Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 154
Filtrar
1.
J Neurosci ; 39(18): 3454-3469, 2019 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-30804093

RESUMO

Vagus nerve stimulation (VNS) has been shown to enhance learning and memory, yet the mechanisms behind these enhancements are unknown. Here, we present evidence that epigenetic modulation underlies VNS-induced improvements in cognition. We show that VNS enhances novelty preference (NP); alters the hippocampal, cortical, and blood epigenetic transcriptomes; and epigenetically modulates neuronal plasticity and stress-response signaling genes in male Sprague Dawley rats. Brain-behavior analysis revealed structure-specific relationships between NP test performance (NPTP) and epigenetic alterations. In the hippocampus, NPTP correlated with decreased histone deacetylase 11 (HDAC11), a transcriptional repressor enriched in CA1 cells important for memory consolidation. In the cortex, the immediate early gene (IEG) ARC was increased in VNS rats and correlated with transcription of plasticity genes and epigenetic regulators, including HDAC3. For rats engaged in NPTP, ARC correlated with performance. Interestingly, blood ARC transcripts decreased in VNS rats performing NPTP, but increased in VNS-only rats. Because DNA double-strand breaks (DSBs) facilitate transcription of IEGs, we investigated phosphorylated H2A.X (γH2A.X), a histone modification known to colocalize with DSBs. In agreement with reduced cortical stress-response transcription factor NF-κB1, chromatin immunoprecipitation revealed reduced γH2A.X in the ARC promoter. Surprisingly, VNS did not significantly reduce transcription of cortical or hippocampal proinflammatory cytokines. However, TNFRSF11B (osteoprotegerin) correlated with NPTP as well as plasticity, stress-response signaling, and epigenetic regulation transcripts in both hippocampus and cortex. Together, our findings provide the first evidence that VNS induces widespread changes in the cognitive epigenetic landscape and specifically affects epigenetic modulators associated with NPTP, stress-response signaling, memory consolidation, and cortical neural remodeling.SIGNIFICANCE STATEMENT Recent studies have implicated vagus nerve stimulation (VNS) in enhanced learning and memory. However, whereas epigenetic modifications are known to play an important role in memory, the particular mechanisms involved in VNS-enhanced cognition are unknown. In this study, we examined brain and behavior changes in VNS and sham rats performing a multiday novelty preference (NP) task. We found that VNS activated specific histone modifications and DNA methylation changes at important stress-response signaling and plasticity genes. Both cortical and hippocampal plasticity changes were predictive of NP test performance. Our results reveal important epigenetic alterations associated with VNS cognitive improvements, as well as new potential pharmacological targets for enhancing cortical and hippocampal plasticity.


Assuntos
Cognição/fisiologia , Epigênese Genética , Estimulação do Nervo Vago , Animais , Sinalização do Cálcio , Córtex Cerebral/metabolismo , Comportamento Exploratório , Hipocampo/metabolismo , Masculino , Plasticidade Neuronal , Ratos Sprague-Dawley , Estresse Fisiológico , Transcriptoma
2.
Horm Behav ; 126: 104848, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32918873

RESUMO

The increased prevalence of neurodevelopmental disorders during the last half-century led us to investigate the potential for intergenerational detrimental neurodevelopmental effects of synthetic female gonadal hormones, typically used in contraceptive pills. We examined 3 separate cohorts of mice over the span of 2 years, a total of 150 female F0 mice and over 300 male and female rodents from their F1 progeny. We demonstrate that F1 male offsprings of female mice previously exposed to the synthetic estrogen 17α-ethinylestradiol (EE2) in combination with the synthetic progestin Norethindrone, exhibit neurodevelopmental and behavioral differences compared to control mice. Because the EE2 + Norethindrone administration resulted in gene expression changes in the exposed F0 mice ovaries persisting after the end of treatment, it is likely that the synthetic hormone treatment caused changes in the germline cells and that led to altered neurodevelopment in the offsprings. An altered gene expression pattern was discovered in the frontal cortex of male mice from the first offspring (F1.1) at infancy and an ADHD-like hyperactive locomotor behavior was exhibited in young male mice from the second offspring (F1.2) of female mice treated with contraceptive pill doses of EE2 + Norethindrone prior to pregnancy. The intergenerational neurodevelopmental effects of EE2 + Norethindrone treatment were sex specific, predominantly affecting males. Our observations in mice support the hypothesis that the use of synthetic contraceptive hormones is a potential environmental factor impacting the prevalence of human neurodevelopmental disorders. Additionally, our results indicate that contraceptive hormone drug safety assessments may need to be extended to F1 offspring.


Assuntos
Encéfalo/embriologia , Contraceptivos Hormonais/efeitos adversos , Congêneres do Estradiol/efeitos adversos , Exposição Materna/efeitos adversos , Animais , Comportamento Animal/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Encéfalo/crescimento & desenvolvimento , Cognição/efeitos dos fármacos , Etinilestradiol/efeitos adversos , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transtornos do Neurodesenvolvimento/induzido quimicamente , Transtornos do Neurodesenvolvimento/fisiopatologia , Gravidez
3.
Nature ; 515(7528): 582-6, 2014 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-25219850

RESUMO

Memory formation is a multi-stage process that initially requires cellular consolidation in the hippocampus, after which memories are downloaded to the cortex for maintenance, in a process termed systems consolidation. Epigenetic mechanisms regulate both types of consolidation, but histone variant exchange, in which canonical histones are replaced with their variant counterparts, is an entire branch of epigenetics that has received limited attention in the brain and has never, to our knowledge, been studied in relation to cognitive function. Here we show that histone H2A.Z, a variant of histone H2A, is actively exchanged in response to fear conditioning in the hippocampus and the cortex, where it mediates gene expression and restrains the formation of recent and remote memory. Our data provide evidence for H2A.Z involvement in cognitive function and specifically implicate H2A.Z as a negative regulator of hippocampal consolidation and systems consolidation, probably through downstream effects on gene expression. Moreover, alterations in H2A.Z binding at later stages of systems consolidation suggest that this histone has the capacity to mediate stable molecular modifications required for memory retention. Overall, our data introduce histone variant exchange as a novel mechanism contributing to the molecular basis of cognitive function and implicate H2A.Z as a potential therapeutic target for memory disorders.


Assuntos
Epigênese Genética , Histonas/genética , Histonas/metabolismo , Memória/fisiologia , Animais , Cognição/fisiologia , Condicionamento Psicológico/fisiologia , Medo/fisiologia , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Hipocampo/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica
4.
Neurobiol Learn Mem ; 161: 149-157, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31002880

RESUMO

Transcriptional changes in the hippocampus are required for memory formation, and these changes are regulated by numerous post-translational modifications of chromatin-associated proteins. One of the epigenetic marks that has been implicated in memory formation is histone 3 lysine 4 trimethylation (H3K4me3), and this modification is found at the promoters of actively transcribed genes. The total levels of H3K4me3 are increased in the CA1 region of the hippocampus during memory formation, and genetic perturbation of the K4 methyltransferases and demethylases interferes with forming memories. Previous chromatin immunoprecipitation followed by deep sequencing (ChIP-seq) analyses failed to detect changes in H3K4me3 levels at the promoters of memory-linked genes. Since the breadth of H3K4me3 marks was recently reported to be associated with the transcriptional outcome of a gene, we re-analyzed H3K4me3 ChIP-seq data sets to identify the role of H3K4me3 broad domains in CA1 neurons, as well as identify differences in breadth that occur during contextual fear conditioning. We found that, under baseline conditions, broad H3K4me3 peaks mark important learning and memory genes and are often regulated by super-enhancers. The peaks at many learning-associated genes become broader during novel environment exposure and memory formation. Furthermore, the important learning- and memory-associated lysine methyltransferases, Kmt2a and Kmt2b, are involved in maintaining H3K4me3 peak width. Our findings highlight the importance of analyzing H3K4me3 peak shape, and demonstrate that breadth of H3K4me3 marks in neurons of the hippocampus is regulated during memory formation.


Assuntos
Região CA1 Hipocampal/metabolismo , Condicionamento Clássico/fisiologia , Epigênese Genética/fisiologia , Histonas/metabolismo , Memória/fisiologia , Transcrição Gênica/fisiologia , Ativação Transcricional/fisiologia , Animais , Medo/fisiologia , Feminino , Histona-Lisina N-Metiltransferase/metabolismo , Masculino , Metilação , Camundongos , Camundongos Endogâmicos C57BL , Proteína de Leucina Linfoide-Mieloide/metabolismo
5.
Nurs Res ; 68(2): 145-155, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30586060

RESUMO

BACKGROUND: Multiple cell signaling pathways are implicated in the development, progression, and persistence of cisplatin-induced peripheral neuropathy. Although advances have been made in terms of understanding specific neurotoxic mechanisms, there are few predictive factors identified that can help inform the clinician approach to symptom prevention or management. OBJECTIVE: We investigate the differential sensitivity to cisplatin-induced peripheral neuropathy and examine the contribution of dorsal root ganglion (DRG) transcriptional profiles across two inbred strains of mice. METHODS: Cisplatin (4 mg/kg intraperitoneal or vehicle control) was administered twice a week for 4 weeks to adult female C57BL/6J and A/J mice-the C57BL/6J strain of mice characterized by a robust mechanical allodynia and the A/J with a mild largely resistant allodynia phenotype. Peripheral nerve conduction velocities (NCVs), electrophysiological evaluation of wide dynamic range (WDR) neurons, morphological examination of DRG neurons, and microarray analysis of spinal cord tissues were compared across the 4 weeks. RESULTS: The A/J strain presents with an early, mild nocifensive response to cisplatin with reduced neuronal activity in WDR neurons and small changes in cross-sectional nucleus size in DRG neurons at 4 weeks. The more nocifensive-sensitive C57BL/6J strain presents with no early changes in WDR neuron responsiveness; however, there were significant changes in DRG size. Both strains demonstrate a drop in NCV after 4 weeks of treatment, with the greatest reduction present in the A/J strain. Transcriptome data implicate neuroimmune modulation in the differential response to cisplatin in the DRGs of A/J and C57BL/6J mice. DISCUSSION: Nocifensive responses in both strains implicate involvement of small myelinated and unmyelinated fibers in neurotoxic cisplatin response, whereas reductions in NCV reflect involvement of the largest myelinated fibers in the peripheral nerves. Microarray data analysis identifies neuropathy-relevant gene sets with differential activation of pathways, suggesting a role for antigen presentation in the differential neurotoxic response to cisplatin across strains. Further research is indicated to determine the relative contributions of each of these potential pathological mechanisms to both the neurotoxic response to cisplatin and to the potential for targeted therapy.


Assuntos
Antineoplásicos/farmacologia , Cisplatino/farmacologia , Canais de Cátion Regulados por Nucleotídeos Cíclicos/metabolismo , Neuralgia/fisiopatologia , Doenças do Sistema Nervoso Periférico/induzido quimicamente , Receptores de Fator de Crescimento Neural/metabolismo , Animais , Apoptose/efeitos dos fármacos , Gânglios Espinais/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL
6.
Crit Rev Biochem Mol Biol ; 51(3): 185-94, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26915423

RESUMO

Over the past decade, since epigenetic mechanisms were first implicated in memory formation and synaptic plasticity, dynamic DNA methylation reactions have been identified as integral to long-term memory formation, maintenance, and recall. This review incorporates various new findings that DNA methylation mechanisms are important regulators of non-Hebbian plasticity mechanisms, suggesting that these epigenetic mechanisms are a fundamental link between synaptic plasticity and metaplasticity. Because the field of neuroepigenetics is so young and the biochemical tools necessary to probe gene-specific questions are just now being developed and used, this review also speculates about the direction and potential of therapeutics that target epigenetic mechanisms in the central nervous system and the unique pharmacokinetic and pharmacodynamic properties that epigenetic therapies may possess. Mapping the dynamics of the epigenome in response to experiential learning, even a single epigenetic mark in isolation, remains a significant technical and bioinformatic hurdle facing the field, but will be necessary to identify changes to the methylome that govern memory-associated gene expression and effectively drug the epigenome.


Assuntos
Metilação de DNA , Epigênese Genética , Memória , Animais , Metilação de DNA/efeitos dos fármacos , Descoberta de Drogas , Epigênese Genética/efeitos dos fármacos , Genômica , Humanos , Memória/efeitos dos fármacos , Plasticidade Neuronal/efeitos dos fármacos
7.
Annu Rev Pharmacol Toxicol ; 55: 591-611, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25340930

RESUMO

In this review, we discuss the potential pharmacological targeting of a set of powerful epigenetic mechanisms: DNA methylation control systems in the central nervous system (CNS). Specifically, we focus on the possible use of these targets for novel future treatments for learning and memory disorders. We first describe several unique pharmacological attributes of epigenetic mechanisms, especially DNA cytosine methylation, as potential drug targets. We then present an overview of the existing literature regarding DNA methylation control pathways and enzymes in the nervous system, particularly as related to synaptic function, plasticity, learning and memory. Lastly, we speculate upon potential categories of CNS cognitive disorders that might be amenable to methylomic targeting.


Assuntos
Fármacos do Sistema Nervoso Central/uso terapêutico , Metilação de DNA/efeitos dos fármacos , Descoberta de Drogas/métodos , Epigênese Genética/efeitos dos fármacos , Transtornos Mentais/tratamento farmacológico , Terapia de Alvo Molecular/métodos , Animais , Cognição/efeitos dos fármacos , Metilases de Modificação do DNA/antagonistas & inibidores , Metilases de Modificação do DNA/química , Metilases de Modificação do DNA/metabolismo , Inibidores Enzimáticos/uso terapêutico , Inativação Gênica/efeitos dos fármacos , Humanos , Memória/efeitos dos fármacos , Transtornos Mentais/genética , Transtornos Mentais/psicologia , Conformação de Ácido Nucleico , Conformação Proteica , Relação Estrutura-Atividade
8.
Mol Vis ; 24: 153-164, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29463953

RESUMO

Purpose: Epigenetic and transcriptional mechanisms have been shown to contribute to long-lasting functional changes in adult neurons. The purpose of this study was to identify any such modifications in diseased retinal tissues from a mouse model of rhodopsin mutation-associated autosomal dominant retinitis pigmentosa (ADRP), Q344X, relative to age-matched wild-type (WT) controls. Methods: We performed RNA sequencing (RNA-seq) at poly(A) selected RNA to profile the transcriptional patterns in 3-week-old ADRP mouse model rhodopsin Q344X compared to WT controls. Differentially expressed genes were determined by DESeq2 using the Benjamini & Hochberg p value adjustment and an absolute log2 fold change cutoff. Quantitative western blots were conducted to evaluate protein expression levels of histone H3 phosphorylated at serine 10 and histone H4. qRT-PCR was performed to validate the expression patterns of differentially expressed genes. Results: We observed significant differential expression in 2151 genes in the retina of Q344X mice compared to WT controls, including downregulation in the potassium channel gene, Kcnv2, and differential expression of histone genes, including the H1 family histone member, H1foo; the H3 histone family 3B, H3f3b; and the histone deacetylase 9, Hdac9. Quantitative western blots revealed statistically significant decreased protein expression of both histone H3 phosphorylated at serine 10 and histone H4 in 3-week-old Q344X retinas. Furthermore, qRT-PCR performed on select differentially expressed genes based on our RNA-seq results revealed matched expression patterns of up or downregulation. Conclusions: These findings provide evidence that transcriptomic alterations occur in the ADRP mouse model rhodopsin Q344X retina and that these processes may contribute to the dysfunction and neurodegeneration seen in this animal model.


Assuntos
Substituição de Aminoácidos , Cromatina/metabolismo , Transtornos Cromossômicos/genética , Retinose Pigmentar/genética , Rodopsina/genética , Transcrição Gênica , Animais , Cromatina/química , Transtornos Cromossômicos/metabolismo , Transtornos Cromossômicos/patologia , Feminino , Perfilação da Expressão Gênica , Técnicas de Introdução de Genes , Genes Dominantes , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Mutação , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Retinose Pigmentar/metabolismo , Retinose Pigmentar/patologia , Rodopsina/metabolismo
9.
Nature ; 551(7681): 448-449, 2017 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-29168827
10.
Learn Mem ; 24(7): 278-288, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28620075

RESUMO

Using a hippocampus-dependent contextual threat learning and memory task, we report widespread, coordinated DNA methylation changes in CA1 hippocampus of Sprague-Dawley rats specific to threat learning at genes involved in synaptic transmission. Experience-dependent alternations in gene expression and DNA methylation were observed as early as 1 h following memory acquisition and became more pronounced after 24 h. Gene ontology analysis revealed significant enrichment of functional categories related to synaptic transmission in genes that were hypomethylated at 24 h following threat learning. Integration of these data sets with previously characterized epigenetic and transcriptional changes in brain disease states suggested significant overlap between genes regulated by memory formation and genes altered in memory-related neurological and neuropsychiatric diseases. These findings provide a comprehensive resource to aid in the identification of memory-relevant therapeutic targets. Our results shed new light on the gene expression and DNA methylation changes involved in memory formation, confirming that these processes are dynamic and experience-dependent. Finally, this work provides a roadmap for future studies to identify linkage of memory-associated genes to altered disease states.


Assuntos
Condicionamento Clássico/fisiologia , Epigenômica/métodos , Regulação da Expressão Gênica/fisiologia , Hipocampo/metabolismo , Memória/fisiologia , Animais , Ilhas de CpG/fisiologia , Metilação de DNA/fisiologia , Ontologia Genética , Masculino , Aprendizagem em Labirinto , Modelos Moleculares , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Transmissão Sináptica/genética , Fatores de Tempo
11.
J Neurosci ; 36(4): 1324-35, 2016 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-26818519

RESUMO

Aberrant gene expression within the hippocampus has recently been implicated in the pathogenesis of obesity-induced memory impairment. Whether a dysregulation of epigenetic modifications mediates this disruption in gene transcription has yet to be established. Here we report evidence of obesity-induced alterations in DNA methylation of memory-associated genes, including Sirtuin 1 (Sirt1), within the hippocampus, and thus offer a novel mechanism by which SIRT1 expression within the hippocampus is suppressed during obesity. Forebrain neuron-specific Sirt1 knock-out closely recapitulated the memory deficits exhibited by obese mice, consistent with the hypothesis that the high-fat diet-mediated reduction of hippocampal SIRT1 could be responsible for obesity-linked memory impairment. Obese mice fed a diet supplemented with the SIRT1-activating molecule resveratrol exhibited increased hippocampal SIRT1 activity and preserved hippocampus-dependent memory, further strengthening this conclusion. Thus, our findings suggest that the memory-impairing effects of diet-induced obesity may potentially be mediated by neuroepigenetic dysregulation of SIRT1 within the hippocampus. SIGNIFICANCE STATEMENT: Previous studies have implicated transcriptional dysregulation within the hippocampus as being a relevant pathological concomitant of obesity-induced memory impairment, yet a deeper understanding of the basis for, and etiological significance of, transcriptional dysregulation in this context is lacking. Here we present the first evidence of epigenetic dysregulation (i.e., altered DNA methylation and hydroxymethylation) of memory-related genes, including Sirt1, within the hippocampus of obese mice. Furthermore, experiments using transgenic and pharmacological approaches strongly implicate reduced hippocampal SIRT1 as being a principal pathogenic mediator of obesity-induced memory impairment. This paper offers a novel working model that may serve as a conceptual basis for the development of therapeutic interventions for obesity-induced memory impairment.


Assuntos
Hipocampo/metabolismo , Transtornos da Memória/etiologia , Neurônios/metabolismo , Obesidade/complicações , Obesidade/fisiopatologia , Sirtuína 1/metabolismo , Animais , Antioxidantes/farmacologia , Metilação de DNA/efeitos dos fármacos , Metilação de DNA/genética , Dieta Hiperlipídica/efeitos adversos , Suplementos Nutricionais , Modelos Animais de Doenças , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/genética , Comportamento Exploratório/efeitos dos fármacos , Comportamento Exploratório/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Insulina/metabolismo , Masculino , Transtornos da Memória/dietoterapia , Transtornos da Memória/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Obesidade/induzido quimicamente , Prosencéfalo/patologia , Reconhecimento Psicológico/efeitos dos fármacos , Reconhecimento Psicológico/fisiologia , Resveratrol , Sirtuína 1/genética , Memória Espacial/efeitos dos fármacos , Memória Espacial/efeitos da radiação , Estilbenos/farmacologia , Fatores de Tempo
12.
J Neurochem ; 137(3): 312-30, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26849493

RESUMO

Hebbian plasticity, including long-term potentiation and long-term depression, has long been regarded as important for local circuit refinement in the context of memory formation and stabilization. However, circuit development and stabilization additionally relies on non-Hebbian, homeostatic, forms of plasticity such as synaptic scaling. Synaptic scaling is induced by chronic increases or decreases in neuronal activity. Synaptic scaling is associated with cell-wide adjustments in postsynaptic receptor density, and can occur in a multiplicative manner resulting in preservation of relative synaptic strengths across the entire neuron's population of synapses. Both active DNA methylation and demethylation have been validated as crucial regulators of gene transcription during learning, and synaptic scaling is known to be transcriptionally dependent. However, it has been unclear whether homeostatic forms of plasticity such as synaptic scaling are regulated via epigenetic mechanisms. This review describes exciting recent work that has demonstrated a role for active changes in neuronal DNA methylation and demethylation as a controller of synaptic scaling and glutamate receptor trafficking. These findings bring together three major categories of memory-associated mechanisms that were previously largely considered separately: DNA methylation, homeostatic plasticity, and glutamate receptor trafficking. This review describes exciting recent work that has demonstrated a role for active changes in neuronal DNA methylation and demethylation as a controller of synaptic scaling and glutamate receptor trafficking. These findings bring together three major categories of memory-associated mechanisms that were previously considered separately: glutamate receptor trafficking, DNA methylation, and homeostatic plasticity.


Assuntos
Metilação de DNA/fisiologia , Receptores de Glutamato/fisiologia , Sinapses/fisiologia , Animais , Humanos , Memória/fisiologia , Plasticidade Neuronal/fisiologia , Receptores de Glutamato/metabolismo
13.
J Neurochem ; 139 Suppl 2: 179-199, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-26875778

RESUMO

This brief review summarizes 60 years of conceptual advances that have demonstrated a role for active changes in neuronal connectivity as a controller of behavior and behavioral change. Seminal studies in the first phase of the six-decade span of this review firmly established the cellular basis of behavior - a concept that we take for granted now, but which was an open question at the time. Hebbian plasticity, including long-term potentiation and long-term depression, was then discovered as being important for local circuit refinement in the context of memory formation and behavioral change and stabilization in the mammalian central nervous system. Direct demonstration of plasticity of neuronal circuit function in vivo, for example, hippocampal neurons forming place cell firing patterns, extended this concept. However, additional neurophysiologic and computational studies demonstrated that circuit development and stabilization additionally relies on non-Hebbian, homoeostatic, forms of plasticity, such as synaptic scaling and control of membrane intrinsic properties. Activity-dependent neurodevelopment was found to be associated with cell-wide adjustments in post-synaptic receptor density, and found to occur in conjunction with synaptic pruning. Pioneering cellular neurophysiologic studies demonstrated the critical roles of transmembrane signal transduction, NMDA receptor regulation, regulation of neural membrane biophysical properties, and back-propagating action potential in critical time-dependent coincidence detection in behavior-modifying circuits. Concerning the molecular mechanisms underlying these processes, regulation of gene transcription was found to serve as a bridge between experience and behavioral change, closing the 'nature versus nurture' divide. Both active DNA (de)methylation and regulation of chromatin structure have been validated as crucial regulators of gene transcription during learning. The discovery of protein synthesis dependence on the acquisition of behavioral change was an influential discovery in the neurochemistry of behavioral modification. Higher order cognitive functions such as decision making and spatial and language learning were also discovered to hinge on neural plasticity mechanisms. The role of disruption of these processes in intellectual disabilities, memory disorders, and drug addiction has recently been clarified based on modern genetic techniques, including in the human. The area of neural plasticity and behavior has seen tremendous advances over the last six decades, with many of those advances being specifically in the neurochemistry domain. This review provides an overview of the progress in the area of neuroplasticity and behavior over the life-span of the Journal of Neurochemistry. To organize the broad literature base, the review collates progress into fifteen broad categories identified as 'conceptual advances', as viewed by the author. The fifteen areas are delineated in the figure above. This article is part of the 60th Anniversary special issue.


Assuntos
Comportamento/fisiologia , Encéfalo/fisiologia , Aprendizagem/fisiologia , Plasticidade Neuronal/fisiologia , Animais , Humanos , Potenciação de Longa Duração/fisiologia , Transtornos da Memória/metabolismo , Transtornos da Memória/psicologia
14.
Learn Mem ; 20(2): 61-74, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23322554

RESUMO

Understanding the cellular and molecular mechanisms underlying the formation and maintenance of memories is a central goal of the neuroscience community. It is well regarded that an organism's ability to lastingly adapt its behavior in response to a transient environmental stimulus relies on the central nervous system's capability for structural and functional plasticity. This plasticity is dependent on a well-regulated program of neurotransmitter release, post-synaptic receptor activation, intracellular signaling cascades, gene transcription, and subsequent protein synthesis. In the last decade, epigenetic markers like DNA methylation and post-translational modifications of histone tails have emerged as important regulators of the memory process. Their ability to regulate gene transcription dynamically in response to neuronal activation supports the consolidation of long-term memory. Furthermore, the persistent and self-propagating nature of these mechanisms, particularly DNA methylation, suggests a molecular mechanism for memory maintenance. In this review, we will examine the evidence that supports a role of epigenetic mechanisms in learning and memory. In doing so, we hope to emphasize (1) the widespread involvement of these mechanisms across different behavioral paradigms and distinct brain regions, (2) the temporal and genetic specificity of these mechanisms in response to upstream signaling cascades, and (3) the functional outcome these mechanisms may have on structural and functional plasticity. Finally, we consider the future directions of neuroepigenetic research as it relates to neuronal storage of information.


Assuntos
Encéfalo/fisiologia , Epigênese Genética , Epigenômica , Memória/fisiologia , Animais , Metilação de DNA/genética , Humanos , Transdução de Sinais/genética , Fatores de Tempo
15.
J Neurosci ; 32(48): 17059-66, 2012 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-23197699

RESUMO

Dynamic epigenetic mechanisms including histone and DNA modifications regulate animal behavior and memory. While numerous enzymes regulating these mechanisms have been linked to memory formation, the regulation of active DNA demethylation (i.e., cytosine-5 demethylation) has only recently been investigated. New discoveries aim toward the Growth arrest and DNA damage-inducible 45 (Gadd45) family, particularly Gadd45b, in activity-dependent demethylation in the adult CNS. This study found memory-associated expression of gadd45b in the hippocampus and characterized the behavioral phenotype of gadd45b(-/-) mice. Results indicate normal baseline behaviors and initial learning but enhanced persisting memory in mutants in tasks of motor performance, aversive conditioning and spatial navigation. Furthermore, we showed facilitation of hippocampal long-term potentiation in mutants. These results implicate Gadd45b as a learning-induced gene and a regulator of memory formation and are consistent with its potential role in active DNA demethylation in memory.


Assuntos
Tonsila do Cerebelo/fisiologia , Antígenos de Diferenciação/genética , Metilação de DNA/genética , Hipocampo/fisiologia , Potenciação de Longa Duração/genética , Memória de Longo Prazo/fisiologia , Sinapses/genética , Tonsila do Cerebelo/citologia , Animais , Antígenos de Diferenciação/metabolismo , Comportamento Animal/fisiologia , Células Cultivadas , Regulação da Expressão Gênica , Hipocampo/citologia , Masculino , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Knockout , Neurônios/citologia , Neurônios/fisiologia , Comportamento Espacial/fisiologia , Sinapses/metabolismo
16.
J Neurochem ; 124(1): 109-22, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23113835

RESUMO

Protease-activated receptor-1 (PAR1) is an unusual G-protein coupled receptor (GPCR) that is activated through proteolytic cleavage by extracellular serine proteases. Although previous work has shown that inhibiting PAR1 activation is neuroprotective in models of ischemia, traumatic injury, and neurotoxicity, surprisingly little is known about PAR1's contribution to normal brain function. Here, we used PAR1-/- mice to investigate the contribution of PAR1 function to memory formation and synaptic function. We demonstrate that PAR1-/- mice have deficits in hippocampus-dependent memory. We also show that while PAR1-/- mice have normal baseline synaptic transmission at Schaffer collateral-CA1 synapses, they exhibit severe deficits in N-methyl-d-aspartate receptor (NMDAR)-dependent long-term potentiation (LTP). Mounting evidence indicates that activation of PAR1 leads to potentiation of NMDAR-mediated responses in CA1 pyramidal cells. Taken together, this evidence and our data suggest an important role for PAR1 function in NMDAR-dependent processes subserving memory formation and synaptic plasticity.


Assuntos
Hipocampo/citologia , Potenciação de Longa Duração/genética , Memória/fisiologia , Receptor PAR-1/metabolismo , Sinapses/genética , 6-Ciano-7-nitroquinoxalina-2,3-diona/farmacologia , Potenciais de Ação/efeitos dos fármacos , Potenciais de Ação/genética , Animais , Biofísica , Condicionamento Psicológico/fisiologia , Estimulação Elétrica , Antagonistas de Aminoácidos Excitatórios/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Potenciais Pós-Sinápticos Excitadores/genética , Medo/fisiologia , Técnicas In Vitro , Potenciação de Longa Duração/efeitos dos fármacos , Transtornos da Memória/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/metabolismo , Receptor PAR-1/deficiência , Sinapses/efeitos dos fármacos , Sinapses/fisiologia
17.
Adv Exp Med Biol ; 793: 81-119, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24104475

RESUMO

The growth arrest and DNA damage-inducible (Gadd)45 proteins have been associated with numerous cellular mechanisms including cell-cycle control, DNA damage sensation and repair, genotoxic stress, neoplasia, and molecular epigenetics. The genes were originally identified in in vitro screens of irradiation- and interleukin-induced transcription and have since been implicated in a host of normal and aberrant central nervous system processes. These include early and postnatal development, injury, cancer, memory, aging, and neurodegenerative and psychiatric disease states. The proteins act through a variety of molecular signaling cascades including the MAPK cascade, cell-cycle control mechanisms, histone regulation, and epigenetic DNA demethylation. In this review, we provide a comprehensive discussion of the literature implicating each of the three members of the Gadd45 family in these processes.


Assuntos
Antígenos de Diferenciação/genética , Proteínas de Ciclo Celular/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Sistema Nervoso/metabolismo , Proteínas Nucleares/genética , Transtornos Psicóticos/metabolismo , Traumatismos do Sistema Nervoso/metabolismo , Animais , Antígenos de Diferenciação/metabolismo , Apoptose , Pontos de Checagem do Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Metilação de DNA , Epigênese Genética , Histonas/genética , Histonas/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Sistema Nervoso/fisiopatologia , Neurogênese/genética , Proteínas Nucleares/metabolismo , Transtornos Psicóticos/genética , Transtornos Psicóticos/fisiopatologia , Transdução de Sinais , Traumatismos do Sistema Nervoso/genética , Traumatismos do Sistema Nervoso/fisiopatologia
18.
Proc Natl Acad Sci U S A ; 107(39): 16994-8, 2010 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-20837545

RESUMO

Learning and memory have been closely linked to strengthening of synaptic connections between neurons (i.e., synaptic plasticity) within the dentate gyrus (DG)-CA3-CA1 trisynaptic circuit of the hippocampus. Conspicuously absent from this circuit is area CA2, an intervening hippocampal region that is poorly understood. Schaffer collateral synapses on CA2 neurons are distinct from those on other hippocampal neurons in that they exhibit a perplexing lack of synaptic long-term potentiation (LTP). Here we demonstrate that the signaling protein RGS14 is highly enriched in CA2 pyramidal neurons and plays a role in suppression of both synaptic plasticity at these synapses and hippocampal-based learning and memory. RGS14 is a scaffolding protein that integrates G protein and H-Ras/ERK/MAP kinase signaling pathways, thereby making it well positioned to suppress plasticity in CA2 neurons. Supporting this idea, deletion of exons 2-7 of the RGS14 gene yields mice that lack RGS14 (RGS14-KO) and now express robust LTP at glutamatergic synapses in CA2 neurons with no impact on synaptic plasticity in CA1 neurons. Treatment of RGS14-deficient CA2 neurons with a specific MEK inhibitor blocked this LTP, suggesting a role for ERK/MAP kinase signaling pathways in this process. When tested behaviorally, RGS14-KO mice exhibited marked enhancement in spatial learning and in object recognition memory compared with their wild-type littermates, but showed no differences in their performance on tests of nonhippocampal-dependent behaviors. These results demonstrate that RGS14 is a key regulator of signaling pathways linking synaptic plasticity in CA2 pyramidal neurons to hippocampal-based learning and memory but distinct from the canonical DG-CA3-CA1 circuit.


Assuntos
Região CA2 Hipocampal/fisiologia , Aprendizagem , Plasticidade Neuronal , Células Piramidais/fisiologia , Proteínas RGS/metabolismo , Sinapses/fisiologia , Animais , Região CA2 Hipocampal/metabolismo , Memória , Camundongos , Camundongos Knockout , Células Piramidais/metabolismo , Sinapses/metabolismo
19.
Neurobiol Learn Mem ; 98(1): 25-32, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22542746

RESUMO

Mounting evidence has established that diet-induced obesity (DIO) is associated with deficits in hippocampus-dependent memory. The bulk of research studies dealing with this topic have utilized rats fed a high-fat diet as an experimental model. To date, there has been a paucity of research studies that have established whether the memory deficits exhibited in DIO rats can be recapitulated in mice. Moreover, the majority of experiments that have evaluated memory performance in rodent models of DIO have utilized memory tests that are essentially aversive in nature (i.e., Morris water maze). The current study sought to fill an empirical void by determining if mice maintained on a high-fat diet exhibit deficits in two non-aversive memory paradigms: novel object recognition (NOR) and object location memory (OLM). Here we report that mice fed a high-fat diet over 23 weeks exhibit intact NOR, albeit a marked impairment in hippocampus-dependent OLM. We also determined the existence of corresponding aberrations in gene expression within the hippocampus of DIO mice. DIO mice exhibited significant reductions in both SIRT1 and PP1 mRNA within the hippocampus. Our data suggest that mice maintained on a high-fat diet present with impaired hippocampus-dependent spatial memory and a corresponding alteration in the expression of genes that have been implicated in memory consolidation.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Hipocampo/metabolismo , Aprendizagem em Labirinto/fisiologia , Transtornos da Memória/etiologia , Sirtuína 1/genética , Animais , Medo/fisiologia , Hipocampo/fisiopatologia , Masculino , Memória/fisiologia , Transtornos da Memória/genética , Transtornos da Memória/metabolismo , Transtornos da Memória/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/genética , Obesidade/metabolismo , Sirtuína 1/metabolismo
20.
Mol Cell Neurosci ; 46(1): 45-54, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20708080

RESUMO

Regulation of forebrain cellular structure and function by small GTPase pathways is crucial for normal and pathological brain development and function. Kalirin is a brain-specific activator of Rho-like small GTPases implicated in neuropsychiatric disorders. We have recently demonstrated key roles for kalirin in cortical synaptic transmission, dendrite branching, spine density, and working memory. However, little is known about the impact of the complete absence of kalirin on the hippocampus in mice. We thus investigated hippocampal function, structure, and associated behavioral phenotypes in KALRN knockout (KO) mice we have recently generated. Here we show that KALRN KO mice had modest impairments in hippocampal LTP, but normal hippocampal synaptic transmission. In these mice, both context and cue-dependent fear conditioning were impaired. Spine density and dendrite morphology in hippocampal pyramidal neurons were not significantly affected in the KALRN KO mice, but small alterations in the gross morphology of the hippocampus were detected. These data suggest that hippocampal structure and function are more resilient to the complete loss of kalirin, and reveal impairments in fear learning. These studies allow the comparison of the phenotypes of different kalirin mutant mice and shed light on the brain region-specific functions of small GTPase signaling.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/metabolismo , Hipocampo/fisiologia , Animais , Comportamento Animal/fisiologia , Células Cultivadas , Medo/fisiologia , Fatores de Troca do Nucleotídeo Guanina/genética , Hipocampo/citologia , Humanos , Potenciação de Longa Duração/fisiologia , Masculino , Camundongos , Camundongos Knockout , Plasticidade Neuronal/fisiologia , Fenótipo , Transmissão Sináptica/fisiologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa