Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.039
Filter
1.
Eur J Psychotraumatol ; 15(1): 2364441, 2024.
Article in English | MEDLINE | ID: mdl-38973398

ABSTRACT

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.


Subject(s)
Extinction, Psychological , Glucocorticoids , Hypothalamo-Hypophyseal System , Pituitary-Adrenal System , Stress Disorders, Post-Traumatic , Humans , Stress Disorders, Post-Traumatic/drug therapy , Extinction, Psychological/drug effects , Extinction, Psychological/physiology , Glucocorticoids/pharmacology , Hypothalamo-Hypophyseal System/drug effects , Pituitary-Adrenal System/drug effects , Hydrocortisone , Male , Adult , Female , Magnetic Resonance Imaging
2.
Biol Psychiatry ; 95(8): 785-799, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38952926

ABSTRACT

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.


Subject(s)
Disease Models, Animal , Extinction, Psychological , Fear , Mice, Knockout , Neurons , Oxytocin , Prader-Willi Syndrome , Somatostatin , Vasopressins , Animals , Oxytocin/pharmacology , Somatostatin/pharmacology , Somatostatin/metabolism , Fear/drug effects , Fear/physiology , Extinction, Psychological/drug effects , Extinction, Psychological/physiology , Neurons/drug effects , Neurons/metabolism , Mice , Prader-Willi Syndrome/physiopathology , Prader-Willi Syndrome/drug therapy , Vasopressins/metabolism , Aggression/drug effects , Aggression/physiology , Male , Social Behavior , Septal Nuclei/drug effects , Septal Nuclei/metabolism , Optogenetics , Mice, Inbred C57BL , Intracellular Signaling Peptides and Proteins , Intrinsically Disordered Proteins
3.
Commun Biol ; 7(1): 728, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38877285

ABSTRACT

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.


Subject(s)
Benzodiazepines , Extinction, Psychological , Fear , Hippocampus , Midline Thalamic Nuclei , Fear/drug effects , Animals , Benzodiazepines/pharmacology , Hippocampus/drug effects , Hippocampus/physiology , Hippocampus/metabolism , Extinction, Psychological/drug effects , Male , Midline Thalamic Nuclei/drug effects , Midline Thalamic Nuclei/physiology , Midline Thalamic Nuclei/metabolism , Rats , Anti-Anxiety Agents/pharmacology , Mice
4.
BMC Psychol ; 12(1): 358, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38890761

ABSTRACT

BACKGROUND: Fear overgeneralization constitutes a susceptibility factor contributing to the development and maintenance of anxiety spectrum disorders. Extant research has demonstrated that exposure to positive and supportive social relationships attenuates fear acquisition and promotes the extinction of conditioned fear responses. However, the literature lacks investigation into the effect of secure attachment priming on inhibiting the generalization of conditioned fear. METHODS: In this study, college students were recruited via online platforms to voluntarily engage in the experimental procedures, resulting in 57 subjects whose data were deemed suitable for analysis. The experimental protocol consisted of four consecutive phases: pre-acquisition, acquisition, priming, and generalization. The priming phase consisted of two experimental conditions: secure attachment priming (experimental group) and positive emotion priming (control group). This study adopted the perceptual discrimination fear conditioning paradigm, employing subjective expectancy of shock ratings and skin conductance responses as primary assessment indices. Individual difference variables were measured using corresponding psychological measurement scales. RESULTS: In terms of generalization degree, a notable divergence surfaced in the skin conductance responses across various generalization materials between the secure attachment priming group and the control group. Similarly, during generalization extinction, a significant disparity emerged in the skin conductance responses across different generalization phases between the secure attachment priming group and the control group. In addition, individual differences analyses revealed that the inhibitory effect of secure attachment priming on fear generalization was not affected by intolerance of uncertainty and attachment orientations. Conversely, slope analyses confirmed that as intolerance of uncertainty increased, the inhibitory effect of positive emotion priming on fear generalization was attenuated. CONCLUSION: The findings suggest that activating participants' representations of secure attachment via imagination effectively attenuates the generalization of perceptual fear at the physiological level. The inhibitory effect of secure attachment priming appears to be distinct from positive emotional modulation and remains unaffected by individual trait attachment styles. These results offer novel insights and avenues for the prevention and clinical intervention of anxiety spectrum disorders.


Subject(s)
Conditioning, Classical , Fear , Generalization, Psychological , Object Attachment , Humans , Fear/psychology , Male , Female , Young Adult , Adult , Conditioning, Classical/physiology , Galvanic Skin Response/physiology , Extinction, Psychological/physiology , Adolescent
5.
Elife ; 132024 Jun 24.
Article in English | MEDLINE | ID: mdl-38913410

ABSTRACT

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.


Subject(s)
Fear , Prefrontal Cortex , Transcranial Magnetic Stimulation , Humans , Fear/physiology , Prefrontal Cortex/physiology , Male , Young Adult , Female , Adult , Extinction, Psychological/physiology
6.
BMC Psychol ; 12(1): 324, 2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38831468

ABSTRACT

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.


Subject(s)
Diffusion Tensor Imaging , Extinction, Psychological , White Matter , Humans , Male , Female , Diffusion Tensor Imaging/methods , White Matter/diagnostic imaging , Adult , Young Adult , Extinction, Psychological/physiology , Learning/physiology , Neural Pathways/diagnostic imaging , Gyrus Cinguli/diagnostic imaging , Gyrus Cinguli/anatomy & histology , Anisotropy
7.
PLoS One ; 19(6): e0305066, 2024.
Article in English | MEDLINE | ID: mdl-38843228

ABSTRACT

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).


Subject(s)
Avoidance Learning , Corpus Striatum , Extinction, Psychological , Rats, Wistar , Tetrodotoxin , Animals , Male , Extinction, Psychological/drug effects , Extinction, Psychological/physiology , Rats , Avoidance Learning/drug effects , Avoidance Learning/physiology , Corpus Striatum/physiology , Corpus Striatum/drug effects , Tetrodotoxin/pharmacology , Memory Consolidation/drug effects , Memory Consolidation/physiology , Amnesia/physiopathology , Amnesia/prevention & control , Electroshock
8.
Trials ; 25(1): 396, 2024 Jun 19.
Article in English | MEDLINE | ID: mdl-38898471

ABSTRACT

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.


Subject(s)
Extinction, Psychological , Fear , Randomized Controlled Trials as Topic , Sleep Initiation and Maintenance Disorders , Transcranial Magnetic Stimulation , Humans , Sleep Initiation and Maintenance Disorders/therapy , Sleep Initiation and Maintenance Disorders/physiopathology , Sleep Initiation and Maintenance Disorders/psychology , Transcranial Magnetic Stimulation/methods , Adult , Treatment Outcome , Middle Aged , Female , Male , Memory , Young Adult , Time Factors , Adolescent , Sleep
10.
Transl Psychiatry ; 14(1): 242, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38844463

ABSTRACT

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.


Subject(s)
Fear , Receptors, Oxytocin , Social Interaction , Animals , Fear/physiology , Male , Rats , Receptors, Oxytocin/metabolism , Female , Memory/physiology , Extinction, Psychological/physiology , Receptors, Dopamine/metabolism , Conditioning, Classical/physiology , Reward , Rats, Wistar , Memory Consolidation/physiology
11.
Behav Ther ; 55(4): 724-737, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38937046

ABSTRACT

Prior research has demonstrated that conducting acquisition in multiple contexts results in more responding to the point that it can even nullify the benefit of subsequent extinction in multiple contexts on reducing renewal of excitatory responding. The underlying mechanism to explain why this happens has not been systematically examined. Using self-reported expectancy of the outcome, the current study investigates three mechanisms that potentially explain why acquisition in multiple contexts results in more responding-greater generalization, stronger acquisition learning, or slower extinction learning. Participants (N = 180) received discriminative training with a conditioned stimulus (CS+) and outcome pairing and a CS- → noOutcome pairing in either one or three contexts. This was followed by either extinction treatment in a novel context or no extinction. Finally, testing occurred in the acquisition context, the extinction context, or a novel context. Stronger renewal of extinguished conditioned expectation was observed for participants who received CS+ → Outcome pairings in three contexts relative to one context. There was no effect of the number of contexts on the strength of the excitatory CS+ → Outcome association or degree of inhibitory learning that occurred during extinction. This suggests that generalization is the mechanism responsible for the adverse impact to extinction learning when acquisition is conducted in multiple contexts.


Subject(s)
Conditioning, Classical , Extinction, Psychological , Generalization, Psychological , Humans , Extinction, Psychological/physiology , Male , Female , Young Adult , Conditioning, Classical/physiology , Adult , Adolescent , Discrimination Learning/physiology
12.
Behav Neurosci ; 138(3): 152-163, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38934919

ABSTRACT

Here, we describe the efforts we dedicated to the challenge of modifying entrenched emotionally laden memories. In recent years, through a number of collaborations and using a combination of behavioral, molecular, and computational approaches, we: (a) developed novel approaches to fear attenuation that engage mechanisms that differ from those engaged during extinction (Monfils), (b) examined whether our approaches can generalize to other reinforcers (Lee, Gonzales, Chaudhri, Cofresi, and Monfils), (c) derived principled explanations for the differential outcomes of our approaches (Niv, Gershman, Song, and Monfils), (d) developed better assessment metrics to evaluate outcome success (Shumake and Monfils), (e) identified biomarkers that can explain significant variance in our outcomes of interest (Shumake and Monfils), and (f) developed better basic research assays and translated efforts to the clinic (Smits, Telch, Otto, Shumake, and Monfils). We briefly highlight each of these milestones and conclude with final remarks and extracted principles. (PsycInfo Database Record (c) 2024 APA, all rights reserved).


Subject(s)
Extinction, Psychological , Fear , Animals , Humans , Extinction, Psychological/physiology , Fear/physiology , Translational Research, Biomedical/methods
13.
Elife ; 132024 May 21.
Article in English | MEDLINE | ID: mdl-38770736

ABSTRACT

Pavlovian fear conditioning has been extensively used to study the behavioral and neural basis of defensive systems. In a typical procedure, a cue is paired with foot shock, and subsequent cue presentation elicits freezing, a behavior theoretically linked to predator detection. Studies have since shown a fear conditioned cue can elicit locomotion, a behavior that - in addition to jumping, and rearing - is theoretically linked to imminent or occurring predation. A criticism of studies observing fear conditioned cue-elicited locomotion is that responding is non-associative. We gave rats Pavlovian fear discrimination over a baseline of reward seeking. TTL-triggered cameras captured 5 behavior frames/s around cue presentation. Experiment 1 examined the emergence of danger-specific behaviors over fear acquisition. Experiment 2 examined the expression of danger-specific behaviors in fear extinction. In total, we scored 112,000 frames for nine discrete behavior categories. Temporal ethograms show that during acquisition, a fear conditioned cue suppresses reward seeking and elicits freezing, but also elicits locomotion, jumping, and rearing - all of which are maximal when foot shock is imminent. During extinction, a fear conditioned cue most prominently suppresses reward seeking, and elicits locomotion that is timed to shock delivery. The independent expression of these behaviors in both experiments reveals a fear conditioned cue to orchestrate a temporally organized suite of behaviors.


Knowing that an animal is fearful is crucial for many psychology and neuroscience studies. For instance, this knowledge allows researchers to examine the brain pathways involved in processing and responding to fear. Typically, researchers consider that a rodent is experiencing fear if it 'freezes' ­ a response which, in the wild, helps to evade detection by predators. In Pavlovian fear conditioning experiments, for example, rats and mice freeze when exposed to a stimulus (often a specific sound) previously associated with unpleasant sensations. However, rodents can also respond more actively to threats, for instance by running or jumping away. It remains unclear whether the 'fearful stimuli' used in Pavlovian approaches specifically elicits only freezing, or other fear-related behaviors as well. To investigate this, Chu et al. used high-speed cameras to record rats' responses to a sound cue they had 'learned' to associate with a mild foot shock. In addition to freezing, the animals ran, jumped, stood on their hind legs and stopped their usual reward-seeking behavior in response to the cue. Crucially, these reactions were absent when the rats were exposed to sound cues not associated with pain. Overall, these experiments demonstrate that Pavlovian conditioning can elicit a full range of fear-related behaviors beyond freezing. Understanding the neural activity behind these diverse responses could lead to more targeted therapies and interventions addressing the various ways stress and anxiety manifest in people.


Subject(s)
Behavior, Animal , Conditioning, Classical , Cues , Fear , Animals , Fear/physiology , Rats , Behavior, Animal/physiology , Male , Locomotion/physiology , Extinction, Psychological/physiology
14.
Psychoneuroendocrinology ; 166: 107083, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38788461

ABSTRACT

In mammals, some physiological conditions are associated with the high brain oxytocin (OXT) system activity. These include lactation in females and mating in males and females, both of which have been linked to reduced stress responsiveness and anxiolysis. Also, in a murine model of social fear conditioning (SFC), enhanced brain OXT signaling in lactating mice, specifically in the lateral septum (LS), was reported to underlie reduced social fear expression. Here, we studied the effects of mating in male mice on anxiety-related behaviour, social (and cued) fear expression and its extinction, and the activity of OXT neurons reflected by cFos expression and OXT release in the LS and amygdala. We further focused on the involvement of brain OXT in the mating-induced facilitation of social fear extinction. We could confirm the anxiolytic effect of mating in male mice irrespective of the occurrence of ejaculation. Further, we found that only successful mating resulting in ejaculation (Ej+) facilitated social fear extinction, whereas mating without ejaculation (Ej-) did not. In contrast, mating did not affect cues fear expression. Using the cellular activity markers cFos and pErk, we further identified the ventral LS (vLS) as a potential region participating in the effect of ejaculation on social fear extinction. In support, microdialysis experiments revealed a rise in OXT release within the LS, but not the amygdala, during mating. Finally, infusion of an OXT receptor antagonist into the LS before mating or into the lateral ventricle (icv) after mating demonstrated a significant role of brain OXT receptor-mediated signaling in the mating-induced facilitation of social fear extinction.


Subject(s)
Amygdala , Extinction, Psychological , Fear , Oxytocin , Sexual Behavior, Animal , Animals , Fear/physiology , Oxytocin/metabolism , Male , Extinction, Psychological/physiology , Mice , Female , Sexual Behavior, Animal/physiology , Amygdala/metabolism , Social Behavior , Anxiety/metabolism , Receptors, Oxytocin/metabolism , Septal Nuclei/metabolism , Septal Nuclei/drug effects , Ejaculation/physiology , Copulation/physiology , Septum of Brain/metabolism , Septum of Brain/physiology , Mice, Inbred C57BL , Behavior, Animal/physiology , Behavior, Animal/drug effects
15.
Neurobiol Learn Mem ; 212: 107937, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38735637

ABSTRACT

Systemic manipulations that enhance dopamine (DA) transmission around the time of fear extinction can strengthen fear extinction and reduce conditioned fear relapse. Prior studies investigating the brain regions where DA augments fear extinction focus on targets of mesolimbic and mesocortical DA systems originating in the ventral tegmental area, given the role of these DA neurons in prediction error. The dorsal striatum (DS), a primary target of the nigrostriatal DA system originating in the substantia nigra (SN), is implicated in behaviors beyond its canonical role in movement, such as reward and punishment, goal-directed action, and stimulus-response associations, but whether DS DA contributes to fear extinction is unknown. We have observed that chemogenetic stimulation of SN DA neurons during fear extinction prevents the return of fear in contexts different from the extinction context, a form of relapse called renewal. This effect of SN DA stimulation is mimicked by a DA D1 receptor (D1R) agonist injected into the DS, thus implicating DS DA in fear extinction. Different DS subregions subserve unique functions of the DS, but it is unclear where in the DS D1R agonist acts during fear extinction to reduce renewal. Furthermore, although fear extinction increases neural activity in DS subregions, whether neural activity in DS subregions is causally involved in fear extinction is unknown. To explore the role of DS subregions in fear extinction, adult, male Long-Evans rats received microinjections of either the D1R agonist SKF38393 or a cocktail consisting of GABAA/GABAB receptor agonists muscimol/baclofen selectively into either dorsomedial (DMS) or dorsolateral (DLS) DS subregions immediately prior to fear extinction, and extinction retention and renewal were subsequently assessed drug-free. While increasing D1R signaling in the DMS during fear extinction did not impact fear extinction retention or renewal, DMS inactivation reduced later renewal. In contrast, DLS inactivation had no effect on fear extinction retention or renewal but increasing D1R signaling in the DLS during extinction reduced fear renewal. These data suggest that DMS and DLS activity during fear extinction can have opposing effects on later fear renewal, with the DMS promoting renewal and the DLS opposing renewal. Mechanisms through which the DS could influence the contextual gating of fear extinction are discussed.


Subject(s)
Corpus Striatum , Extinction, Psychological , Fear , Receptors, Dopamine D1 , Animals , Fear/physiology , Fear/drug effects , Extinction, Psychological/drug effects , Extinction, Psychological/physiology , Male , Rats , Corpus Striatum/drug effects , Corpus Striatum/physiology , Corpus Striatum/metabolism , Receptors, Dopamine D1/physiology , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D1/antagonists & inhibitors , Dopamine Agonists/pharmacology , Conditioning, Classical/drug effects , Conditioning, Classical/physiology , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/physiology , Substantia Nigra/drug effects , Substantia Nigra/physiology , Rats, Long-Evans , Dopamine/metabolism , Dopamine/physiology
16.
J Anxiety Disord ; 104: 102870, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38733644

ABSTRACT

Exposure therapy is an evidence-based treatment option for anxiety-related disorders. Many patients also take medication that could, in principle, affect exposure therapy efficacy. Clinical and laboratory evidence indeed suggests that benzodiazepines may have detrimental effects. Large clinical trials with propranolol, a common beta-blocker, are currently lacking, but several preclinical studies do indicate impaired establishment of safety memories. Here, we investigated the effects of propranolol given prior to extinction training in 9 rat studies (N = 215) and one human study (N = 72). A Bayesian meta-analysis of our rat studies provided strong evidence against propranolol-induced extinction memory impairment during a drug-free test, and the human study found no significant difference with placebo. Two of the rat studies actually suggested a small beneficial effect of propranolol. Lastly, two rat studies with a benzodiazepine (midazolam) group provided some evidence for a harmful effect on extinction memory, i.e., impaired extinction retention. In conclusion, our midazolam findings are in line with prior literature (i.e., an extinction retention impairment), but this is not the case for the 10 studies with propranolol. Our data thus support caution regarding the use of benzodiazepines during exposure therapy, but argue against a harmful effect of propranolol on extinction learning.


Subject(s)
Adrenergic beta-Antagonists , Extinction, Psychological , Fear , Memory , Midazolam , Propranolol , Propranolol/pharmacology , Propranolol/administration & dosage , Animals , Fear/drug effects , Extinction, Psychological/drug effects , Rats , Humans , Adrenergic beta-Antagonists/pharmacology , Adrenergic beta-Antagonists/administration & dosage , Male , Memory/drug effects , Midazolam/pharmacology , Midazolam/administration & dosage , Midazolam/adverse effects , Adult , Bayes Theorem , Female , Conditioning, Classical/drug effects , Young Adult
17.
Drug Alcohol Depend ; 260: 111328, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38776581

ABSTRACT

RATIONALE: A return to cocaine use following abstinence frequently occurs in a social context, and the presence of other individuals using cocaine may contribute to the likelihood of use. Previous studies have reported that chronic d-amphetamine treatment decreases cocaine self-administration in laboratory animals and reduces a return to cocaine use following abstinence in humans. OBJECTIVE: The purpose of this study was to examine the effects of chronic d-amphetamine treatment on the reacquisition of cocaine use in rats self-administering cocaine in different social contexts. METHODS: Male and female rats were implanted with intravenous catheters and trained to self-administer cocaine during daily 6-hr sessions. After 14 days, cocaine self-administration was extinguished by substituting saline for the cocaine stimulus. At this time, rats were randomized to receive chronic treatment with either d-amphetamine or saline. After 9 days of extinction, cocaine was again made available during daily 6-hr sessions. At this time, rats were further randomized into three social conditions: (1) rats continued self-administering cocaine in isolation, (2) rats self-administered cocaine in the presence of a same-sex partner that also self-administered cocaine, or (3) rats self-administered cocaine in the presence of a same-sex partner that did not have access to cocaine. Daily treatment with d-amphetamine or saline continued for the duration of reacquisition testing. RESULTS: Chronic treatment with d-amphetamine decreased cocaine intake during reacquisition, but these effects were not influenced by the social context. No sex differences were observed. CONCLUSION: These data support previous studies reporting that d-amphetamine decreases cocaine intake and demonstrate its efficacy across social contexts.


Subject(s)
Cocaine , Dextroamphetamine , Self Administration , Animals , Male , Female , Rats , Cocaine/administration & dosage , Central Nervous System Stimulants/administration & dosage , Central Nervous System Stimulants/pharmacology , Extinction, Psychological/drug effects , Cocaine-Related Disorders/drug therapy , Cocaine-Related Disorders/psychology , Rats, Sprague-Dawley , Social Behavior , Social Environment
18.
PeerJ ; 12: e17262, 2024.
Article in English | MEDLINE | ID: mdl-38737738

ABSTRACT

Although exposure-based therapy has been found to be effective at alleviating symptoms of social anxiety disorder, it often does not lead to full remission, and relapse after treatment is common. Exposure therapy is based on theoretical principles of extinction of conditioned fear responses. However, there are inconsistencies in findings across experiments that have investigated the effect of social anxiety on threat conditioning and extinction processes. This systematic review and meta-analysis aimed to examine whether elevated levels of social anxiety are associated with abnormalities in threat conditioning and extinction processes. A second aim was to examine the sensitivity of various study designs and characteristics to detect social anxiety-related differences in threat conditioning and extinction. A systematic search was conducted, which identified twenty-three experiments for inclusion in the review. The findings did not demonstrate compelling evidence that high levels of social anxiety are associated with atypical threat conditioning or extinction. Further, when systematically examining the data, there was no convincing support that the use of a particular psychophysiological measure, subjective rating, or experimental parameter yields more consistent associations between social anxiety and conditioning processes during threat acquisition or extinction. Meta-analyses demonstrated that during threat extinction, the use of anxiety ratings as a dependent variable, socially relevant unconditioned stimuli, and a higher reinforcement schedule produced more detectable effects of social anxiety on compromised extinction processes compared to any other dependent variable (subjective or physiological) or experimental parameter. Overall, the results of this study suggest that social anxiety is not reliably related to deficits in conditioning and extinction processes in the context of laboratory-based Pavlovian conditioning paradigms.


Subject(s)
Extinction, Psychological , Fear , Phobia, Social , Humans , Fear/psychology , Phobia, Social/psychology , Anxiety/psychology , Conditioning, Classical
19.
Curr Biol ; 34(10): 2247-2255.e5, 2024 05 20.
Article in English | MEDLINE | ID: mdl-38714199

ABSTRACT

Rapid eye movement (REM) sleep is known to facilitate fear extinction and play a protective role against fearful memories.1,2 Consequently, disruption of REM sleep after a traumatic event may increase the risk for developing PTSD.3,4 However, the underlying mechanisms by which REM sleep promotes extinction of aversive memories remain largely unknown. The infralimbic cortex (IL) is a key brain structure for the consolidation of extinction memory.5 Using calcium imaging, we found in mice that most IL pyramidal neurons are intensively activated during REM sleep. Optogenetically suppressing the IL specifically during REM sleep within a 4-h window after auditory-cued fear conditioning impaired extinction memory consolidation. In contrast, REM-specific IL inhibition after extinction learning did not affect the extinction memory. Whole-cell patch-clamp recordings demonstrated that inactivating IL neurons during REM sleep depresses their excitability. Together, our findings suggest that REM sleep after fear conditioning facilitates fear extinction by enhancing IL excitability and highlight the importance of REM sleep in the aftermath of traumatic events for protecting against traumatic memories.


Subject(s)
Extinction, Psychological , Fear , Sleep, REM , Animals , Fear/physiology , Sleep, REM/physiology , Mice , Extinction, Psychological/physiology , Male , Mice, Inbred C57BL , Memory/physiology , Memory Consolidation/physiology , Conditioning, Classical/physiology , Pyramidal Cells/physiology
20.
Curr Biol ; 34(10): R510-R512, 2024 05 20.
Article in English | MEDLINE | ID: mdl-38772341

ABSTRACT

The ability to forget fear-inducing situations is essential for adapting to our environment, but the neural mechanisms underlying 'fear forgetting' remain unclear. Novel findings reveal that the activity of the infralimbic cortex - specifically during REM sleep - contributes to the extinction of fear memory.


Subject(s)
Fear , Memory , Sleep, REM , Fear/physiology , Sleep, REM/physiology , Animals , Memory/physiology , Humans , Extinction, Psychological/physiology , Dreams/physiology , Dreams/psychology
SELECTION OF CITATIONS
SEARCH DETAIL
...