aggression induced by early adverse experiences is a growing issue of social concern and urges the development of effective treatment strategies. Here we report that synergistic interactions between psychosocial and biological factors specifically ameliorate escalated aggression induced by early adverse experiences. Rats reared in isolation from weaning until early adulthood showed abnormal forms of aggression and social deficits that were temporarily ameliorated by re-socialization, but aggression again escalated in a novel environment. We demonstrate that when re-socialization was combined with the antidepressant fluoxetine, which has been shown to reactivate juvenile-like state of plasticity, escalated aggression was greatly attenuated, while neither treatment alone was effective. Early isolation induced a permanent, re-socialization-resistant reduction in Bdnf expression in the amygdala and the infralimbic cortex. Only the combined treatment of fluoxetine and re-socialization was able to recover Bdnf expression via epigenetic regulation. Moreover, the behavior improvement after the combined treatment was dependent on TrkB activity. Combined treatment specifically strengthened the input from the ventral hippocampus to the mPFC, suggesting that this pathway is an important mediator of the beneficial behavioral effects of the combined psychosocial and pharmacological treatment of abnormal aggression. Our findings suggest that synergy between pharmacological induction of plasticity and psychosocial rehabilitation could enhance the efficacy of therapies for pathological aggression.


Assuntos
Agressão/fisiologia , Comportamento Animal/fisiologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Fluoxetina/farmacologia , Plasticidade Neuronal/fisiologia , Córtex Pré-Frontal/metabolismo , Receptor trkB/metabolismo , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Transdução de Sinais/fisiologia , Isolamento Social , Aprendizado Social/fisiologia , Socialização , Agressão/efeitos dos fármacos , Tonsila do Cerebelo/efeitos dos fármacos , Tonsila do Cerebelo/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Fator Neurotrófico Derivado do Encéfalo/efeitos dos fármacos , Epigênese Genética/efeitos dos fármacos , Epigênese Genética/fisiologia , Fluoxetina/administração & dosagem , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Masculino , Plasticidade Neuronal/efeitos dos fármacos , Córtex Pré-Frontal/efeitos dos fármacos , Ratos , Ratos Wistar , Receptor trkB/efeitos dos fármacos , Inibidores Seletivos de Recaptação de Serotonina/administração & dosagem , Transdução de Sinais/efeitos dos fármacos , Aprendizado Social/efeitos dos fármacos
7.
Gene ; 396(1): 39-45, 2007 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-17459613

RESUMO

Long terminal repeat (LTR) retrotransposon gtwin was initially discovered in silico, and then it was isolated as gypsy-homologous sequence from Drosophila melanogaster strain, G32. The presence of ORF3 suggests, that gtwin, like gypsy, may be an endogenous retrovirus, which can leave the cell and infect another one. Therefore, in this study we decided to investigate the distribution of gtwin in different species of the melanogaster subgroup in order to find out whether gtwin can be transferred horizontally as well as vertically. Gtwin was found in all 9 species of this subgroup, hence it seems to have inhabited the host genomes for a long time. In addition, we have shown that in the Drosophila erecta genome two gtwin families are present. The first one has 93% of identity to D. melanogaster element and is likely to be a descendant of gtwin that existed in Drosophila before the divergence of the melanogaster subgroup species. The other one has >99% of identity to D. melanogaster gtwin. The most reasonable explanation is that this element has been recently horizontally transferred between D. melanogaster and D. erecta. The number and variety of gtwin copies from the "infectious" family suggest that after the horizontal transfer into D. erecta genome, gtwin underwent amplification and aberrations, leading to the rise of its diverse variants.


Assuntos
Drosophila/classificação , Drosophila/genética , Amplificação de Genes , Transferência Genética Horizontal , Retroelementos/genética , Homologia de Sequência do Ácido Nucleico , Sequências Repetidas Terminais/genética , Animais , Células Clonais , DNA/isolamento & purificação , Genoma de Inseto/genética , Modelos Genéticos , Fases de Leitura Aberta/genética , Filogenia , Mapeamento por Restrição , Especificidade da Espécie
8.
Curr Top Med Chem ; 17(7): 771-793, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27086781

RESUMO

Neuronal network and plasticity change as a function of experience. Altered neural connectivity leads to distinct transcriptional programs of neuronal plasticity-related genes. The environmental challenges throughout life may promote long-lasting reprogramming of gene expression and the development of brain disorders. The modifications in neuronal epigenome mediate gene-environmental interactions and are required for activity-dependent regulation of neuronal differentiation, maturation and plasticity. Here, we highlight the latest advances in understanding the role of the main players of epigenetic machinery (DNA methylation and demethylation, histone modifications, chromatin-remodeling enzymes, transposons, and non-coding RNAs) in activity-dependent and long- term neural and synaptic plasticity. The review focuses on both the transcriptional and post-transcriptional regulation of gene expression levels, including the processes of promoter activation, alternative splicing, regulation of stability of gene transcripts by natural antisense RNAs, and alternative polyadenylation. Further, we discuss the epigenetic aspects of impaired neuronal plasticity and the pathogenesis of neurodevelopmental (Rett syndrome, Fragile X Syndrome, genomic imprinting disorders, schizophrenia, and others), stressrelated (mood disorders) and neurodegenerative Alzheimer's, Parkinson's and Huntington's disorders. The review also highlights the pharmacological compounds that modulate epigenetic programming of gene expression, the potential treatment strategies of discussed brain disorders, and the questions that should be addressed during the development of effective and safe approaches for the treatment of brain disorders.


Assuntos
Epigênese Genética , Plasticidade Neuronal , Humanos , Processamento Pós-Transcricional do RNA , RNA não Traduzido/genética , Transcrição Gênica
9.
Int J Dev Neurosci ; 44: 55-64, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25997908

RESUMO

The in utero exposure to common chemical stressors, environmental pollutant methylmercury and antidepressant fluoxetine, results in behavioral impairments persistent into adulthood. Modulation of critical periods in brain development may alter proper network formation and lastingly impair brain function. To investigate whether early-life stressors can modulate critical periods, we analyzed the development of parvalbumin (PV) and perineuronal nets (PNNs) in the dentate gyrus and CA1 area of the hippocampus and the basolateral amygdala in mice perinatally exposed to either fluoxetine or methylmercury. The number of PV and PNN neurons, and PV intensity, were analyzed by fluorescent immunohistochemistry at the postnatal ages P17 (ongoing critical period) and P24 (closing critical period). The exposure to fluoxetine did not affect the number of PV cells and PV intensity but decreased PNN formation around the cells at P17 and P24 in all tissues. In contrast, perinatal methylmercury inhibited the development of PV interneurons and PV expression at P17 only, but at P24 these parameters were restored. Methylmercury strongly increased PNN formation from P17 to P24 in the amygdala only. We suggest that perinatal fluoxetine and methylmercury might delay the closure and the onset, respectively, of the critical periods in the amygdala and hippocampus.


Assuntos
Encéfalo , Fluoxetina/toxicidade , Compostos de Metilmercúrio/toxicidade , Rede Nervosa/patologia , Parvalbuminas/metabolismo , Efeitos Tardios da Exposição Pré-Natal/patologia , Inibidores Seletivos de Recaptação de Serotonina/toxicidade , Fatores Etários , Animais , Animais Recém-Nascidos , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Encéfalo/patologia , Contagem de Células , Proteínas da Matriz Extracelular/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Rede Nervosa/metabolismo , Gravidez
10.
Neuropharmacology ; 76 Pt C: 709-18, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23587647

RESUMO

Brain-derived neurotrophic factor (BDNF) is a key mediator of the activity-dependent processes in the brain that have a major impact on neuronal development and plasticity. Impaired control of neuronal activity-induced BDNF expression mediates the pathogenesis of various neurological and psychiatric disorders. Different environmental stimuli, such as the use of pharmacological compounds, physical and learning exercises or stress exposure, lead to activation of specific neuronal networks. These processes entail tight temporal and spatial transcriptional control of numerous BDNF splice variants through epigenetic mechanisms. The present review highlights recent findings on the dynamic and long-term epigenetic programming of BDNF gene expression by the DNA methylation, histone-modifying and microRNA machineries. The review also summarizes the current knowledge on the activity-dependent BDNF mRNA trafficking critical for rapid local regulation of BDNF levels and synaptic plasticity. Current data open novel directions for discovery of new promising therapeutic targets for treatment of neuropsychiatric disorders. This article is part of the Special Issue entitled 'BDNF Regulation of Synaptic Structure, Function, and Plasticity'.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/genética , Epigênese Genética , Plasticidade Neuronal/genética , Neurônios/metabolismo , Animais , Encéfalo/citologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Metilação de DNA/genética , MicroRNAs/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , RNA Mensageiro/metabolismo , Sinapses
11.
Front Behav Neurosci ; 8: 257, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25147512

RESUMO

Several behavioral interventions, based on social enrichment and observational learning are applied in treatment of neuropsychiatric disorders. However, the mechanism of such modulatory effect and the safety of applied methods on individuals involved in social support need further investigation. We took advantage of known differences between inbred mouse strains to reveal the effect of social enrichment on behavior and neurobiology of animals with different behavioral phenotypes. C57BL/6 and DBA/2 female mice displaying multiple differences in cognitive, social, and emotional behavior were group-housed either in same-strain or in mixed-strain conditions. Comprehensive behavioral phenotyping and analysis of expression of several plasticity- and stress-related genes were done to measure the reciprocal effects of social interaction between the strains. Contrary to our expectation, mixed housing did not change the behavior of DBA/2 mice. Nevertheless, the level of serum corticosterone and the expression of glucocorticoid receptor Nr3c1 in the brain were increased in mixed housed DBA/2 as compared with those of separately housed DBA/2 mice. In contrast, socially active C57BL/6 animals were more sensitive to the mixed housing, displaying several signs of stress: alterations in learning, social, and anxiety-like behavior and anhedonia. These behavioral impairments were accompanied by the elevated serum corticosterone and the reduced expression of Nr3c1, as well as the elevated Bdnf levels in the cortex and hippocampus. Our results demonstrate the importance of social factors in modulation of both behavior and the underlying neurobiological mechanisms in stress response, and draw attention to the potential negative impact of social interventions for individuals involved in social support.

12.
Eur Neuropsychopharmacol ; 24(7): 1162-74, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24837571

RESUMO

The antidepressant fluoxetine induces synaptic plasticity in the visual and fear networks and promotes the structural remodeling of neuronal circuits, which is critical for experience-dependent plasticity in response to an environmental stimulus. We recently demonstrated that chronic fluoxetine administration together with extinction training in adult mice reduced fear in a context-independent manner. Fear conditioning and extinction alter excitatory and inhibitory transmissions within the fear circuitry. In this study, we investigated whether fluoxetine, extinction or their combination produced distinct long-lasting changes in the synaptic protein profile in the amygdala, hippocampus and prefrontal cortex of conditioned mice. We determined that extinction induced synaptophysin expression and down-regulated the GluA1:GluA2 ratio throughout the fear network in water- and fluoxetine-treated mice, suggesting a common fluoxetine-independent mechanism for increased synaptic transmission and re-arrangement of AMPA-receptors by extinction training. In contrast to common changes, the presynaptic vesicular neurotransmitter transporters VGAT and Vglut1 were upregulated after extinction in water- and fluoxetine-treated mice, respectively. The cortical levels of the GABA transporter Gat1 were reduced in high-freezing water-drinking mice, suggesting a maladaptive increase of GABA spillover at cortical inhibitory synapses. Fear conditioning decreased, and extinction induced the expression of GABA-receptor alpha1 and alpha2 subunits in water- and fluoxetine-treated mice, respectively. Only a combination of fluoxetine with extinction enhanced GluN2A expression in the amygdala and hippocampus, emphasizing the role of this NMDA-receptor subunit in the successful erasure of fear memories. Our finding provides novel data that may become helpful in developing beneficial pharmacological fear-reducing treatment strategies.


Assuntos
Extinção Psicológica/fisiologia , Medo/efeitos dos fármacos , Fluoxetina/farmacologia , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Sinapses/metabolismo , Animais , Condicionamento Clássico/efeitos dos fármacos , Condicionamento Clássico/fisiologia , Extinção Psicológica/efeitos dos fármacos , Medo/fisiologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Camundongos , Plasticidade Neuronal/efeitos dos fármacos , Plasticidade Neuronal/fisiologia , Subunidades Proteicas/metabolismo , Receptores de AMPA/metabolismo , Sinapses/efeitos dos fármacos , Sinaptofisina/metabolismo
13.
Front Behav Neurosci ; 8: 315, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25309367

RESUMO

Developmental exposure to low dose of methylmercury (MeHg) has a long-lasting effect on memory and attention deficits in humans, as well as cognitive performance, depression-like behavior and the hippocampal levels of the brain-derived neurotrophic factor (Bdnf)in mice. The Bdnf receptor TrkB is a key player of Bdnf signaling. Using transgenic animals, here we analyzed the effect of the full-length TrkB overexpression (TK+) on behavior impairments induced by perinatal MeHg. TK overexpression in the MeHg-exposed mice enhanced generalized anxiety and cue memory in the fear conditioning (FC) test. Early exposure to MeHg induced deficits in reversal spatial memory in the Morris water maze (MWM) test and depression-like behavior in the forced swim test (FST) in only wild-type (WT) mice but did not affect these parameters in TK+ mice. These changes were associated with TK+ effect on the increase in Bdnf 2, 3, 4 and 6 transcription in the hippocampus as well as with interaction of TK+ and MeHg factors for Bdnf 1, 9a and truncated TrkB.T1 transcripts in the prefrontal cortex. However, the MeHg-induced anxiety-like behavior in the elevated plus maze (EPM) and open field (OF) tests was ameliorated by TK+ background only in the OF test. Moreover, TK overexpression in the MeHg mice did not prevent significant stress-induced weight loss during the period of adaptation to individual housing in metabolic cages. These TK genotype-independent changes were primarily accompanied by the MeHg-induced hippocampal deficits in the activity-dependent Bdnf 1, 4 and 9a variants, TrkB.T1, and transcripts for important antioxidant enzymes glyoxalases Glo1 and Glo2 and glutathione reductase Gsr. Our data suggest a role of full-length TrkB in buffering against memory deficits and depression-like behavior in the MeHg mice but propose the involvement of additional pathways, such as the antioxidant system or TrkB.T1 signaling, in stress- or anxiety-related responses induced by developmental MeHg exposure.

14.
PLoS One ; 8(7): e68722, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23844236

RESUMO

Brain-derived neurotrophic factor (BDNF) importantly regulates learning and memory and supports the survival of injured neurons. Reduced BDNF levels have been detected in the brains of Alzheimer's disease (AD) patients but the exact role of BDNF in the pathophysiology of the disorder remains obscure. We have recently shown that reduced signaling of BDNF receptor TrkB aggravates memory impairment in APPswe/PS1dE9 (APdE9) mice, a model of AD. The present study examined the influence of Bdnf gene deficiency (heterozygous knockout) on spatial learning, spontaneous exploratory activity and motor coordination/balance in middle-aged male and female APdE9 mice. We also studied brain BDNF protein levels in APdE9 mice in different ages showing progressive amyloid pathology. Both APdE9 and Bdnf mutations impaired spatial learning in males and showed a similar trend in females. Importantly, the effect was additive, so that double mutant mice performed the worst. However, APdE9 and Bdnf mutations influenced spontaneous locomotion in contrasting ways, such that locomotor hyperactivity observed in APdE9 mice was normalized by Bdnf deficiency. Obesity associated with Bdnf deficiency did not account for the reduced hyperactivity in double mutant mice. Bdnf deficiency did not alter amyloid plaque formation in APdE9 mice. Before plaque formation (3 months), BDNF protein levels where either reduced (female) or unaltered (male) in the APdE9 mouse cortex. Unexpectedly, this was followed by an age-dependent increase in mature BDNF protein. Bdnf mRNA and phospho-TrkB levels remained unaltered in the cortical tissue samples of middle-aged APdE9 mice. Immunohistological studies revealed increased BDNF immunoreactivity around amyloid plaques indicating that the plaques may sequester BDNF protein and prevent it from activating TrkB. If similar BDNF accumulation happens in human AD brains, it would suggest that functional BDNF levels in the AD brains are even lower than reported, which could partially contribute to learning and memory problems of AD patients.


Assuntos
Doença de Alzheimer/genética , Doença de Alzheimer/patologia , Fator Neurotrófico Derivado do Encéfalo/genética , Transtornos da Memória/etiologia , Doença de Alzheimer/complicações , Precursor de Proteína beta-Amiloide/genética , Animais , Peso Corporal/genética , Encéfalo/metabolismo , Encéfalo/patologia , Fator Neurotrófico Derivado do Encéfalo/deficiência , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Modelos Animais de Doenças , Meio Ambiente , Feminino , Humanos , Hipercinese/genética , Masculino , Aprendizagem em Labirinto , Memória , Camundongos , Camundongos Transgênicos , Placa Amiloide , Presenilina-1/genética , Desempenho Psicomotor
15.
Neurobiol Aging ; 33(6): 1122.e23-39, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22209410

RESUMO

Brain-derived neurotrophic factor (BDNF) plays an important role in neuronal plasticity, learning, and memory. Levels of BDNF and its main receptor TrkB (TrkB.TK) have been reported to be decreased while the levels of the truncated TrkB (TrkB.T1) are increased in Alzheimer's disease. We show here that incubation with amyloid-ß increased TrkB.T1 receptor levels and decreased TrkB.TK levels in primary neurons. In vivo, APPswe/PS1dE9 transgenic mice (APdE9) showed an age-dependent relative increase in cortical but not hippocampal TrkB.T1 receptor levels compared with TrkB.TK. To investigate the role of TrkB isoforms in Alzheimer's disease, we crossed AP mice with mice overexpressing the truncated TrkB.T1 receptor (T1) or the full-length TrkB.TK isoform. Overexpression of TrkB.T1 in APdE9 mice exacerbated their spatial memory impairment while the overexpression of TrkB.TK alleviated it. These data suggest that amyloid-ß changes the ratio between TrkB isoforms in favor of the dominant-negative TrkB.T1 isoform both in vitro and in vivo and supports the role of BDNF signaling through TrkB in the pathophysiology and cognitive deficits of Alzheimer's disease.


Assuntos
Precursor de Proteína beta-Amiloide/genética , Transtornos da Memória/metabolismo , Presenilina-1/genética , Receptor trkB/antagonistas & inibidores , Transdução de Sinais/genética , Precursor de Proteína beta-Amiloide/biossíntese , Animais , Células Cultivadas , Feminino , Masculino , Transtornos da Memória/genética , Transtornos da Memória/psicologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Presenilina-1/biossíntese , Receptor trkB/biossíntese , Receptor trkB/genética
16.
Neurosci Lett ; 495(3): 168-72, 2011 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-21420470

RESUMO

During early postnatal phase, the environment deeply affects developmental trajectories through epigenetic mechanisms that control the levels of key molecules for brain function, such as neurotrophins. Indeed, it has been shown that adverse early experiences induce epigenetic modifications leading to decreased brain derived neurotrophic factor (BDNF) levels at adulthood. However, no data about the effects of enriching early experiences are available. Here we exploit the mouse Communal Nest (CN) paradigm in order to investigate the effects of a highly stimulating early social environment on BDNF epigenetic modifications and protein expression at adulthood. CN, which consists of a single nest where three mothers keep their pups together and share care-giving behavior until weaning, is characterized by high levels of maternal behavior and peer interactions. Our results show that CN leads to high levels of histone acetylation at the BDNF gene at adulthood, which is more permissive to expression. However, such epigenetic modification is associated to increased BDNF protein expression only 1h after an environmental challenge and not at baseline or 3h after the challenge, suggesting that the epigenetic modifications do not affect expression under steady-state conditions but allow a fast increase in BDNF levels following stimulation. The present findings corroborate the role of epigenetic modifications in mediating the effects of the early social environment on adult brain function and behavior. In addition, these show, for the first time, an association between an epigenetic modification and a change in the rapidity of induction of protein expression, expanding the knowledge on the mechanisms by which epigenetic changes modify brain function.


Assuntos
Fator Neurotrófico Derivado do Encéfalo/metabolismo , Encéfalo/metabolismo , Epigenômica , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Meio Social , Análise de Variância , Animais , Animais Recém-Nascidos , Fator Neurotrófico Derivado do Encéfalo/genética , Imunoprecipitação da Cromatina/métodos , Feminino , Histonas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos ICR , Fatores de Tempo
17.
Science ; 334(6063): 1731-4, 2011 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-22194582

RESUMO

Antidepressant drugs and psychotherapy combined are more effective in treating mood disorders than either treatment alone, but the neurobiological basis of this interaction is unknown. To investigate how antidepressants influence the response of mood-related systems to behavioral experience, we used a fear-conditioning and extinction paradigm in mice. Combining extinction training with chronic fluoxetine, but neither treatment alone, induced an enduring loss of conditioned fear memory in adult animals. Fluoxetine treatment increased synaptic plasticity, converted the fear memory circuitry to a more immature state, and acted through local brain-derived neurotrophic factor. Fluoxetine-induced plasticity may allow fear erasure by extinction-guided remodeling of the memory circuitry. Thus, the pharmacological effects of antidepressants need to be combined with psychological rehabilitation to reorganize networks rendered more plastic by the drug treatment.


Assuntos
Antidepressivos de Segunda Geração/uso terapêutico , Transtornos de Ansiedade/terapia , Terapia Comportamental , Extinção Psicológica , Medo , Fluoxetina/uso terapêutico , Plasticidade Neuronal/efeitos dos fármacos , Tonsila do Cerebelo/citologia , Tonsila do Cerebelo/efeitos dos fármacos , Tonsila do Cerebelo/fisiologia , Animais , Antidepressivos de Segunda Geração/farmacologia , Fator Neurotrófico Derivado do Encéfalo/genética , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Terapia Combinada , Condicionamento Clássico , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Fluoxetina/farmacologia , Interneurônios/efeitos dos fármacos , Interneurônios/fisiologia , Masculino , Memória , Camundongos , Camundongos Endogâmicos C57BL , Rede Nervosa/efeitos dos fármacos , Rede Nervosa/fisiologia , Neurônios/citologia , Neurônios/efeitos dos fármacos , Transmissão Sináptica/efeitos dos fármacos
18.
Eur Neuropsychopharmacol ; 19(2): 97-108, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18973993

RESUMO

There is evidence that antidepressant drug treatment during a critical period of postnatal development renders mice susceptible to depression- and anxiety-related behaviour in adulthood. The mechanism of how early antidepressant treatment brings about long-term effects in emotional behaviour is not yet understood, but neurotrophins, particularly brain-derived neurotrophic factor (BDNF), have been implicated in this context. We examined the long-term effects of a transient early postnatal fluoxetine treatment on depression- and anxiety-related behaviours as well as gene expression of BDNF and its receptor TrkB in C57BL/6J mice. Treatment with fluoxetine between postnatal days P4 and P21 resulted in a significant loss of body weight and long-lasting behavioural inhibition in adult mice in response to stressful events such as the light-dark or open field tests. Postnatal fluoxetine exposure also decreased behavioural despair in the forced swim test. Both body weight and behavioural alterations were restored by chronic fluoxetine treatment in adulthood. The behavioral alterations were accompanied by changes in hippocampal BDNF mRNA. Specifically, we show that early-life fluoxetine exposure resulted in the long-term upregulation of BDNF expression in adult mice. However, chromatin immunoprecipitation studies did not reveal any changes in the acetylation or trimethylation of histone H3 at the BDNF promoters. Our experiments show that behavioural and molecular changes induced by early postnatal fluoxetine administration are reversed by chronic fluoxetine treatment of adult mice to control levels.


Assuntos
Animais Recém-Nascidos/psicologia , Comportamento Animal/efeitos dos fármacos , Química Encefálica/efeitos dos fármacos , Fluoxetina/farmacologia , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Animais , Ansiedade/psicologia , Peso Corporal/efeitos dos fármacos , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Cromatina/metabolismo , Escuridão , Depressão/psicologia , Comportamento Exploratório/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Imunoprecipitação , Luz , Masculino , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Receptor trkB/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Natação/psicologia
19.
Mol Genet Genomics ; 279(5): 463-72, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18253751

RESUMO

Mobile genetic elements constitute a substantial part of eukaryotic genome and play an important role in its organization and functioning. Co-evolution of retrotransposons and their hosts resulted in the establishment of control systems employing mechanisms of RNA interference that seem to be impossible to evade. However, "active" copies of endogenous retrovirus gypsy escape cellular control in some cases, while its evolutionary elder "inactive" variants do not. To clarify the evolutionary relationship between "active" and "inactive" gypsy we combined two approaches: the analysis of gypsy sequences, isolated from G32 Drosophila melanogaster strain and from different Drosophila species of the melanogaster subgroup, as well as the study of databases, available on the Internet. No signs of "intermediate" (between "active" and "inactive") gypsy form were found in GenBank, and four full-size G32 gypsy copies demonstrated a convergence that presumably involves gene conversion. No "active" gypsy were revealed among PCR generated gypsy ORF3 sequences from the various Drosophila species indicating that "active" gypsy appeared in some population of D. melanogaster and then started to spread out. Analysis of sequences flanking gypsy variants in G32 revealed their predominantly heterochromatic location. Discrepancy between the structure of actual gypsy sites in G32 and corresponding sequences in database might indicate significant inter-strain heterochromatin diversity.


Assuntos
Evolução Biológica , Sequência Conservada , Drosophila melanogaster/genética , Retrovirus Endógenos/genética , Polimorfismo Genético , Animais , Sequência de Bases , Biblioteca Gênica , Dados de Sequência Molecular , Fases de Leitura Aberta/genética , Filogenia , Retroelementos/genética , Retroelementos/fisiologia , Homologia de Sequência do Ácido Nucleico , Especificidade da Espécie
20.
Cell Motil Cytoskeleton ; 63(5): 301-12, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16518797

RESUMO

In this study we describe a novel Drosophila protein Jupiter, which shares properties with several structural microtubule-associated proteins (MAPs) including TAU, MAP2, MAP4. Jupiter is a soluble unfolded molecule with the high net positive charge, rich in Glycine. It possesses two degenerated repeats around the sequence PPGG, separated by a Serine-rich region. Jupiter associates with microtubules in vitro and, fused with the green fluorescent protein (GFP), is an excellent marker to follow microtubule dynamics in vivo. In a jupiter transgenic Drosophila strain generated by the "protein-trap" technique, Jupiter:GFP fusion protein localizes to the microtubule network through the cell cycle at the different stages of development. We found particularly high Jupiter:GFP concentrations in the young embryo, larval nervous system, precursors of eye photoreceptors and adult ovary. Moreover, from jupiter:gfp embryos we have established two permanent cell lines presenting strongly fluorescent microtubules during the whole cell cycle. In these cells, the distribution of the Jupiter:GFP fusion protein reproduces microtubule behavior upon treatment by the drugs colchicine and taxol. The jupiter cell lines and fly strain should be of wide interest for biologists interested in in vivo analysis of microtubule dynamics.


Assuntos
Proteínas de Drosophila/análise , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Proteínas dos Microtúbulos/análise , Proteínas dos Microtúbulos/metabolismo , Microtúbulos/química , Sequência de Aminoácidos , Animais , Proteínas de Drosophila/genética , Drosophila melanogaster/citologia , Drosophila melanogaster/genética , Embrião não Mamífero/química , Embrião não Mamífero/citologia , Genes de Insetos , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Microscopia de Fluorescência , Proteínas dos Microtúbulos/genética , Microtúbulos/metabolismo , Dados de Sequência Molecular , Proteínas Recombinantes de Fusão/análise , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA