Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
eNeuro ; 11(6)2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38902023

RESUMEN

Formation and retrieval of remote contextual memory depends on cortical engram neurons that are defined during learning. Manipulation of astrocytic Gq and Gi associated G-protein coupled receptor (GPCR) signaling has been shown to affect memory processing, but little is known about the role of cortical astrocytic Gs-GPCR signaling in remote memory acquisition and the functioning of cortical engram neurons. We assessed this by chemogenetic manipulation of astrocytes in the medial prefrontal cortex (mPFC) of male mice, during either encoding or consolidation of a contextual fear memory, while simultaneously labeling cortical engram neurons. We found that stimulation of astrocytic Gs signaling during memory encoding and consolidation did not alter remote memory expression. In line with this, the size of the mPFC engram population and the recall-induced reactivation of these neurons was unaffected. Hence, our data indicate that activation of Gs-GPCR signaling in cortical astrocytes is not sufficient to alter memory performance and functioning of cortical engram neurons.


Asunto(s)
Astrocitos , Miedo , Neuronas , Corteza Prefrontal , Transducción de Señal , Animales , Astrocitos/metabolismo , Masculino , Corteza Prefrontal/metabolismo , Corteza Prefrontal/fisiología , Transducción de Señal/fisiología , Neuronas/metabolismo , Neuronas/fisiología , Miedo/fisiología , Ratones Endogámicos C57BL , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Ratones , Memoria/fisiología , Memoria a Largo Plazo/fisiología
2.
Front Behav Neurosci ; 16: 893572, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35832291

RESUMEN

Post-reactivation amnesia of contextual fear memories by blockade of noradrenergic signaling has been shown to have limited replicability in rodents. This is usually attributed to several boundary conditions that gate the destabilization of memory during its retrieval. How these boundary conditions can be overcome, and what neural mechanisms underlie post-reactivation changes in contextual fear memories remain largely unknown. Here, we report a series of experiments in a contextual fear-conditioning paradigm in mice, that were aimed at solving these issues. We first attempted to obtain a training paradigm that would consistently result in contextual fear memory that could be destabilized upon reactivation, enabling post-retrieval amnesia by the administration of propranolol. Unexpectedly, our attempts were unsuccessful to this end. Specifically, over a series of experiments in which we varied different parameters of the fear acquisition procedure, at best small and inconsistent effects were observed. Additionally, we found that propranolol did not alter retrieval-induced neural activity, as measured by the number of c-Fos+ cells in the hippocampal dentate gyrus. To determine whether propranolol was perhaps ineffective in interfering with reactivated contextual fear memories, we also included anisomycin (i.e., a potent and well-known amnesic drug) in several experiments, and measures of synaptic glutamate receptor subunit GluA2 (i.e., a marker of memory destabilization). No post-retrieval amnesia by anisomycin and no altered GluA2 expression by reactivation was observed, suggesting that the memories did not undergo destabilization. The null findings are surprising, given that the training paradigms we implemented were previously shown to result in memories that could be modified upon reactivation. Together, our observations illustrate the elusive nature of reactivation-dependent changes in non-human fear memory.

3.
Biol Psychiatry ; 91(12): 1029-1038, 2022 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34715992

RESUMEN

BACKGROUND: Cocaine-associated environments (i.e., contexts) evoke persistent memories of cocaine reward and thereby contribute to the maintenance of addictive behavior in cocaine users. From a therapeutic perspective, enhancing inhibitory control over cocaine-conditioned responses is of pivotal importance but requires a more detailed understanding of the neural circuitry that can suppress context-evoked cocaine memories, e.g., through extinction learning. The ventral medial prefrontal cortex (vmPFC) and dorsal medial prefrontal cortex (dmPFC) are thought to bidirectionally regulate responding to cocaine cues through their projections to other brain regions. However, whether these mPFC subregions interact to enable adaptive responding to cocaine-associated contextual stimuli has remained elusive. METHODS: We used antero- and retrograde tracing combined with chemogenetic intervention to examine the role of vmPFC-to-dmPFC projections in extinction of cocaine-induced place preference in mice. In addition, electrophysiological recordings and optogenetics were used to determine whether parvalbumin-expressing inhibitory interneurons and pyramidal neurons in the dmPFC are innervated by vmPFC projections. RESULTS: We found that vmPFC-to-dmPFC projecting neurons are activated during unreinforced re-exposure to a cocaine-associated context, and selective suppression of these cells impairs extinction learning. Parvalbumin-expressing inhibitory interneurons in the dmPFC receive stronger monosynaptic excitatory input from vmPFC projections than local dmPFC pyramidal neurons, consequently resulting in disynaptic inhibition of pyramidal neurons. In line with this, we show that chemogenetic suppression of dmPFC parvalbumin-expressing inhibitory interneurons impairs extinction learning. CONCLUSIONS: Our data reveal that vmPFC projections mediate extinction of a cocaine-associated contextual memory through recruitment of feed-forward inhibition in the dmPFC, thereby providing a novel neuronal substrate that promotes extinction-induced inhibitory control.


Asunto(s)
Cocaína , Animales , Cocaína/farmacología , Extinción Psicológica/fisiología , Ratones , Parvalbúminas , Corteza Prefrontal/fisiología , Recompensa
4.
Biol Psychiatry ; 90(7): 494-504, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34503674

RESUMEN

BACKGROUND: Traumatic experiences, such as conditioned threat, are coded as enduring memories that are frequently subject to generalization, which is characterized by (re-) expression of fear in safe environments. However, the neurobiological mechanisms underlying threat generalization after a traumatic experience and the role of stress hormones in this process remain poorly understood. METHODS: We examined the influence of glucocorticoid hormones on the strength and specificity of conditioned fear memory at the level of sparsely distributed dentate gyrus (DG) engram cells in male mice. RESULTS: We found that elevating glucocorticoid hormones after fear conditioning induces a generalized contextual fear response. This was accompanied by a selective and persistent increase in the excitability and number of activated DG granule cells. Selective chemogenetic suppression of these sparse cells in the DG prevented glucocorticoid-induced fear generalization and restored contextual memory specificity, while leaving expression of auditory fear memory unaffected. CONCLUSIONS: These results implicate the sparse ensemble of DG engram cells as a critical cellular substrate underlying fear generalization induced by glucocorticoid stress hormones.


Asunto(s)
Giro Dentado , Glucocorticoides , Animales , Miedo , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas
5.
Sci Rep ; 11(1): 15486, 2021 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-34326412

RESUMEN

Hibernation induces neurodegeneration-like changes in the brain, which are completely reversed upon arousal. Hibernation-induced plasticity may therefore be of great relevance for the treatment of neurodegenerative diseases, but remains largely unexplored. Here we show that a single torpor and arousal sequence in mice does not induce dendrite retraction and synapse loss as observed in seasonal hibernators. Instead, it increases hippocampal long-term potentiation and contextual fear memory. This is accompanied by increased levels of key postsynaptic proteins and mitochondrial complex I and IV proteins, indicating mitochondrial reactivation and enhanced synaptic plasticity upon arousal. Interestingly, a single torpor and arousal sequence was also sufficient to restore contextual fear memory in an APP/PS1 mouse model of Alzheimer's disease. Our study demonstrates that torpor in mice evokes an exceptional state of hippocampal plasticity and that naturally occurring plasticity mechanisms during torpor provide an opportunity to identify unique druggable targets for the treatment of cognitive impairment.


Asunto(s)
Enfermedad de Alzheimer/fisiopatología , Memoria/fisiología , Sinapsis/fisiología , Letargo/fisiología , Animales , Cognición/fisiología , Modelos Animales de Enfermedad , Miedo , Hibernación/fisiología , Hipocampo/fisiología , Potenciación a Largo Plazo , Masculino , Trastornos de la Memoria/fisiopatología , Ratones , Ratones Endogámicos C57BL , Mitocondrias/fisiología , Plasticidad Neuronal , Neuronas/fisiología , Estaciones del Año , Temperatura
6.
Cells ; 10(6)2021 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-34199502

RESUMEN

The metabotropic glutamate receptor 5 (mGluR5) is an essential modulator of synaptic plasticity, learning and memory; whereas in pathological conditions, it is an acknowledged therapeutic target that has been implicated in multiple brain disorders. Despite robust pre-clinical data, mGluR5 antagonists failed in several clinical trials, highlighting the need for a better understanding of the mechanisms underlying mGluR5 function. In this study, we dissected the molecular synaptic modulation mediated by mGluR5 using genetic and pharmacological mouse models to chronically and acutely reduce mGluR5 activity. We found that next to dysregulation of synaptic proteins, the major regulation in protein expression in both models concerned specific processes in mitochondria, such as oxidative phosphorylation. Second, we observed morphological alterations in shape and area of specifically postsynaptic mitochondria in mGluR5 KO synapses using electron microscopy. Third, computational and biochemical assays suggested an increase of mitochondrial function in neurons, with increased level of NADP/H and oxidative damage in mGluR5 KO. Altogether, our observations provide diverse lines of evidence of the modulation of synaptic mitochondrial function by mGluR5. This connection suggests a role for mGluR5 as a mediator between synaptic activity and mitochondrial function, a finding which might be relevant for the improvement of the clinical potential of mGluR5.


Asunto(s)
Mitocondrias/metabolismo , NADP/metabolismo , Receptor del Glutamato Metabotropico 5/metabolismo , Sinapsis/metabolismo , Animales , Masculino , Ratones , Ratones Noqueados , Mitocondrias/genética , NADP/genética , Oxidación-Reducción , Receptor del Glutamato Metabotropico 5/genética , Sinapsis/genética
7.
Sci Adv ; 6(19): eaax7060, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32494694

RESUMEN

Alcohol use disorder is characterized by a high risk of relapse during periods of abstinence. Relapse is often triggered by retrieval of persistent alcohol memories upon exposure to alcohol-associated environmental cues, but little is known about the neuronal circuitry that supports the long-term storage of alcohol cue associations. We found that a small ensemble of neurons in the medial prefrontal cortex (mPFC) of mice was activated during cue-paired alcohol self-administration (SA) and that selective suppression of these neurons 1 month later attenuated cue-induced relapse to alcohol seeking. Inhibition of alcohol seeking was specific to these neurons as suppression of a non-alcohol-related or sucrose SA-activated mPFC ensemble did not affect relapse behavior. Hence, the mPFC neuronal ensemble activated during cue-paired alcohol consumption functions as a lasting memory trace that mediates cue-evoked relapse long after cessation of alcohol intake, thereby providing a potential target for treatment of alcohol relapse vulnerability.

8.
Front Cell Neurosci ; 13: 465, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31749686

RESUMEN

G-protein-coupled receptor 158 (Gpr158) is highly expressed in striatum, hippocampus and prefrontal cortex. It gained attention as it was implicated in physiological responses to stress and depression. Recently, Gpr158 has been shown to act as a pathway-specific synaptic organizer in the hippocampus, required for proper mossy fiber-CA3 neurocircuitry establishment, structure, and function. Although rodent Gpr158 expression is highest in CA3, considerable expression occurs in CA1 especially after the first postnatal month. Here, we combined hippocampal-dependent behavioral paradigms with subsequent electrophysiological and morphological analyses from the same group of mice to assess the effects of Gpr158 deficiency on CA1 physiology and function. We demonstrate deficits in spatial memory acquisition and retrieval in the Morris water maze paradigm, along with deficits in the acquisition of extinction memory in the passive avoidance test in Gpr158 KO mice. Electrophysiological recordings from CA1 pyramidal neurons revealed normal basal excitatory and inhibitory synaptic transmission, however, Schaffer collateral stimulation yielded dramatically reduced post-synaptic currents. Interestingly, intrinsic excitability of CA1 pyramidals was found increased, potentially acting as a compensatory mechanism to the reductions in Schaffer collateral-mediated drive. Both ex vivo and in vitro, neurons deficient for or with lowered levels of Gpr158 exhibited robust reductions in dendritic architecture and complexity, i.e., reduced length, surface, bifurcations, and branching. This effect was localized in the apical but not basal dendrites of adult CA1 pyramidals, indicative of compartment-specific alterations. A significant positive correlation between spatial memory acquisition and extent of complexity of CA1 pyramidals was found. Taken together, we provide first evidence of significant disruptions in hippocampal CA1 neuronal dendritic architecture and physiology, driven by Gpr158 deficiency. Importantly, the hippocampal neuronal morphology deficits appear to support the impairments in spatial memory acquisition observed in Gpr158 KO mice.

9.
Nat Commun ; 10(1): 2232, 2019 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-31110186

RESUMEN

Sparse populations of neurons in the dentate gyrus (DG) of the hippocampus are causally implicated in the encoding of contextual fear memories. However, engram-specific molecular mechanisms underlying memory consolidation remain largely unknown. Here we perform unbiased RNA sequencing of DG engram neurons 24 h after contextual fear conditioning to identify transcriptome changes specific to memory consolidation. DG engram neurons exhibit a highly distinct pattern of gene expression, in which CREB-dependent transcription features prominently (P = 6.2 × 10-13), including Atf3 (P = 2.4 × 10-41), Penk (P = 1.3 × 10-15), and Kcnq3 (P = 3.1 × 10-12). Moreover, we validate the functional relevance of the RNAseq findings by establishing the causal requirement of intact CREB function specifically within the DG engram during memory consolidation, and identify a novel group of CREB target genes involved in the encoding of long-term memory.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas del Citoesqueleto/metabolismo , Giro Dentado/fisiología , Consolidación de la Memoria/fisiología , Proteínas del Tejido Nervioso/metabolismo , Factor de Transcripción Activador 3/genética , Factor de Transcripción Activador 3/metabolismo , Animales , Condicionamiento Psicológico/fisiología , Giro Dentado/citología , Encefalinas/genética , Encefalinas/metabolismo , Miedo/fisiología , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/fisiología , Canal de Potasio KCNQ3/genética , Canal de Potasio KCNQ3/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Animales , Neuronas/metabolismo , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Análisis de Secuencia de ARN , Técnicas Estereotáxicas
10.
Nat Commun ; 10(1): 2315, 2019 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-31127098

RESUMEN

Encoding and retrieval of contextual memories is initially mediated by sparsely activated neurons, so-called engram cells, in the hippocampus. Subsequent memory persistence is thought to depend on network-wide changes involving progressive contribution of cortical regions, a process referred to as systems consolidation. Using a viral-based TRAP (targeted recombination in activated populations) approach, we studied whether consolidation of contextual fear memory by neurons in the medial prefrontal cortex (mPFC) is modulated by memory strength and CREB function. We demonstrate that activity of a small subset of mPFC neurons is sufficient and necessary for remote memory expression, but their involvement depends on the strength of conditioning. Furthermore, selective disruption of CREB function in mPFC engram cells after mild conditioning impairs remote memory expression. Together, our data demonstrate that memory consolidation by mPFC engram cells requires CREB-mediated transcription, with the functionality of this network hub being gated by memory strength.


Asunto(s)
Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Miedo/fisiología , Consolidación de la Memoria/fisiología , Memoria a Largo Plazo/fisiología , Corteza Prefrontal/fisiología , Animales , Conducta Animal/fisiología , Condicionamiento Psicológico/fisiología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/antagonistas & inhibidores , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Dependovirus/genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Microinyecciones , Modelos Animales , Neuronas/metabolismo , Técnicas de Placa-Clamp , Corteza Prefrontal/citología , Técnicas Estereotáxicas
12.
Elife ; 62017 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-29199957

RESUMEN

Glutamatergic synapses rely on AMPA receptors (AMPARs) for fast synaptic transmission and plasticity. AMPAR auxiliary proteins regulate receptor trafficking, and modulate receptor mobility and its biophysical properties. The AMPAR auxiliary protein Shisa7 (CKAMP59) has been shown to interact with AMPARs in artificial expression systems, but it is unknown whether Shisa7 has a functional role in glutamatergic synapses. We show that Shisa7 physically interacts with synaptic AMPARs in mouse hippocampus. Shisa7 gene deletion resulted in faster AMPAR currents in CA1 synapses, without affecting its synaptic expression. Shisa7 KO mice showed reduced initiation and maintenance of long-term potentiation of glutamatergic synapses. In line with this, Shisa7 KO mice showed a specific deficit in contextual fear memory, both short-term and long-term after conditioning, whereas auditory fear memory and anxiety-related behavior were normal. Thus, Shisa7 is a bona-fide AMPAR modulatory protein affecting channel kinetics of AMPARs, necessary for synaptic hippocampal plasticity, and memory recall.


Asunto(s)
Proteínas Portadoras/metabolismo , Hipocampo/fisiología , Proteínas de la Membrana/metabolismo , Memoria , Receptores AMPA/metabolismo , Sinapsis/fisiología , Animales , Proteínas Portadoras/genética , Técnicas de Inactivación de Genes , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Unión Proteica , Mapeo de Interacción de Proteínas
13.
Sci Rep ; 6: 20569, 2016 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-26837469

RESUMEN

Alzheimer's disease (AD) is characterised by amyloid-beta (Aß) protein deposition in the brain. Posttranslational modifications in Aß play an important role in Aß deposition. Tissue transglutaminase (tTG) is an enzyme involved in posttranslational cross-linking of proteins. tTG levels and activity are increased in AD brains, and tTG is associated with Aß deposits and lesion-associated astrocytes in AD cases. Furthermore, Aß is a substrate of tTG-catalysed cross-linking. To study the role of tTG in Aß pathology, we compared tTG distribution and activity in both the APPSWE/PS1ΔE9 and APP23 mice models with human AD. Using immunohistochemistry, we found association of both tTG and in situ active tTG with Aß plaques and vascular Aß, in early and late stages of Aß deposition. In addition, tTG staining colocalised with Aß-associated reactive astrocytes. Thus, alike human AD cases, tTG was associated with Aß depositions in these AD models. Although, distribution pattern and spatial overlay of both tTG and its activity with Aß pathology was substantially different from human AD cases, our findings provide evidence for an early role of tTG in Aß pathology. Yet, species differences should be taken into account when using these models to study the role of tTG in Aß pathology.


Asunto(s)
Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Astrocitos/metabolismo , Transglutaminasas/metabolismo , Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/metabolismo , Animales , Dominio Catalítico , Modelos Animales de Enfermedad , Activación Enzimática , Humanos , Ratones , Placa Amiloide/metabolismo , Transglutaminasas/química
14.
Hippocampus ; 25(11): 1250-61, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25708624

RESUMEN

A change in efficacy of hippocampal synapses is critical for memory formation. So far, the molecular analysis of synapses during learning has focused on small groups of proteins, whereas the dynamic global changes at these synapses have remained unknown. Here, we analyzed the temporal changes of the mouse hippocampal synaptic membrane proteome 1 and 4 h after contextual fear learning, comparing two groups; (1) a fear memory forming "delayed-shock" group and (2) a fear memory-deficient "immediate-shock" group. No changes in protein expression were observed 1 h after conditioning between the two experimental groups. However, 423 proteins were significantly regulated 4 h later of which 164 proteins showed a temporal regulation after a delayed shock and 273 proteins after the stress of an immediate shock. From the proteins that were differentially regulated between the delayed- and the immediate-shock groups at 4 h, 48 proteins, most prominently representing endocytosis, (amphiphysin, dynamin, and synaptojanin1), glutamate signaling (glutamate [NMDA] receptor subunit epsilon-1, disks large homolog 3), and neurotransmitter metabolism (excitatory amino acid transporter 1, excitatory amino acid transporter 2, sodium- and chloride-dependent GABA transporter 3) were regulated in both protocols, but in opposite directions, pointing toward an interaction of learning and stress. Taken together, this data set yields novel insight into diverse and dynamic changes that take place at hippocampal synapses over the time course of contextual fear-memory learning.


Asunto(s)
Conducta Animal/fisiología , Condicionamiento Psicológico/fisiología , Miedo/fisiología , Hipocampo/metabolismo , Proteoma/metabolismo , Estrés Psicológico/metabolismo , Membranas Sinápticas/metabolismo , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Factores de Tiempo
15.
Biol Psychiatry ; 76(8): 648-55, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24703509

RESUMEN

BACKGROUND: A deficit in impulse control is a prominent, heritable symptom in several psychiatric disorders, such as addiction, attention-deficit/hyperactivity disorder, and schizophrenia. Here, we aimed to identify genes regulating impulsivity, specifically of impulsive action, in mice. METHODS: Using the widely used 5-choice serial reaction time task, we measured impulsive action in 1) a panel of 41 BXD recombinant inbred strains of mice (n = 13.7 ± .8 per strain; n = 654 total) to detect underlying genetic loci; 2) congenic mice (n = 23) to replicate the identified locus; 3) mice overexpressing the Nrg3 candidate gene in the medial prefrontal cortex (n = 21); and 4) a Nrg3 loss-of-function mutant (n = 59) to functionally implicate the Nrg3 candidate gene in impulsivity. RESULTS: Genetic mapping of impulsive action in the BXD panel identified a locus on chromosome 14 (34.5-41.4 Mb), syntenic with the human 10q22-q23 schizophrenia-susceptibility locus. Congenic mice carrying the impulsivity locus (Impu1) confirmed its influence on impulsive action. Increased impulsivity was associated with increased Nrg3 gene expression in the medial prefrontal cortex (mPFC). Viral overexpression of Nrg3 in the mPFC increased impulsivity, whereas a constitutive Nrg3 loss-of-function mutation decreased it. CONCLUSIONS: The causal relation between Nrg3 expression in the mPFC and level of impulsive action shown here provides a mechanism by which polymorphism in NRG3 in humans contributes to a specific cognitive deficit seen in several psychiatric diseases, such as addiction, attention-deficit/hyperactivity disorder, and schizophrenia.


Asunto(s)
Trastornos del Conocimiento/genética , Conducta Impulsiva/fisiología , Péptidos y Proteínas de Señalización Intracelular/genética , Corteza Prefrontal/patología , Animales , Conducta de Elección/fisiología , Trastornos del Conocimiento/patología , Modelos Animales de Enfermedad , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación/genética , Neurregulinas , Oligodesoxirribonucleótidos Antisentido/farmacología , Sitios de Carácter Cuantitativo , Tiempo de Reacción/genética , Estadísticas no Paramétricas
16.
Nat Neurosci ; 14(10): 1302-8, 2011 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-21909089

RESUMEN

Upon retrieval, fear memories are rendered labile and prone to modification, necessitating a restabilization process of reconsolidation to persist further. This process is also crucial for modulating both strength and content of an existing memory and forms a promising therapeutic target for fear-related disorders. However, the molecular and cellular mechanism of adaptive reconsolidation still remains obscure. Here we show that retrieval of fear memory induces a biphasic temporal change in GluA2-containing AMPA-type glutamate receptor (AMPAR) membrane expression and synaptic strength in the mouse dorsal hippocampus. Blockade of retrieval-induced, regulated, GluA2-dependent endocytosis enhanced subsequent expression of fear. In addition, this blockade prevented the loss of fear response after reconsolidation-update of fear memory content in the long-term. Thus, endocytosis of GluA2-containing AMPARs allows plastic changes at the synaptic level that exerts an inhibitory constraint on memory strengthening and underlies the loss of fear response by reinterpretation of memory content during adaptive reconsolidation.


Asunto(s)
Condicionamiento Clásico/fisiología , Endocitosis/fisiología , Miedo/fisiología , Recuerdo Mental/fisiología , Receptores AMPA/metabolismo , Sinapsis/metabolismo , Adaptación Fisiológica/efectos de los fármacos , Adaptación Fisiológica/fisiología , Análisis de Varianza , Animales , Anisomicina/farmacología , Conducta Animal , Biotinilación , Condicionamiento Clásico/efectos de los fármacos , Electrochoque/efectos adversos , Endocitosis/efectos de los fármacos , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Miedo/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Hipocampo/citología , Técnicas In Vitro , Masculino , Recuerdo Mental/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Técnicas de Placa-Clamp , Péptidos/farmacología , Inhibidores de la Síntesis de la Proteína/farmacología , Transporte de Proteínas/efectos de los fármacos , Receptores AMPA/química , Sinapsis/efectos de los fármacos , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA