Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 12(1): 4156, 2021 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-34230461

RESUMO

Fear extinction is an adaptive process whereby defensive responses are attenuated following repeated experience of prior fear-related stimuli without harm. The formation of extinction memories involves interactions between various corticolimbic structures, resulting in reduced central amygdala (CEA) output. Recent studies show, however, the CEA is not merely an output relay of fear responses but contains multiple neuronal subpopulations that interact to calibrate levels of fear responding. Here, by integrating behavioural, in vivo electrophysiological, anatomical and optogenetic approaches in mice we demonstrate that fear extinction produces reversible, stimulus- and context-specific changes in neuronal responses to conditioned stimuli in functionally and genetically defined cell types in the lateral (CEl) and medial (CEm) CEA. Moreover, we show these alterations are absent when extinction is deficient and that selective silencing of protein kinase C delta-expressing (PKCδ) CEl neurons impairs fear extinction. Our findings identify CEA inhibitory microcircuits that act as critical elements within the brain networks mediating fear extinction.


Assuntos
Núcleo Central da Amígdala/fisiologia , Extinção Psicológica/fisiologia , Medo/fisiologia , Animais , Comportamento Animal , Condicionamento Clássico/fisiologia , Masculino , Memória , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/metabolismo
2.
Int J Neuropsychopharmacol ; 22(4): 317-328, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30668714

RESUMO

BACKGROUND: Pathological anxiety originates from a complex interplay of genetic predisposition and environmental factors, acting via epigenetic mechanisms. Epigenetic processes that can counteract detrimental genetic risk towards innate high anxiety are not well characterized. METHODS: We used female mouse lines of selectively bred high (HAB)- vs low (LAB)-innate anxiety-related behavior and performed select environmental and pharmacological manipulations to alter anxiety levels as well as brain-specific manipulations and immunohistochemistry to investigate neuronal mechanisms associated with alterations in anxiety-related behavior. RESULTS: Inborn hyperanxiety of high anxiety-like phenotypes was effectively reduced by environmental enrichment exposure. c-Fos mapping revealed that hyperanxiety in high anxiety-like phenotypes was associated with blunted challenge-induced neuronal activation in the cingulate-cortex, which was normalized by environmental enrichment. Relating this finding with epigenetic modifications, we found that high anxiety-like phenotypes (compared with low-innate anxiety phenotypes) showed reduced acetylation in the hypoactivated cingulate-cortex neurons following a mild emotional challenge, which again was normalized by environmental enrichment. Paralleling the findings using environmental enrichment, systemic administration of histone-deacetylase-inhibitor MS-275 elicited an anxiolytic-like effect, which was correlated with increased acetylated-histone-3 levels within cingulate-cortex. Finally, as a proof-of-principle, local MS-275 injection into cingulate-cortex rescued enhanced innate anxiety and increased acetylated-histone-3 within the cingulate-cortex, suggesting this epigenetic mark as a biomarker for treatment success. CONCLUSIONS: Taken together, the present findings provide the first causal evidence that the attenuation of high innate anxiety-like behavior via environmental/pharmacological manipulations is epigenetically mediated via acetylation changes within the cingulate-cortex. Finally, histone-3 specific histone-deacetylase-inhibitor could be of therapeutic importance in anxiety disorders.


Assuntos
Ansiolíticos/farmacologia , Ansiedade , Comportamento Animal , Meio Ambiente , Epigênese Genética , Giro do Cíngulo , Inibidores de Histona Desacetilases/farmacologia , Animais , Ansiedade/tratamento farmacológico , Ansiedade/metabolismo , Ansiedade/fisiopatologia , Ansiedade/reabilitação , Comportamento Animal/efeitos dos fármacos , Comportamento Animal/fisiologia , Benzamidas/farmacologia , Feminino , Giro do Cíngulo/efeitos dos fármacos , Giro do Cíngulo/metabolismo , Giro do Cíngulo/fisiopatologia , Histonas/efeitos dos fármacos , Histonas/metabolismo , Instinto , Masculino , Camundongos , Camundongos Endogâmicos , Estudo de Prova de Conceito , Piridinas/farmacologia
3.
Biol Psychiatry ; 81(12): 979-989, 2017 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-28104225

RESUMO

BACKGROUND: MicroRNA (miRNA)-mediated control of gene expression suggests that miRNAs are interesting targets and/or biomarkers in the treatment of anxiety- and trauma-related disorders, where often memory-associated gene expression is adversely affected. METHODS: The role of miRNAs in the rescue of impaired fear extinction was assessed using the 129S1/SvlmJ (S1) mouse model of impaired fear extinction. miRNA microarray analysis, reverse transcription polymerase chain reaction, fluorescent in situ hybridization, lentiviral overexpression, and Luciferase reporter assays were used to gain insight into the mechanisms underlying miRNA-mediated normalization of deficient fear extinction. RESULTS: Rescuing impaired fear extinction via dietary zinc restriction was associated with differential expression of miRNAs in the amygdala. One candidate, miR-144-3p, robustly expressed in the basolateral amygdala, showed specific extinction-induced, but not fear-induced, increased expression in both extinction-rescued S1 mice and extinction-intact C57BL/6 (BL6) mice. miR-144-3p upregulation and effects on subsequent behavioral adaption was assessed in S1 and BL6 mice. miR-144-3p overexpression in the basolateral amygdala rescued impaired fear extinction in S1 mice, led to enhanced fear extinction acquisition in BL6 mice, and furthermore protected against fear renewal in BL6 mice. miR-144-3p targets a number of genes implicated in the control of plasticity-associated signaling cascades, including Pten, Spred1, and Notch1. In functional interaction studies, we revealed that the miR-144-3p target, PTEN, colocalized with miR-144-3p in the basolateral amygdala and showed functional downregulation following successful fear extinction in S1 mice. CONCLUSIONS: These findings identify a fundamental role of miR-144-3p in the rescue of impaired fear extinction and suggest this miRNA as a viable target in developing novel treatments for posttraumatic stress disorder and related disorders.


Assuntos
Extinção Psicológica/fisiologia , Medo , Memória/fisiologia , MicroRNAs/fisiologia , Tonsila do Cerebelo/metabolismo , Animais , Regulação para Baixo , Masculino , Camundongos , MicroRNAs/genética , PTEN Fosfo-Hidrolase/biossíntese , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/fisiologia , Transdução de Sinais/fisiologia , Regulação para Cima , Zinco/deficiência
4.
Elife ; 52016 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-27897969

RESUMO

SATB2 is a risk locus for schizophrenia and encodes a DNA-binding protein that regulates higher-order chromatin configuration. In the adult brain Satb2 is almost exclusively expressed in pyramidal neurons of two brain regions important for memory formation, the cerebral cortex and the CA1-hippocampal field. Here we show that Satb2 is required for key hippocampal functions since deletion of Satb2 from the adult mouse forebrain prevents the stabilization of synaptic long-term potentiation and markedly impairs long-term fear and object discrimination memory. At the molecular level, we find that synaptic activity and BDNF up-regulate Satb2, which itself binds to the promoters of coding and non-coding genes. Satb2 controls the hippocampal levels of a large cohort of miRNAs, many of which are implicated in synaptic plasticity and memory formation. Together, our findings demonstrate that Satb2 is critically involved in long-term plasticity processes in the adult forebrain that underlie the consolidation and stabilization of context-linked memory.


Assuntos
Regulação da Expressão Gênica , Hipocampo/fisiologia , Proteínas de Ligação à Região de Interação com a Matriz/metabolismo , Memória de Longo Prazo , MicroRNAs/biossíntese , Fatores de Transcrição/metabolismo , Animais , Técnicas de Inativação de Genes , Proteínas de Ligação à Região de Interação com a Matriz/genética , Camundongos , Camundongos Knockout , Fatores de Transcrição/genética
5.
Artigo em Inglês | MEDLINE | ID: mdl-26625894

RESUMO

BACKGROUND: Despite its success in treating specific anxiety disorders, the effect of exposure therapy is limited by problems with tolerability, treatment resistance, and fear relapse after initial response. The identification of novel drug targets facilitating fear extinction in clinically relevant animal models may guide improved treatment strategies for these disorders in terms of efficacy, acceleration of fear extinction, and return of fear. METHODS: The extinction-facilitating potential of neuropeptide S, D-cycloserine, and a benzodiazepine was investigated in extinction-impaired high anxiety HAB rats and 129S1/SvImJ mice using a classical cued fear conditioning paradigm followed by extinction training and several extinction test sessions to study fear relapse. RESULTS: Administration of D-cycloserine improved fear extinction in extinction-limited, but not in extinction-deficient, rodents compared with controls. Preextinction neuropeptide S caused attenuated fear responses in extinction-deficient 129S1/SvImJ mice at extinction training onset and further reduced freezing during this session. While the positive effects of either D-cycloserine or neuropeptide S were not persistent in 129S1/SvImJ mice after 10 days, the combination of preextinction neuropeptide S with postextinction D-cycloserine rendered the extinction memory persistent and context independent up to 5 weeks after extinction training. This dual pharmacological adjunct to extinction learning also protected against fear reinstatement in 129S1/SvImJ mice. CONCLUSIONS: By using the potentially nonsedative anxiolytic neuropeptide S and the cognitive enhancer D-cycloserine to facilitate deficient fear extinction, we provide here the first evidence of a purported efficacy of a dual over a single drug approach. This approach may render exposure sessions less aversive and more efficacious for patients, leading to enhanced protection from fear relapse in the long term.


Assuntos
Ansiolíticos/farmacologia , Ansiedade/terapia , Comportamento Animal/efeitos dos fármacos , Ciclosserina/farmacologia , Extinção Psicológica/efeitos dos fármacos , Medo/efeitos dos fármacos , Terapia Implosiva/métodos , Neuropeptídeos/farmacologia , Animais , Ansiedade/genética , Ansiedade/psicologia , Cognição/efeitos dos fármacos , Condicionamento Clássico/efeitos dos fármacos , Modelos Animais de Doenças , Quimioterapia Combinada , Masculino , Camundongos da Linhagem 129 , Ratos Endogâmicos , Recidiva , Fatores de Tempo
6.
Front Behav Neurosci ; 9: 313, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26635563

RESUMO

Successful attenuation of fearful memories is a cognitive process requiring initiation of highly coordinated transcription programs. Chromatin-modulating mechanisms such as DNA methylation and histone modifications, including acetylation, are key regulators of these processes. However, knowledge concerning the role of ATP-dependent chromatin remodeling factors (ChRFs) being required for successful fear extinction is lacking. Underscoring the potential importance of these factors that alter histone-DNA contacts within nucleosomes are recent genome-wide association studies linking several ChRFs to various human cognitive and psychiatric disorders. To better understand the role of ChRFs in the brain, and since to date little is known about ChRF expression in the brain, we performed a comprehensive survey of expression levels of 24 ATP-dependent remodelers across different brain areas, and we identified several distinct high molecular weight complexes by chromatographic methods. We next aimed to gain novel insight into the potential regulation of ChRFs in different brain regions in association with normal and impaired fear extinction learning. To this end, we established the 129S1/SvImJ (S1) laboratory mouse strain as a model for compromised contextual fear extinction learning that can be rescued by dietary zinc restriction (ZnR). Using this model along with genetically related but fear extinction-competent 129S6/SvEv (S6) mice as controls, we found that impaired fear extinction in S1 was associated with enhanced ventral hippocampal expression of CHD1 and reduced expression of CHD5 that was normalized following successful rescue of impaired fear extinction. Moreover, a select reduction in CHD3 expression was observed in the ventral hippocampus (vHC) following successful rescue of fear extinction in S1 mice. Taken together, these data provide novel insight into the regulation of specific ChRFs following an impaired cognitive process and its rescue, and they suggest that imbalance of CHD-type remodeler levels, which consequently may lead to changes of transcriptional programs, may be an underlying mechanism involved in impaired fear extinction learning and its therapeutic rescue.

7.
Sci Adv ; 1(6)2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26504902

RESUMO

Persistent anxiety after a psychological trauma is a hallmark of many anxiety disorders. However, the neural circuits mediating the extinction of traumatic fear memories remain incompletely understood. We show that selective, in vivo stimulation of the ventromedial prefrontal cortex (vmPFC)-amygdala pathway facilitated extinction memory formation, but not retrieval. Conversely, silencing the vmPFC-amygdala pathway impaired extinction formation and reduced extinction-induced amygdala activity. Our data demonstrate a critical instructional role for the vmPFC-amygdala circuit in the formation of extinction memories. These findings advance our understanding of the neural basis of persistent fear, with implications for posttraumatic stress disorder and other anxiety disorders.

8.
Brain Struct Funct ; 220(4): 2209-21, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24807818

RESUMO

Reduced daily intake of magnesium (Mg(2+)) is suggested to contribute to depression. Indeed, preclinical studies show dietary magnesium restriction (MgR) elicits enhanced depression-like behaviour establishing a causal relationship. Amongst other mechanisms, Mg(2+) gates the activity of N-methyl-D-asparte (NMDA) receptors; however, it is not known whether reduced dietary Mg(2+) intake can indeed affect brain NMDA receptor complexes. Thus, the aim of the current study was to reveal whether MgR induces changes in brain NMDA receptor subunit composition that would indicate altered NMDA receptor regulation. The results revealed that enhanced depression-like behaviour elicited by MgR was associated with reduced amygdala-hypothalamic protein levels of GluN1-containing NMDA complexes. No change in GluN1 mRNA levels was observed indicating posttranslational changes were induced by dietary Mg(2+) restriction. To reveal possible protein interaction partners, GluN1 immunoprecipitation and proximity ligation assays were carried out revealing the expected GluN1 subunit association with GluN2A, GluN2B, but also novel interactions with GluA1, GluA2 in addition to known downstream signalling proteins. Chronic paroxetine treatment in MgR mice normalized enhanced depression-like behaviour, but did not alter protein levels of GluN1-containing NMDA receptors, indicating targets downstream of the NMDA receptor. Collectively, present data demonstrate that dietary MgR alters brain levels of GluN1-containing NMDA receptor complexes, containing GluN2A, GluN2B, AMPA receptors GluA1, GluA2 and several protein kinases. These data indicate that the modulation of dietary Mg(2+) intake may alter the function and signalling of this receptor complex indicating its involvement in the enhanced depression-like behaviour elicited by MgR.


Assuntos
Tonsila do Cerebelo/metabolismo , Depressão/complicações , Hipotálamo/metabolismo , Deficiência de Magnésio , Magnésio/efeitos adversos , Proteínas do Tecido Nervoso/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Análise de Variância , Animais , Depressão/tratamento farmacológico , Dieta/efeitos adversos , Modelos Animais de Doenças , Regulação da Expressão Gênica/fisiologia , Magnésio/metabolismo , Deficiência de Magnésio/complicações , Deficiência de Magnésio/etiologia , Deficiência de Magnésio/patologia , Masculino , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/genética , Paroxetina/uso terapêutico , Receptores de N-Metil-D-Aspartato/genética , Inibidores Seletivos de Recaptação de Serotonina/uso terapêutico
10.
Biochem Soc Trans ; 42(2): 569-81, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24646280

RESUMO

A novel strategy to treat anxiety and fear-related disorders such as phobias, panic and PTSD (post-traumatic stress disorder) is combining CBT (cognitive behavioural therapy), including extinction-based exposure therapy, with cognitive enhancers. By targeting and boosting mechanisms underlying learning, drug development in this field aims at designing CBT-augmenting compounds that help to overcome extinction learning deficits, promote long-term fear inhibition and thus support relapse prevention. Progress in revealing the role of epigenetic regulation of specific genes associated with extinction memory generation has opened new avenues in this direction. The present review examines recent evidence from pre-clinical studies showing that increasing histone acetylation, either via genetic or pharmacological inhibition of HDACs (histone deacetylases) by e.g. vorinostat/SAHA (suberoylanilide hydroxamic acid), entinostat/MS-275, sodium butyrate, TSA (trichostatin A) or VPA (valproic acid), or by targeting HATs (histone acetyltransferases), augments fear extinction and, importantly, generates a long-term extinction memory that can protect from return of fear phenomena. The molecular mechanisms and pathways involved including BDNF (brain-derived neurotrophic factor) and NMDA (N-methyl-D-aspartate) receptor signalling are just beginning to be revealed. First studies in healthy humans are in support of extinction-facilitating effects of HDAC inhibitors. Very recent evidence that HDAC inhibitors can rescue deficits in extinction-memory-impaired rodents indicates a potential clinical utility of this approach also for exposure therapy-resistant patients. Important future work includes investigation of the long-term safety aspects of HDAC inhibitor treatment, as well as design of isotype(s)-specific inhibitors. Taken together, HDAC inhibitors display promising potential as pharmacological adjuncts to augment the efficacy of exposure-based approaches in anxiety and trauma therapy.


Assuntos
Ansiedade/tratamento farmacológico , Medo/efeitos dos fármacos , Inibidores de Histona Desacetilases/uso terapêutico , Nootrópicos/uso terapêutico , Humanos , Ferimentos e Lesões/tratamento farmacológico
11.
Neurobiol Learn Mem ; 113: 69-81, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24231425

RESUMO

The neural circuitry mediating fear extinction has been increasingly well studied and delineated. The rodent infralimbic subregion (IL) of the ventromedial prefrontal cortex (vmPFC) has been found to promote extinction, whereas the prelimbic cortex (PL) demonstrates an opposing, pro-fear, function. Studies employing in vivo electrophysiological recordings have observed that while increased IL single-unit firing and bursting predicts robust extinction retrieval, increased PL firing can correlate with sustained fear and poor extinction. These relationships between single-unit firing and extinction do not hold under all experimental conditions, however. In the current study, we further investigated the relationship between vmPFC and PL single-unit firing and extinction using inbred mouse models of intact (C57BL/6J, B6) and deficient (129S1/SvImJ, S1) extinction strains. Simultaneous single-unit recordings were made in the PL and vmPFC (encompassing IL) as B6 and S1 mice performed extinction training and retrieval. Impaired extinction retrieval in S1 mice was associated with elevated PL single-unit firing, as compared to firing in extinguishing B6 mice, consistent with the hypothesized pro-fear contribution of PL. Analysis of local field potentials also revealed significantly higher gamma power in the PL of S1 than B6 mice during extinction training and retrieval. In the vmPFC, impaired extinction in S1 mice was also associated with exaggerated single-unit firing, relative to B6 mice. This is in apparent contradiction to evidence that IL activity promotes extinction, but could reflect a (failed) compensatory effort by the vmPFC to mitigate fear-promoting activity in other regions, such as the PL or amygdala. In support of this hypothesis, augmenting IL activity via direct infusion of the GABAA receptor antagonist picrotoxin rescued impaired extinction retrieval in S1 mice. Chronic fluoxetine treatment produced modest reductions in fear during extinction retrieval and increased the number of Zif268-labeled cells in layer II of IL, but failed to increase vmPFC single-unit firing. Collectively, these findings further support the important contribution these cortical regions play in determining the balance between robust extinction on the one hand, and sustained fear on the other. Elucidating the precise nature of these roles could help inform understanding of the pathophysiology of fear-related anxiety disorders.


Assuntos
Extinção Psicológica/fisiologia , Medo/fisiologia , Fluoxetina/farmacologia , Antagonistas GABAérgicos/farmacologia , Picrotoxina/farmacologia , Córtex Pré-Frontal/fisiologia , Inibidores Seletivos de Recaptação de Serotonina/farmacologia , Animais , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Extinção Psicológica/efeitos dos fármacos , Medo/efeitos dos fármacos , Fluoxetina/administração & dosagem , Antagonistas GABAérgicos/administração & dosagem , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Técnicas de Patch-Clamp/métodos , Picrotoxina/administração & dosagem , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/fisiopatologia , Inibidores Seletivos de Recaptação de Serotonina/administração & dosagem
12.
Biol Mood Anxiety Disord ; 3(1): 13, 2013 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-23830244

RESUMO

BACKGROUND: Various neuropsychiatric conditions, including posttraumatic stress disorder (PTSD), are characterized by deficient fear extinction, but individuals differ greatly in risk for these. While there is growing evidence that fear extinction is influenced by certain procedural variables, it is unclear how these influences might vary across individuals and subpopulations. To model individual differences in fear extinction, prior studies identified a strain of inbred mouse, 129S1/SvImJ (S1), which exhibits a profound deficit in fear extinction, as compared to other inbred strains, such as C57BL/6J (B6). METHODS: Here, we assessed the effects of procedural variables on the impaired extinction phenotype of the S1 strain and, by comparison, the extinction-intact B6 strain. The variables studied were 1) the interval between conditioning and extinction, 2) the interval between cues during extinction training, 3) single-cue exposure before extinction training, and 4) extinction of a second-order conditioned cue. RESULTS: Conducting extinction training soon after ('immediately') conditioning attenuated fear retrieval in S1 mice and impaired extinction in B6 mice. Spacing cue presentations with long inter-trial intervals during extinction training augmented fear in S1 and B6 mice. The effect of spacing was lost with one-trial fear conditioning in B6, but not S1 mice. A single exposure to a conditioned cue before extinction training did not alter extinction retrieval, either in B6 or S1 mice. Both the S1 and B6 strains exhibited robust second-order fear conditioning, in which a cue associated with footshock was sufficient to serve as a conditioned exciter to condition a fear association to a second cue. B6 mice extinguished the fear response to the second-order conditioned cue, but S1 mice failed to do so. CONCLUSIONS: These data provide further evidence that fear extinction is strongly influenced by multiple procedural variables and is so in a highly strain-dependent manner. This suggests that the efficacy of extinction-based behavioral interventions, such as exposure therapy, for trauma-related anxiety disorders will be determined by the procedural parameters employed and the degree to which the patient can extinguish.

13.
Neuropharmacology ; 64: 414-23, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22722028

RESUMO

Anxiety disorders are characterized by persistent, excessive fear. Therapeutic interventions that reverse deficits in fear extinction represent a tractable approach to treating these disorders. We previously reported that 129S1/SvImJ (S1) mice show no extinction learning following normal fear conditioning. We now demonstrate that weak fear conditioning does permit fear reduction during massed extinction training in S1 mice, but reveals specific deficiency in extinction memory consolidation/retrieval. Rescue of this impaired extinction consolidation/retrieval was achieved with d-cycloserine (N-methly-d-aspartate partial agonist) or MS-275 (histone deacetylase (HDAC) inhibitor), applied after extinction training. We next examined the ability of different drugs and non-pharmacological manipulations to rescue the extreme fear extinction deficit in S1 following normal fear conditioning with the ultimate aim to produce low fear levels in extinction retrieval tests. Results showed that deep brain stimulation (DBS) by applying high frequency stimulation to the nucleus accumbens (ventral striatum) during extinction training, indeed significantly reduced fear during extinction retrieval compared to sham stimulation controls. Rescue of both impaired extinction acquisition and deficient extinction consolidation/retrieval was achieved with prior extinction training administration of valproic acid (a GABAergic enhancer and HDAC inhibitor) or AMN082 [metabotropic glutamate receptor 7 (mGlu7) agonist], while MS-275 or PEPA (AMPA receptor potentiator) failed to affect extinction acquisition in S1 mice. Collectively, these data identify potential beneficial effects of DBS and various drug treatments, including those with HDAC inhibiting or mGlu7 agonism properties, as adjuncts to overcome treatment resistance in exposure-based therapies. This article is part of a Special Issue entitled 'Cognitive Enhancers'.


Assuntos
Transtornos de Ansiedade/terapia , Estimulação Encefálica Profunda , Modelos Animais de Doenças , Agonistas de Aminoácidos Excitatórios/uso terapêutico , Extinção Psicológica , Agonistas GABAérgicos/uso terapêutico , Inibidores de Histona Desacetilases/uso terapêutico , Animais , Ansiolíticos/uso terapêutico , Transtornos de Ansiedade/tratamento farmacológico , Compostos Benzidrílicos/uso terapêutico , Extinção Psicológica/efeitos dos fármacos , Medo/efeitos dos fármacos , Masculino , Camundongos , Camundongos da Linhagem 129 , Terapia de Alvo Molecular , Nootrópicos/uso terapêutico , Núcleo Accumbens , Distribuição Aleatória , Receptores de Glutamato Metabotrópico/agonistas , Receptores de Glutamato Metabotrópico/metabolismo , Ácido Valproico/uso terapêutico
14.
J Neurosci ; 31(13): 5131-44, 2011 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-21451049

RESUMO

Although extinction-based therapies are among the most effective treatments for anxiety disorders, the neural bases of fear extinction remain still essentially unclear. Recent evidence suggests that the intercalated cell masses of the amygdala (ITCs) are critical structures for fear extinction. However, the neuronal organization of ITCs and how distinct clusters contribute to different fear states are still entirely unknown. Here, by combining whole-cell patch-clamp recordings and biocytin labeling with full anatomical reconstruction of the filled neurons and ultrastructural analysis of their synaptic contacts, we have elucidated the cellular organization and efferent connections of one of the main ITC clusters in mice. Our data showed an unexpected heterogeneity in the axonal pattern of medial paracapsular ITC (Imp) neurons and the presence of three distinct neuronal subtypes. Functionally, we observed that the Imp was preferentially activated during fear expression, whereas extinction training and extinction retrieval activated the main ITC nucleus (IN), as measured by quantifying Zif268 expression. This can be explained by the IPSPs evoked in the IN after Imp stimulation, most likely through the GABAergic monosynaptic innervation of IN neurons by one subtype of Imp cells, namely the medial capsular-projecting (MCp)-Imp neurons. MCp-Imp neurons also target large ITC cells that surround ITC clusters and express the metabotropic glutamate receptor 1α. These findings reveal a distinctive participation of ITC clusters to different fear states and the underlying anatomical circuitries, hence shedding new light on ITC networks and providing a novel framework to elucidate their role in fear expression and extinction.


Assuntos
Tonsila do Cerebelo/fisiologia , Comunicação Celular/fisiologia , Medo/fisiologia , Interneurônios/fisiologia , Rede Nervosa/fisiologia , Tonsila do Cerebelo/citologia , Animais , Axônios/fisiologia , Axônios/ultraestrutura , Medo/psicologia , Interneurônios/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Rede Nervosa/citologia
15.
Amino Acids ; 40(4): 1231-48, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21312047

RESUMO

There is evidence to suggest that low levels of magnesium (Mg) are associated with affective disorders, however, causality and central neurobiological mechanisms of this link are largely unproven. We have recently shown that mice fed a low Mg-containing diet (10% of daily requirement) display enhanced depression-like behavior sensitive to chronic antidepressant treatment. The aim of the present study was to utilize this model to gain insight into underlying mechanisms by quantifying amygdala/hypothalamus protein expression using gel-based proteomics and correlating changes in protein expression with changes in depression-like behavior. Mice fed Mg-restricted diet displayed reduced brain Mg tissue levels and altered expression of four proteins, N(G),N(G)-dimethylarginine dimethylaminohydrolase 1 (DDAH1), manganese-superoxide dismutase (MnSOD), glutamate dehydrogenase 1 (GDH1) and voltage-dependent anion channel 1. The observed alterations in protein expression may indicate increased nitric oxide production, increased anti-oxidant response to increased oxidative stress and potential alteration in energy metabolism. Aberrant expressions of DDAH1, MnSOD and GDH1 were normalized by chronic paroxetine treatment which also normalized the enhanced depression-like behavior, strengthening the link between the changes in these proteins and depression-like behavior. Collectively, these findings provide first evidence of low magnesium-induced alteration in brain protein levels and biochemical pathways, contributing to central dysregulation in affective disorders.


Assuntos
Amidoidrolases/metabolismo , Depressão/genética , Glutamato Desidrogenase/metabolismo , Superóxido Dismutase/metabolismo , Canal de Ânion 1 Dependente de Voltagem/metabolismo , Amidoidrolases/genética , Tonsila do Cerebelo/enzimologia , Tonsila do Cerebelo/fisiopatologia , Animais , Antidepressivos/administração & dosagem , Antidepressivos/uso terapêutico , Depressão/tratamento farmacológico , Depressão/etiologia , Depressão/metabolismo , Depressão/fisiopatologia , Dieta/efeitos adversos , Eletroforese em Gel Bidimensional , Perfilação da Expressão Gênica , Glutamato Desidrogenase/genética , Hipotálamo/enzimologia , Hipotálamo/fisiopatologia , Deficiência de Magnésio/complicações , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico/biossíntese , Estresse Oxidativo , Paroxetina/administração & dosagem , Paroxetina/uso terapêutico , Proteômica , Superóxido Dismutase/genética , Espectrometria de Massas em Tandem , Canal de Ânion 1 Dependente de Voltagem/genética
16.
J Neurosci ; 30(41): 13586-96, 2010 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-20943900

RESUMO

Fear extinction is impaired in neuropsychiatric disorders, including posttraumatic stress disorder. Identifying drugs that facilitate fear extinction in animal models provides leads for novel pharmacological treatments for these disorders. Zinc (Zn) is expressed in neurons in a cortico-amygdala circuit mediating fear extinction, and modulates neurotransmitter systems regulating extinction. We previously found that the 129S1/SvImJ mouse strain (S1) exhibited a profound impairment in fear extinction, coupled with abnormalities in the activation of the extinction circuit. Here, we tested the role of Zn in fear extinction in S1 and C57BL/6N reference strain (B6) by feeding the mice a Zn-restricted diet (ZnR) and testing for fear extinction, as well as neuronal activation of the extinction circuit via quantification of the immediate-early genes c-Fos and Zif268. Results showed that (preconditioning or postconditioning) ZnR completely rescued deficient extinction learning and long-term extinction retrieval in S1 and expedited extinction learning in B6, without affecting fear acquisition or fear expression. The extinction-facilitating effects of ZnR were associated with the normalization of Zif268 and/or c-Fos expression in cortico-amygdala regions of S1. Specifically, ZnR increased activity in infralimbic cortex, lateral and basolateral amygdala nuclei, and lateral central amygdala nucleus, and decreased activity in prelimbic and insular cortices and medial central amygdala nucleus. ZnR also increased activation in the main intercalated nucleus and decreased activation of the medial paracapsular intercalated mass in S1. Our findings reveal a novel role for Zn in fear extinction and further support the utility of the S1 model for identifying extinction facilitating drugs.


Assuntos
Tonsila do Cerebelo/fisiopatologia , Córtex Cerebral/fisiopatologia , Extinção Psicológica/fisiologia , Medo/fisiologia , Rede Nervosa/fisiopatologia , Zinco/administração & dosagem , Tonsila do Cerebelo/metabolismo , Análise de Variância , Animais , Córtex Cerebral/metabolismo , Condicionamento Psicológico/fisiologia , Reação de Congelamento Cataléptica/fisiologia , Camundongos , Camundongos Transgênicos , Rede Nervosa/metabolismo , Neurônios/fisiologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Zinco/metabolismo
17.
Toxicon ; 56(5): 668-73, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19538985

RESUMO

Ciguatera is a food poisoning identified as the principal risk factor in the consumption of tropical fish in Oceania. The syndrome, which follows ingestion of ciguatoxin-contaminated ciguateric fishes, is characterised by an array of gastrointestinal and neurological features. In this report we examine forensic samples associated with a human fatality using a (3)H-brevetoxin binding assay and reversed-phase HPLC/MS and HPLC/MS/MS. Three Pacific ciguatoxins (P-CTX) were detected in the implicated fish flesh sample by LC-MS/MS, implicating multiple P-CTXs in the fatal case. Additionally, ciguatoxin was identified in a liver sample obtained at post-mortem. The level of ciguatoxin detected (0.14 ppb P-CTX-1 equivalents by binding assay) indicated that at least 10% of the ingested P-CTX-1 remained in the human liver 6 days after the toxic fish was consumed. This study confirms the potential of tropical reef fish to accumulate sufficient P-CTX to be lethal to humans, especially if the liver and viscera are consumed as part of the meal.


Assuntos
Ciguatera/mortalidade , Animais , Cromatografia Líquida de Alta Pressão , Humanos , Espectrometria de Massas/métodos , Oceano Pacífico , Ensaio Radioligante
18.
Neuropharmacology ; 57(5-6): 556-66, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19607848

RESUMO

The serotonin(1A) receptor (5-HT(1A) R) knock-out mouse (KO) is a widely used animal model for anxiety and cognitive function and regulation of signaling cascades by this receptor has been reported. We aimed to determine individual representatives of signaling cascades in order to screen 5-HT(1A) R-dependent signaling proteins (SPs). Hippocampal proteins from wild type and 5-HT(1A) R KO mice were extracted, run on two-dimensional gel electrophoresis, proteins were identified by MALDI and nano-ESI-LC-MS/MS and SPs were quantified by specific software. Nucleoside diphosphate kinase A (NDK A, synonym: nm23), Dual specificity mitogen-activated protein kinase kinase 1 (MAPKK1, synonym: MEK), Serine/threonine-protein phosphatase PP1-gamma catalytic subunit (PP-1G), Septin-5, were reduced in the KO mice. Novel phosphorylation sites at T386 on MAPKK1 and at S225 and Y265 on Septin-5 were observed. MAPKK1 and PP-1G are known 5-HT(1A) R-dependent signaling compounds and are in agreement with receptor knock-out and septin-5 is involved in serotonin transport, although regulation by 5-HT(1A) R has not been reported. 5-HT(1A) R - dependent levels for NDK A have not been demonstrated so far and we herewith propose a role for NDK A in 5-HT(1A) R signaling. Reduced SP levels along with findings of two novel phosphorylation sites may be relevant for interpretation of previous and the design of future studies on this receptor system.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Hipocampo/metabolismo , MAP Quinase Quinase 1/metabolismo , Nucleosídeo NM23 Difosfato Quinases/metabolismo , Proteína Fosfatase 1/metabolismo , Receptor 5-HT1A de Serotonina/metabolismo , Sequência de Aminoácidos , Análise de Variância , Animais , Western Blotting , Proteínas do Citoesqueleto/genética , Bases de Dados Genéticas , Eletroforese em Gel Bidimensional , Proteínas de Ligação ao GTP/genética , MAP Quinase Quinase 1/genética , Camundongos , Camundongos Knockout , Fosforilação , Receptor 5-HT1A de Serotonina/genética , Septinas , Transdução de Sinais , Software , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
19.
J Neurosci ; 28(32): 8074-85, 2008 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-18685032

RESUMO

Fear extinction is a form of new learning that results in the inhibition of conditioned fear. Trait deficits in fear extinction are a risk factor for anxiety disorders. There are few examples of naturally occurring animal models of impaired extinction. The present study compared fear extinction in a panel of inbred mouse strains. This strain survey revealed an impairment in fear extinction in 129/SvImJ (129S1). The phenotypic specificity of this deficit was evaluated by comparing 129S1 and C57BL/6J for one-trial and multitrial fear conditioning, nociception, and extinction of conditioned taste aversion and an appetitive instrumental response. 129S1 were tested for sensitivity to the extinction-facilitating effects of extended training, as well as d-cycloserine and yohimbine treatment. To elucidate the neural basis of impaired 129S1 fear extinction, c-Fos and Zif268 expression was mapped after extinction recall. Results showed that impaired fear extinction in 129S1 was unrelated to altered fear conditioning or nociception, and was dissociable from intact appetitive extinction. Yohimbine treatment facilitated extinction in 129S1, but neither extended extinction training nor d-cycloserine treatment improved 129S1 extinction. After extinction recall, 129S1 showed reduced c-Fos and Zif268 expression in the infralimbic cortex and basolateral amygdala, and elevated c-Fos or Zif268 expression in central nucleus of the amygdala and medial paracapsular intercalated cell mass, relative to C57BL/6J. Collectively, these data demonstrate a deficit in fear extinction in 129S1 associated with a failure to properly engage corticolimbic extinction circuitry. This common inbred strain provides a novel model for studying impaired fear extinction in anxiety disorders.


Assuntos
Tonsila do Cerebelo/anormalidades , Extinção Psicológica , Medo , Aprendizagem , Malformações do Sistema Nervoso/psicologia , Córtex Pré-Frontal/anormalidades , Animais , Aprendizagem da Esquiva , Comportamento Animal , Condicionamento Psicológico , Ciclosserina/farmacologia , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Medo/efeitos dos fármacos , Interneurônios/metabolismo , Masculino , Rememoração Mental , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Malformações do Sistema Nervoso/genética , Malformações do Sistema Nervoso/metabolismo , Malformações do Sistema Nervoso/fisiopatologia , Vias Neurais/anormalidades , Nociceptores , Proteínas Proto-Oncogênicas c-fos/metabolismo , Paladar , Ioimbina/farmacologia
20.
Neuropsychopharmacology ; 33(8): 1929-41, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17957216

RESUMO

Antagonists of the substance P (SP) preferring neurokinin 1 receptor (NK1R) represent a promising novel class of drugs for the treatment of stress-related disorders such as depression and anxiety disorders; however, the involved neuronal pathways releasing SP in response to stressors are ill defined. By using in vivo microdialysis in combination with a highly sensitive and selective radioimmunoassay we found that exposure to forced swim stress increased SP release in the rat lateral septum (LS), a key area in processing emotions and stress responses. Acute administration of the selective NK1R antagonist L-822429 injected either systemically or locally into the LS reduced passive and facilitated active stress-coping strategies in the forced swim test. This effect seems to be mediated by enhanced intraseptal serotonergic transmission via serotonin (5-HT)1A receptors since NK1R blockade reversed the swim stress-induced decrease to an increase in extracellular 5-HT efflux, and furthermore the behavioral effects of L-822429 were blocked by intraseptal 5-HT1A receptor antagonism. A direct heterosynaptic regulation by NK1R on 5-HT release from serotonergic fibers was ruled out by immunocytochemistry at the light and electron microscopic level indicating involvement of GABAergic interneuron(s) in this interaction. Taken together, our data identify the LS as a critical brain area for the involvement of SP transmission in the modulation of stress responses and demonstrate that NK1R blockade can elicit a functionally significant facilitatory effect on 5-HT transmission, which does not necessarily involve the previously proposed interaction with neuronal firing at the cell body level of raphe neurons.


Assuntos
Adaptação Psicológica/efeitos dos fármacos , Antagonistas dos Receptores de Neurocinina-1 , Septo do Cérebro/fisiologia , Serotonina/fisiologia , Estresse Psicológico/tratamento farmacológico , Estresse Psicológico/psicologia , Transmissão Sináptica/efeitos dos fármacos , Animais , Comportamento Animal/efeitos dos fármacos , Imuno-Histoquímica , Masculino , Microdiálise , Microscopia Eletrônica , Neurônios Aferentes/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Receptor 5-HT1A de Serotonina/efeitos dos fármacos , Septo do Cérebro/efeitos dos fármacos , Serotonina/metabolismo , Estereoisomerismo , Substância P/metabolismo , Substância P/fisiologia , Natação/psicologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA