Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-38691149

ABSTRACT

RATIONALE: Therapeutic approaches to mitigating traumatic memories have often faced resistance. Exploring safe reconsolidation blockers, drugs capable of reducing the emotional valence of the memory upon brief retrieval and reactivation, emerges as a promising pharmacological strategy. Towards this objective, preclinical investigations should focus on aversive memories resulting in maladaptive outcomes and consider sex-related differences to enhance their translatability. OBJECTIVES: After selecting a relatively high training magnitude leading to the formation of a more intense and generalized fear memory in adult female and male rats, we investigated whether two clinically approved drugs disrupting its reconsolidation remain effective. RESULTS: We found resistant reconsolidation impairment by the α2-adrenergic receptor agonist clonidine or cannabidiol, a major non-psychotomimetic Cannabis sativa component. However, pre-retrieval administration of D-cycloserine, a partial agonist at the glycine-binding site of the N-methyl-D-aspartate (NMDA) receptor complex, facilitated their impairing effects on reconsolidation. A similar reconsolidation blockade by clonidine or cannabidiol was achieved following exposure to a non-conditioned but generalized context after D-cycloserine administration. This suggests that sufficient memory destabilization can accompany generalized fear expression. Combining clonidine with cannabidiol without potentiating memory destabilization by D-cycloserine was ineffective. CONCLUSIONS: These findings highlight the importance of NMDA receptor signaling in memory destabilization and underscore the efficacy of a dual-step pharmacological intervention in attenuating traumatic-like memories, even in a context different from the original learning environment.

2.
Neurosci Biobehav Rev ; 147: 105101, 2023 04.
Article in English | MEDLINE | ID: mdl-36804263

ABSTRACT

Fear conditioning and avoidance tasks usually elicit adaptive aversive memories. Traumatic memories are more intense, generalized, inflexible, and resistant to attenuation via extinction- and reconsolidation-based strategies. Inducing and assessing these dysfunctional, maladaptive features in the laboratory are crucial to interrogating posttraumatic stress disorder's neurobiology and exploring innovative treatments. Here we analyze over 350 studies addressing this question in adult rats and mice. There is a growing interest in modeling several qualitative and quantitative memory changes by exposing already stressed animals to freezing- and avoidance-related tests or using a relatively high aversive training magnitude. Other options combine aversive/fearful tasks with post-acquisition or post-retrieval administration of one or more drugs provoking neurochemical or epigenetic alterations reported in the trauma aftermath. It is potentially instructive to integrate these procedures and incorporate the measurement of autonomic and endocrine parameters. Factors to consider when defining the organismic and procedural variables, partially neglected aspects (sex-dependent differences and recent vs. remote data comparison) and suggestions for future research (identifying reliable individual risk and treatment-response predictors) are discussed.


Subject(s)
Extinction, Psychological , Stress Disorders, Post-Traumatic , Rats , Mice , Animals , Extinction, Psychological/physiology , Rodentia , Conditioning, Psychological , Fear/physiology
3.
Neuroscience ; 497: 118-133, 2022 08 10.
Article in English | MEDLINE | ID: mdl-34560200

ABSTRACT

Systemic administration of drugs that activate the noradrenergic or glucocorticoid system potentiates aversive memory consolidation and reconsolidation. The opposite happens with the stimulation of endocannabinoid signaling under certain conditions. An unbalance of these interacting neurotransmitters can lead to the formation and maintenance of traumatic memories, whose strength and specificity attributes are often maladaptive. Here we aimed to investigate whether originally low-intensity and precise contextual fear memories would turn similar to traumatic ones in rats systemically administered with adrenaline, corticosterone, and/or the cannabinoid type-1 receptor antagonist/inverse agonist AM251 during consolidation or reconsolidation. The high dose of each pharmacological agent evaluated significantly increased freezing times at test in the conditioning context one and nine days later when given alone post-acquisition or post-retrieval. Their respective low dose produced no relative changes when given separately, but co-treatment of adrenaline with corticosterone or AM251 and the three drugs combined, but not corticosterone with AM251, produced results equivalent to those mentioned initially. Neither the high nor the low dose of adrenaline, corticosterone, or AM251 altered freezing times at test in a novel, neutral context two and ten days later. In contrast, animals receiving the association of their low dose exhibited significantly higher freezing times than controls. Together, the results indicate that newly acquired and destabilized threat memory traces become more intense and generalized after a combined interference acting synergistically and mimicking that reported in patients presenting stress-related psychiatric conditions.


Subject(s)
Endocannabinoids , Glucocorticoids , Animals , Cannabinoid Receptor Agonists , Corticosterone/pharmacology , Epinephrine , Fear/physiology , Glucocorticoids/pharmacology , Memory/physiology , Norepinephrine/physiology , Rats , Rats, Wistar
4.
Neuropharmacology ; 205: 108913, 2022 03 01.
Article in English | MEDLINE | ID: mdl-34864001

ABSTRACT

Growing evidence indicates that cannabidiol (CBD), a substance present in the Cannabis sativa plant, has potential therapeutic value to regulate abnormal emotional memories associated with post-traumatic stress and drug use disorders. CBD can attenuate their valence after retrieval (i.e., during reconsolidation) or potentiate their suppression by extinction. Pharmacological research has now focused on elucidating how it acts. Systemic antagonism of cannabinoid type-1 (CB1) receptors has often prevented the abovementioned effects of CBD. However, it is unknown in which brain regions CBD stimulates CB1 receptors and how it interferes with local activity-related plasticity to produce these effects. The present study addressed these questions considering the reconsolidation of contextual fear memories in rats. We focused on the medial prefrontal cortex (mPFC), which comprises the anterior cingulate (AC), prelimbic (PL), and infralimbic (IL) subregions, as local activity or plasticity has been associated with the process to-be-investigated. Animals that received post-retrieval systemic CBD treatment presented relatively fewer cells expressing Zif268/Egr1 protein, a proxy for synaptic plasticity related to reconsolidation, in the AC and PL. At the same time, there were no significant differences in the IL. Pretreatment with the CB1 receptor antagonist/inverse agonist AM251 into the AC, PL, or IL prevented the impairing effects of systemic CBD treatment on reconsolidation. CBD also caused reconsolidation impairments when injected directly into the AC or PL but not the IL. Together, these findings show complementary mechanisms through which CBD may hinder the reconsolidation of destabilized aversive memories along the dorsoventral axis of the mPFC.


Subject(s)
Cannabidiol/adverse effects , Cannabinoid Receptor Agonists/adverse effects , Cannabinoid Receptor Antagonists/pharmacology , Memory Consolidation/drug effects , Memory Disorders/chemically induced , Memory Disorders/drug therapy , Neuronal Plasticity/drug effects , Prefrontal Cortex/drug effects , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Male , Rats , Receptor, Cannabinoid, CB1/antagonists & inhibitors
7.
Eur Neuropsychopharmacol ; 25(6): 958-65, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25799920

ABSTRACT

Δ(9)-tetrahydrocannabinol (THC) and cannabidiol (CBD) are the major constituents of the Cannabis sativa plant, which is frequently consumed by subjects exposed to life-threatening situations to relief their symptomatology. It is still unknown, however, whether THC could also affect the maintenance of an aversive memory formed at that time when taken separately and/or in conjunction with CBD. The present study sought to investigate this matter at a preclinical level. We report that THC (0.3-10mg/kg, i.p.) was able to disrupt the reconsolidation of a contextual fear memory, resulting in reduced conditioned freezing expression for over 22 days. This effect was dependent on activation of cannabinoid type-1 receptors located in prelimbic subregion of the medial prefrontal cortex and on memory retrieval/reactivation. Since CBD may counteract the negative psychotropic effects induced by THC and has been shown to be a reconsolidation blocker, we then investigated and demonstrated that associating sub-effective doses of these two compounds was equally effective in attenuating fear memory maintenance in an additive fashion and in a dose ratio of 10 to 1, which contrasts with that commonly found in C. sativa recreational samples. Of note, neither THC alone nor CBD plus THC interfered with anxiety-related behaviors and locomotor activity, as assessed in the elevated plus-maze test, at a time point coinciding with that used to evaluate their effects on memory reconsolidation. Altogether, present findings suggest a potential therapeutic value of using THC and/or CBD to mitigate a dysfunctional aversive memory through reconsolidation disruption in post-traumatic stress disorder patients.


Subject(s)
Cannabidiol/pharmacology , Dronabinol/pharmacology , Fear/drug effects , Memory Disorders/chemically induced , Memory/drug effects , Analysis of Variance , Animals , Cannabidiol/adverse effects , Disease Models, Animal , Dose-Response Relationship, Drug , Dronabinol/adverse effects , Drug Combinations , Male , Maze Learning/drug effects , Rats , Rats, Wistar , Time Factors
8.
Int J Neuropsychopharmacol ; 18(1)2014 Oct 31.
Article in English | MEDLINE | ID: mdl-25539509

ABSTRACT

BACKGROUND: Traumatic memories have been resilient to therapeutic approaches targeting their permanent attenuation. One of the potentially promising pharmacological strategies under investigation is the search for safe reconsolidation blockers. However, preclinical studies focusing on this matter have scarcely addressed abnormal aversive memories and related outcomes. METHODS: By mimicking the enhanced noradrenergic activity reported after traumatic events in humans, here we sought to generate a suitable condition to establish whether some clinically approved drugs able to disrupt the reconsolidation of conditioned fear memories in rodents would still be effective. RESULTS: We report that the α2-adrenoceptor antagonist yohimbine was able to induce an inability to restrict behavioral (fear) and cardiovascular (increased systolic blood pressure) responses to the paired context when administered immediately after acquisition, but not 6h later, indicating the formation of a generalized fear memory, which endured for over 29 days and was less susceptible to suppression by extinction. It was also resistant to reconsolidation disruption by the α2-adrenoceptor agonist clonidine or cannabidiol, the major non-psychotomimetic component of Cannabis sativa. Since signaling at N-methyl-D-aspartate (NMDA) receptors is important for memory labilization and because a dysfunctional memory may be less labile than is necessary to trigger reconsolidation on its brief retrieval and reactivation, we then investigated and demonstrated that pre-retrieval administration of the partial NMDA agonist D-cycloserine allowed the disrupting effects of clonidine and cannabidiol on reconsolidation. CONCLUSIONS: These findings highlight the effectiveness of a dual-step pharmacological intervention to mitigate an aberrant and enduring aversive memory similar to that underlying the post-traumatic stress disorder.


Subject(s)
Fear/drug effects , Memory/drug effects , Psychotropic Drugs/pharmacology , Adrenergic alpha-2 Receptor Agonists/pharmacology , Adrenergic alpha-2 Receptor Antagonists/pharmacology , Animals , Blood Pressure/drug effects , Cannabidiol/pharmacology , Clonidine/pharmacology , Cycloserine/pharmacology , Excitatory Amino Acid Agents/pharmacology , Extinction, Psychological/drug effects , Fear/physiology , Male , Random Allocation , Rats, Wistar , Stress Disorders, Post-Traumatic/drug therapy , Yohimbine/pharmacology
9.
Learn Mem ; 21(1): 14-20, 2013 Dec 16.
Article in English | MEDLINE | ID: mdl-24344180

ABSTRACT

The prelimbic cortex has been implicated in the consolidation of previously learned fear. Herein, we report that temporarily inactivating this medial prefrontal cortex subregion with the GABAA agonist muscimol (4.0 nmol in 0.2 µL per hemisphere) was able to equally disrupt 1-, 7-, and 21-d-old contextual fear memories after their brief retrieval in rats. In all cases, this effect was prevented when memory reactivation was omitted. These results indicate that recent and remote fear memories are susceptible to reconsolidation blockade induced by prelimbic cortex inactivation. It was also demonstrated that the disrupting effect of prelimbic cortex inactivation on fear memory persisted over 11 d, and did not show extinction-related features, such as reinstatement. Infusing the same dose and volume of muscimol bilaterally into the infralimbic cortex after brief retrieval/reactivation of the fear memory did not disrupt it, as seen in prelimbic cortex-inactivated animals. The expression of Zif268/Egr1, the product of an immediate early gene related to memory reconsolidation, was also less pronounced in the infralimbic cortex than in prelimbic cortex following memory retrieval/reactivation. Altogether, the present findings highlight that activity in the prelimbic cortex may reestablish reactivated aversive memories and, therefore, contribute to their maintenance over time.


Subject(s)
Cerebral Cortex/physiology , Conditioning, Classical/physiology , Fear/physiology , Memory/physiology , Analysis of Variance , Animals , Cerebral Cortex/drug effects , Conditioning, Classical/drug effects , Dose-Response Relationship, Drug , Early Growth Response Protein 1/metabolism , Extinction, Psychological/drug effects , Fear/drug effects , Functional Laterality , GABA-A Receptor Agonists/pharmacology , Male , Memory/drug effects , Muscimol/pharmacology , Rats , Rats, Wistar , Time Factors
10.
Learn Mem ; 20(4): 210-9, 2013 Mar 19.
Article in English | MEDLINE | ID: mdl-23512937

ABSTRACT

Consolidation and reconsolidation are phases of memory stabilization that diverge slightly. Noradrenaline is known to influence both processes, but the relative contribution of α1- and ß-adrenoceptors is unclear. The present study sought to investigate this matter by comparing their recruitment to consolidate and/or reconsolidate a contextual fear memory trace under enhanced noradrenergic activity induced by yohimbine. We report that this α2-adrenoceptor antagonist was able to potentiate fear memory trace consolidation or reconsolidation when administered immediately after acquisition or retrieval, respectively, resulting in increased freezing expression. In either case, generalization of this response to an unpaired context was also seen when it achieved a ceiling level in the paired context. These effects endured for over 7 d and relied on action at central rather than peripheral sites, but were prevented when a memory trace was not acquired, when memory reactivation was omitted, or when administration of yohimbine was delayed until 6 h after acquiring or retrieving the memory trace. The ß-adrenoceptor antagonist propranolol was able to prevent the above-mentioned effects of yohimbine, while pretreatment with the α1-adrenoceptor antagonist prazosin blocked only its facilitating effects on memory reconsolidation. These results highlight a differential participation of α1- and ß-adrenoceptors in fear memory processing. Moreover, it was shown that the α2-adrenoceptor agonist clonidine, as opposed to yohimbine, mitigates fear expression by weakening memory consolidation or reconsolidation.


Subject(s)
Conditioning, Psychological/physiology , Fear/physiology , Memory/physiology , Norepinephrine/physiology , Receptors, Adrenergic, alpha-1/physiology , Receptors, Adrenergic, beta/physiology , Adrenergic Agents/pharmacology , Analysis of Variance , Animals , Avoidance Learning/drug effects , Avoidance Learning/physiology , Conditioning, Psychological/drug effects , Extinction, Psychological/drug effects , Extinction, Psychological/physiology , Fear/drug effects , Freezing Reaction, Cataleptic/drug effects , Freezing Reaction, Cataleptic/physiology , Male , Memory/drug effects , Rats , Rats, Wistar
11.
Neuropsychopharmacology ; 37(9): 2132-42, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22549120

ABSTRACT

The search for reconsolidation blockers may uncover clinically relevant drugs for disrupting memories of significant stressful life experiences, such as those underlying the posttraumatic stress disorder. Considering the safety of systemically administered cannabidiol (CBD), the major non-psychotomimetic component of Cannabis sativa, to animals and humans, the present study sought to investigate whether and how this phytocannabinoid (3-30 mg/kg intraperitoneally; i.p.) could mitigate an established memory, by blockade of its reconsolidation, evaluated in a contextual fear-conditioning paradigm in rats. We report that CBD is able to disrupt 1- and 7-days-old memories when administered immediately, but not 6 h, after their retrieval for 3 min, with the dose of 10 mg/kg being the most effective. This effect persists in either case for at least 1 week, but is prevented when memory reactivation was omitted, or when the cannabinoid type-1 receptors were antagonized selectively with AM251 (1.0 mg/kg). Pretreatment with the serotonin type-1A receptor antagonist WAY100635, however, failed to block CBD effects. These results highlight that recent and older fear memories are equally vulnerable to disruption induced by CBD through reconsolidation blockade, with a consequent long-lasting relief in contextual fear-induced freezing. Importantly, this CBD effect is dependent on memory reactivation, restricted to time window of <6 h, and is possibly dependent on cannabinoid type-1 receptor-mediated signaling mechanisms. We also observed that the fear memories disrupted by CBD treatment do not show reinstatement or spontaneous recovery over 22 days. These findings support the view that reconsolidation blockade, rather than facilitated extinction, accounts for the aforementioned CBD results in our experimental conditions.


Subject(s)
Cannabidiol/pharmacology , Conditioning, Psychological/drug effects , Fear/drug effects , Memory/drug effects , Animals , Conditioning, Psychological/physiology , Evidence-Based Medicine/methods , Extinction, Psychological/drug effects , Extinction, Psychological/physiology , Fear/physiology , Male , Memory/physiology , Random Allocation , Rats , Rats, Wistar , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/physiology
12.
J Ethnopharmacol ; 137(3): 1143-8, 2011 Oct 11.
Article in English | MEDLINE | ID: mdl-21801825

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Trichilia catigua preparations have been popularly used in Brazil as a tonic for the treatment of fatigue, stress, impotence, and deficiency of memory. The aim of the present study was to investigate the possible antidepressant, anxiolytic, motor and cognitive effects of the crude extract (CE) or ethyl-acetate fraction (EAF) of Trichilia catigua. Analyses of the total phenolics and total tannins content, as well as the in vitro antioxidant activity of CE and EAF were also performed. MATERIALS AND METHODS: CE (200-800 mg/kg) and EAF (100-400mg/kg) were orally administered to mice and 1h later the behavioral tests were performed. The free radical scavenging activity was measured by using 2,2-diphenyl-1-picryl-hydrazyl (DPPH) method. RESULTS: Single administration of CE (200-400 mg/kg) or EAF (100-400 mg/kg) did not change the behavior of the animals submitted to the elevated plus maze or their locomotor activity in the open field test. An antidepressant-like effect was detected with EAF (400 mg/kg) after acute administration. Both CE (800 mg/kg) and EAF (200 and 400 mg/kg), improve memory in mice as measured by an increased latency in the step-down inhibitory avoidance test. The EAF presented higher total phenolics and total tannins as compared to CE as well as it exhibited the best antioxidant activity. CONCLUSIONS: The present results showed an in vitro antioxidant activity for EAF and suggested that it may be useful for cognitive improvement. It is possible that both functional and chemical activities are related.


Subject(s)
Central Nervous System Agents/pharmacology , Central Nervous System/drug effects , Meliaceae , Plant Extracts/pharmacology , Animals , Anti-Anxiety Agents/pharmacology , Antidepressive Agents/pharmacology , Behavior, Animal/drug effects , Biphenyl Compounds/chemistry , Central Nervous System Agents/chemistry , Central Nervous System Agents/isolation & purification , Central Nervous System Agents/toxicity , Cognition/drug effects , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Free Radical Scavengers/pharmacology , Lethal Dose 50 , Male , Meliaceae/chemistry , Memory/drug effects , Mice , Motor Activity/drug effects , Phenols/analysis , Picrates/chemistry , Plant Bark , Plant Extracts/chemistry , Plant Extracts/isolation & purification , Plant Extracts/toxicity , Plants, Medicinal , Tannins/analysis , Time Factors
13.
Behav Brain Res ; 210(1): 38-45, 2010 Jun 26.
Article in English | MEDLINE | ID: mdl-20138920

ABSTRACT

We investigated the effects of chronic administration of vitamin E on nitric oxide (NO)-producing neurons in the brains of streptozotocin (STZ)-induced diabetic rats using nicotinamide adenine dinucleotide phosphate-diaphorase (NADPH-d) histochemistry. We further evaluated the effects of diabetes and vitamin E treatment on experimental anxiety and memory processes using the elevated plus maze (EPM) Trial 1/2 protocol. Wistar rats were divided into four groups: normoglycemics (N), normoglycemics treated with vitamin E (NVE), diabetics (D), and diabetics treated with vitamin E (DVE). Diabetes mellitus was induced by a single intraperitoneal injection of STZ (35mg/kg). Vitamin E (100mg/kg) or vehicle was administered orally by gavage (1ml/kg) once each day for 7 weeks. After behavioral testing, the dentate gyrus of the hippocampus (DG), striatum, paraventricular nucleus of the hypothalamus (PVN), supraoptic nucleus (SON), and dorsolateral periaqueductal grey (DLPAG) were analyzed for NADPH-d histochemistry. STZ-induced diabetic rats exhibited decreased locomotor activity and cognitive impairment compared with normoglycemic controls. The number of NADPH-d-positive neurons was increased in the DG, striatum, and DLPAG of diabetic rats. An increase in soma area was detected in all structures analyzed (DG, striatum, PVN, SON, and DLPAG) of STZ-induced diabetic animals. The present study showed that chronic administration of vitamin E ameliorates memory in STZ-induced diabetic rats and revealed that NOS-producing neurons have an increased soma area which can be restored, at least partially, by vitamin E treatment. These results suggest the potential use of vitamin E as an adjuvant therapy for the prevention and treatment of diabetic conditions.


Subject(s)
Brain/metabolism , Diabetes Mellitus, Experimental/diet therapy , Learning/physiology , Neurons/metabolism , Nitric Oxide/metabolism , Vitamin E/therapeutic use , Animals , Anxiety/drug therapy , Anxiety/metabolism , Brain/pathology , Cognition Disorders/diet therapy , Cognition Disorders/metabolism , Cognition Disorders/pathology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Male , Maze Learning/physiology , Memory/physiology , Motor Activity/physiology , NADP/metabolism , Neurons/pathology , Random Allocation , Rats , Rats, Wistar , Vitamin E/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...