Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
J Neurosci ; 34(7): 2444-50, 2014 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-24523535

RESUMO

Brain-derived neurotrophic factor (BDNF) is known to have an integral role in establishing stable memories after learning events. The neuroplasticity induced by Pavlovian fear conditioning has likewise been shown to rely on interactions between BDNF and its principal receptor, tyrosine kinase receptor B (TrkB), in the amygdala after training. Although the necessity of amygdala bdnf expression and TrkB activation for associative learning within aversive contexts has been explored, it is unclear to what extent this interaction is involved in appetitive learning. It is also unclear whether the noted increases in amygdala BDNF after fear conditioning are due to local gene transcription and translation or anterograde transmission from cortical regions. To address both of these questions, we used two lentiviral approaches in mice, using both fear conditioning and cocaine-conditioned place preference (CPP), during acquisition and extinction. First, we decreased expression of bdnf mRNA in the amygdala of homozygous floxed mice with a Cre-expressing virus. In a second set of studies, we infused a virus that expressed a dominant-negative TrkB isoform into the same region. These approaches significantly impaired consolidation of fear conditioning and cocaine-CPP, as well as extinction of CPP. Together, these data suggest that BDNF-TrkB signaling is critical for amygdala-dependent learning of both appetitive and aversive emotional memories.


Assuntos
Tonsila do Cerebelo/metabolismo , Apetite , Aprendizagem por Associação/fisiologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Receptor trkB/metabolismo , Transdução de Sinais/fisiologia , Animais , Memória/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Mutantes
2.
J Neurosci ; 34(7): 2464-70, 2014 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-24523537

RESUMO

The excessive accumulation of soluble amyloid peptides (Aß) plays a crucial role in the pathogenesis of Alzheimer's disease (AD), particularly in synaptic dysfunction. The role of the two major chaperone proteins, Hsp70 and Hsp90, in clearing misfolded protein aggregates has been established. Despite their abundant presence in synapses, the role of these chaperones in synapses remains elusive. Here, we report that Hsp90 inhibition by 17-AAG elicited not only a heat shock-like response but also upregulated presynaptic and postsynaptic proteins, such as synapsin I, synaptophysin, and PSD95 in neurons. 17-AAG treatment enhanced high-frequency stimulation-evoked LTP and protected neurons from synaptic damage induced by soluble Aß. In AD transgenic mice, the daily administration of 17-AAG over 7 d resulted in a marked increase in PSD95 expression in hippocampi. 17-AAG treatments in wild-type C57BL/6 mice challenged by soluble Aß significantly improved contextual fear memory. Further, we demonstrate that 17-AAG activated synaptic protein expression via transcriptional mechanisms through the heat shock transcription factor HSF1. Together, our findings identify a novel function of Hsp90 inhibition in regulating synaptic plasticity, in addition to the known neuroprotective effects of the chaperones against Aß and tau toxicity, thus further supporting the potential of Hsp90 inhibitors in treating neurodegenerative diseases.


Assuntos
Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/toxicidade , Benzoquinonas/farmacologia , Proteínas de Choque Térmico HSP90/metabolismo , Lactamas Macrocíclicas/farmacologia , Potenciação de Longa Duração/efeitos dos fármacos , Sinapses/metabolismo , Doença de Alzheimer/patologia , Animais , Western Blotting , Modelos Animais de Doenças , Imuno-Histoquímica , Potenciação de Longa Duração/fisiologia , Memória/efeitos dos fármacos , Memória/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Ratos , Sinapses/efeitos dos fármacos , Sinapses/patologia
3.
Physiol Genomics ; 47(8): 308-17, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25991709

RESUMO

Alcoholism, stress, and anxiety are strongly interacting heritable, polygenetic traits. In a previous study, we identified a quantitative trait locus (QTL) on murine chromosome (Chr) 1 between 23.0 and 31.5 Mb that modulates genetic differences in the effects of ethanol on anxiety-related phenotypes. The goal of the present study was to extend the analysis of this locus with a focus on identifying candidate genes using newly available data and tools. Anxiety-like behavior was evaluated with an elevated zero maze following saline or ethanol injections (1.8 g/kg) in C57BL/6J, DBA2J, and 72 BXD strains. We detected significant effects of strain and treatment and their interaction on anxiety-related behaviors, although surprisingly, sex was not a significant factor. The Chr1 QTL is specific to the ethanol-treated cohort. Candidate genes in this locus were evaluated using now standard bioinformatic criteria. Collagen 19a1 (Col19a1) and family sequence 135a (Fam135a) met most criteria but have lower expression levels and lacked biological verification and, therefore, were considered less likely candidates. In contrast, two other genes, the prenylated protein tyrosine phosphate family member Ptp4a1 (protein tyrosine phosphate 4a1) and the zinc finger protein Phf3 (plant homeoDomain finger protein 3) met each of our bioinformatic criteria and are thus strong candidates. These findings are also of translational relevance because both Ptp4a1 and Phf3 have been nominated as candidates genes for alcohol dependence in a human genome-wide association study. Our findings support the hypothesis that variants in one or both of these genes modulate heritable differences in the effects of ethanol on anxiety-related behaviors.


Assuntos
Cromossomos de Mamíferos/genética , Etanol/efeitos adversos , Estudos de Associação Genética , Locos de Características Quantitativas/genética , Estresse Fisiológico/genética , Animais , Comportamento Animal , Feminino , Proteínas Imediatamente Precoces/genética , Masculino , Camundongos , Fenótipo , Polimorfismo Genético , Proteínas Tirosina Fosfatases/genética
4.
Int J Mol Sci ; 16(1): 758-87, 2014 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-25561230

RESUMO

We have developed a focal blast model of closed-head mild traumatic brain injury (TBI) in mice. As true for individuals that have experienced mild TBI, mice subjected to 50-60 psi blast show motor, visual and emotional deficits, diffuse axonal injury and microglial activation, but no overt neuron loss. Because microglial activation can worsen brain damage after a concussive event and because microglia can be modulated by their cannabinoid type 2 receptors (CB2), we evaluated the effectiveness of the novel CB2 receptor inverse agonist SMM-189 in altering microglial activation and mitigating deficits after mild TBI. In vitro analysis indicated that SMM-189 converted human microglia from the pro-inflammatory M1 phenotype to the pro-healing M2 phenotype. Studies in mice showed that daily administration of SMM-189 for two weeks beginning shortly after blast greatly reduced the motor, visual, and emotional deficits otherwise evident after 50-60 psi blasts, and prevented brain injury that may contribute to these deficits. Our results suggest that treatment with the CB2 inverse agonist SMM-189 after a mild TBI event can reduce its adverse consequences by beneficially modulating microglial activation. These findings recommend further evaluation of CB2 inverse agonists as a novel therapeutic approach for treating mild TBI.


Assuntos
Benzofenonas/farmacologia , Lesões Encefálicas/tratamento farmacológico , Atividade Motora/efeitos dos fármacos , Receptor CB2 de Canabinoide/agonistas , Animais , Lesões Encefálicas/complicações , Lesões Encefálicas/patologia , Proteínas de Ligação ao Cálcio/metabolismo , Células Cultivadas , Quimiocinas/metabolismo , Citocinas/metabolismo , Depressão/etiologia , Depressão/patologia , Modelos Animais de Doenças , Agonismo Inverso de Drogas , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas dos Microfilamentos/metabolismo , Microglia/citologia , Microglia/efeitos dos fármacos , Microglia/metabolismo , Fenótipo , Receptor CB2 de Canabinoide/metabolismo , Transtornos da Visão/etiologia , Transtornos da Visão/patologia
5.
Learn Mem ; 20(9): 482-90, 2013 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-23955171

RESUMO

Experience-induced changes associated with odor learning are mediated by a number of signaling molecules, including nitric oxide (NO), which is predominantly synthesized by neuronal nitric oxide synthase (nNOS) in the brain. In the current study, we investigated the role of nNOS in the acquisition and retention of conditioned olfactory fear. Mice lacking nNOS received six training trials, each consisting of an odor-CS co-terminating with a foot shock-US. Mice showed reduced freezing responses to the trained odor 24 h and 7 d after training, compared to wild-type mice. Pretraining systemic injections of the NO donor, molsidomine, rescued fear retention in nNOS knockout mice. In wild-type mice, pretraining systemic injections of L-NAME, a nonspecific nNOS blocker, disrupted odor-CS fear retention in a dose-dependent manner. To evaluate whether NO signaling is involved in generalization of fear memories, nNOS knockout mice and wild-type mice receiving L-NAME were trained to one odor and tested with a series of similar odors. In both cases, we found increased generalization, as measured by increased freezing to similar, unpaired odors. Despite the impairment in fear memory retention and generalization, neither mice receiving injections of L-NAME nor nNOS knockout mice showed any deficits in either novel odor investigation time or odor habituation, suggesting intact olfactory perception and short-term memory olfactory learning. These results support a necessary role for neuronal NO signaling in the normal expression and generalization of olfactory conditioned fear.


Assuntos
Condicionamento Psicológico/fisiologia , Memória de Longo Prazo/fisiologia , Óxido Nítrico Sintase Tipo I/fisiologia , Olfato , Tonsila do Cerebelo/fisiologia , Animais , Medo , Feminino , Hipocampo/fisiologia , Masculino , Camundongos , Camundongos Knockout , NG-Nitroarginina Metil Éster/farmacologia , Neurônios/enzimologia , Óxido Nítrico Sintase Tipo I/antagonistas & inibidores , Óxido Nítrico Sintase Tipo I/genética , Odorantes
6.
J Virol ; 86(1): 373-81, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22013043

RESUMO

Here we report a novel viral glycoprotein created by replacing a natural receptor-binding sequence of the ecotropic Moloney murine leukemia virus envelope glycoprotein with the peptide ligand somatostatin. This new chimeric glycoprotein, which has been named the Sst receptor binding site (Sst-RBS), gives targeted transduction based on three criteria: (i) a gain of the use of a new entry receptor not used by any known virus; (ii) targeted entry at levels comparable to gene delivery by wild-type ecotropic Moloney murine leukemia virus and vesicular stomatitis virus (VSV) G glycoproteins; and (iii) a loss of the use of the natural ecotropic virus receptor. Retroviral vectors coated with Sst-RBS gained the ability to bind and transduce human 293 cells expressing somatostatin receptors. Their infection was specific to target somatostatin receptors, since a synthetic somatostatin peptide inhibited infection in a dose-dependent manner and the ability to transduce mouse cells bearing the natural ecotropic receptor was effectively lost. Importantly, vectors coated with the Sst-RBS glycoprotein gave targeted entry of up to 1 × 10(6) transducing U/ml, a level comparable to that seen with infection of vectors coated with the parental wild-type ecotropic Moloney murine leukemia virus glycoprotein through the ecotropic receptor and approaching that of infection of VSV G-coated vectors through the VSV receptor. To our knowledge, this is the first example of a glycoprotein that gives targeted entry of retroviral vectors at levels comparable to the natural capacity of viral envelope glycoproteins.


Assuntos
Técnicas de Transferência de Genes/instrumentação , Vírus da Leucemia Murina de Moloney/genética , Receptores de Somatostatina/metabolismo , Somatostatina/genética , Proteínas do Envelope Viral/genética , Internalização do Vírus , Animais , Sítios de Ligação , Linhagem Celular , Marcação de Genes/instrumentação , Vetores Genéticos/química , Vetores Genéticos/genética , Vetores Genéticos/fisiologia , Humanos , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Vírus da Leucemia Murina de Moloney/química , Vírus da Leucemia Murina de Moloney/fisiologia , Ligação Proteica , Engenharia de Proteínas , Receptores de Somatostatina/química , Receptores de Somatostatina/genética , Receptores Virais/genética , Receptores Virais/metabolismo , Somatostatina/química , Somatostatina/metabolismo , Proteínas do Envelope Viral/química , Proteínas do Envelope Viral/metabolismo
7.
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
8.
J Neurosci ; 30(21): 7139-51, 2010 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-20505082

RESUMO

The heterogeneity and distribution of GABA(A) receptor subunits mediates differential roles in behavior. It is thought that particular behavioral responses to benzodiazepine (BZ) ligands might be associated with an action at a regionally defined receptor subtype. However, the role of specific GABA(A) receptor subtypes in particular brain regions is less clear. Such detailed knowledge of regional alpha1-GABA(A) receptor function will advance our understanding of the neural circuitry underlying the role of GABA(A) receptors and the effects of GABA(A)-modulating drugs on behavior. By combining inducible, site-specific alpha1 subunit deletion, using a lentivirus expressing Cre-recombinase in mice with the alpha1 subunit gene flanked by loxP sites, we examine baseline and pharmacological effects of deletion of amygdala alpha1-GABA(A) receptors. We find that amygdala-specific reduction of alpha1 receptor subunits does not affect mRNA or protein levels of amygdala alpha2 or alpha3 subunit receptors. Nor does this inducible reduction affect baseline locomotion or measures of anxiety. However, we also find that this inducible, site-specific deletion does disrupt the normal sedative-locomotor inhibition as well as the anticonvulsive effects, of two distinct BZ-site ligands, diazepam and zolpidem, which is relatively alpha1-subunit selective. These data, using inducible, region and subunit-specific deletion, combined with pharmacogenetic approaches, demonstrate that amygdala expression of the alpha1-GABA(A) receptor subunit is required for normal BZ effects on sedation, locomotion, and seizure inhibition, but not for anxiolysis.


Assuntos
Tonsila do Cerebelo/fisiologia , Anticonvulsivantes/farmacologia , Benzodiazepinas/farmacologia , Hipnóticos e Sedativos/farmacologia , Locomoção/efeitos dos fármacos , Receptores de GABA-A/metabolismo , Tonsila do Cerebelo/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Ansiolíticos/farmacologia , Linhagem Celular Transformada , Células Cultivadas , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Epilepsia/induzido quimicamente , Epilepsia/tratamento farmacológico , Comportamento Exploratório/efeitos dos fármacos , Comportamento Exploratório/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Proteínas de Fluorescência Verde/genética , Hipocampo/citologia , Humanos , Locomoção/genética , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pentilenotetrazol , Piridinas/farmacologia , RNA Mensageiro/metabolismo , Receptores de GABA-A/genética , Transfecção/métodos , Zolpidem
9.
Behav Pharmacol ; 20(7): 584-95, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19675456

RESUMO

Gerbils show a neurokinin (NK)1 receptor pharmacological profile, which is similar to that observed in humans, and thus have become a commonly used species to test efficacy of NK1 receptor antagonists. The aim of this study was to determine whether systemic administration of the NK1 receptor antagonist GR-205171 produced anxiolytic-like effects in the elevated plus maze and in a novel contextual conditioned fear test using fear-potentiated startle (FPS). On the elevated plus maze, treatment with GR-205171 at 0, 0.3, 1.0, and 5.0 mg/kg doses, 30 min before testing produced anxiolytic-like effects in an increasing dose-response manner as measured by the percentage of open arm time and percentage of open arm entries. For contextual fear conditioning, gerbils were given 10 unsignaled footshocks (0.6 mA) at a 2-min variable interstimulus interval in a distinctive training context. Twenty-four hours after training, gerbils received treatment of GR-205171 at 0, 0.3, 1.0, and 5.0 mg/kg doses, 30 min before testing in which startle was elicited in the same context in which they were trained. Contextual FPS was defined as an increase in startle over pretraining baseline values. All drug dose levels (0.3, 1.0, and 5.0 mg/kg) significantly attenuated contextual FPS when compared with the vehicle control group. A control group, which received testing in a different context, showed little FPS. These findings support other evidence for anxiolytic activity of NK1 receptor antagonists and provide a novel conditioned fear test that may be an appropriate procedure to test other NK1 antagonists for preclinical anxiolytic activity in gerbils.


Assuntos
Ansiolíticos/uso terapêutico , Ansiedade/tratamento farmacológico , Medo/efeitos dos fármacos , Aprendizagem em Labirinto/efeitos dos fármacos , Antagonistas dos Receptores de Neurocinina-1 , Piperidinas/uso terapêutico , Reflexo de Sobressalto/efeitos dos fármacos , Tetrazóis/uso terapêutico , Animais , Ansiolíticos/farmacologia , Relação Dose-Resposta a Droga , Gerbillinae , Masculino , Modelos Animais , Piperidinas/farmacologia , Tetrazóis/farmacologia
10.
Behav Brain Res ; 187(2): 262-72, 2008 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-17963852

RESUMO

Neglect is a complex human cognitive spatial disorder typically induced by damage to prefrontal or posterior parietal association cortices. Behavioral treatments for neglect rarely generalize outside of the therapeutic context or across tasks within the same therapeutic context. Recovery, when it occurs, is spontaneous over the course of weeks to months, but often it is incomplete. A number of studies have indicated that anti-Nogo-A antibodies can be used to enhance plasticity and behavioral recovery following damage to motor cortex, and spinal cord. In the present studies the anti-Nogo-A antibodies IN-1, 7B12, or 11C7 were applied intraventricularly to adult rats demonstrating severe neglect produced by unilateral medial agranular cortex lesions in rats. The three separate anti-Nogo-A antibody groups were treated immediately following the medial agranular cortex lesions. Each of the three antibodies induced dramatic significant behavioral recovery from neglect relative to controls. Severing the corpus callosum to destroy inputs from the contralesional hemisphere resulted in reinstatement of severe neglect, pointing to a possible role of interhemispheric mechanisms in behavioral recovery from neglect.


Assuntos
Córtex Cerebral/fisiologia , Proteínas da Mielina/fisiologia , Plasticidade Neuronal/fisiologia , Transtornos da Percepção/fisiopatologia , Recuperação de Função Fisiológica/fisiologia , Análise de Variância , Animais , Corpo Caloso/fisiologia , Lateralidade Funcional/fisiologia , Atividade Motora/fisiologia , Proteínas Nogo , Distribuição Aleatória , Ratos , Estatísticas não Paramétricas
11.
Eur J Neurosci ; 26(12): 3631-44, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18088283

RESUMO

Previous work suggests the gamma-aminobutyric acid (GABA)ergic system may be dynamically regulated during emotional learning. In the current study we examined training-induced changes in the expression of GABA(A)-related genes and the binding of GABA receptor radioligands in the amygdala after the acquisition and extinction of Pavlovian fear. Using in situ hybridization, we examined the expression pattern changes of mRNAs for GABAergic markers in the lateral, basolateral and central subdivisions of the amygdala in C57Bl/6J mice. These markers included GABA-synthesizing enzymes (GAD67 and GAD65), major GABA(A) receptor subunits (alpha1, alpha2, alpha3, alpha5, beta2 and gamma2) and the expression of mRNAs that are involved in a variety of GABA-related intracellular processes, including GABA transporter-1 (GAT1), GABA(A) receptor-associated protein and the GABA(A) clustering protein, gephyrin. With fear conditioning, we found decreased mRNA levels of alpha1, alpha5 and GAD67, as well as deceased benzodiazepine binding in the amygdala. Fear extinction induced an increase in mRNA levels of alpha2, beta2, GAD67 and gephyrin, as well as a decrease in GAT1. Together, these findings indicate that the acquisition of fear induced a downregulation of mRNA markers related to a decrease in amygdala GABAergic function, whereas the acquisition of fear extinction produced an upregulation of GABAergic markers related to enhanced GABAergic transmission.


Assuntos
Tonsila do Cerebelo/metabolismo , Condicionamento Clássico/fisiologia , Extinção Psicológica/fisiologia , Medo , Receptores de GABA-A/metabolismo , Animais , Biomarcadores/metabolismo , Proteínas de Transporte/genética , Medo/fisiologia , Proteínas da Membrana Plasmática de Transporte de GABA/genética , Expressão Gênica , Glutamato Descarboxilase/genética , Hibridização In Situ , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Isoformas de Proteínas/metabolismo , RNA Mensageiro/metabolismo , Reflexo de Sobressalto , Retenção Psicológica , Distribuição Tecidual
12.
eNeuro ; 4(4)2017.
Artigo em Inglês | MEDLINE | ID: mdl-28828401

RESUMO

Mild traumatic brain injury (mTBI) can cause severe long-term cognitive and emotional deficits, including impaired memory, depression, and persevering fear, but the neuropathological basis of these deficits is uncertain. As medial prefrontal cortex (mPFC) and hippocampus play important roles in memory and emotion, we used multi-site, multi-electrode recordings of oscillatory neuronal activity in local field potentials (LFPs) in awake, head-fixed mice to determine if the functioning of these regions was abnormal after mTBI, using a closed-skull focal cranial blast model. We evaluated mPFC, hippocampus CA1, and primary somatosensory/visual cortical areas (S1/V1). Although mTBI did not alter the power of oscillations, it did cause increased coherence of θ (4-10 Hz) and ß (10-30 Hz) oscillations within mPFC and S1/V1, reduced CA1 sharp-wave ripple (SWR)-evoked LFP activity in mPFC, downshifted SWR frequencies in CA1, and enhanced θ-γ phase-amplitude coupling (PAC) within mPFC. These abnormalities might be linked to the impaired memory, depression, and persevering fear seen after mTBI. Treatment with the cannabinoid type-2 (CB2) receptor inverse agonist SMM-189 has been shown to mitigate functional deficits and neuronal injury after mTBI in mice. We found that SMM-189 also reversed most of the observed neurophysiological abnormalities. This neurophysiological rescue is likely to stem from the previously reported reduction in neuron loss and/or the preservation of neuronal function and connectivity resulting from SMM-189 treatment, which appears to stem from the biasing of microglia from the proinflammatory M1 state to the prohealing M2 state by SMM-189.


Assuntos
Benzofenonas/uso terapêutico , Concussão Encefálica/tratamento farmacológico , Concussão Encefálica/patologia , Encéfalo/efeitos dos fármacos , Agonistas de Receptores de Canabinoides/uso terapêutico , Potenciais de Ação/efeitos dos fármacos , Animais , Encéfalo/patologia , Mapeamento Encefálico , Ondas Encefálicas/efeitos dos fármacos , Modelos Animais de Doenças , Eletroencefalografia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Análise de Componente Principal , Receptor CB2 de Canabinoide/metabolismo , Fatores de Tempo
13.
Brain Res ; 1073-1074: 229-39, 2006 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-16442084

RESUMO

The habenula complex modulates the activity of dopamine and serotonin systems in the brain. An important question remains whether there is a link between habenula dysfunction and monoamine-related disorders, such as schizophrenia. In this study, we describe an interaction between habenula lesions and stress that produces long-lasting effects on behavior. Mice received control lesions or bilateral electrolytic lesions of the habenula and were tested for fear-potentiated startle and freezing measures of conditioned fear. They were also tested for prepulse inhibition (PPI) and locomotor activity in the presence or absence of a dopaminergic agonist (apomorphine) or an atypical antipsychotic with mixed dopamine/serotonin antagonist properties (clozapine). There were no detectable effects of habenula lesions on fear conditioning and no effects on PPI in the absence of stress. However, following conditioned fear stress, habenula-lesioned animals showed decreased PPI which normalized with clozapine. Lesioned animals also showed diminished activity at baseline, with hyperlocomotion following apomorphine. These data support the hypothesis that the habenula may be normally involved in stress-dependent regulation of monoamine systems.


Assuntos
Dopamina/metabolismo , Habenula/fisiopatologia , Locomoção/fisiologia , Inibição Neural/fisiologia , Estresse Fisiológico/fisiopatologia , Análise de Variância , Animais , Apomorfina/administração & dosagem , Comportamento Animal , Eletrólise/efeitos adversos , Comportamento Exploratório/efeitos dos fármacos , Comportamento Exploratório/fisiologia , Medo/psicologia , Reação de Congelamento Cataléptica/fisiologia , Habenula/lesões , Locomoção/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Reflexo de Sobressalto/fisiologia , Estresse Fisiológico/etiologia
14.
Front Cell Neurosci ; 10: 39, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26973458

RESUMO

Although it is generally recognized that certain α-subunits of γ-aminobutyric acid type A receptors (GABAARs) form enriched clusters on the axonal initial segment (AIS), the degree to which these clusters vary in different brain areas is not well known. In the current study, we quantified the density, size, and enrichment ratio of fluorescently labeled α1-, α2-, or α3-subunits aggregates co-localized with the AIS-marker ankyrin G and compared them to aggregates in non-AIS locations among different brain areas including hippocampal subfields, basal lateral amygdala (BLA), prefrontal cortex (PFC), and sensory cortex (CTX). We found regional differences in the enrichment of GABAAR α-subunits on the AIS. Significant enrichment was identified in the CA3 of hippocampus for α1-subunits, in the CA1, CA3, and BLA for α2-subunits, and in the BLA for α3-subunits. Using α-subunit knock-out (KO) mice, we found that BLA enrichment of α2- and α3-subunits were physiologically independent of each other, as the enrichment of one subunit was unaffected by the genomic deletion of the other. To further investigate the unique pattern of α-subunit enrichment in the BLA, we examined the association of α2- and α3-subunits with the presynaptic vesicular GABA transporter (vGAT) and the anchoring protein gephyrin (Geph). As expected, both α2- and α3-subunits on the AIS within the BLA received prominent GABAergic innervation from vGAT-positive terminals. Further, we found that the association of α2- and α3-subunits with Geph was weaker in AIS versus non-AIS locations, suggesting that Geph might be playing a lesser role in the enrichment of α2- and α3-subunits on the AIS. Overall, these observations suggest that GABAARs on the AIS differ in subunit composition across brain regions. As with somatodendritic GABAARs, the distinctive expression pattern of AIS-located GABAAR α-subunits in the BLA, and other brain areas, likely contribute to unique forms of GABAergic inhibitory transmission and pharmacological profiles seen in different brain areas.

15.
Elife ; 5: e14120, 2016 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-26971710

RESUMO

Recent findings indicate a high level of specialization at the level of microcircuits and cell populations within brain structures with regards to the control of fear and anxiety. The hippocampus, however, has been treated as a unitary structure in anxiety and fear research despite mounting evidence that different hippocampal subregions have specialized roles in other cognitive domains. Using novel cell-type- and region-specific conditional knockouts of the GABAA receptor α2 subunit, we demonstrate that inhibition of the principal neurons of the dentate gyrus or CA3 via α2-containing GABAA receptors (α2GABAARs) is required to suppress anxiety, while the inhibition of CA1 pyramidal neurons is required to suppress fear responses. We further show that the diazepam-modulation of hippocampal theta activity shows certain parallels with our behavioral findings, suggesting a possible mechanism for the observed behavioral effects. Thus, our findings demonstrate a double dissociation in the regulation of anxiety versus fear by hippocampal microcircuitry.


Assuntos
Ansiedade , Mapeamento Encefálico , Medo , Hipocampo/fisiologia , Animais , Camundongos , Rede Nervosa , Receptores de GABA-A/genética
16.
Behav Neurosci ; 119(1): 329-35, 2005 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15727538

RESUMO

This study demonstrates that mice display olfactory-cued fear as measured with both freezing and fear-potentiated startle. Following a preconditioning test to measure any unconditioned responses to odor, mice received 5 pairings of a 10-s odor with a 0.25-s, 0.4-mA footshock. The next day, startle and freezing were measured in the presence and absence of the odor. Both fear measures increased after training with amyl acetate (Experiment 1) and acetophenone (Experiment 2). The enhancement of startle did not occur when the same number of odors and shocks were presented in an unpaired fashion (Experiment 3). Furthermore, mice were able to discriminate between an odor paired with shock and a nonreinforced odor (Experiment 4).


Assuntos
Aprendizagem da Esquiva , Medo , Olfato , Acetofenonas/farmacologia , Animais , Temperatura Baixa , Condicionamento Clássico , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Odorantes , Pentanóis/farmacologia , Reflexo de Sobressalto
17.
Behav Neurosci ; 129(1): 50-61, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25621792

RESUMO

Nitric oxide (NO) is an important molecule for the proper development and function of the central nervous system. In this study, we investigated the behavioral alterations in the neuronal NO synthase knockout mice (NOS1 KO) with a deficient NO production mechanism in the brain, characterizing it as a potential rodent model for attention deficit hyperactivity disorder (ADHD). NOS1 KO exhibited higher locomotor activity than their wildtype counterparts in a novel environment, as measured by open field (OF) test. In a 2-way active avoidance paradigm (TWAA), we found sex-dependent effects, where male KO displayed deficits in avoidance and escape behavior, sustained higher incidences of shuttle crossings, and higher incidences of intertrial interval crossings, suggesting learning, and/or performance impairments. On the other hand, female KO demonstrated few deficits in TWAA. Molsidomine (MSD), a NO donor, rescued TWAA deficits in male KO when acutely administered before training. In a passive avoidance paradigm, KO of both sexes displayed significantly shorter step-through latencies after training. Further, abnormal spontaneous motor activity rhythms were found in the KO during the dark phase of the day, indicating dysregulation of rhythmic activities. These data indicate that NOS1 KO mimics certain ADHD-like behaviors and could potentially serve as a novel rodent model for ADHD.


Assuntos
Transtorno do Deficit de Atenção com Hiperatividade/fisiopatologia , Transtorno do Deficit de Atenção com Hiperatividade/psicologia , Modelos Animais de Doenças , Óxido Nítrico Sintase Tipo I/fisiologia , Animais , Transtorno do Deficit de Atenção com Hiperatividade/prevenção & controle , Aprendizagem da Esquiva/efeitos dos fármacos , Aprendizagem da Esquiva/fisiologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Molsidomina/administração & dosagem , Atividade Motora/efeitos dos fármacos , Óxido Nítrico Sintase Tipo I/genética
18.
Neuropsychopharmacology ; 29(9): 1610-9, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15114343

RESUMO

Recent postmortem studies in humans suggest that defects in GABAergic neurotransmission might contribute to the neuropathology associated with schizophrenia. Disturbances in GABAergic systems may also contribute to the sensorimotor gating deficits classically observed in schizophrenic patients, including deficits in prepulse inhibition (PPI). To explore the relationship, the current study examined the integrity of PPI and startle habituation in knockout (KO) mice that lack the GABA synthesizing enzyme glutamic acid decarboxylase 65 (GAD 65). GAD65 KO mice displayed normal baseline and habituated startle responses, which did not differ from GAD65 wild-type (WT) or heterozygous (HET) mice. However, GAD65 KO mice showed robust deficits in PPI which were reversed by the atypical antipsychotic agent clozapine. These results lend support to the view that abnormalities in GABAergic systems might contribute to the basic pathophysiological mechanisms in schizophrenia.


Assuntos
Antipsicóticos/farmacologia , Glutamato Descarboxilase/genética , Glutamato Descarboxilase/fisiologia , Isoenzimas/genética , Isoenzimas/fisiologia , Reflexo de Sobressalto/genética , Estimulação Acústica , Animais , Clozapina/farmacologia , Primers do DNA , Deleção de Genes , Genótipo , Habituação Psicofisiológica/fisiologia , Hibridização In Situ , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transmissão Sináptica/efeitos dos fármacos , Ácido gama-Aminobutírico/fisiologia
19.
Behav Brain Res ; 144(1-2): 175-85, 2003 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-12946608

RESUMO

The inferior colliculus (IC) is the major source of auditory information involved in processing the behavioral significance of acoustic stimuli. In the current study, we assessed whether the IC is a critical source of information which mediates the expression of fear and the inhibition of fear conditioned to an auditory stimulus. Fear and the inhibition of fear were tested by measuring fear-potentiated startle. In Experiment 1, we demonstrated that rats which received electrolytic lesions of the IC failed to show fear-potentiated startle in the presence of a noise previously conditioned to elicit fear. In Experiment 2, we demonstrated that rats with similarly placed lesions of the IC failed to inhibit fear-potentiated startle in the presence of a noise previously conditioned to inhibit fear to a light. Thus, in both Experiments 1 and 2, lesions of the IC disrupted the behavioral significance of the noise stimulus. Together with previous findings, these results are consistent with the view that the IC is a common source of diverging auditory information used to mediate the fear eliciting and safety signal properties conditioned to auditory stimuli.


Assuntos
Condicionamento Psicológico , Medo/psicologia , Colículos Inferiores/fisiopatologia , Inibição Psicológica , Estimulação Acústica/métodos , Animais , Comportamento Animal , Constrição , Luz/efeitos adversos , Masculino , Ruído/efeitos adversos , Ratos , Ratos Sprague-Dawley , Reflexo de Sobressalto/fisiologia
20.
Psychopharmacology (Berl) ; 231(9): 1865-96, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24563183

RESUMO

RATIONALE: Zolpidem is a positive allosteric modulator of γ-aminobutyric acid (GABA) with preferential binding affinity and efficacy for α1-subunit containing GABA(A) receptors (α1-GABA(A)Rs). Over the last three decades, a variety of animal models and experimental procedures have been used in an attempt to relate the behavioral profile of zolpidem and classic benzodiazepines (BZs) to their interaction with α1-GABA(A)Rs. OBJECTIVES: This paper reviews the results of rodent and non-human primate studies that have evaluated the effects of zolpidem on motor behaviors, anxiety, memory, food and fluid intake, and electroencephalogram (EEG) sleep patterns. Also included are studies that examined zolpidem's discriminative, reinforcing, and anticonvulsant effects as well as behavioral signs of tolerance and withdrawal. RESULTS: The literature reviewed indicates that α1-GABA(A)Rs play a principle role in mediating the hypothermic, ataxic-like, locomotor- and memory-impairing effects of zolpidem and BZs. Evidence also suggests that α1-GABA(A)Rs play partial roles in the hypnotic, EEG sleep, anticonvulsant effects, and anxiolytic-like of zolpidem and diazepam. These studies also indicate that α1-GABA(A)Rs play a more prominent role in mediating the discriminative stimulus, reinforcing, hyperphagic, and withdrawal effects of zolpidem and BZs in primates than in rodents. CONCLUSIONS: The psychopharmacological data from both rodents and non-human primates suggest that zolpidem has a unique pharmacological profile when compared with classic BZs. The literature reviewed here provides an important framework for studying the role of different GABA(A)R subtypes in the behavioral effects of BZ-type drugs and helps guide the development of new pharmaceutical agents for disorders currently treated with BZ-type drugs.


Assuntos
Comportamento Animal/efeitos dos fármacos , Agonistas de Receptores de GABA-A/farmacologia , Piridinas/farmacologia , Receptores de GABA-A/metabolismo , Animais , Comportamento Animal/fisiologia , Agonistas de Receptores de GABA-A/farmacocinética , Humanos , Piridinas/farmacocinética , Zolpidem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA