Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 10.060
Filtrer
1.
Transl Psychiatry ; 14(1): 319, 2024 Aug 03.
Article de Anglais | MEDLINE | ID: mdl-39097609

RÉSUMÉ

Nicotine intake is linked to the maintenance and development of anxiety disorders and impairs adaptive discrimination of threat and safety in rodents and humans. Yet, it is unclear if nicotine exerts a causal pharmacological effect on the affective and neural mechanisms that underlie aversive learning. We conducted a pre-registered, pseudo-randomly and double-blinded pharmacological fMRI study to investigate the effect of acute nicotine on Fear Acquisition and Extinction in non-smokers (n = 88). Our results show that nicotine administration led to decreased discrimination between threat and safety in subjective fear. Nicotine furthermore decreased differential (threat vs. safety) activation in the hippocampus, which was functionally coupled with Nucleus Accumbens and amygdala, compared to placebo controls. Additionally, nicotine led to enhanced physiological arousal to learned threats and overactivation of the ventral tegmental area. This study provides mechanistic evidence that single doses of nicotine impair neural substrates of adaptive aversive learning in line with the risk for the development of pathological anxiety.


Sujet(s)
Amygdale (système limbique) , Peur , Hippocampe , Imagerie par résonance magnétique , Nicotine , Noyau accumbens , Humains , Nicotine/pharmacologie , Nicotine/effets indésirables , Nicotine/administration et posologie , Noyau accumbens/effets des médicaments et des substances chimiques , Noyau accumbens/imagerie diagnostique , Mâle , Hippocampe/effets des médicaments et des substances chimiques , Peur/effets des médicaments et des substances chimiques , Adulte , Amygdale (système limbique)/effets des médicaments et des substances chimiques , Amygdale (système limbique)/imagerie diagnostique , Femelle , Jeune adulte , Méthode en double aveugle , /effets des médicaments et des substances chimiques , Agonistes nicotiniques/pharmacologie , Agonistes nicotiniques/effets indésirables , Agonistes nicotiniques/administration et posologie , Extinction (psychologie)/effets des médicaments et des substances chimiques
2.
Transl Psychiatry ; 14(1): 283, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38997258

RÉSUMÉ

Return to use, or relapse, is a major challenge in the treatment of opioid use disorder (OUD). Relapse can be precipitated by several factors, including exposure to drug-conditioned cues. Identifying successful treatments to mitigate cue-induced relapse has been challenging, perhaps due to extinction memory recall (EMR) deficits. Previously, inhibition of estradiol (E2) signaling in the basolateral amygdala (BLA) impaired heroin-cue EMR. This effect was recapitulated by antagonism of BLA estrogen receptors (ER) in a sex-specific manner such that blocking ERα in males, but ERß in females, impaired EMR. However, it is unclear whether increased E2 signaling, in the BLA or systemically, enhances heroin-cue EMR. We hypothesized that ERß agonism would enhance heroin-cue EMR in a sex- and region-specific manner. To determine the capacity of E2 signaling to improve EMR, we pharmacologically manipulated ERß across several translationally designed experiments. First, male and female rats acquired heroin or sucrose self-administration. Next, during a cued extinction session, we administered diarylpropionitrile (DPN, an ERß agonist) and tested anxiety-like behavior on an open field. Subsequently, we assessed EMR in a cue-induced reinstatement test and, finally, measured ERß expression in several brain regions. Across all experiments, females took more heroin and sucrose than males and had greater responses during heroin-cued extinction. Administration of DPN in the BLA enhanced EMR in females only, driven by ERß's impacts on memory consolidation. Interestingly, however, systemic DPN administration improved EMR for heroin cues in both sexes across several different tests, but did not impact sucrose-cue EMR. Immunohistochemical analysis of ERß expression across several different brain regions showed that females only had greater expression of ERß in the basal nucleus of the BLA. Here, in several preclinical experiments, we demonstrated that ERß agonism enhances heroin-cue EMR and has potential utility in combatting cue-induced relapse.


Sujet(s)
Signaux , Récepteur bêta des oestrogènes , Extinction (psychologie) , Héroïne , Rappel mnésique , Animaux , Mâle , Femelle , Récepteur bêta des oestrogènes/agonistes , Récepteur bêta des oestrogènes/métabolisme , Héroïne/pharmacologie , Rats , Extinction (psychologie)/effets des médicaments et des substances chimiques , Extinction (psychologie)/physiologie , Rappel mnésique/effets des médicaments et des substances chimiques , Rappel mnésique/physiologie , Nitriles/pharmacologie , Groupe nucléaire basolatéral/métabolisme , Groupe nucléaire basolatéral/effets des médicaments et des substances chimiques , Propionates/pharmacologie , Facteurs sexuels , Autoadministration , Rat Sprague-Dawley , Dépendance à l'héroïne/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques
3.
Int J Mol Sci ; 25(13)2024 Jun 28.
Article de Anglais | MEDLINE | ID: mdl-39000204

RÉSUMÉ

Fear conditioning evokes a physiologic release of glucocorticoids that assists learning. As a cochaperone in the glucocorticoid receptor complex, FKBP51 modulates stress-induced glucocorticoid signaling and may influence conditioned fear responses. This study combines molecular and behavioral approaches to examine whether locally reducing FKBP51 expression in the ventral hippocampus is sufficient to affect fear-related behaviors. We hypothesized that reducing FKBP51 expression in the VH would increase glucocorticoid signaling to alter auditory fear conditioning. Adult male rats were injected with an adeno-associated virus (AAV) vector expressing short hairpin - RNAs (shRNA) targeting FKBP5 into the ventral hippocampus to reduce FKBP5 levels or a control AAV. Infusion of FKBP5-shRNA into the ventral hippocampus decreased auditory fear acquisition and recall. Although animals injected with FKBP5-shRNA showed less freezing during extinction recall, the difference was due to a reduced fear recall rather than improved extinction. Reducing ventral hippocampus FKBP51 did not affect exploratory behavior in either the open field test or the elevated zero maze test but did increase passive behavior in the forced swim test, suggesting that the reduction in auditory fear recall was not due to more active responses to acute stress. Furthermore, lower ventral hippocampus FKBP51 levels did not alter corticosterone release in response to restraint stress, suggesting that the reduced fear recall was not due to lower corticosterone release. Our findings suggest FKBP51 in the ventral hippocampus plays a selective role in modulating fear-learning processes and passive behavioral responses to acute stress rather than hypothalamic-pituitary-adrenal axis reactivity or exploratory responses.


Sujet(s)
Peur , Hippocampe , Protéines de liaison au tacrolimus , Animaux , Mâle , Peur/physiologie , Protéines de liaison au tacrolimus/métabolisme , Protéines de liaison au tacrolimus/génétique , Hippocampe/métabolisme , Rats , Corticostérone/métabolisme , Corticostérone/sang , Rat Sprague-Dawley , Petit ARN interférent/métabolisme , Petit ARN interférent/génétique , Récepteurs aux glucocorticoïdes/métabolisme , Extinction (psychologie)/physiologie
4.
Eur J Psychotraumatol ; 15(1): 2364441, 2024.
Article de Anglais | MEDLINE | ID: mdl-38973398

RÉSUMÉ

Background: Trauma-focused treatments for post-traumatic stress disorder (PTSD) are effective for many patients. However, relapse may occur when acquired extinction memories fail to generalize beyond treatment contexts. A subgroup of PTSD patients - potentially with substantial exposure to early-life adversity (ELA) - show dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis, which results in lower cortisol levels. Glucocorticoids, including cortisol, appear to facilitate strength and generalization of emotional memories.Objective: We describe the protocol of an integrated PTSD study. We investigate (A) associations between HPA-axis dysregulation, ELA, epigenetic markers, and PTSD treatment outcome (observational study); and (B) effects of exogenous glucocorticoids on strength and generalization of extinction memories and associated neural mechanisms [pharmacological intervention study with functional magnetic resonance imaging (fMRI)]. The objective is to provide proof of concept that PTSD patients with HPA-axis dysregulation often experienced ELA and may show improved strength and generalization of extinction learning after glucocorticoid administration.Method: The observational study (n = 160 PTSD group, n = 30 control group) assesses ELA, follow-up PTSD symptoms, epigenetic markers, and HPA-axis characteristics (salivary cortisol levels during low-dose dexamethasone suppression test and socially evaluated cold-pressor test). The pharmacological intervention study (n = 80 PTSD group, with and without HPA-axis dysregulation) is a placebo-controlled fMRI study with a crossover design. To investigate strength and generalization of extinction memories, we use a differential fear acquisition, extinction, and extinction recall task with spatial contexts within a virtual environment. Prior to extinction learning, 20 mg hydrocortisone or placebo is administered. During next-day recall, strength of the extinction memory is determined by recovery of skin conductance and pupil dilation differential responding, whereas generalization is assessed by comparing responses between different spatial contexts.Conclusion: The integrated study described in the current protocol paper could inform a personalized treatment approach in which these PTSD patients may receive glucocorticoids as a treatment enhancer in trauma-focused therapies.Trial registration: The research project is registered in the European Union Drug Regulating Authorities Clinical Trials (EudraCT) database, https://eudract.ema.europa.eu/, EudraCT number 2020-000712-30.


This protocol reports a proof-of-concept study for glucocorticoids as an enhancer for PTSD treatment.The study examines whether glucocorticoids enhance the strength and generalization of extinction memory.A further aim is to identify HPA-axis-related PTSD subgroups that may particularly benefit.


Sujet(s)
Extinction (psychologie) , Glucocorticoïdes , Axe hypothalamohypophysaire , Axe hypophyso-surrénalien , Troubles de stress post-traumatique , Humains , Troubles de stress post-traumatique/traitement médicamenteux , Extinction (psychologie)/effets des médicaments et des substances chimiques , Extinction (psychologie)/physiologie , Glucocorticoïdes/pharmacologie , Axe hypothalamohypophysaire/effets des médicaments et des substances chimiques , Axe hypophyso-surrénalien/effets des médicaments et des substances chimiques , Hydrocortisone , Mâle , Adulte , Femelle , Imagerie par résonance magnétique
5.
Elife ; 132024 Jul 19.
Article de Anglais | MEDLINE | ID: mdl-39027985

RÉSUMÉ

How is new information organized in memory? According to latent state theories, this is determined by the level of surprise, or prediction error, generated by the new information: a small prediction error leads to the updating of existing memory, large prediction error leads to encoding of a new memory. We tested this idea using a protocol in which rats were first conditioned to fear a stimulus paired with shock. The stimulus was then gradually extinguished by progressively reducing the shock intensity until the stimulus was presented alone. Consistent with latent state theories, this gradual extinction protocol (small prediction errors) was better than standard extinction (large prediction errors) in producing long-term suppression of fear responses, and the benefit of gradual extinction was due to updating of the conditioning memory with information about extinction. Thus, prediction error determines how new information is organized in memory, and latent state theories adequately describe the ways in which this occurs.


Sujet(s)
Encéphale , Peur , Mémoire , Animaux , Rats , Mémoire/physiologie , Peur/physiologie , Encéphale/physiologie , Mâle , Extinction (psychologie)/physiologie , Conditionnement classique/physiologie
6.
Int Rev Neurobiol ; 177: 235-250, 2024.
Article de Anglais | MEDLINE | ID: mdl-39029986

RÉSUMÉ

Cannabidiol (CBD) modulates aversive memory and its extinction, with potential implications for treating anxiety- and stress-related disorders. Here, we summarize and discuss scientific evidence showing that CBD administered after the acquisition (consolidation) and retrieval (reconsolidation) of fear memory attenuates it persistently in rats and mice. CBD also reduces fear expression and enhances fear extinction. These effects involve the activation of cannabinoid type-1 (CB1) receptors in the dorsal hippocampus, bed nucleus of stria terminalis, and medial prefrontal cortex, comprising the anterior cingulate, prelimbic, and infralimbic subregions. Serotonin type-1A (5-HT1A) receptors also mediate some CBD effects on fear memory. CBD effects on fear memory acquisition vary, depending on the aversiveness of the conditioning procedure. While rodent findings are relatively consistent and encouraging, human studies investigating CBD's efficacy in modulating aversive/traumatic memories are still limited. More studies are needed to investigate CBD's effects on maladaptive, traumatic memories, particularly in post-traumatic stress disorder patients.


Sujet(s)
Cannabidiol , Peur , Troubles de stress post-traumatique , Animaux , Cannabidiol/pharmacologie , Peur/effets des médicaments et des substances chimiques , Peur/physiologie , Troubles de stress post-traumatique/traitement médicamenteux , Troubles de stress post-traumatique/physiopathologie , Humains , Extinction (psychologie)/effets des médicaments et des substances chimiques , Extinction (psychologie)/physiologie
7.
Biol Psychiatry ; 95(8): 785-799, 2024 Apr 15.
Article de Anglais | MEDLINE | ID: mdl-38952926

RÉSUMÉ

Background: Responding to social signals by expressing the correct behavior is not only challenged in autism, but also in diseases with high prevalence of autism, like Prader-Willi Syndrome (PWS). Clinical evidence suggests aberrant pro-social behavior in patients can be regulated by intranasal oxytocin (OXT) or vasopressin (AVP). However, what neuronal mechanisms underlie impaired behavioral responses in a socially-aversive context, and how can they be corrected, remains largely unknown. Methods: Using the Magel2 knocked-out (KO) mouse model of PWS (crossed with CRE-dependent transgenic lines), we devised optogenetic, physiological and pharmacological strategies in a social-fear-conditioning paradigm. Pathway specific roles of OXT and AVP signaling were investigated converging on the lateral septum (LS), a region which receives dense hypothalamic inputs. Results: OXT and AVP signaling promoted inhibitory synaptic transmission in the LS, which failure in Magel2KO mice disinhibited somatostatin (SST) neurons and disrupted social-fear extinction. The source of OXT and AVP deficits mapped specifically in the supraoptic nucleus→LS pathway of Magel2KO mice disrupting social-fear extinction, which could be corrected by optogenetic or pharmacological inhibition of SST-neurons in the LS. Interestingly, LS SST-neurons also gated the expression of aggressive behavior, possibly as part of functional units operating beyond local septal circuits. Conclusions: SST cells in the LS play a crucial role in integration and expression of disrupted neuropeptide signals in autism, thereby altering the balance in expression of safety versus fear. Our results uncover novel mechanisms underlying dysfunction in a socially-aversive context, and provides a new framework for future treatments in autism-spectrum disorders.


Sujet(s)
Modèles animaux de maladie humaine , Extinction (psychologie) , Peur , Souris knockout , Neurones , Ocytocine , Syndrome de Prader-Willi , Somatostatine , Vasopressines , Animaux , Ocytocine/pharmacologie , Somatostatine/pharmacologie , Somatostatine/métabolisme , Peur/effets des médicaments et des substances chimiques , Peur/physiologie , Extinction (psychologie)/effets des médicaments et des substances chimiques , Extinction (psychologie)/physiologie , Neurones/effets des médicaments et des substances chimiques , Neurones/métabolisme , Souris , Syndrome de Prader-Willi/physiopathologie , Syndrome de Prader-Willi/traitement médicamenteux , Vasopressines/métabolisme , Agressivité/effets des médicaments et des substances chimiques , Agressivité/physiologie , Mâle , Comportement social , Noyaux du septum/effets des médicaments et des substances chimiques , Noyaux du septum/métabolisme , Optogénétique , Souris de lignée C57BL , Protéines et peptides de signalisation intracellulaire , Protéines intrinsèquement désordonnées
8.
Adv Neurobiol ; 38: 149-161, 2024.
Article de Anglais | MEDLINE | ID: mdl-39008015

RÉSUMÉ

Fear attenuation is an etiologically relevant process for animal survival, since once acquired information needs to be continuously updated in the face of changing environmental contingencies. Thus, when situations are encountered that were originally perceived as fearful but are no longer so, fear must be attenuated, otherwise, it risks becoming maladaptive. But what happens to the original memory trace of fear during fear attenuation? In this chapter, we review the studies that have started to approach this question from an engram perspective. We find evidence pointing to both the original memory trace of fear being suppressed, as well as it being updated towards safety. These seemingly conflicting results reflect a well-established dichotomy in the field of fear memory attenuation, namely whether fear attenuation is mediated by an inhibitory mechanism that suppresses fear expression, called extinction, or by an updating mechanism that allows the fear memory to reconsolidate in a different form, called reconsolidation-updating. Which of these scenarios takes the upper hand is ultimately influenced by the behavioral paradigms used to induce fear attenuation, but is an important area for further study as the precise cell populations underlying fear attenuation and the molecular mechanisms therein can now be understood at unprecedented resolution.


Sujet(s)
Extinction (psychologie) , Peur , Mémoire , Animaux , Humains , Consolidation de la mémoire/physiologie
9.
Neurobiol Learn Mem ; 213: 107960, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39004160

RÉSUMÉ

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.


Sujet(s)
Conditionnement classique , Peur , Propranolol , Peur/effets des médicaments et des substances chimiques , Peur/physiologie , Animaux , Mâle , Propranolol/pharmacologie , Femelle , Souris , Conditionnement classique/effets des médicaments et des substances chimiques , Consolidation de la mémoire/effets des médicaments et des substances chimiques , Consolidation de la mémoire/physiologie , Souris de lignée C57BL , Mémoire/effets des médicaments et des substances chimiques , Mémoire/physiologie , /effets des médicaments et des substances chimiques , /physiologie , Extinction (psychologie)/effets des médicaments et des substances chimiques
10.
Commun Biol ; 7(1): 728, 2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38877285

RÉSUMÉ

Benzodiazepines, commonly used for anxiolytics, hinder conditioned fear extinction, and the underlying circuit mechanisms are unclear. Utilizing remimazolam, an ultra-short-acting benzodiazepine, here we reveal its impact on the thalamic nucleus reuniens (RE) and interconnected hippocamposeptal circuits during fear extinction. Systemic or RE-specific administration of remimazolam impedes fear extinction by reducing RE activation through A type GABA receptors. Remimazolam enhances long-range GABAergic inhibition from lateral septum (LS) to RE, underlying the compromised fear extinction. RE projects to ventral hippocampus (vHPC), which in turn sends projections characterized by feed-forward inhibition to the GABAergic neurons of the LS. This is coupled with long-range GABAergic projections from the LS to RE, collectively constituting an overall positive feedback circuit construct that promotes fear extinction. RE-specific remimazolam negates the facilitation of fear extinction by disrupting this circuit. Thus, remimazolam in RE disrupts fear extinction caused by hippocamposeptal intermediation, offering mechanistic insights for the dilemma of combining anxiolytics with extinction-based exposure therapy.


Sujet(s)
Benzodiazépines , Extinction (psychologie) , Peur , Hippocampe , Noyaux médians du thalamus , Peur/effets des médicaments et des substances chimiques , Animaux , Benzodiazépines/pharmacologie , Hippocampe/effets des médicaments et des substances chimiques , Hippocampe/physiologie , Hippocampe/métabolisme , Extinction (psychologie)/effets des médicaments et des substances chimiques , Mâle , Noyaux médians du thalamus/effets des médicaments et des substances chimiques , Noyaux médians du thalamus/physiologie , Noyaux médians du thalamus/métabolisme , Rats , Anxiolytiques/pharmacologie , Souris
11.
PLoS One ; 19(6): e0305066, 2024.
Article de Anglais | MEDLINE | ID: mdl-38843228

RÉSUMÉ

A large body of evidence has shown that treatments that interfere with memory consolidation become ineffective when animals are subjected to an intense learning experience; this effect has been observed after systemic and local administration of amnestic drugs into several brain areas, including the striatum. However, the effects of amnestic treatments on the process of extinction after intense training have not been studied. Previous research demonstrated increased spinogenesis in the dorsomedial striatum, but not in the dorsolateral striatum after intense training, indicating that the dorsomedial striatum is involved in the protective effect of intense training. To investigate this issue, male Wistar rats, previously trained with low, moderate, or high levels of foot shock, were used to study the effect of tetrodotoxin inactivation of dorsomedial striatum on memory consolidation and subsequent extinction of inhibitory avoidance. Performance of the task was evaluated during seven extinction sessions. Tetrodotoxin produced a marked deficit of memory consolidation of inhibitory avoidance trained with low and moderate intensities of foot shock, but normal consolidation occurred when a relatively high foot shock was used. The protective effect of intense training was long-lasting, as evidenced by the high resistance to extinction exhibited throughout the extinction sessions. We discuss the possibility that increased dendritic spinogenesis in dorsomedial striatum may underly this protective effect, and how this mechanism may be related to the resilient memory typical of post-traumatic stress disorder (PTSD).


Sujet(s)
Apprentissage par évitement , Corps strié , Extinction (psychologie) , Rat Wistar , Tétrodotoxine , Animaux , Mâle , Extinction (psychologie)/effets des médicaments et des substances chimiques , Extinction (psychologie)/physiologie , Rats , Apprentissage par évitement/effets des médicaments et des substances chimiques , Apprentissage par évitement/physiologie , Corps strié/physiologie , Corps strié/effets des médicaments et des substances chimiques , Tétrodotoxine/pharmacologie , Consolidation de la mémoire/effets des médicaments et des substances chimiques , Consolidation de la mémoire/physiologie , Amnésie/physiopathologie , Amnésie/prévention et contrôle , Électrochoc
12.
Trials ; 25(1): 396, 2024 Jun 19.
Article de Anglais | MEDLINE | ID: mdl-38898471

RÉSUMÉ

BACKGROUND: Fear memory extinction is closely related to insomnia. Repetitive transcranial magnetic stimulation (rTMS) is safe and effective for treating insomnia disorder (ID), and it has been shown to be an efficient method for modulating fear extinction. However, whether rTMS can improve fear extinction memory in ID patients remains to be studied. In this study, we specifically aim to (1) show that 1 Hz rTMS stimulation could improve fear extinction memory in ID patients and (2) examine whether changes in sleep mediate this impact. METHODS AND DESIGN: We propose a parallel group randomised controlled trial of 62 ID participants who meet the inclusion criteria. Participants will be assigned to a real rTMS group or a sham rTMS group. The allocation ratio will be 1:1, with 31 subjects in each group. Interventions will be administered five times per week over a 4-week period. The assessments will take place at baseline (week 0), post-intervention (week 4), and 8-week follow-up (week 8). The primary outcome measure of this study will be the mean change in the Pittsburgh Sleep Quality Index (PSQI) scores from baseline to post-intervention at week 4. The secondary outcome measures include the mean change in skin conductance response (SCR), fear expectation during fear extinction, Insomnia Severity Index (ISI), Zung Self-Rating Anxiety Scale (SAS), and the Zung Self-Rating Depression Scale (SDS). DISCUSSION: This study will be the first examination of the impact of rTMS on fear memory extinction in ID patients. TRIAL REGISTRATION: Chinese Clinical Trials Register ChiCTR2300076097. Registered on 25 September 2021.


Sujet(s)
Extinction (psychologie) , Peur , Essais contrôlés randomisés comme sujet , Troubles de l'endormissement et du maintien du sommeil , Stimulation magnétique transcrânienne , Humains , Troubles de l'endormissement et du maintien du sommeil/thérapie , Troubles de l'endormissement et du maintien du sommeil/physiopathologie , Troubles de l'endormissement et du maintien du sommeil/psychologie , Stimulation magnétique transcrânienne/méthodes , Adulte , Résultat thérapeutique , Adulte d'âge moyen , Femelle , Mâle , Mémoire , Jeune adulte , Facteurs temps , Adolescent , Sommeil
13.
Elife ; 132024 Jun 24.
Article de Anglais | MEDLINE | ID: mdl-38913410

RÉSUMÉ

Downregulating emotional overreactions toward threats is fundamental for developing treatments for anxiety and post-traumatic disorders. The prefrontal cortex (PFC) is critical for top-down modulatory processes, and despite previous studies adopting repetitive transcranial magnetic stimulation (rTMS) over this region provided encouraging results in enhancing extinction, no studies have hitherto explored the effects of stimulating the medial anterior PFC (aPFC, encompassing the Brodmann area 10) on threat memory and generalization. Here we showed that rTMS over the aPFC applied before threat memory retrieval immediately decreases implicit reactions to learned and novel stimuli in humans. These effects enduringly persisted 1 week later in the absence of rTMS. No effects were detected on explicit recognition. Critically, rTMS over the aPFC resulted in a more pronounced reduction of defensive responses compared to rTMS targeting the dorsolateral PFC. These findings reveal a previously unexplored prefrontal region, the modulation of which can efficiently and durably inhibit implicit reactions to learned threats. This represents a significant advancement toward the long-term deactivation of exaggerated responses to threats.


Sujet(s)
Peur , Cortex préfrontal , Stimulation magnétique transcrânienne , Humains , Peur/physiologie , Cortex préfrontal/physiologie , Mâle , Jeune adulte , Femelle , Adulte , Extinction (psychologie)/physiologie
15.
Transl Psychiatry ; 14(1): 242, 2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38844463

RÉSUMÉ

It has been well established that a consolidated memory can be updated during the plastic state induced by reactivation. This updating process opens the possibility to modify maladaptive memory. In the present study, we evaluated whether fear memory could be updated to less-aversive level by incorporating hedonic information during reactivation. Thus, male rats were fear conditioned and, during retrieval, a female was presented as a social rewarding stimulus. We found that memory reactivation with a female (but not a male) reduces fear expression within-session and in the test, without presenting reinstatement or spontaneous recovery. Interestingly, this intervention impaired extinction. Finally, we demonstrated that this emotional remodeling to eliminate fear expression requires the activation of dopamine and oxytocin receptors during retrieval. Hence, these results shed new lights on the memory updating process and suggests that the exposure to natural rewarding information such as a female during retrieval reduces a previously consolidated fear memory.


Sujet(s)
Peur , Récepteurs à l'ocytocine , Interaction sociale , Animaux , Peur/physiologie , Mâle , Rats , Récepteurs à l'ocytocine/métabolisme , Femelle , Mémoire/physiologie , Extinction (psychologie)/physiologie , Récepteurs dopaminergiques/métabolisme , Conditionnement classique/physiologie , Récompense , Rat Wistar , Consolidation de la mémoire/physiologie
16.
BMC Psychol ; 12(1): 324, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38831468

RÉSUMÉ

Cognitive functions, such as learning and memory processes, depend on effective communication between brain regions which is facilitated by white matter tracts (WMT). We investigated the microstructural properties and the contribution of WMT to extinction learning and memory in a predictive learning task. Forty-two healthy participants completed an extinction learning paradigm without a fear component. We examined differences in microstructural properties using diffusion tensor imaging to identify underlying neural connectivity and structural correlates of extinction learning and their potential implications for the renewal effect. Participants with good acquisition performance exhibited higher fractional anisotropy (FA) in WMT including the bilateral inferior longitudinal fasciculus (ILF) and the right temporal part of the cingulum (CNG). This indicates enhanced connectivity and communication between brain regions relevant to learning and memory resulting in better learning performance. Our results suggest that successful acquisition and extinction performance were linked to enhanced structural connectivity. Lower radial diffusivity (RD) in the right ILF and right temporal part of the CNG was observed for participants with good acquisition learning performance. This observation suggests that learning difficulties associated with increased RD may potentially be due to less myelinated axons in relevant WMT. Also, participants with good acquisition performance were more likely to show a renewal effect. The results point towards a potential role of structural integrity in extinction-relevant WMT for acquisition and extinction.


Sujet(s)
Imagerie par tenseur de diffusion , Extinction (psychologie) , Substance blanche , Humains , Mâle , Femelle , Imagerie par tenseur de diffusion/méthodes , Substance blanche/imagerie diagnostique , Adulte , Jeune adulte , Extinction (psychologie)/physiologie , Apprentissage/physiologie , Voies nerveuses/imagerie diagnostique , Gyrus du cingulum/imagerie diagnostique , Gyrus du cingulum/anatomie et histologie , Anisotropie
17.
Neurosci Lett ; 836: 137888, 2024 Jul 27.
Article de Anglais | MEDLINE | ID: mdl-38945352

RÉSUMÉ

There are currently no FDA-approved treatments for cocaine use disorder. Recent preclinical and clinical studies showed that deep brain stimulation (DBS) in limbic regions reduced drug seeking behavior. Our previous work indicated that DBS of the nucleus accumbens shell attenuated reinstatement of cocaine seeking, a model of relapse, in male rats. The current experiments were designed to evaluate the effect of electrical DBS on cocaine reinstatement in female rats across the estrous cycle. Rats were allowed to self-administer cocaine and lever responding was subsequently extinguished. Cocaine seeking was reinstated by an acute injection of experimenter-delivered cocaine. The effect of nucleus accumbens shell DBS vs. sham stimulation on cocaine-primed reinstatement was evaluated in female and male rats using a within-subjects counterbalanced design. Consistent with previous work, accumbens shell DBS suppressed cocaine seeking in male rats. In sharp contrast, accumbens shell DBS had no effect on cocaine reinstatement in female rats evaluated in either the estrus or non-estrus phases. These results suggest that changes across the estrous cycle are not responsible for the differences in the effect of DBS on cocaine reinstatement between female and male rats.


Sujet(s)
Cocaïne , Stimulation cérébrale profonde , Comportement de recherche de substances , Cycle oestral , Noyau accumbens , Autoadministration , Animaux , Femelle , Mâle , Stimulation cérébrale profonde/méthodes , Rats , Noyau accumbens/effets des médicaments et des substances chimiques , Cocaïne/administration et posologie , Comportement de recherche de substances/physiologie , Comportement de recherche de substances/effets des médicaments et des substances chimiques , Cycle oestral/physiologie , Troubles liés à la cocaïne/thérapie , Troubles liés à la cocaïne/psychologie , Rat Sprague-Dawley , Extinction (psychologie)/effets des médicaments et des substances chimiques , Caractères sexuels
18.
J Appl Behav Anal ; 57(3): 615-634, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38825878

RÉSUMÉ

Extinction bursts, or temporary increases in rates and intensities of behavior during extinction, can preclude the inclusion of extinction in intervention packages meant to suppress severe challenging behavior. To identify underlying behavioral mechanisms responsible for response persistence and bursting, 69 adults with developmental disabilities completed a low-stakes translational investigation employing a 2 × 2 factorial, crossed, and randomized matched blocks design, with batched randomization logic. In each of the four test groups, we made distinct antecedent manipulations with two value parameters commonly studied through behavioral economics (i.e., demand intensity, Pmax) and evaluated the extent to which each of these manipulations influenced target responding during extinction. Although we found statistically significant differences attributable to both parameters, variations in reinforcer consumption relative to demand intensity were most influential across all dependent variables. This outcome implicates consumption relative to demand intensity as both a mitigating and exacerbating preextinction factor that influences the prevalence of adverse collateral extinction effects (e.g., bursts).


Sujet(s)
Extinction (psychologie) , , Humains , Mâle , Femelle , Adulte , Incapacités de développement/psychologie , Incapacités de développement/rééducation et réadaptation , Jeune adulte , Adulte d'âge moyen , Adolescent
19.
Neurosci Biobehav Rev ; 163: 105751, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38838877

RÉSUMÉ

A growing literature has sought to include mental imagery in fear conditioning studies. Imaginal extinction and imagery rescripting are mental imagery-based interventions that reduce conditioned fear. In the current study, we reviewed the recent findings on the efficacy of imaginal extinction and imagery rescripting as interventions to attenuate conditioned fear responses among healthy individuals. In accordance with the PRISMA guidelines, we conducted a literature search in four databases, PubMed, Scopus, Science Direct, and Web of Science to find published original empirical articles involving imagery-based interventions using a fear conditioning paradigm. The inclusion criteria were (i) use of an imagery-based intervention (either imaginal extinction or imagery rescripting), and (ii) use of a differential fear conditioning paradigm. 13 original articles reporting 15 experimental studies were included in the review. The review revealed that imagery-based interventions are effective in reducing conditioned fear. Although studies have shown that imaginal extinction and standard extinction have comparable effects in fear extinction, many studies have not been conducted to confirm the findings, or explore the underlying mechanisms. We also found the need for a standardized intervention protocol to enhance experimental control in intervention-based fear conditioning studies.


Sujet(s)
Extinction (psychologie) , Peur , , Humains , Peur/physiologie , Extinction (psychologie)/physiologie , /méthodes , Conditionnement classique/physiologie , Imagination/physiologie , Conditionnement psychologique/physiologie
20.
Neurosci Biobehav Rev ; 163: 105762, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38857666

RÉSUMÉ

The reuniens (Re) nucleus is located in the ventral midline thalamus. It has fostered increasing interest, not only for its participation in a variety of cognitive functions (e.g., spatial working memory, systemic consolidation, reconsolidation, extinction of fear or generalization), but also for its neuroanatomical positioning as a bidirectional relay between the prefrontal cortex (PFC) and the hippocampus (HIP). In this review we compile and discuss recent studies having tackled a possible implication of the Re nucleus in behavioral flexibility, a major PFC-dependent executive function controlling goal-directed behaviors. Experiments considered explored a possible role for the Re nucleus in perseveration, reversal learning, fear extinction, and set-shifting. They point to a contribution of this nucleus to behavioral flexibility, mainly by its connections with the PFC, but possibly also by those with the hippocampus, and even with the amygdala, at least for fear-related behavior. As such, the Re nucleus could be a crucial crossroad supporting a PFC-orchestrated ability to cope with new, potentially unpredictable environmental contingencies, and thus behavioral flexibility and adaption.


Sujet(s)
Noyaux médians du thalamus , Animaux , Noyaux médians du thalamus/physiologie , Humains , Peur/physiologie , Cortex préfrontal/physiologie , Extinction (psychologie)/physiologie , Hippocampe/physiologie , Fonction exécutive/physiologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE