Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 458
Filtrar
Mais filtros

País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 184(22): 5622-5634.e25, 2021 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-34610277

RESUMO

Disinhibitory neurons throughout the mammalian cortex are powerful enhancers of circuit excitability and plasticity. The differential expression of neuropeptide receptors in disinhibitory, inhibitory, and excitatory neurons suggests that each circuit motif may be controlled by distinct neuropeptidergic systems. Here, we reveal that a bombesin-like neuropeptide, gastrin-releasing peptide (GRP), recruits disinhibitory cortical microcircuits through selective targeting and activation of vasoactive intestinal peptide (VIP)-expressing cells. Using a genetically encoded GRP sensor, optogenetic anterograde stimulation, and trans-synaptic tracing, we reveal that GRP regulates VIP cells most likely via extrasynaptic diffusion from several local and long-range sources. In vivo photometry and CRISPR-Cas9-mediated knockout of the GRP receptor (GRPR) in auditory cortex indicate that VIP cells are strongly recruited by novel sounds and aversive shocks, and GRP-GRPR signaling enhances auditory fear memories. Our data establish peptidergic recruitment of selective disinhibitory cortical microcircuits as a mechanism to regulate fear memories.


Assuntos
Córtex Auditivo/metabolismo , Bombesina/metabolismo , Medo/fisiologia , Memória/fisiologia , Rede Nervosa/metabolismo , Sequência de Aminoácidos , Animais , Cálcio/metabolismo , Sinalização do Cálcio , Condicionamento Clássico , Peptídeo Liberador de Gastrina/química , Peptídeo Liberador de Gastrina/metabolismo , Regulação da Expressão Gênica , Genes Precoces , Células HEK293 , Humanos , Espaço Intracelular/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Receptores da Bombesina/metabolismo , Som , Peptídeo Intestinal Vasoativo/metabolismo
2.
J Neurosci ; 43(40): 6816-6829, 2023 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-37625855

RESUMO

Dysfunctions in growth hormone (GH) secretion increase the prevalence of anxiety and other neuropsychiatric diseases. GH receptor (GHR) signaling in the amygdala has been associated with fear memory, a key feature of posttraumatic stress disorder. However, it is currently unknown which neuronal population is targeted by GH action to influence the development of neuropsychiatric diseases. Here, we showed that approximately 60% of somatostatin (SST)-expressing neurons in the extended amygdala are directly responsive to GH. GHR ablation in SST-expressing cells (SSTΔGHR mice) caused no alterations in energy or glucose metabolism. Notably, SSTΔGHR male mice exhibited increased anxiety-like behavior in the light-dark box and elevated plus maze tests, whereas SSTΔGHR females showed no changes in anxiety. Using auditory Pavlovian fear conditioning, both male and female SSTΔGHR mice exhibited a significant reduction in fear memory. Conversely, GHR ablation in SST neurons did not affect memory in the novel object recognition test. Gene expression was analyzed in a micro punch comprising the central nucleus of the amygdala (CEA) and basolateral (BLA) complex. GHR ablation in SST neurons caused sex-dependent changes in the expression of factors involved in synaptic plasticity and function. In conclusion, GHR expression in SST neurons is necessary to regulate anxiety in males, but not female mice. GHR ablation in SST neurons also decreases fear memory and affects gene expression in the amygdala, although marked sex differences were observed. Our findings identified for the first time a neurochemically-defined neuronal population responsible for mediating the effects of GH on behavioral aspects associated with neuropsychiatric diseases.SIGNIFICANCE STATEMENT Hormone action in the brain regulates different neurological aspects, affecting the predisposition to neuropsychiatric disorders, like depression, anxiety, and posttraumatic stress disorder. Growth hormone (GH) receptor is widely expressed in the brain, but the exact function of neuronal GH action is not fully understood. Here, we showed that mice lacking the GH receptor in a group of neurons that express the neuropeptide somatostatin exhibit increased anxiety. However, this effect is only observed in male mice. In contrast, the absence of the GH receptor in somatostatin-expressing neurons decreases fear memory, a key feature of posttraumatic stress disorder, in males and females. Thus, our study identified a specific group of neurons in which GH acts to affect the predisposition to neuropsychiatric diseases.


Assuntos
Hormônio do Crescimento , Somatostatina , Feminino , Masculino , Camundongos , Animais , Somatostatina/metabolismo , Hormônio do Crescimento/metabolismo , Ansiedade , Medo , Receptores da Somatotropina/genética , Receptores da Somatotropina/metabolismo , Neurônios/metabolismo
3.
Physiol Genomics ; 56(4): 327-342, 2024 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-38314698

RESUMO

This study investigated the interaction between genetic differences in stress reactivity/coping and environmental challenges, such as acute stress during adolescence on adult contextual fear memory and anxiety-like behaviors. Fischer 344 (F344) and the inbred F344;WKY-Stresp3/Eer congenic strain (congenic), in which chromosomal regions from the Wistar-Kyoto (WKY) strain were introgressed into the F344 background, were exposed to a modified forced swim test during adolescence, while controls were undisturbed. In adulthood, fear learning and memory, assessed by contextual fear conditioning, were significantly greater in congenic animals compared with F344 animals, and stress during adolescence increased them even further in males of both strains. Anxiety-like behavior, measured by the open field test, was also greater in congenic than F344 animals, and stress during adolescence increased it further in both strains of adult males. Whole genome sequencing of the F344;WKY-Stresp3/Eer strain revealed an enrichment of WKY genotypes in chromosomes 9, 14, and 15. An example of functional WKY sequence variations in the congenic strain, cannabinoid receptor interacting protein 1 (Cnrip1) had a Cnrip1 transcript isoform that lacked two exons. Although the original hypothesis that the genetic predisposition to increased anxiety of the WKY donor strain would exaggerate fear memory relative to the background strain was confirmed, the consequences of adolescent stress were strain independent but sex dependent in adulthood. Molecular genomic approaches combined with genetic mapping of WKY sequence variations in chromosomes 9, 14, and 15 could aid in finding quantitative trait genes contributing to the variation in fear memory.NEW & NOTEWORTHY This study found that 1) whole genome sequencing of congenic strains should be a criterion for their recognition; 2) sequence variations between Wistar-Kyoto and Fischer 344 strains at regions of chromosomes 9, 14, and 15 contribute to differences in contextual fear memory and anxiety-like behaviors; and 3) stress during adolescence affects these behaviors in males, but not females, and is independent of strain.


Assuntos
Ansiedade , Medo , Masculino , Ratos , Animais , Ratos Endogâmicos WKY , Ratos Endogâmicos F344 , Ansiedade/genética , Cromossomos , Animais Congênicos , Proteínas de Transporte/genética
4.
Eur J Neurosci ; 59(7): 1696-1722, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38269959

RESUMO

Vitamin D deficiency is a worldwide health concern, especially in the elderly population. Much remains unknown about the relationship between vitamin D deficiency (VDD), stress-induced cognitive dysfunctions and depressive-like behaviour. In this study, 4-month-old male C57Bl/6J mice were fed with control or vitamin D free diet for 6 months, followed by unpredictable chronic stress (UCMS) for 8 weeks. VDD induced cognitive impairment and reduced grooming behaviour, but did not induce depressive-like behaviour. While UCMS in vitamin D sufficient mice induced expected depressive-like phenotype and impairments in the contextual fear memory, chronic stress did not manifest as an additional risk factor for memory impairments and depressive-like behaviour in VDD mice. In fact, UCMS restored self-care behaviour in VDD mice. At the histopathological level, VDD mice exhibited cell loss in the granule cell layer, reduced survival of newly generated cells, accompanied with an increased number of apoptotic cells and alterations in glial morphology in the hippocampus; however, these effects were not exacerbated by UCMS. Interestingly, UCMS reversed VDD induced loss of microglial cells. Moreover, tyrosine hydroxylase levels decreased in the striatum of VDD mice, but not in stressed VDD mice. These findings indicate that long-term VDD in adulthood impairs cognition but does not augment behavioural response to UCMS in middle-aged mice. While VDD caused cell loss and altered glial response in the DG of the hippocampus, these effects were not exacerbated by UCMS and could contribute to mechanisms regulating altered stress response.


Assuntos
Deficiência de Vitamina D , Vitamina D , Idoso , Humanos , Animais , Camundongos , Masculino , Pessoa de Meia-Idade , Lactente , Hipocampo , Encéfalo , Transtornos da Memória/etiologia , Deficiência de Vitamina D/complicações , Camundongos Endogâmicos C57BL , Estresse Psicológico/complicações , Modelos Animais de Doenças
5.
Neurobiol Learn Mem ; 213: 107960, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39004160

RESUMO

Labilization-reconsolidation, which relies on retrieval, has been considered an opportunity to attenuate the negative aspects of traumatic memories. A therapeutic strategy based on reconsolidation blockade is deemed more effective than current therapies relying on memory extinction. Nevertheless, extremely stressful memories frequently prove resistant to this process. Here, after inducing robust fear memory in mice through strong fear conditioning, we examined the possibility of rendering it susceptible to pharmacological modulation based on the degree of generalized fear (GF). To achieve this, we established an ordered gradient of GF, determined by the perceptual similarity between the associated context (CA) and non-associated contexts (CB, CC, CD, and CE) to the aversive event. We observed that as the exposure context became less similar to CA, the defensive pattern shifted from passive to active behaviors in both male and female mice. Subsequently, in conditioned animals, we administered propranolol after exposure to the different contexts (CA, CB, CC, CD or CE). In males, propranolol treatment resulted in reduced freezing time and enhanced risk assessment behaviors when administered following exposure to CA or CB, but not after CC, CD, or CE, compared to the control group. In females, a similar change in behavioral pattern was observed with propranolol administered after exposure to CC, but not after the other contexts. These results highlight the possibility of indirectly manipulating a robust contextual fear memory by controlling the level of generalization during recall. Additionally, it was demonstrated that the effect of propranolol on reconsolidation would not lead to a reduction in fear memory per se, but rather to its reorganization resulting in greater behavioral flexibility (from passive to active behaviors). Finally, from a clinical viewpoint, this would be of considerable relevance since following this strategy could make the treatment of psychiatric disorders associated with traumatic memory formation more effective and less stressful.


Assuntos
Condicionamento Clássico , Medo , Propranolol , Medo/efeitos dos fármacos , Medo/fisiologia , Animais , Masculino , Propranolol/farmacologia , Feminino , Camundongos , Condicionamento Clássico/efeitos dos fármacos , Consolidação da Memória/efeitos dos fármacos , Consolidação da Memória/fisiologia , Camundongos Endogâmicos C57BL , Memória/efeitos dos fármacos , Memória/fisiologia , Generalização Psicológica/efeitos dos fármacos , Generalização Psicológica/fisiologia , Extinção Psicológica/efeitos dos fármacos
6.
Behav Brain Funct ; 20(1): 21, 2024 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-39182120

RESUMO

BACKGROUND: The Duchenne and Becker muscular dystrophies (DMD, BMD) are neuromuscular disorders commonly associated with diverse cognitive and behavioral comorbidities. Genotype-phenotype studies suggest that severity and risk of central defects in DMD patients increase with cumulative loss of different dystrophins produced in CNS from independent promoters of the DMD gene. Mutations affecting all dystrophins are nevertheless rare and therefore the clinical evidence on the contribution of the shortest Dp71 isoform to cognitive and behavioral dysfunctions is limited. In this study, we evaluated social, emotional and locomotor functions, and fear-related learning in the Dp71-null mouse model specifically lacking this short dystrophin. RESULTS: We demonstrate the presence of abnormal social behavior and ultrasonic vocalization in Dp71-null mice, accompanied by slight changes in exploratory activity and anxiety-related behaviors, in the absence of myopathy and alterations of learning and memory of aversive cue-outcome associations. CONCLUSIONS: These results support the hypothesis that distal DMD gene mutations affecting Dp71 may contribute to the emergence of social and emotional problems that may relate to the autistic traits and executive dysfunctions reported in DMD. The present alterations in Dp71-null mice may possibly add to the subtle social behavior problems previously associated with the loss of the Dp427 dystrophin, in line with the current hypothesis that risk and severity of behavioral problems in patients increase with cumulative loss of several brain dystrophin isoforms.


Assuntos
Distrofina , Camundongos Knockout , Comportamento Social , Animais , Distrofina/genética , Distrofina/deficiência , Camundongos , Masculino , Emoções/fisiologia , Medo/fisiologia , Medo/psicologia , Comportamento Animal/fisiologia , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças , Ansiedade/genética , Ansiedade/psicologia
7.
Cereb Cortex ; 33(8): 5007-5024, 2023 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-36218820

RESUMO

Social support during exposure-based psychotherapy is believed to diminish fear and improve therapy outcomes. However, some clinical trials challenge that notion. Underlying mechanisms remain unknown, hindering the understanding of benefits and pitfalls of such approach. To study social buffering during fear extinction, we developed a behavioral model in which partner's presence decreases response to fear-associated stimuli. To identify the neuronal background of this phenomenon, we combined behavioral testing with c-Fos mapping, optogenetics, and chemogenetics. We found that the presence of a partner during fear extinction training causes robust inhibition of freezing; the effect, however, disappears in subjects tested individually on the following day. It is accompanied by lowered activation of the prelimbic (PL) and anterior cingulate (ACC) but not infralimbic (IL) cortex. Accordingly, blocking of IL activity left social buffering intact. Similarly, inhibition of the ventral hippocampus-PL pathway, suppressing fear response after prolonged extinction training, did not diminish the effect. In contrast, inhibition of the ACC-central amygdala pathway, modulating social behavior, blocked social buffering. By reporting that social modulation of fear inhibition is transient and insensitive to manipulation of the fear extinction-related circuits, we show that the mechanisms underlying social buffering during extinction are different from those of individual extinction.


Assuntos
Tonsila do Cerebelo , Córtex Pré-Frontal , Humanos , Tonsila do Cerebelo/fisiologia , Córtex Pré-Frontal/fisiologia , Extinção Psicológica/fisiologia , Medo/fisiologia , Córtex Cerebral/metabolismo
8.
Cogn Emot ; : 1-17, 2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38625561

RESUMO

Despite the salient experience of encoding threatening events, these memories are prone to distortions and often non-veridical from encoding to recall. Further, threat has been shown to preferentially disrupt the binding of event details and enhance goal-relevant information. While extensive work has characterised distinctive features of emotional memory, research has not fully explored the influence threat has on temporal memory, a process putatively supported by the binding of event details into a temporal context. Two primary competing hypotheses have been proposed; that threat can impair or enhance temporal memory. We analysed two datasets to assess temporal memory for an in-person haunted house experience. In study 1, we examined the temporal structure of memory by characterising memory contiguity in free recall as a function of individual levels of heart rate as a proxy of threat. In study 2, we replicated marginal findings of threat-related increases in memory contiguity found in study 1. We extended these findings by showing threat-related increases in recency discriminations, an explicit test of temporal memory. Together, these findings demonstrate that threat enhances temporal memory regarding free recall structure and during explicit memory judgments.

9.
Int J Neurosci ; : 1-12, 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38598305

RESUMO

INTRODUCTION: Social inequality conditions induce aversion and affect brain functions and mood. This study investigated the effects of chronic social equality and inequality (CSE and CSI, respectively) conditions on passive avoidance memory and post-traumatic stress disorder (PTSD)-like behaviors in rats under chronic empathic stress. METHODS: Rats were divided into different groups, including control, sham-observer, sham-demonstrator, observer, demonstrator, and co-demonstrator groups. Chronic stress (2 h/day) was administered to all stressed groups for 21 days. Fear learning, fear memory, memory consolidation, locomotor activity, and PTSD-like behaviors were evaluated using the passive avoidance test. Apart from the hippocampal weight, the correlations of memory and right hippocampal weight with serum corticosterone (CORT) levels were separately assessed for all experimental groups. RESULTS: Latency was significantly higher in the demonstrator and sham-demonstrator groups compared to the control group. It was decreased significantly in other groups compared to the control group. Latency was also decreased in the observer and co-demonstrator groups compared to the demonstrator group. Moreover, the right hippocampal weight was significantly decreased in the demonstrator and sham-demonstrator groups compared to the control group. Pearson's correlation of memory and hippocampal weight with serum CORT levels supported the present findings. CONCLUSION: Maladaptive fear responses occurred in demonstrators and sham-demonstrators. Also, extremely high levels of psychological stress, especially under CSI conditions (causing abnormal fear learning) led to heightened fear memory and PTSD-like behaviors. Right hippocampal atrophy confirmed the potential role of CSI conditions in promoting PTSD-like behaviors. Compared to inequality conditions, the abnormal fear memory was reduced under equality conditions.

10.
J Neurosci ; 42(5): 877-893, 2022 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-34876468

RESUMO

The retrieval of recent and remote memories are thought to rely on distinct brain circuits and mechanisms. The retrosplenial cortex (RSC) is robustly activated during the retrieval of remotely acquired contextual fear memories (CFMs), but the contribution of particular subdivisions [granular (RSG) vs agranular retrosplenial area (RSA)] and the circuit mechanisms through which they interact to retrieve remote memories remain unexplored. In this study, using both anterograde and retrograde viral tracing approaches, we identified excitatory projections from layer 5 pyramidal neurons of the RSG to the CA1 stratum radiatum/lacunosum-moleculare of the dorsal hippocampus and the superficial layers of the RSA in male mice. We found that chemogenetic or optogenetic inhibition of the RSG-to-CA1, but not the RSG-to-RSA, pathway selectively impairs the retrieval of remote CFMs. Collectively, our results uncover a specific role for the RSG in remote CFM recall and provide circuit evidence that RSG-mediated remote CFM retrieval relies on direct RSG-to-CA1 connectivity. The present study provides a better understanding of brain circuit mechanisms underlying the retrieval of remote CFMs and may help guide the development of therapeutic strategies to attenuate remote traumatic memories that lead to mental health issues such as post-traumatic stress disorder.SIGNIFICANCE STATEMENT The RSC is implicated in contextual information processing and remote recall. However, how different subdivisions of the RSC and circuit mechanisms through which they interact to underlie remote memory recall remain unexplored. This study shows that granular subdivision of the RSC and its input to hippocampal area CA1 contributes to the retrieval of remote contextual fear memories. Our results support the hypothesis that the RSC and hippocampus require each other to preserve fear memories and may provide a novel therapeutic avenue to attenuate remote traumatic memories in patients with post-traumatic stress disorder.


Assuntos
Medo , Giro do Cíngulo/fisiologia , Rememoração Mental , Células Piramidais/fisiologia , Animais , Giro do Cíngulo/citologia , Hipocampo/citologia , Hipocampo/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
11.
Hippocampus ; 33(7): 862-871, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-36709413

RESUMO

Perineuronal nets (PNNs) which mostly surround the parvalbumin (PV) neurons, have been shown to play critical roles in neural plasticity. Recently, PNNs have been shown to regulate fear-associated memory, but the molecular mechanism is still unclear. In this study, we found that removal of PNNs in vivo using chondroitinase ABC (ChABC) injection resulted in reduced firing rate of PV neurons and decreased inhibitory synaptic transmission in both PV neurons and excitatory neurons in the CA1 hippocampus. Interestingly, altered synaptic transmission appears to be mediated by presynaptic changes. Furthermore, ChABC treatment disrupts long-term contextual fear memory retention. These results suggest PNNs might alter fear memory by reducing the presynaptic GABA release.


Assuntos
Matriz Extracelular , Neurônios , Neurônios/metabolismo , Matriz Extracelular/metabolismo , Hipocampo/metabolismo , Parvalbuminas/metabolismo , Medo , Ácido gama-Aminobutírico
12.
Biochem Biophys Res Commun ; 665: 118-123, 2023 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-37156049

RESUMO

The process of memory consolidation involves the synthesis of new proteins, and interfering with protein synthesis through anisomycin can impair memory. Memory deficits due to aging and sleep disorders may also result from a reduction in protein synthesis. Rescuing memory deficits caused by protein synthesis deficiency is therefore an important issue that needs to be addressed. Our study focused on the effects of cordycepin on fear memory deficits induced by anisomycin using contextual fear conditioning. We observed that cordycepin was able to attenuate these deficits and restore BDNF levels in the hippocampus. The behavioral effects of cordycepin were dependent on the BDNF/TrkB pathway, as demonstrated by the use of ANA-12. Cordycepin had no significant impact on locomotor activity, anxiety or fear memory. Our findings provide the first evidence that cordycepin can prevent anisomycin-induced memory deficits by regulating BDNF expression in the hippocampus.


Assuntos
Fator Neurotrófico Derivado do Encéfalo , Medo , Humanos , Anisomicina/farmacologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Medo/fisiologia , Transtornos da Memória/induzido quimicamente , Transtornos da Memória/tratamento farmacológico , Transtornos da Memória/metabolismo , Hipocampo/metabolismo
13.
Cogn Affect Behav Neurosci ; 23(5): 1374-1383, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37420092

RESUMO

Posttraumatic stress disorder (PTSD), a disabling and chronic condition after exposure to an extreme traumatic event, affects approximately 8% of the population worldwide. However, the underlying mechanisms of PTSD are not clear. The ability to manage fear memories is critical for PTSD. Differences in stress responsiveness and coping strategies by age represent an important starting point for the understanding and prevention of PTSD. However, we do not know whether the ability to cope with fear memories is decreased in middle-aged mice. To investigate this, we compared fear memory extinction among different age groups of mice. We found that middle-aged mice exhibited impaired fear memory extinction, which was accompanied by sustained enhanced long-term potentiation (LTP) induction in the extinction process. Most interestingly, ketamine treatment restored the impaired fear memory extinction in middle-aged mice. Moreover, ketamine could ameliorate the increased LTP during the extinction process through a presynaptic mechanism. Altogether, our results indicated that middle-aged mice were unable to extinguish fear memories, which could be treated with ketamine via presynaptic-mediated synaptic plasticity in middle-aged mice, suggesting that ketamine administration may be a new strategy for the treatment of PTSD.


Assuntos
Ketamina , Transtornos de Estresse Pós-Traumáticos , Camundongos , Animais , Ketamina/farmacologia , Extinção Psicológica , Medo , Potenciação de Longa Duração , Plasticidade Neuronal , Transtornos de Estresse Pós-Traumáticos/tratamento farmacológico , Transtornos da Memória/tratamento farmacológico
14.
Neurobiol Learn Mem ; 205: 107821, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37666411

RESUMO

Destabilization of previously consolidated memories places them in a labile state in which they are open to modification. However, strongly encoded fear memories tend to be destabilization-resistant and the conditions required to destabilize such memories remain poorly understood. Our lab has previously shown that exposure to salient novel contextual cues during memory reactivation can destabilize strongly encoded object location memories and that activity at muscarinic cholinergic receptors is critical for this effect. In the current study, we similarly targeted destabilization-resistant fear memories, hypothesizing that exposure to salient novelty at the time of reactivation would induce destabilization of strongly encoded fear memories in a muscarinic receptor-dependent manner. First, we show that contextual fear memories induced by 3 context-shock pairings readily destabilize upon memory reactivation, and that this destabilization is blocked by systemic (ip) administration of the muscarinic receptor antagonist scopolamine (0.3 mg/kg) in male rats. Following that, we confirm that this effect is dorsal hippocampus (dHPC)-dependent by targeting M1 receptors in the CA1 region with pirenzepine. Next, we show that more strongly encoded fear memories (induced with 5 context-shock pairings) resist destabilization. Consistent with our previous work, however, we report that salient novelty (a change in floor texture) presented during the reactivation session promotes destabilization of resistant contextual fear memories in a muscarinic receptor-dependent manner. Finally, the effect of salient novelty on memory destabilization was mimicked by stimulating muscarinic receptors with the selective M1 agonist CDD-0102A (ip, 0.3 mg/kg). These findings reveal further generalizability of our previous results implicating novel cues and M1 muscarinic signaling in promoting destabilization of resistant memories and suggest possible therapeutic options for disorders characterized by persistent, maladaptive fear memories such as PTSD and phobias.


Assuntos
Memória , Receptor Muscarínico M1 , Ratos , Masculino , Animais , Memória/fisiologia , Medo/fisiologia , Antagonistas Muscarínicos/farmacologia , Escopolamina/farmacologia
15.
Neurobiol Learn Mem ; 203: 107797, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37385522

RESUMO

Glucocorticoid receptors (GRs) of the basolateral amygdala (BLA) play an important role in memory reconsolidation. The present study investigated the role of the BLA GRs in the late reconsolidation of fear memory using an inhibitory avoidance (IA) task in male Wistar rats. Stainless steel cannulae were implanted bilaterally into the BLA of the rats. After 7 days of recovery, the animals were trained in a one-trial IA task (1 mA, 3 s). In Experiment One, 48 h after the training session, the animals received 3 systemic doses of corticosterone (CORT; 1, 3, or 10 mg/kg, i.p.) followed by an intra-BLA microinjection of the vehicle (0.3 µl/side) at different time points (immediately, 12, or 24 h) after memory reactivation. Memory reactivation was performed by returning the animals to the light compartment while the sliding door was open. No shock was delivered during memory reactivation. CORT (10 mg/kg) injection 12 h after memory reactivation most effectively impaired the late memory reconsolidation (LMR). In the second part of Experiment One, immediately, 12, or 24 h after memory reactivation, GR antagonist RU38486 (RU; 1 ng/0.3 µl/side) was injected into BLA following a systemic injection of CORT (10 mg/kg) to examine whether it would block the CORT effect. RU inhibited the impairing effects of CORT on LMR. In Experiment Two, the animals received CORT (10 mg/kg) with time windows immediately, 3, 6, 12, and 24 h after memory reactivation. Again, CORT (10 mg/kg) injection 12 h after memory reactivation impaired LMR. Memory reactivation was performed in the third Experiment, 7, 14, 28, or 56 days after the training session. Injection of CORT (10 mg/kg) 12 h later had no significant effect on the LMR. The impairing effect of CORT was seen only in 2-day-old but not 7, 14, 28, and 56-day-old memories. GRs located in BLA seem to play an important role in the LMR of young memory, as with increasing the age of memories, they become less sensitive to manipulation.


Assuntos
Complexo Nuclear Basolateral da Amígdala , Ratos , Masculino , Animais , Receptores de Glucocorticoides/fisiologia , Corticosterona/farmacologia , Ratos Wistar , Medo
16.
Neurobiol Learn Mem ; 205: 107829, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37734437

RESUMO

Glucocorticoid administration, before or after fear memory reactivation, impairs subsequent fear memory expression, but the underlying mechanisms are not well understood. The present study examined the role of basolateral amygdala (BLA) ß-adrenoceptors in the effects of intra-BLA corticosterone injection on fear memory in rats. Bilateral cannulae were implanted in the BLA of Wistar male rats. The rats were trained and tested using an inhibitory avoidance task (1 mA footshock for 3 s). Forty-eight hours after training, corticosterone (CORT, 5, 10, or 20 ng/0.5 µl/side) and the ß2-adrenoceptor agonist clenbuterol (CLEN, 10 or 20 ng/0.5 µl/side) or the ß-adrenoceptor antagonist propranolol (PROP, 250 or 500 ng/0.5 µl/side) were injected into the BLA before or right after memory reactivation (retrieval, Test 1). We performed subsequent tests 2 (Test 2), 5 (Test 3), 7 (Test 4), and 9 (Test 5) days after Test 1. The results demonstrated that CORT injection before Test 1 disrupted memory retrieval and reduced fear expression in Tests 2-5, possibly due to enhanced extinction or impaired reconsolidation. CORT injection after Test 1 also impaired reconsolidation and reduced fear expression in Tests 2-5. CLEN prevented, but PROP exacerbated, the effects of CORT on fear expression. The reminder shock did not recover fear memory in CORT-treated animals, suggesting that reconsolidation, not extinction, was affected. These results indicate that glucocorticoids and ß-adrenoceptors in the BLA jointly modulate fear memory reconsolidation and expression. Comprehending the neurobiology of stress and the impact of glucocorticoids on fear memory may lead to new treatments for stress and trauma-induced disorders such as PTSD.


Assuntos
Complexo Nuclear Basolateral da Amígdala , Glucocorticoides , Ratos , Masculino , Animais , Glucocorticoides/farmacologia , Corticosterona/metabolismo , Complexo Nuclear Basolateral da Amígdala/metabolismo , Ratos Wistar , Tonsila do Cerebelo/fisiologia , Medo/fisiologia , Receptores Adrenérgicos beta/metabolismo
17.
Dev Psychobiol ; 65(7): e22417, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37860902

RESUMO

Learning processes in rats during early development are importantly mediated by the mother, which represents the primary source of environmental information. This study aimed to determine whether aversive early experiences can induce the expression of pups' fear responses toward a non-aversive stimulus as a consequence of a memory process. First, we determined pups' fear responses toward an anesthetized female after being exposed to this stimulus or an empty cage together with their mothers from Postnatal Day (PNDs) 1 to 4. Second, we evaluated if the administration of the protein synthesis inhibitor cycloheximide (CHX; 0.2 mg/kg, subcutaneously (sc).) disrupted the reconsolidation processes and abolished the fear response on PND 9. Only female pups previously exposed to the female intruder expressed fear responses toward an anesthetized female on PND 8. CHX administration to female pups immediately after exposure to an anesthetized female on PND 8 suppressed fear responses on PND 9, indicating that the fear expression was the result of a memory process, probably mediated by the mother. These findings demonstrated that early experiences can shape responses to social stimuli in a sex-dependent manner and emphasize the critical role of the mother in influencing fear learning in a social context.


Assuntos
Memória , Mães , Ratos , Animais , Feminino , Humanos , Medo/fisiologia , Aprendizagem , Afeto
18.
J Integr Neurosci ; 22(4): 100, 2023 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-37519182

RESUMO

BACKGROUND: Training with inescapable shock (IS; uncontrollable stressor) is followed by significant decreases in rapid eye movement sleep (REM). However, controllability is important in the effects of stress. We examined the effects of escapable shock (ES; controllable stressor) on sleep and whether the central nucleus of the amygdala (CNA) plays a role in regulating these effects. METHODS: Six Wistar rats implanted with a cannula located in CNA underwent two days of ES training (20 shock presentations; 0.5 mA; 5.0 s maximum duration; 1.0 min interstimulus interval). Five days later, they were re-exposed to the shock context. RESULTS: Following shock training, REM was significantly increased in both light and dark periods. Non-REM (NREM) and total sleep (TS) duration were decreased during the light period. Similar effects on REM and NREM were observed following re-exposure to the training context alone. Microinjections of saline into CNA immediately following ES also produced similar increases in REM, whereas microinjections of muscimol (MUS; GABAA (γ-aminobutyric acid) antagonist) subsequent to ES blocked the increases in REM. CONCLUSIONS: These data, along with previous work with ES and IS, demonstrate that stressor controllability is important in determining how stress impacts sleep. Moreover, the results of the microinjection study indicate that the effects of ES on REM are regulated through the CNA.


Assuntos
Núcleo Central da Amígdala , Sono REM , Ratos , Animais , Sono REM/fisiologia , Ratos Wistar , Sono/fisiologia , Muscimol/farmacologia , Eletroencefalografia/métodos
19.
J Neurosci ; 41(6): 1288-1300, 2021 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-33293359

RESUMO

The retrieval of fear memory induces two opposite memory process, i.e., reconsolidation and extinction. Brief retrieval induces reconsolidation to maintain or enhance fear memory, while prolonged retrieval extinguishes this memory. Although the mechanisms of reconsolidation and extinction have been investigated, it remains unknown how fear memory phases are switched from reconsolidation to extinction during memory retrieval. Here, we show that an extracellular signal-regulated kinase (ERK)-dependent memory transition process after retrieval regulates the switch of memory phases from reconsolidation to extinction by preventing induction of reconsolidation in an inhibitory avoidance (IA) task in male mice. First, the transition memory phase, which cancels the induction of reconsolidation, but is insufficient for the acquisition of extinction, was identified after reconsolidation, but before extinction phases. Second, the reconsolidation, transition, and extinction phases after memory retrieval showed distinct molecular and cellular signatures through cAMP responsive element binding protein (CREB) and ERK phosphorylation in the amygdala, hippocampus, and medial prefrontal cortex (mPFC). The reconsolidation phase showed increased CREB phosphorylation, while the extinction phase displayed several neural populations with various combinations of CREB and/or ERK phosphorylation, in these brain regions. Interestingly, the three memory phases, including the transition phase, showed transient ERK activation immediately after retrieval. Most importantly, the blockade of ERK in the amygdala, hippocampus, or mPFC at the transition memory phase disinhibited reconsolidation-induced enhancement of IA memory. These observations suggest that the ERK-signaling pathway actively regulates the transition of memory phase from reconsolidation to extinction and this process functions as a switch that cancels reconsolidation of fear memory.SIGNIFICANCE STATEMENT Retrieval of fear memory induces two opposite memory process; reconsolidation and extinction. Reconsolidation maintains/enhances fear memory, while extinction weakens fear memory. It remains unknown how memory phases are switched from reconsolidation to extinction during retrieval. Here, we identified an active memory transition process functioning as a switch that inhibits reconsolidation. This memory transition phase showed a transient increase of extracellular signal-regulated kinase (ERK) phosphorylation in the amygdala, hippocampus and medial prefrontal cortex (mPFC). Interestingly, inhibition of ERK in these regions at the transition phase disinhibited the reconsolidation-mediated enhancement of inhibitory avoidance (IA) memory. These findings suggest that the transition memory process actively regulates the switch of fear memory phases of fear memory by preventing induction of reconsolidation through the activation of the ERK-signaling pathway.


Assuntos
Tonsila do Cerebelo/enzimologia , Extinção Psicológica/fisiologia , Hipocampo/enzimologia , Sistema de Sinalização das MAP Quinases/fisiologia , Consolidação da Memória/fisiologia , Córtex Pré-Frontal/enzimologia , Animais , Medo , Masculino , Memória/fisiologia , Camundongos , Camundongos Endogâmicos C57BL
20.
J Neurosci ; 41(6): 1174-1190, 2021 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-33303681

RESUMO

The BAD-BAX-caspase-3 cascade is a canonical apoptosis pathway. Macroautophagy ("autophagy" hereinafter) is a process by which organelles and aggregated proteins are delivered to lysosomes for degradation. Here, we report a new function of the BAD-BAX-caspase-3 cascade and autophagy in the control of synaptic vesicle pools. We found that, in hippocampal neurons of male mice, the BAD-BAX-caspase-3 pathway regulates autophagy, which in turn limits the size of synaptic vesicle pools and influences the kinetics of activity-induced depletion and recovery of synaptic vesicle pools. Moreover, the caspase-autophagy pathway is engaged by fear conditioning to facilitate associative fear learning and memory. This work identifies a new mechanism for controlling synaptic vesicle pools, and a novel, nonapoptotic, presynaptic function of the BAD-BAX-caspase-3 cascade.SIGNIFICANCE STATEMENT Despite the importance of synaptic vesicles for neurons, little is known about how the size of synaptic vesicle pools is maintained under basal conditions and regulated by neural activity. This study identifies a new mechanism for the control of synaptic vesicle pools, and a new, nonapoptotic function of the BAD-BAX-caspase-3 pathway in presynaptic terminals. Additionally, it indicates that autophagy is not only a homeostatic mechanism to maintain the integrity of cells and tissues, but also a process engaged by neural activity to regulate synaptic vesicle pools for optimal synaptic responses, learning, and memory.


Assuntos
Autofagia/fisiologia , Caspase 3/deficiência , Transdução de Sinais/fisiologia , Vesículas Sinápticas/metabolismo , Proteína X Associada a bcl-2/deficiência , Proteína de Morte Celular Associada a bcl/deficiência , Animais , Caspase 3/genética , Células Cultivadas , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Knockout , Imagem Molecular/métodos , Técnicas de Cultura de Órgãos , Vesículas Sinápticas/genética , Vesículas Sinápticas/ultraestrutura , Proteína X Associada a bcl-2/genética , Proteína de Morte Celular Associada a bcl/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA