Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Neurobiol Learn Mem ; 107: 93-100, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24291571

RESUMO

We have previously shown that auditory Pavlovian fear conditioning is associated with an increase in DNA methyltransferase (DNMT) expression in the lateral amygdala (LA) and that intra-LA infusion or bath application of an inhibitor of DNMT activity impairs the consolidation of an auditory fear memory and long-term potentiation (LTP) at thalamic and cortical inputs to the LA, in vitro. In the present study, we use awake behaving neurophysiological techniques to examine the role of DNMT activity in memory-related neurophysiological changes accompanying fear memory consolidation and reconsolidation in the LA, in vivo. We show that auditory fear conditioning results in a training-related enhancement in the amplitude of short-latency auditory-evoked field potentials (AEFPs) in the LA. Intra-LA infusion of a DNMT inhibitor impairs both fear memory consolidation and, in parallel, the consolidation of training-related neural plasticity in the LA; that is, short-term memory (STM) and short-term training-related increases in AEFP amplitude in the LA are intact, while long-term memory (LTM) and long-term retention of training-related increases in AEFP amplitudes are impaired. In separate experiments, we show that intra-LA infusion of a DNMT inhibitor following retrieval of an auditory fear memory has no effect on post-retrieval STM or short-term retention of training-related changes in AEFP amplitude in the LA, but significantly impairs both post-retrieval LTM and long-term retention of AEFP amplitude changes in the LA. These findings are the first to demonstrate the necessity of DNMT activity in the consolidation and reconsolidation of memory-associated neural plasticity, in vivo.


Assuntos
Tonsila do Cerebelo/enzimologia , Tonsila do Cerebelo/fisiologia , Metilases de Modificação do DNA/fisiologia , Memória/fisiologia , Plasticidade Neuronal/fisiologia , Estimulação Acústica , Animais , Azacitidina/análogos & derivados , Azacitidina/farmacologia , Metilases de Modificação do DNA/antagonistas & inibidores , Decitabina , Inibidores Enzimáticos/farmacologia , Medo/fisiologia , Masculino , Memória/efeitos dos fármacos , Rememoração Mental/efeitos dos fármacos , Rememoração Mental/fisiologia , Plasticidade Neuronal/efeitos dos fármacos , Ftalimidas/farmacologia , Ratos , Ratos Sprague-Dawley , Triptofano/análogos & derivados , Triptofano/farmacologia
2.
Learn Mem ; 20(2): 109-19, 2013 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-23328899

RESUMO

Modifications in chromatin structure have been widely implicated in memory and cognition, most notably using hippocampal-dependent memory paradigms including object recognition, spatial memory, and contextual fear memory. Relatively little is known, however, about the role of chromatin-modifying enzymes in amygdala-dependent memory formation. Here, we use a combination of biochemical, behavioral, and neurophysiological methods to systematically examine the role of p300/CBP histone acetyltransferase (HAT) activity in the consolidation and reconsolidation of auditory Pavlovian fear memories. We show that local infusions of c646, a selective pharmacological inhibitor of p300/CBP activity, shortly following either fear conditioning or fear memory retrieval impair training and retrieval-related regulation of histone acetylation in the lateral nucleus of the amygdala (LA). Furthermore, we show that intra-LA infusion of c646 significantly impairs fear memory consolidation, reconsolidation, and associated neural plasticity in the LA. Our findings collectively suggest that p300/CBP HAT activity is critical for the consolidation and reconsolidation of amygdala-dependent Pavlovian fear memories.


Assuntos
Tonsila do Cerebelo/fisiologia , Condicionamento Clássico/fisiologia , Medo , Memória/fisiologia , Fatores de Transcrição de p300-CBP/metabolismo , Acetilação/efeitos dos fármacos , Estimulação Acústica , Tonsila do Cerebelo/efeitos dos fármacos , Análise de Variância , Animais , Condicionamento Clássico/efeitos dos fármacos , Eletrodos Implantados , Eletrochoque , Inibidores Enzimáticos/efeitos adversos , Inibidores Enzimáticos/farmacologia , Potenciais Evocados Auditivos/efeitos dos fármacos , Potenciais Evocados Auditivos/fisiologia , Masculino , Memória/efeitos dos fármacos , Transtornos da Memória/induzido quimicamente , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
3.
J Neurosci ; 31(19): 7073-82, 2011 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-21562269

RESUMO

The activity-regulated cytoskeletal-associated protein (Arc/Arg3.1) is an immediate-early gene that has been widely implicated in synaptic plasticity and in the consolidation of a variety of hippocampal- and amygdala-dependent memory tasks. The functional role of Arc/Arg3.1 in memory reconsolidation processes, however, has not been systematically studied. In the present study, we examined the role of Arc/Arg3.1 in the reconsolidation of an amygdala-dependent auditory pavlovian fear memory. We show that Arc/Arg3.1 protein is regulated in the lateral nucleus of the amygdala (LA) by retrieval of an auditory fear memory. Next, we show that antisense knockdown of Arc/Arg3.1 in the LA impairs fear memory reconsolidation of both a recent (1-d-old) as well as a well-consolidated (2-week-old) fear memory; that is, post-retrieval short-term memory, tested at 3 h after retrieval, is intact, whereas post-retrieval long-term memory, tested approximately 24 h after retrieval, is significantly impaired. The effect of Arc/Arg3.1 knockdown was observed to be time limited and specific to an actively reactivated fear memory. Moreover, the reconsolidation deficit induced by Arc/Arg3.1 knockdown was not found to be sensitive to spontaneous recovery, reinstatement, or a shift in the testing context, suggesting that our behavioral effects are not attributable to facilitated extinction. Collectively, our findings provide the first comprehensive look at the functional role of Arc/Arg3.1 in memory reconsolidation processes in the mammalian brain.


Assuntos
Tonsila do Cerebelo/metabolismo , Condicionamento Clássico/fisiologia , Proteínas do Citoesqueleto/metabolismo , Medo/fisiologia , Memória/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Análise de Variância , Animais , Comportamento Animal/efeitos dos fármacos , Western Blotting , Imuno-Histoquímica , Masculino , Ratos , Ratos Sprague-Dawley
4.
Learn Mem ; 18(1): 24-38, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21177377

RESUMO

The immediate-early gene early growth response gene-1 (EGR-1, zif-268) has been extensively studied in synaptic plasticity and memory formation in a variety of memory systems. However, a convincing role for EGR-1 in amygdala-dependent memory consolidation processes has yet to emerge. In the present study, we have examined the role of EGR-1 in the consolidation and reconsolidation of amygdala-dependent auditory Pavlovian fear conditioning. In our first series of experiments, we show that EGR-1 is regulated following auditory fear conditioning in the lateral nucleus of the amygdala (LA). Next, we use antisense oligodeoxynucleotide (ODN) knockdown of EGR-1 in the LA to show that training-induced expression of EGR-1 is required for memory consolidation of auditory fear conditioning; that is, long-term memory (LTM) is significantly impaired while acquisition and short-term memory (STM) are intact. In a second set of experiments, we show that EGR-1 is regulated in the LA by retrieval of an auditory fear memory. We then show that retrieval-induced expression of EGR-1 in the LA is required for memory reconsolidation of auditory fear conditioning; that is, post-retrieval (PR)-LTM is significantly impaired while memory retrieval and PR-STM are intact. Additional experiments show these effects to be restricted to the LA, to be temporally graded, and unlikely to be due to nonspecific toxicity within the LA. Collectively, our findings strongly implicate a role for EGR-1 in both the initial consolidation and in the reconsolidation of auditory fear memories in the LA.


Assuntos
Tonsila do Cerebelo/metabolismo , Condicionamento Clássico/fisiologia , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Medo , Memória/fisiologia , Estimulação Acústica/efeitos adversos , Tonsila do Cerebelo/efeitos dos fármacos , Animais , Aprendizagem por Associação/efeitos dos fármacos , Comportamento Animal , Condicionamento Clássico/efeitos dos fármacos , Proteína 1 de Resposta de Crescimento Precoce/genética , Eletrochoque/efeitos adversos , Medo/efeitos dos fármacos , Masculino , Memória/efeitos dos fármacos , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Ratos , Ratos Sprague-Dawley , Tempo de Reação/efeitos dos fármacos , Fatores de Tempo
5.
Learn Mem ; 18(9): 579-93, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21868438

RESUMO

Epigenetic mechanisms have been widely implicated in synaptic plasticity and in memory consolidation, yet little is known about the role of epigenetic mechanisms in memory reconsolidation processes. In the present study, we systematically examine the role of histone acetylation and DNA methylation in the reconsolidation of an amygdala-dependent Pavlovian fear memory. We first show that the acetylation of histone 3 (H3), but not histone 4 (H4), is regulated following auditory fear memory retrieval in the lateral nucleus of the amygdala (LA). We next show that histone deacetylase (HDAC) inhibition in the LA enhances both retrieval-induced histone acetylation and reconsolidation of an auditory fear memory. Conversely, inhibition of DNA methytransferase (DNMT) activity in the LA significantly impairs both retrieval-related H3 acetylation and fear memory reconsolidation. The effects of HDAC and DNMT inhibitors on fear memory reconsolidation were observed to be time-limited and were not evident in the absence of memory reactivation. Further, memories lost following DNMT inhibition were not observed to be vulnerable to spontaneous recovery, reinstatement, or to a shift in testing context, suggesting that memory impairment was not the result of facilitated extinction. Finally, pretreatment with the HDAC inhibitor was observed to rescue the reconsolidation deficit induced by the DNMT inhibitor. These findings collectively suggest that histone acetylation and DNA methylation are critical for reconsolidation of fear memories in the LA.


Assuntos
Tonsila do Cerebelo/metabolismo , Condicionamento Clássico/fisiologia , Epigênese Genética/genética , Epigenômica , Medo/fisiologia , Histonas/metabolismo , Acetilação/efeitos dos fármacos , Estimulação Acústica/métodos , Tonsila do Cerebelo/efeitos dos fármacos , Animais , Condicionamento Clássico/efeitos dos fármacos , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA/efeitos dos fármacos , Relação Dose-Resposta a Droga , Eletrochoque/efeitos adversos , Inibidores Enzimáticos/farmacologia , Epigênese Genética/efeitos dos fármacos , Medo/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Histona Desacetilases/metabolismo , Histonas/genética , Masculino , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
6.
Nat Neurosci ; 10(4): 414-6, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17351634

RESUMO

When reactivated, memories enter a labile, protein synthesis-dependent state, a process referred to as reconsolidation. Here, we show in rats that fear memory retrieval produces a synaptic potentiation in the lateral amygdala that is selective to the reactivated memory, and that disruption of reconsolidation is correlated with a reduction of synaptic potentiation in the lateral amygdala. Thus, both retrieval and reconsolidation alter memories via synaptic plasticity at selectively targeted synapses.


Assuntos
Tonsila do Cerebelo/fisiologia , Condicionamento Clássico/fisiologia , Medo , Memória/fisiologia , Sinapses/fisiologia , Estimulação Acústica/métodos , Tonsila do Cerebelo/citologia , Tonsila do Cerebelo/efeitos dos fármacos , Análise de Variância , Animais , Comportamento Animal , Butadienos/farmacologia , Condicionamento Clássico/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Masculino , Memória/efeitos dos fármacos , Nitrilas/farmacologia , Ratos , Ratos Sprague-Dawley , Tempo de Reação/fisiologia , Tempo de Reação/efeitos da radiação , Sinapses/efeitos dos fármacos
7.
Learn Mem ; 17(4): 221-35, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20351057

RESUMO

We have recently hypothesized that NO-cGMP-PKG signaling in the lateral nucleus of the amygdala (LA) during auditory fear conditioning coordinately regulates ERK-driven transcriptional changes in both auditory thalamic (MGm/PIN) and LA neurons that serve to promote pre- and postsynaptic alterations at thalamo-LA synapses, respectively. In the present series of experiments, we show that N-methyl-D-aspartate receptor (NMDAR)-driven synaptic plasticity and NO-cGMP-PKG signaling in the LA regulate the training-induced expression of ERK and the ERK-driven immediate early genes (IEGs) Arc/Arg3.1, c-Fos, and EGR-1 in the LA and the MGm/PIN. Rats receiving intra-LA infusion of the NR2B selective antagonist Ifenprodil, the NOS inhibitor 7-Ni, or the PKG inhibitor Rp-8-Br-PET-cGMPS exhibited significant decreases in ERK activation and in the training-induced expression of all three IEGs in the LA and MGm/PIN while intra-LA infusion of the PKG activator 8-Br-cGMP had the opposite effect. Remarkably, those rats given intra-LA infusion of the membrane impermeable NO scavenger c-PTIO exhibited significant decreases in ERK activation and ERK-driven IEG expression in the MGm/PIN, but not in the LA. Together with our previous experiments, these results suggest that synaptic plasticity and the NO-cGMP-PKG signaling pathway promote fear memory consolidation, in part, by regulating ERK-driven transcription in both the LA and the MGm/PIN. They further suggest that synaptic plasticity in the LA during fear conditioning promotes ERK-driven transcription in MGm/PIN neurons via NO-driven "retrograde signaling."


Assuntos
Tonsila do Cerebelo/fisiologia , Condicionamento Clássico/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Medo , Regulação da Expressão Gênica/fisiologia , Plasticidade Neuronal/fisiologia , Transdução de Sinais/fisiologia , Tálamo/fisiologia , Estimulação Acústica/efeitos adversos , Tonsila do Cerebelo/efeitos dos fármacos , Animais , Comportamento Animal , Condicionamento Clássico/efeitos dos fármacos , GMP Cíclico/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Inibidores Enzimáticos/farmacologia , Antagonistas de Aminoácidos Excitatórios/farmacologia , Medo/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Modelos Biológicos , Plasticidade Neuronal/efeitos dos fármacos , Plasticidade Neuronal/genética , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Tálamo/efeitos dos fármacos , Fatores de Tempo
8.
J Neurochem ; 112(3): 636-50, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19912470

RESUMO

Most recent studies aimed at defining the cellular and molecular mechanisms of Pavlovian fear conditioning have focused on protein kinase signaling pathways and the transcription factor cAMP-response element binding protein (CREB) that promote fear memory consolidation in the lateral nucleus of the amygdala (LA). Despite this progress, there still remains a paucity of information regarding the genes downstream of CREB that are required for long-term fear memory formation in the LA. We have adopted a strategy of using microarray technology to initially identify genes induced within the dentate gyrus following in vivo long-term potentiation (LTP) followed by analysis of whether these same genes are also regulated by fear conditioning within the LA. In the present study, we first identified 34 plasticity-associated genes that are induced within 30 min following LTP induction utilizing a combination of DNA microarray, qRT-PCR, and in situ hybridization. To determine whether these genes are also induced in the LA following Pavlovian fear conditioning, we next exposed rats to an auditory fear conditioning protocol or to control conditions that do not support fear learning followed by qRT-PCR on mRNA from microdissected LA samples. Finally, we asked whether identified genes induced by fear learning in the LA are downstream of the extracellular-regulated kinase/mitogen-activated protein kinase signaling cascade. Collectively, our findings reveal a comprehensive list of genes that represent the first wave of transcription following both LTP induction and fear conditioning that largely belong to a class of genes referred to as 'neuronal activity dependent genes' that are likely calcium, extracellular-regulated kinase/mitogen-activated protein kinase, and CREB-dependent.


Assuntos
Tonsila do Cerebelo/metabolismo , Condicionamento Clássico/fisiologia , Medo , Regulação da Expressão Gênica/fisiologia , Potenciação de Longa Duração/fisiologia , Aminoacetonitrila/análogos & derivados , Aminoacetonitrila/farmacologia , Análise de Variância , Animais , Biofísica , Proteína de Ligação a CREB/genética , Proteína de Ligação a CREB/metabolismo , Giro Denteado/fisiologia , Estimulação Elétrica/métodos , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Microdissecção , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Inibidores de Proteases/farmacologia , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
9.
Neural Plast ; 2010: 540940, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21461354

RESUMO

Long-term potentiation (LTP) at thalamic input synapses to the lateral nucleus of the amygdala (LA) has been proposed as a cellular mechanism of the formation of auditory fear memories. We have previously shown that signaling via ERK/MAPK in both the LA and the medial division of the medial geniculate nucleus/posterior intralaminar nucleus (MGm/PIN) is critical for LTP at thalamo-LA synapses. Here, we show that LTP-inducing stimulation of thalamo-LA inputs regulates the activation of ERK and the expression of ERK-driven immediate early genes (IEGs) in both the LA and MGm/PIN. Further, we show that pharmacological blockade of NMDAR-driven synaptic plasticity, NOS activation, or PKG signaling in the LA significantly impairs high-frequency stimulation-(HFS-) induced ERK activation and IEG expression in both regions, while blockade of extracellular NO signaling in the LA impairs HFS-induced ERK activation and IEG expression exclusively in the MGm/PIN. These findings suggest that NMDAR-driven synaptic plasticity and NO-cGMP-PKG signaling within the LA coordinately regulate ERK-driven gene expression in both the LA and the MGm/PIN following LTP induction at thalamo-LA synapses, and that synaptic plasticity in the LA promotes ERK-driven transcription in MGm/PIN neurons via NO-driven "retrograde signaling".


Assuntos
Tonsila do Cerebelo/enzimologia , Proteínas Quinases Dependentes de GMP Cíclico/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/genética , Potenciação de Longa Duração/fisiologia , Óxido Nítrico/fisiologia , Transdução de Sinais/fisiologia , Tálamo/enzimologia , Tonsila do Cerebelo/fisiologia , Animais , GMP Cíclico/fisiologia , Estimulação Elétrica/métodos , Regulação Enzimológica da Expressão Gênica/fisiologia , Potenciação de Longa Duração/genética , Masculino , Vias Neurais/enzimologia , Vias Neurais/fisiologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/genética , Transmissão Sináptica/genética , Tálamo/fisiologia
10.
J Neurosci ; 28(47): 12383-95, 2008 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-19020031

RESUMO

The activity-regulated cytoskeletal-associated protein (Arc/Arg3.1) is an immediate early gene that has been widely implicated in hippocampal-dependent learning and memory and is believed to play an integral role in synapse-specific plasticity. Here, we examined the role of Arc/Arg3.1 in amygdala-dependent Pavlovian fear conditioning. We first examined the regulation of Arc/Arg3.1 mRNA and protein after fear conditioning and LTP-inducing stimulation of thalamic inputs to the lateral amygdala (LA). Quantitative real-time PCR, in situ hybridization, Western blotting and immunohistochemistry revealed a significant upregulation of Arc/Arg3.1 mRNA and protein in the LA relative to controls. In behavioral experiments, intra-LA infusion of an Arc/Arg3.1 antisense oligodeoxynucleotide (ODN) was observed to be anatomically restricted to the LA, taken up by LA cells, and to promote significant knockdown of Arc/Arg3.1 protein. Rats given intra-LA infusions of multiple doses of the Arc/Arg3.1 ODN showed an impairment of LTM (tested approximately 24 later), but no deficit in STM (tested 3 h later) relative to controls infused with scrambled ODN. Finally, to determine whether upregulation of Arc/Arg3.1 occurs downstream of ERK/MAPK activation, we examined Arc/Arg3.1 expression in rats given intra-LA infusion of the MEK inhibitor U0126. Relative to vehicle controls, infusion of U0126 impaired training-induced increases in Arc/Arg3.1 expression. These findings suggest that Arc/Arg3.1 expression in the amygdala is required for fear memory consolidation, and further suggest that Arc/Arg3.1 regulation in the LA is downstream of the ERK/MAPK signaling pathway.


Assuntos
Tonsila do Cerebelo/fisiologia , Condicionamento Clássico/fisiologia , Proteínas do Citoesqueleto/metabolismo , Medo , Memória/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Análise de Variância , Animais , Comportamento Animal/efeitos dos fármacos , Butadienos/farmacologia , Condicionamento Clássico/efeitos dos fármacos , Proteínas do Citoesqueleto/genética , Relação Dose-Resposta a Droga , Relação Dose-Resposta à Radiação , Estimulação Elétrica/métodos , Inibidores Enzimáticos/farmacologia , Potenciação de Longa Duração/efeitos dos fármacos , Potenciação de Longa Duração/fisiologia , Potenciação de Longa Duração/efeitos da radiação , Masculino , Memória/efeitos dos fármacos , Proteínas do Tecido Nervoso/genética , Nitrilas/farmacologia , Oligodesoxirribonucleotídeos Antissenso/farmacologia , RNA Mensageiro , Ratos , Fatores de Tempo
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.
Learn Mem ; 15(2): 55-62, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18230673

RESUMO

We have previously shown that the extracellular signal-regulated kinase/mitogen-activated protein kinase (ERK/ MAPK) is transiently activated in anatomically restricted regions of the lateral amygdala (LA) following Pavlovian fear conditioning and that blockade of ERK/MAPK activation in the LA impairs both fear memory consolidation and long-term potentiation (LTP) in the amygdala, in vitro. The present experiments evaluated the role of the ERK/MAPK signaling cascade in LTP at thalamo-LA input synapses, in vivo. We first show that ERK/MAPK is transiently activated/phosphorylated in the LA at 5 min, but not 15 or 60 min, after high-frequency, but not low-frequency, stimulation of the auditory thalamus. ERK activation induced by LTP-inducing stimulation was anatomically restricted to the same regions of the LA previously shown to exhibit ERK regulation following fear conditioning. We next show that intra-LA infusion of U0126, an inhibitor of ERK/MAPK activation, impairs LTP at thalamo-LA input synapses. Collectively, results demonstrate that ERK/MAPK activation is necessary for synaptic plasticity in anatomically defined regions of the LA, in vivo.


Assuntos
Tonsila do Cerebelo/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Potenciação de Longa Duração/fisiologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Tonsila do Cerebelo/enzimologia , Animais , Vias Auditivas/fisiologia , Butadienos/administração & dosagem , Butadienos/farmacologia , Estimulação Elétrica/métodos , Ativação Enzimática/fisiologia , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Imuno-Histoquímica , Potenciação de Longa Duração/efeitos dos fármacos , Masculino , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Plasticidade Neuronal/fisiologia , Nitrilas/administração & dosagem , Nitrilas/farmacologia , Fosforilação , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/fisiologia , Sinapses/fisiologia , Tálamo/fisiologia , Fatores de Tempo , Distribuição Tecidual
13.
Learn Mem ; 15(10): 792-805, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18832566

RESUMO

Recent studies have shown that nitric oxide (NO) signaling plays a crucial role in memory consolidation of Pavlovian fear conditioning and in synaptic plasticity in the lateral amygdala (LA). In the present experiments, we examined the role of the cGMP-dependent protein kinase (PKG), a downstream effector of NO, in fear memory consolidation and long-term potentiation (LTP) at thalamic and cortical input pathways to the LA. In behavioral experiments, rats given intra-LA infusions of either the PKG inhibitor Rp-8-Br-PET-cGMPS or the PKG activator 8-Br-cGMP exhibited dose-dependent impairments or enhancements of fear memory consolidation, respectively. In slice electrophysiology experiments, bath application of Rp-8-Br-PET-cGMPS or the guanylyl cyclase inhibitor LY83583 impaired LTP at thalamic, but not cortical inputs to the LA, while bath application of 8-Br-cGMP or the guanylyl cyclase activator YC-1 resulted in enhanced LTP at thalamic inputs to the LA. Interestingly, YC-1-induced enhancement of LTP in the LA was reversed by concurrent application of the MEK inhibitor U0126, suggesting that the NO-cGMP-PKG signaling pathway may promote synaptic plasticity and fear memory formation in the LA, in part by activating the ERK/MAPK signaling cascade. As a test of this hypothesis, we next showed that rats given intra-LA infusion of the PKG inhibitor Rp-8-Br-PET-cGMPS or the PKG activator 8-Br-cGMP exhibit impaired or enhanced activation, respectively, of ERK/MAPK in the LA after fear conditioning. Collectively, our findings suggest that an NO-cGMP-PKG-dependent form of synaptic plasticity at thalamic input synapses to the LA may underlie memory consolidation of Pavlovian fear conditioning, in part, via activation of the ERK/MAPK signaling cascade.


Assuntos
Tonsila do Cerebelo/fisiologia , Potenciação de Longa Duração/fisiologia , Memória/fisiologia , Óxido Nítrico/metabolismo , Transdução de Sinais/fisiologia , Animais , Western Blotting , Condicionamento Clássico , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Medo/fisiologia , Masculino , Técnicas de Cultura de Órgãos , Técnicas de Patch-Clamp , Ratos , Ratos Sprague-Dawley , Sinapses/fisiologia
14.
Neuropsychopharmacology ; 44(4): 733-742, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30542090

RESUMO

Chronic exposure to stress is a well-known risk factor for the development of mood and anxiety disorders. Promoting resilience to stress may prevent the development of these disorders, but resilience-enhancing compounds are not yet clinically available. One compound that has shown promise in the clinical setting is curcumin, a polyphenol compound found in the rhizome of the turmeric plant (Curcuma longa) with known anti-inflammatory and antidepressant properties. Here, we tested the efficacy of 1.5% dietary curcumin at promoting resilience to chronic social defeat stress (CSDS) in 129/SvEv mice, a strain that we show is highly susceptible to this type of stress. We found that administration of curcumin during CSDS produced a 4.5-fold increase in stress resilience, as measured by the social interaction test. Although the overall effects of curcumin were striking, we identified two distinct responses to curcumin. While 64% of defeated mice on curcumin were resilient (responders), the remaining 36% of mice were susceptible to the effects of stress (non-responders). Interestingly, responders released less corticosterone following acute restraint stress and had lower levels of peripheral IL-6 than nonresponders, implicating a role for the NF-κB pathway in treatment response. Importantly, curcumin also prevented anxiety-like behavior in both responders and non-responders in the elevated-plus maze and open field test. Collectively, our findings provide the first preclinical evidence that curcumin promotes resilience to CSDS and suggest that curcumin may prevent the emergence of a range of anxiety-like symptoms when given to individuals during exposure to chronic social stress.


Assuntos
Ansiedade/prevenção & controle , Comportamento Animal/efeitos dos fármacos , Fármacos do Sistema Nervoso Central/farmacologia , Curcumina/farmacologia , Resiliência Psicológica/efeitos dos fármacos , Estresse Psicológico/prevenção & controle , Animais , Ansiedade/sangue , Ansiedade/dietoterapia , Ansiedade/fisiopatologia , Fármacos do Sistema Nervoso Central/administração & dosagem , Corticosterona/sangue , Curcumina/administração & dosagem , Interleucina-6/sangue , Masculino , Camundongos , Camundongos da Linhagem 129 , Estresse Psicológico/sangue , Estresse Psicológico/dietoterapia , Estresse Psicológico/fisiopatologia
15.
Neuron ; 44(1): 75-91, 2004 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-15450161

RESUMO

Fear conditioning is a valuable behavioral paradigm for studying the neural basis of emotional learning and memory. The lateral nucleus of the amygdala (LA) is a crucial site of neural changes that occur during fear conditioning. Pharmacological manipulations of the LA, strategically timed with respect to training and testing, have shed light on the molecular events that mediate the acquisition of fear associations and the formation and maintenance of long-term memories of those associations. Similar mechanisms have been found to underlie long-term potentiation (LTP) in LA, an artificial means of inducing synaptic plasticity and a physiological model of learning and memory. Thus, LTP-like changes in synaptic plasticity may underlie fear conditioning. Given that the neural circuit underlying fear conditioning has been implicated in emotional disorders in humans, the molecular mechanisms of fear conditioning are potential targets for psychotherapeutic drug development.


Assuntos
Tonsila do Cerebelo/fisiologia , Emoções/fisiologia , Aprendizagem/fisiologia , Animais , Condicionamento Psicológico/fisiologia , Humanos , Memória/fisiologia
16.
J Neurosci ; 26(48): 12387-96, 2006 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-17135400

RESUMO

In the standard model of pavlovian fear learning, sensory input from neutral and aversive stimuli converge in the lateral nucleus of the amygdala (LA), in which alterations in synaptic transmission encode the association. During fear expression, the LA is thought to engage the central nucleus of the amygdala (CE), which serves as the principal output nucleus for the expression of conditioned fear responses. In the present study, we reexamined the roles of LA and CE. Specifically, we asked whether CE, like LA, might also be involved in fear learning and memory consolidation. Using functional inactivation methods, we first show that CE is involved not only in the expression but also the acquisition of fear conditioning. Next, we show that inhibition of protein synthesis in CE after training impairs fear memory consolidation. These findings indicate that CE is not only involved in fear expression but, like LA, is also involved in the learning and consolidation of pavlovian fear conditioning.


Assuntos
Tonsila do Cerebelo/fisiologia , Condicionamento Clássico/fisiologia , Medo/fisiologia , Rede Nervosa/fisiologia , Pensamento/fisiologia , Animais , Medo/psicologia , Masculino , Ratos , Ratos Sprague-Dawley
17.
18.
Nat Commun ; 8: 13920, 2017 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-28067224

RESUMO

Pavlovian aversive conditioning requires learning of the association between a conditioned stimulus (CS) and an unconditioned, aversive stimulus (US) but also involves encoding the time interval between the two stimuli. The neurobiological bases of this time interval learning are unknown. Here, we show that in rats, the dorsal striatum and basal amygdala belong to a common functional network underlying temporal expectancy and learning of a CS-US interval. Importantly, changes in coherence between striatum and amygdala local field potentials (LFPs) were found to couple these structures during interval estimation within the lower range of the theta rhythm (3-6 Hz). Strikingly, we also show that a change to the CS-US time interval results in long-term changes in cortico-striatal synaptic efficacy under the control of the amygdala. Collectively, this study reveals physiological correlates of plasticity mechanisms of interval timing that take place in the striatum and are regulated by the amygdala.


Assuntos
Tonsila do Cerebelo/fisiologia , Condicionamento Clássico/fisiologia , Corpo Estriado/fisiologia , Plasticidade Neuronal/fisiologia , Percepção do Tempo/fisiologia , Tonsila do Cerebelo/anatomia & histologia , Animais , Corpo Estriado/anatomia & histologia , Eletrodos Implantados , Medo/fisiologia , Masculino , Memória/fisiologia , Ratos , Ratos Sprague-Dawley , Ritmo Teta/fisiologia
19.
J Neurosci ; 25(43): 10010-4, 2005 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-16251449

RESUMO

Although it is believed that different types of memories are localized in discreet regions of the brain, concrete experimental evidence of the existence of such engrams is often elusive. Despite being one of the best characterized memory systems of the brain, the question of where fear memories are localized in the brain remains a hotly debated issue. Here, we combine site-specific behavioral pharmacology with multisite electrophysiological recording techniques to show that the lateral nucleus of the amygdala, long thought to be critical for the acquisition of fear memories, is also an essential locus of fear memory storage.


Assuntos
Tonsila do Cerebelo/fisiologia , Medo/fisiologia , Memória/fisiologia , Tonsila do Cerebelo/efeitos dos fármacos , Animais , Comportamento Animal , Butadienos/farmacologia , Inibidores Enzimáticos/farmacologia , Potenciais Evocados/efeitos dos fármacos , Potenciais Evocados/fisiologia , Potenciais Evocados/efeitos da radiação , Reação de Congelamento Cataléptica/efeitos dos fármacos , Masculino , Memória/efeitos dos fármacos , Nitrilas/farmacologia , Ratos , Ratos Sprague-Dawley
20.
J Neurosci ; 25(24): 5730-9, 2005 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-15958739

RESUMO

In the present study, we examined the role of the auditory thalamus [medial division of the medial geniculate nucleus and the adjacent posterior intralaminar nucleus (MGm/PIN)] in auditory pavlovian fear conditioning using pharmacological manipulation of intracellular signaling pathways. In the first experiment, rats were given intrathalamic infusions of the MEK (mitogen-activated protein kinase-kinase) inhibitor 1,4-diamino-2,3-dicyano-1,4-bis(o-aminophenylmercapto) butadiene (U0126) before fear conditioning. Findings revealed that long-term memory (assessed at 24 h) was impaired, whereas short-term memory (assessed at 1-3 h) of fear conditioning was intact. In the second experiment, rats received immediate posttraining intrathalamic infusion of U0126, the mRNA synthesis inhibitor 5,6-dichloro-1-beta-D-ribofuranosylbenzimidazole (DRB), or infusion of the protein synthesis inhibitor anisomycin. Posttraining infusion of either U0126 or DRB significantly impaired long-term retention of fear conditioning, whereas infusion of anisomycin had no effect. In the final experiment, rats received intrathalamic infusion of U0126 before long-term potentiation (LTP)-inducing stimulation of thalamic inputs to the lateral nucleus of the amygdala (LA). Findings revealed that thalamic infusion of U0126 impaired LTP in the LA. Together, these results suggest the possibility that MGm/PIN cells that project to the LA contribute to memory formation via ERK (extracellular signal-regulated kinase)-mediated transcription, but that they do so by promoting protein synthesis-dependent plasticity locally in the LA.


Assuntos
Tonsila do Cerebelo/fisiologia , Condicionamento Psicológico/fisiologia , Medo/fisiologia , Potenciação de Longa Duração/fisiologia , Sistema de Sinalização das MAP Quinases/fisiologia , Tálamo/fisiologia , Estimulação Acústica , Animais , Butadienos/farmacologia , Condicionamento Psicológico/efeitos dos fármacos , Eletrochoque , Inibidores Enzimáticos/farmacologia , Masculino , Modelos Animais , Plasticidade Neuronal , Nitrilas/farmacologia , Ratos , Ratos Sprague-Dawley , Sinapses/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA