Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 170(6): 1209-1223.e20, 2017 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-28823556

RESUMO

Fragile X syndrome (FXS) is a leading genetic cause of intellectual disability and autism. FXS results from the loss of function of fragile X mental retardation protein (FMRP), which represses translation of target transcripts. Most of the well-characterized target transcripts of FMRP are synaptic proteins, yet targeting these proteins has not provided effective treatments. We examined a group of FMRP targets that encode transcriptional regulators, particularly chromatin-associated proteins. Loss of FMRP in mice results in widespread changes in chromatin regulation and aberrant gene expression. To determine if targeting epigenetic factors could reverse phenotypes associated with the disorder, we focused on Brd4, a BET protein and chromatin reader targeted by FMRP. Inhibition of Brd4 function alleviated many of the phenotypes associated with FXS. We conclude that loss of FMRP results in significant epigenetic misregulation and that targeting transcription via epigenetic regulators like Brd4 may provide new treatments for FXS.


Assuntos
Azepinas/farmacologia , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Síndrome do Cromossomo X Frágil/tratamento farmacológico , Síndrome do Cromossomo X Frágil/metabolismo , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/metabolismo , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/metabolismo , Triazóis/farmacologia , Animais , Células Cultivadas , Epigênese Genética , Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Histonas/metabolismo , Camundongos , Camundongos Knockout , Naftiridinas/farmacologia , Neurônios/metabolismo , Fenazinas , Transcrição Gênica
2.
Mol Psychiatry ; 25(12): 3322-3336, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-31363163

RESUMO

The cognitive mechanisms underlying attention-deficit hyperactivity disorder (ADHD), a highly heritable disorder with an array of candidate genes and unclear genetic architecture, remain poorly understood. We previously demonstrated that mice overexpressing CK1δ (CK1δ OE) in the forebrain show hyperactivity and ADHD-like pharmacological responses to D-amphetamine. Here, we demonstrate that CK1δ OE mice exhibit impaired visual attention and a lack of D-amphetamine-induced place preference, indicating a disruption of the dopamine-dependent reward pathway. We also demonstrate the presence of abnormalities in the frontostriatal circuitry, differences in synaptic ultra-structures by electron microscopy, as well as electrophysiological perturbations of both glutamatergic and GABAergic transmission, as observed by altered frequency and amplitude of mEPSCs and mIPSCs. Furthermore, gene expression profiling by next-generation sequencing alone, or in combination with bacTRAP technology to study specifically Drd1a versus Drd2 medium spiny neurons, revealed that developmental CK1δ OE alters transcriptional homeostasis in the striatum, including specific alterations in Drd1a versus Drd2 neurons. These results led us to perform a fine molecular characterization of targeted gene networks and pathway analysis. Importantly, a large fraction of 92 genes identified by GWAS studies as associated with ADHD in humans are significantly altered in our mouse model. The multiple abnormalities described here might be responsible for synaptic alterations and lead to complex behavioral abnormalities. Collectively, CK1δ OE mice share characteristics typically associated with ADHD and should represent a valuable model to investigate the disease in vivo.


Assuntos
Transtorno do Deficit de Atenção com Hiperatividade , Caseína Quinase Idelta/genética , Animais , Transtorno do Deficit de Atenção com Hiperatividade/genética , Corpo Estriado , Dopamina , Camundongos , Neurônios , Receptores de Dopamina D2/genética
3.
Mol Psychiatry ; 25(7): 1364-1381, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32439846

RESUMO

Selective serotonin reuptake inhibitors (SSRIs) are the most widely prescribed drugs for mood disorders. While the mechanism of SSRI action is still unknown, SSRIs are thought to exert therapeutic effects by elevating extracellular serotonin levels in the brain, and remodel the structural and functional alterations dysregulated during depression. To determine their precise mode of action, we tested whether such neuroadaptive processes are modulated by regulation of specific gene expression programs. Here we identify a transcriptional program regulated by activator protein-1 (AP-1) complex, formed by c-Fos and c-Jun that is selectively activated prior to the onset of the chronic SSRI response. The AP-1 transcriptional program modulates the expression of key neuronal remodeling genes, including S100a10 (p11), linking neuronal plasticity to the antidepressant response. We find that AP-1 function is required for the antidepressant effect in vivo. Furthermore, we demonstrate how neurochemical pathways of BDNF and FGF2, through the MAPK, PI3K, and JNK cascades, regulate AP-1 function to mediate the beneficial effects of the antidepressant response. Here we put forth a sequential molecular network to track the antidepressant response and provide a new avenue that could be used to accelerate or potentiate antidepressant responses by triggering neuroplasticity.


Assuntos
Anexina A2/metabolismo , Antidepressivos/farmacologia , Proteínas S100/metabolismo , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Fator de Transcrição AP-1/metabolismo , Animais , Anexina A2/genética , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Linhagem Celular , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Camundongos , Plasticidade Neuronal/efeitos dos fármacos , Plasticidade Neuronal/genética , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-fos/metabolismo , RNA Mensageiro/genética , Ratos , Proteínas S100/genética , Serotonina/metabolismo
4.
Mol Psychiatry ; 25(5): 1035-1049, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-30760886

RESUMO

Genetic polymorphisms of the L-type voltage-gated calcium channel (VGCC) are associated with psychiatric disorders including major depressive disorder. Alterations of S100A10 (p11) level are also implicated in the etiology of major depressive disorder. However, the existence of an endogenous regulator in the brain regulating p11, L-type VGCC, and depressive behavior has not been known. Here we report that Ahnak, whose function in the brain has been obscure, stabilizes p11 and Anxa2 proteins in the hippocampus and prefrontal cortex in the rodent brain. Protein levels of Ahnak, p11, and Anxa2 are highly and positively correlated in the brain. Together these data suggest the existence of an Ahnak/p11/Anxa2 protein complex. Ahnak is expressed in p11-positive as well as p11-negative neurons. Ahnak, through its N-terminal region, scaffolds the L-type pore-forming α1 subunit and, through its C-terminal region, scaffolds the ß subunit of VGCC and the p11/Anxa2 complex. Cell surface expression of the α1 subunits and L-type calcium current are significantly reduced in primary cultures of Ahnak knockout (KO) neurons compared to wild-type controls. A decrease in the L-type calcium influx is observed in both glutamatergic neurons and parvalbumin (PV) GABAergic interneurons of Ahnak KO mice. Constitutive Ahnak KO mice or forebrain glutamatergic neuron-selective Ahnak KO mice display a depression-like behavioral phenotype similar to that of constitutive p11 KO mice. In contrast, PV interneuron-selective Ahnak KO mice display an antidepressant-like behavioral phenotype. Our results demonstrate L-type VGCC as an effector of the Ahnak/p11/Anxa2 complex, revealing a novel molecular connection involved in the control of depressive behavior.


Assuntos
Anexina A2/metabolismo , Encéfalo/metabolismo , Canais de Cálcio Tipo L/metabolismo , Transtorno Depressivo Maior/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas S100/metabolismo , Animais , Encéfalo/patologia , Encéfalo/fisiopatologia , Depressão/metabolismo , Transtorno Depressivo Maior/fisiopatologia , Modelos Animais de Doenças , Feminino , Hipocampo/metabolismo , Hipocampo/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/fisiopatologia
5.
Proc Natl Acad Sci U S A ; 113(19): 5418-23, 2016 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-27114526

RESUMO

Cellular trafficking and recycling machineries belonging to late secretory compartments have been associated with increased Alzheimer's disease (AD) risk. We have shown that coat protein complex I (COPI)-dependent trafficking, an early step in Golgi-to-endoplasmic reticulum retrograde transport, affects amyloid precursor protein subcellular localization, cell-surface expression, as well as its metabolism. We present here a set of experiments demonstrating that, by targeting subunit δ-COP function, the moderation of the COPI-dependent trafficking in vivo leads to a significant decrease in amyloid plaques in the cortex and hippocampus of neurological 17 mice crossed with the 2xTg AD mouse model. Remarkably, an improvement of the memory impairments was also observed. Importantly, human genetic association studies of different AD cohorts led to the identification of 12 SNPs and 24 mutations located in COPI genes linked to an increased AD risk. These findings further demonstrate in vivo the importance of early trafficking steps in AD pathogenesis and open new clinical perspectives.


Assuntos
Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Complexo I de Proteína do Envoltório/metabolismo , Progressão da Doença , Placa Amiloide/metabolismo , Frações Subcelulares/metabolismo , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transporte Proteico/fisiologia
6.
PLoS Biol ; 12(8): e1001923, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25093460

RESUMO

STEP (STriatal-Enriched protein tyrosine Phosphatase) is a neuron-specific phosphatase that regulates N-methyl-D-aspartate receptor (NMDAR) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) trafficking, as well as ERK1/2, p38, Fyn, and Pyk2 activity. STEP is overactive in several neuropsychiatric and neurodegenerative disorders, including Alzheimer's disease (AD). The increase in STEP activity likely disrupts synaptic function and contributes to the cognitive deficits in AD. AD mice lacking STEP have restored levels of glutamate receptors on synaptosomal membranes and improved cognitive function, results that suggest STEP as a novel therapeutic target for AD. Here we describe the first large-scale effort to identify and characterize small-molecule STEP inhibitors. We identified the benzopentathiepin 8-(trifluoromethyl)-1,2,3,4,5-benzopentathiepin-6-amine hydrochloride (known as TC-2153) as an inhibitor of STEP with an IC50 of 24.6 nM. TC-2153 represents a novel class of PTP inhibitors based upon a cyclic polysulfide pharmacophore that forms a reversible covalent bond with the catalytic cysteine in STEP. In cell-based secondary assays, TC-2153 increased tyrosine phosphorylation of STEP substrates ERK1/2, Pyk2, and GluN2B, and exhibited no toxicity in cortical cultures. Validation and specificity experiments performed in wild-type (WT) and STEP knockout (KO) cortical cells and in vivo in WT and STEP KO mice suggest specificity of inhibitors towards STEP compared to highly homologous tyrosine phosphatases. Furthermore, TC-2153 improved cognitive function in several cognitive tasks in 6- and 12-mo-old triple transgenic AD (3xTg-AD) mice, with no change in beta amyloid and phospho-tau levels.


Assuntos
Doença de Alzheimer/tratamento farmacológico , Doença de Alzheimer/enzimologia , Transtornos Cognitivos/tratamento farmacológico , Transtornos Cognitivos/enzimologia , Inibidores Enzimáticos/uso terapêutico , Proteínas Tirosina Fosfatases não Receptoras/antagonistas & inibidores , Doença de Alzheimer/complicações , Doença de Alzheimer/patologia , Sequência de Aminoácidos , Animais , Benzotiepinas/farmacologia , Benzotiepinas/uso terapêutico , Domínio Catalítico , Morte Celular/efeitos dos fármacos , Córtex Cerebral/patologia , Transtornos Cognitivos/complicações , Transtornos Cognitivos/patologia , Cisteína/metabolismo , Modelos Animais de Doenças , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Ensaios de Triagem em Larga Escala , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Neurônios/efeitos dos fármacos , Neurônios/patologia , Fosforilação/efeitos dos fármacos , Fosfotirosina/metabolismo , Proteínas Tirosina Fosfatases não Receptoras/química , Proteínas Tirosina Fosfatases não Receptoras/metabolismo , Especificidade por Substrato/efeitos dos fármacos
7.
Behav Pharmacol ; 24(1): 1-9, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23268986

RESUMO

Norepinephrine and epinephrine signaling is thought to facilitate cognitive processes related to emotional events and heightened arousal; however, the specific role of epinephrine in these processes is less known. To investigate the selective impact of epinephrine on arousal and fear-related memory retrieval, mice unable to synthesize epinephrine (phenylethanolamine N-methyltransferase knockout, PNMT-KO) were tested for contextual and cued-fear conditioning. To assess the role of epinephrine in other cognitive and arousal-based behaviors these mice were also tested for acoustic startle, prepulse inhibition, novel object recognition, and open-field activity. Our results show that compared with wild-type mice, PNMT-KO mice showed reduced contextual fear but normal cued fear. Mice exhibited normal memory performance in the short-term version of the novel object recognition task, suggesting that PNMT mice exhibit more selective memory effects on highly emotional and/or long-term memories. Similarly, open-field activity was unaffected by epinephrine deficiency, suggesting that differences in freezing are not related to changes in overall anxiety or exploratory drive. Startle reactivity to acoustic pulses was reduced in PNMT-KO mice, whereas prepulse inhibition was increased. These findings provide further evidence for a selective role of epinephrine in contextual-fear learning and support its potential role in acoustic startle.


Assuntos
Neurônios Adrenérgicos/metabolismo , Epinefrina/fisiologia , Medo , Proteínas do Tecido Nervoso/metabolismo , Feniletanolamina N-Metiltransferase/metabolismo , Reflexo de Sobressalto , Transmissão Sináptica , Animais , Ansiedade/metabolismo , Comportamento Animal , Condicionamento Clássico , Sinais (Psicologia) , Epinefrina/deficiência , Comportamento Exploratório , Masculino , Memória de Curto Prazo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Feniletanolamina N-Metiltransferase/genética , Distribuição Aleatória , Reconhecimento Psicológico
8.
Int J Neuropsychopharmacol ; 14(9): 1179-94, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21205416

RESUMO

Corticotropin-releasing factor (CRF) and norepinephrine (NE) levels are altered in post-traumatic stress disorder and may be related to symptoms of hyperarousal, including exaggerated startle, in these patients. In animals, activation of both systems modulates anxiety behaviours including startle plasticity; however, it is unknown if they exert their actions orthogonally or dependently. We tested the hypothesis that NE receptor activation is required for CRF effects on startle and that CRF1 receptor activation is required for NE effects on startle. The study examined the effects of: (1) α2 agonist clonidine (0.18 mg/kg i.p.), α1 antagonist prazosin (0.8 mg/kg), and ß1/2 antagonist propranolol (0.8, 8.0 mg/kg) pretreatment on ovine-CRF (oCRF)- (0.6 nmol) induced increases in startle reactivity and disruption of prepulse inhibition (PPI); (2) α2 antagonist atipamezole (1-30 mg/kg) and α1 agonist cirazoline (0.025-1.0 mg/kg) treatment on startle; (3) CRF1 antagonist (antalarmin, 14 mg/kg) pretreatment on atipamezole- (10.0 mg/kg) induced increases in startle. oCRF robustly increased startle and reduced PPI. Pretreatment with clonidine or prazosin, but not propranolol, blocked oCRF-induced increases in startle but had no effect on oCRF-induced disruptions in PPI. Atipamezole treatment increased startle, which was partially attenuated by CRF1 antagonist pretreatment. Cirazoline treatment did not increase startle. These findings suggest that CRF modulation of startle, but not PPI, requires activation of α1 adrenergic receptors, while CRF1 activation also contributes to NE modulation of startle. These data support a bi-directional model of CRF-NE modulation of stress responses and suggest that both systems must be activated to induce stress effects on startle reactivity.


Assuntos
Hormônio Liberador da Corticotropina/metabolismo , Norepinefrina/fisiologia , Receptores Adrenérgicos/metabolismo , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Reflexo de Sobressalto , Transmissão Sináptica , Antagonistas de Receptores Adrenérgicos alfa 1/farmacologia , Agonistas de Receptores Adrenérgicos alfa 2/farmacologia , Antagonistas de Receptores Adrenérgicos alfa 2/farmacologia , Animais , Comportamento Animal/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Hormônio Liberador da Corticotropina/administração & dosagem , Infusões Intraventriculares , Locus Cerúleo/efeitos dos fármacos , Locus Cerúleo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/agonistas , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/metabolismo , Inibição Neural/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Receptores Adrenérgicos/química , Receptores de Hormônio Liberador da Corticotropina/agonistas , Receptores de Hormônio Liberador da Corticotropina/antagonistas & inibidores , Reflexo de Sobressalto/efeitos dos fármacos , Carneiro Doméstico , Transtornos de Estresse Pós-Traumáticos/metabolismo , Transmissão Sináptica/efeitos dos fármacos
9.
Behav Brain Funct ; 7: 33, 2011 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-21838910

RESUMO

BACKGROUND: Executive dysfunction may play a major role in cognitive decline with aging because frontal lobe structures are particularly vulnerable to advancing age. Lesion studies in rats and mice have suggested that intradimensional shifts (IDSs), extradimensional shifts (EDSs), and reversal learning are mediated by the anterior cingulate cortex, the medial prefrontal cortex, and the orbitofrontal cortex, respectively. We hypothesized that the latent structure of cognitive performance would reflect functional localization in the brain and would be altered by aging. METHODS: Young (4 months, n = 16) and aged (23 months, n = 18) C57BL/6N mice performed an attentional set-shifting task (ASST) that evaluates simple discrimination (SD), compound discrimination (CD), IDS, EDS, and reversal learning. The performance data were subjected to an exploratory factor analysis to extract the latent structures of ASST performance in young and aged mice. RESULTS: The factor analysis extracted two- and three-factor models. In the two-factor model, the factor associated with SD and CD was clearly separated from the factor associated with the rest of the ASST stages in the young mice only. In the three-factor model, the SD and CD loaded on distinct factors. The three-factor model also showed a separation of factors associated with IDS, EDS, and CD reversal. However, the other reversal learning variables, ID reversal and ED reversal, had somewhat inconsistent factor loadings. CONCLUSIONS: The separation of performance factors in aged mice was less clear than in young mice, which suggests that aged mice utilize neuronal networks more broadly for specific cognitive functions. The result that the factors associated with SD and CD were separated in the three-factor model may suggest that the introduction of an irrelevant or distracting dimension results in the use of a new/orthogonal strategy for better discrimination.


Assuntos
Envelhecimento/fisiologia , Envelhecimento/psicologia , Atenção/fisiologia , Desempenho Psicomotor/fisiologia , Fatores Etários , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL
10.
Psychiatry Res Neuroimaging ; 304: 111137, 2020 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-32731113

RESUMO

Increased corticotroping releasing factor (CRF) contributes to brain circuit abnormalities associated with stress-related disorders including posttraumatic stress disorder. However, the causal relationship between CRF hypersignaling and circuit abnormalities associated with stress disorders is unclear. We hypothesized that increased CRF exposure induces changes in limbic circuit morphology and functions. An inducible, forebrain-specific overexpression of CRF (CRFOE) transgenic mouse line was used to longitudinally investigate its chronic effects on behaviors and microstructural integrity of several brain regions. Behavioral and diffusion tensor imaging studies were performed before treatment, after 3-4 wks of treatment, and again 3 mo after treatment ended to assess recovery. CRFOE was associated with increased perseverative movements only after 3 wks of treatment, as well as reduced fractional anisotropy at 3 wks in the medial prefrontal cortex and increased fractional anisotropy in the ventral hippocampus at 3 mo compared to the control group. In the dorsal hippocampus, mean diffusivity was lower in CRFOE mice both during and after treatment ended. Our data suggest differential response and recovery patterns of cortical and hippocampal subregions in response to CRFOE. Overall these findings support a causal relationship between CRF hypersignaling and microstructural changes in brain regions relevant to stress disorders.


Assuntos
Hormônio Liberador da Corticotropina/metabolismo , Substância Cinzenta/diagnóstico por imagem , Prosencéfalo/diagnóstico por imagem , Prosencéfalo/metabolismo , Animais , Imagem de Tensor de Difusão , Hipocampo/diagnóstico por imagem , Hipocampo/patologia , Humanos , Masculino , Camundongos , Córtex Pré-Frontal/diagnóstico por imagem , Córtex Pré-Frontal/patologia
11.
J Neurosci ; 28(35): 8660-7, 2008 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-18753366

RESUMO

The extracellular signal-regulated kinase (ERK) pathway is critical for various forms of learning and memory, and is activated by the potent estrogen 17beta-estradiol (E(2)). Here, we asked whether E(2) modulates memory via ERK activation and putative membrane-bound estrogen receptors (ERs). Using ovariectomized mice, we first demonstrate that intraperitoneal injection of 0.2 mg/kg E(2) significantly increases dorsal hippocampal levels of phosphorylated ERK protein 1 h after injection. Second, we show that E(2) administered intraperitoneally (0.2 mg/kg) or via intrahippocampal infusion (5.0 microg/side) immediately after training in an object recognition task significantly enhances memory retention, and that the beneficial effect of intraperitoneal E(2) is blocked by dorsal hippocampal inhibition of ERK activation. Third, using bovine serum albumin-conjugated 17beta-estradiol (BSA-E(2)), we demonstrate that E(2) binding at membrane-bound ERs can increase dorsal hippocampal ERK activation and enhance object memory consolidation in an ERK-dependent manner. Fourth, we show that this effect is independent of nuclear ERs, but is dependent on the dorsal hippocampus. By demonstrating that E(2) enhances memory consolidation via dorsal hippocampal ERK activation, this study is the first to identify a specific molecular pathway by which E(2) modulates memory and to demonstrate a novel role for membrane-bound ERs in mediating E(2)-induced improvements in hippocampal memory consolidation.


Assuntos
Estradiol/farmacologia , Estrogênios/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Hipocampo/efeitos dos fármacos , Receptores de Estrogênio/fisiologia , Reconhecimento Psicológico/efeitos dos fármacos , Aminoacetonitrila/análogos & derivados , Aminoacetonitrila/farmacologia , Análise de Variância , Animais , Comportamento Animal/efeitos dos fármacos , Comportamento de Escolha/efeitos dos fármacos , Vias de Administração de Medicamentos , Inibidores Enzimáticos/farmacologia , Comportamento Exploratório/efeitos dos fármacos , Feminino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Muscimol/farmacologia , Ovariectomia/métodos , Fatores de Tempo
12.
Elife ; 82019 12 20.
Artigo em Inglês | MEDLINE | ID: mdl-31860442

RESUMO

Loss of the RNA binding protein FMRP causes Fragile X Syndrome (FXS), the most common cause of inherited intellectual disability, yet it is unknown how FMRP function varies across brain regions and cell types and how this contributes to disease pathophysiology. Here we use conditional tagging of FMRP and CLIP (FMRP cTag CLIP) to examine FMRP mRNA targets in hippocampal CA1 pyramidal neurons, a critical cell type for learning and memory relevant to FXS phenotypes. Integrating these data with analysis of ribosome-bound transcripts in these neurons revealed CA1-enriched binding of autism-relevant mRNAs, and CA1-specific regulation of transcripts encoding circadian proteins. This contrasted with different targets in cerebellar granule neurons, and was consistent with circadian defects in hippocampus-dependent memory in Fmr1 knockout mice. These findings demonstrate differential FMRP-dependent regulation of mRNAs across neuronal cell types that may contribute to phenotypes such as memory defects and sleep disturbance associated with FXS.


Assuntos
Transtorno Autístico/metabolismo , Região CA1 Hipocampal/metabolismo , Proteína do X Frágil da Deficiência Intelectual/genética , Síndrome do Cromossomo X Frágil/genética , Transtornos da Memória/genética , Células Piramidais/metabolismo , Animais , Transtorno Autístico/genética , Transtorno Autístico/fisiopatologia , Região CA1 Hipocampal/citologia , Cerebelo/citologia , Cerebelo/metabolismo , Relógios Circadianos/genética , Relógios Circadianos/fisiologia , Modelos Animais de Doenças , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Síndrome do Cromossomo X Frágil/metabolismo , Síndrome do Cromossomo X Frágil/fisiopatologia , Regulação da Expressão Gênica , Humanos , Transtornos da Memória/metabolismo , Transtornos da Memória/fisiopatologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/metabolismo
13.
Brain Res ; 1160: 91-101, 2007 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-17572392

RESUMO

The present study was designed to examine if 4 weeks of exposure to an enriched housing environment affects the ability of estrogen to facilitate object recognition in young and aged female mice. Object recognition was tested using a novel object recognition task. Ovariectomized young and aged female mice were maintained in standard or enriched housing for 4 weeks prior to and then throughout object recognition testing. Immediately after training, mice were injected intraperitoneally with vehicle or 0.2 mg/kg 17 beta-estradiol and then were re-tested 24 and 48 h later. Among young females, estradiol alone improved object recognition at both delays relative to chance, an effect not present in enriched females treated with estradiol. Enrichment alone had no significant effect on object recognition in young females at either delay. In contrast, enrichment alone in aged females significantly enhanced both 24- and 48-h object recognition relative to chance, an effect not present in mice treated with both enrichment and estradiol. Estradiol alone had no effect on object recognition in aged females at either delay. Together, these data indicate that estradiol and enrichment alone differentially affect object recognition in young and aged females. However, the fact that the combination of estradiol and enrichment treatments did not affect object recognition at either age suggests that co-administration of both treatments is less effective than the most effective single treatment at each age.


Assuntos
Envelhecimento/fisiologia , Meio Ambiente , Estrogênios/administração & dosagem , Reconhecimento Visual de Modelos/efeitos dos fármacos , Análise de Variância , Animais , Comportamento Animal/efeitos dos fármacos , Vias de Administração de Medicamentos , Sistemas de Liberação de Medicamentos , Feminino , Camundongos , Ovariectomia/métodos , Estimulação Luminosa/métodos , Tempo de Reação , Fatores de Tempo , Útero/efeitos dos fármacos
14.
Brain Res ; 1115(1): 135-47, 2006 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-16920082

RESUMO

The manner in which hormone therapy is given to postmenopausal women may significantly influence its ability to reduce age-associated memory loss. To test the hypothesis that a regimen that approximates the timing of estrogen surges in the natural cycle is more beneficial for memory than a regimen that provides continuous levels of estrogen, we examined the effects of continuous and intermittent estrogen regimens on spatial and object memory in aging female mice. Mice (18 months) were treated with 0.2 mg/kg 17beta-estradiol (E(2)) or vehicle (VEH) for 3 months following ovariectomy. A fast-acting water-soluble cyclodextrin-encapsulated E(2) was used to ensure metabolism within 24 h. Vehicle-treated mice received daily injections of 2-hydroxypropyl-beta-cyclodextrin vehicle. The continuous estradiol group (Contin E(2)) was injected daily with estradiol. The intermittent group (Twice/wk E(2)) received estradiol every 4 days and vehicle on all other days. Mice (21 months) were tested in water-escape motivated 8-arm radial arm maze (WRAM) and object recognition tasks. During WRAM acquisition, the Twice/wk E(2) group committed significantly more reference memory errors than VEH and Contin E(2) groups, and tended to make more working memory errors than the VEH group. The Contin E(2) group did not differ from VEH on either WRAM measure. Additionally, the Twice/wk E(2) group tended to exhibit impaired object recognition. Thus, neither treatment improved spatial or object memory. Indeed, intermittent estradiol was detrimental to both types of memory. These results suggest that the timing of administration may play an important role in the mnemonic response of aging females to estrogen.


Assuntos
Envelhecimento/metabolismo , Estrogênios/farmacologia , Transtornos da Memória/tratamento farmacológico , Memória/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Pós-Menopausa/metabolismo , Envelhecimento/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Esquema de Medicação , Estradiol/metabolismo , Estradiol/farmacologia , Estradiol/uso terapêutico , Terapia de Reposição de Estrogênios , Estrogênios/metabolismo , Estrogênios/uso terapêutico , Feminino , Aprendizagem em Labirinto/efeitos dos fármacos , Aprendizagem em Labirinto/fisiologia , Memória/fisiologia , Transtornos da Memória/fisiopatologia , Transtornos da Memória/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Fármacos Neuroprotetores/uso terapêutico , Testes Neuropsicológicos , Tamanho do Órgão/efeitos dos fármacos , Tamanho do Órgão/fisiologia , Ovariectomia , Reconhecimento Psicológico/efeitos dos fármacos , Reconhecimento Psicológico/fisiologia , Resultado do Tratamento , Útero/efeitos dos fármacos , Útero/metabolismo
15.
Pharmacol Biochem Behav ; 84(1): 112-9, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16759685

RESUMO

The present study was designed to determine if post-training injections of a water-soluble form of 17beta-estradiol could enhance spatial and object memory consolidation in young female mice. Young ovariectomized female mice were trained in Morris water maze and object recognition tasks, injected with 0.1, 0.2, or 0.4 mg/kg cyclodextrin-encapsulated 17beta-estradiol or cyclodextrin-conjugated vehicle, and then re-tested after a delay. In the water maze, mice were trained in eight consecutive trials, injected, and memory for the platform location was re-tested after 24 h. All mice learned to find the platform on Day 1, but only mice receiving 0.2 mg/kg estradiol remembered the platform location on Day 2. In the object recognition task, mice were first presented with two identical objects, injected, and then presented with a familiar and novel object after a 24- or 48-h delay. For both delays, the 0.2 and 0.4 mg/kg doses enhanced memory for the familiar object. These data demonstrate that a 0.2 mg/kg dose of estradiol can enhance multiple types of memory consolidation in female mice, and suggest a narrower effective dose range for spatial memory than for object memory.


Assuntos
Estrogênios/farmacologia , Memória/efeitos dos fármacos , Condicionamento Físico Animal , Animais , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Ovariectomia
16.
Nat Med ; 21(9): 1054-9, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26280122

RESUMO

An increase in amyloid-ß (Aß) production is a major pathogenic mechanism associated with Alzheimer's disease (AD), but little is known about possible homeostatic control of the amyloidogenic pathway. Here we report that the amyloid precursor protein (APP) intracellular domain (AICD) downregulates Wiskott-Aldrich syndrome protein (WASP)-family verprolin homologous protein 1 (WAVE1 or WASF1) as part of a negative feedback mechanism to limit Aß production. The AICD binds to the Wasf1 promoter, negatively regulates its transcription and downregulates Wasf1 mRNA and protein expression in Neuro 2a (N2a) cells. WAVE1 interacts and colocalizes with APP in the Golgi apparatus. Experimentally reducing WAVE1 in N2a cells decreased the budding of APP-containing vesicles and reduced cell-surface APP, thereby reducing the production of Aß. WAVE1 downregulation was observed in mouse models of AD. Reduction of Wasf1 gene expression dramatically reduced Aß levels and restored memory deficits in a mouse model of AD. A decrease in amounts of WASF1 mRNA was also observed in human AD brains, suggesting clinical relevance of the negative feedback circuit involved in homeostatic regulation of Aß production.


Assuntos
Peptídeos beta-Amiloides/biossíntese , Precursor de Proteína beta-Amiloide/fisiologia , Transdução de Sinais/fisiologia , Família de Proteínas da Síndrome de Wiskott-Aldrich/fisiologia , Precursor de Proteína beta-Amiloide/química , Animais , Sequência de Bases , Células Cultivadas , Humanos , Masculino , Camundongos , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Família de Proteínas da Síndrome de Wiskott-Aldrich/genética
17.
Behav Neurosci ; 117(6): 1283-91, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14674847

RESUMO

The present studies examined sex differences in object localization and recognition in C57BL/6 mice. Experiment 1 measured responses to spatial novelty (object displacement) and object novelty (object substitution). Males strongly preferred displaced and substituted objects over unchanged objects, whereas females showed a preference in only 1 measure of object novelty. Experiment 2 further examined object recognition by presenting mice with 2 identical objects, followed 24 hr or 7 days later by testing with a familiar and a novel object. After 24 hr, males preferentially explored the novel object, whereas females exhibited no such preference. Neither sex displayed a preference for the novel object after 7 days. The data suggest that male mice are superior to females at localizing and recognizing objects.


Assuntos
Comportamento Exploratório/fisiologia , Percepção Espacial/fisiologia , Comportamento Espacial/fisiologia , Animais , Comportamento Exploratório/classificação , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fatores Sexuais
18.
Brain Res ; 982(1): 98-107, 2003 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-12915244

RESUMO

The present study examined sex differences in spatial working and reference memory in C57BL/6 mice. Males and females were tested in a version of the spatial 8-arm radial arm maze in which the motivating stimulus was escape from water. To test spatial working memory, four arms were baited with submerged escape platforms, each of which was removed after it was found. Four arms that never contained platforms assessed spatial reference memory. In addition to determining the number of working memory and reference memory errors made in each session, working memory errors made in each trial were analyzed to examine performance as the number of arms to be remembered (i.e. the working memory load) increased. Males committed significantly fewer working memory and reference memory errors than females throughout testing. Within a session, males committed fewer working memory errors than females as the working memory load increased. These sex differences were particularly evident during task acquisition. The data indicate that male C57BL/6 mice learn both the working and reference memory components of a water-escape motivated radial arm maze task better than female mice.


Assuntos
Reação de Fuga , Aprendizagem em Labirinto , Memória , Caracteres Sexuais , Percepção Espacial , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Natação
19.
Psychopharmacology (Berl) ; 231(10): 2189-97, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24363077

RESUMO

RATIONALE: Inhibitors of phosphodiesterase 10A (PDE10A), an enzyme highly expressed in medium spiny neurons of the mammalian striatum, enhance activity in direct (dopamine D1 receptor-expressing) and indirect (D2 receptor-expressing striatal output) pathways. The ability of such agents to act to potentiate D1 receptor signaling while inhibiting D2 receptor signaling suggest that PDE10A inhibitors may have a unique antipsychotic-like behavioral profile differentiated from the D2 receptor antagonist-specific antipsychotics currently used in the treatment of schizophrenia. OBJECTIVES: To evaluate the functional consequences of PDE10A inhibitor modulation of D1 and D2 receptor pathway signaling, we compared the effects of a PDE10A inhibitor (TP-10) on D1 and D2 receptor agonist-induced disruptions in prepulse inhibition (PPI), a measure of sensorimotor gating disrupted in patients with schizophrenia. RESULTS: Our results indicate that, in rats: (1) PDE10A inhibition (TP-10, 0.32-10.0 mg/kg) has no effect on PPI disruption resulting from the mixed D1/D2 receptor agonist apomorphine (0.5 mg/kg), confirming previous report; (2) Yet, TP-10 blocked the PPI disruption induced by the D2 receptor agonist quinpirole (0.5 mg/kg); and attenuated apomorphine-induced disruptions in PPI in the presence of the D1 receptor antagonist SCH23390 (0.005 mg/kg). CONCLUSIONS: These findings indicate that TP-10 cannot block dopamine agonist-induced deficits in PPI in the presence of D1 activation and suggest that the effect of PDE10A inhibition on D1 signaling may be counterproductive in some models of antipsychotic activity. These findings, and the contribution of TP-10 effects in the direct pathway on sensorimotor gating in particular, may have implications for the potential antipsychotic efficacy of PDE10A inhibitors.


Assuntos
Agonistas de Dopamina/farmacologia , Inibidores de Fosfodiesterase/farmacologia , Diester Fosfórico Hidrolases/metabolismo , Receptores de Dopamina D1/agonistas , Receptores de Dopamina D2/agonistas , Filtro Sensorial/efeitos dos fármacos , Animais , Apomorfina/farmacologia , Benzazepinas/farmacologia , Antagonistas de Dopamina/farmacologia , Masculino , Quimpirol/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Reflexo de Sobressalto/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
20.
Neuropsychopharmacology ; 39(6): 1409-19, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24326400

RESUMO

Corticotropin releasing factor (CRF) regulates physiological and behavioral responses to stress. Trauma in early life or adulthood is associated with increased CRF in the cerebrospinal fluid and heightened anxiety. Genetic variance in CRF receptors is linked to altered risk for stress disorders. Thus, both heritable differences and environmentally induced changes in CRF neurotransmission across the lifespan may modulate anxiety traits. To test the hypothesis that CRF hypersignaling is sufficient to modify anxiety-related phenotypes (avoidance, startle, and conditioned fear), we induced transient forebrain-specific overexpression of CRF (CRFOE) in mice (1) during development to model early-life stress, (2) in adulthood to model adult-onset stress, or (3) across the entire postnatal lifespan to model heritable increases in CRF signaling. The consequences of these manipulations on CRF peptide levels and behavioral responses were examined in adulthood. We found that transient CRFOE during development decreased startle habituation and prepulse inhibition, and increased avoidance (particularly in females) recapitulating the behavioral effects of lifetime CRFOE despite lower CRF peptide levels at testing. In contrast, CRFOE limited to adulthood reduced contextual fear learning in females and increased startle reactivity in males but did not change avoidance or startle plasticity. These findings suggest that forebrain CRFOE limited to development is sufficient to induce enduring alterations in startle plasticity and anxiety, while forebrain CRFOE during adulthood results in a different phenotype profile. These findings suggest that startle circuits are particularly sensitive to forebrain CRFOE, and that the impact of CRFOE may be dependent on the time of exposure.


Assuntos
Ansiedade/fisiopatologia , Hormônio Liberador da Corticotropina/metabolismo , Prosencéfalo/crescimento & desenvolvimento , Prosencéfalo/fisiopatologia , Reflexo de Sobressalto/fisiologia , Animais , Aprendizagem da Esquiva/fisiologia , Condicionamento Psicológico/fisiologia , Hormônio Liberador da Corticotropina/genética , Modelos Animais de Doenças , Comportamento Exploratório/fisiologia , Medo/fisiologia , Feminino , Habituação Psicofisiológica/fisiologia , Inibição Psicológica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Filtro Sensorial/fisiologia , Fatores Sexuais , Estresse Psicológico/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA