Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 484
Filter
1.
Pharmacol Biochem Behav ; 211: 173296, 2021 12.
Article in English | MEDLINE | ID: mdl-34752797

ABSTRACT

Animal models are important tools for studying neuropsychological disorders. Considering their limitations, a more extensive translational research must encompass data that are generated from several models. Therefore, a comprehensive characterization of these models is needed in terms of behavior and neurophysiology. The present study evaluated the behavioral responses of Carioca Low-conditioned Freezing (CLF) rats to haloperidol and methylphenidate. The CLF breeding line is characterized by low freezing defensive responses to contextual cues that are associated with aversive stimuli. CLF rats exhibited a delayed response to haloperidol at lower doses, needing higher doses to reach similar levels of catatonia as control randomly bred animals. Methylphenidate increased freezing responses to conditioned fear and induced motor effects in the open field. Thus, CLF rats differ from controls in their responses to both haloperidol and methylphenidate. Because of the dopamine-related molecular targets of these drugs, we hypothesize that dopaminergic alterations related to those of animal models of hyperactivity and attention disorders might underlie the observed phenotypes of the CLF line of rats.


Subject(s)
Conditioning, Psychological/drug effects , Fear/drug effects , Freezing Reaction, Cataleptic/drug effects , Haloperidol/pharmacology , Methylphenidate/pharmacology , Animals , Anxiety/drug therapy , Attention Deficit Disorder with Hyperactivity/drug therapy , Behavior, Animal/drug effects , Disease Models, Animal , Dopamine/metabolism , Dopamine Antagonists/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Hyperkinesis/drug therapy , Male , Motor Activity/drug effects , Rats , Rats, Wistar
2.
Acta Neuropsychiatr ; 33(3): 148-155, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33593455

ABSTRACT

OBJECTIVE: Whereas numerous experimental and clinical studies suggest a complex involvement of serotonin in the regulation of anxiety, it remains to be clarified if the dominating impact of this transmitter is best described as anxiety-reducing or anxiety-promoting. The aim of this study was to assess the impact of serotonin depletion on acquisition, consolidation, and expression of conditioned fear. METHODS: Male Sprague-Dawley rats were exposed to foot shocks as unconditioned stimulus and assessed with respect to freezing behaviour when re-subjected to context. Serotonin depletion was achieved by administration of a serotonin synthesis inhibitor, para-chlorophenylalanine (PCPA) (300 mg/kg daily × 3), (i) throughout the period from (and including) acquisition to (and including) expression, (ii) during acquisition but not expression, (iii) after acquisition only, and (iv) during expression only. RESULTS: The time spent freezing was significantly reduced in animals that were serotonin-depleted during the entire period from (and including) acquisition to (and including) expression, as well as in those being serotonin-depleted during either acquisition only or expression only. In contrast, PCPA administrated immediately after acquisition, that is during memory consolidation, did not impact the expression of conditioned fear. CONCLUSION: Intact serotonergic neurotransmission is important for both acquisition and expression of context-conditioned fear.


Subject(s)
Fear/drug effects , Fenclonine/pharmacology , Serotonin Antagonists/pharmacology , Serotonin/metabolism , Animals , Anxiety/metabolism , Behavior, Animal/drug effects , Conditioning, Psychological , Disease Models, Animal , Fear/psychology , Fenclonine/administration & dosage , Freezing Reaction, Cataleptic/drug effects , Male , Rats , Rats, Sprague-Dawley , Serotonin/deficiency , Serotonin Antagonists/administration & dosage
3.
Pharmacol Biochem Behav ; 197: 173019, 2020 10.
Article in English | MEDLINE | ID: mdl-32827503

ABSTRACT

Evidence from clinical and epidemiological studies point towards an association between generalized anxiety disorder (GAD) and alcohol abuse. In the present study we investigated whether a similar relationship could be observed in an animal model of GAD. Specifically, we evaluated the alcohol intake of Carioca High- and Low-conditioned Freezing rats (CHF and CLF, respectively). Sex differences in alcohol drinking behavior were also studied. Male and female rats from randomized crossbreeding populations served as controls (CTL). Free- and forced-choice protocols were used to measure alcohol consumption, and quinine and saccharin were used as taste control solutions. Our results indicate that CHF rats consumed more alcohol than CLF and CTL ones in both the free-choice (6 and 10% concentrations) and the forced-choice (10% concentration) conditions. CHF female rats exhibited the highest amount of alcohol intake in the forced-choice condition. CHF females also consumed more quinine than CHF male rats. Finally, CHF rats exhibited lower saccharin consumption compared to CLF and CTL animals. Altogether, these results support the hypothesis that there is a positive relationship between anxiety and alcohol intake, and provide further evidence for the use of CHF rats as a model of GAD.


Subject(s)
Alcohol Drinking , Alcoholism/complications , Anxiety Disorders/complications , Conditioning, Psychological/drug effects , Disease Models, Animal , Freezing Reaction, Cataleptic/drug effects , Animals , Ethanol , Fear , Female , Male , Phenotype , Quinine , Rats , Rats, Wistar , Saccharin , Taste Perception
4.
Synapse ; 74(12): e22180, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32644234

ABSTRACT

Pharmacological magnetic resonance imaging (phMRI) allows the visualization of brain pharmacological effects of drugs using functional MRI (fMRI). phMRI can help us facilitate central nervous system (CNS) drug development. However, there have been few studies demonstrating the dose relationship of the fMRI response induced by CNS drugs to underlying target engagement or behavioral efficacy. To clarify these relationships, we examined receptor occupancy measurements using positron emission tomography (PET) (n = 3~5), fMRI (n = 5~8) and a cataleptic behavior (n = 6) with raclopride, a dopamine D2 receptor antagonist (8, 20, and 200 µg/kg) on Wistar rats. Dopamine D2 receptor occupancy was increased dose dependently by raclopride (41.8 ± 2.7%, 8 µg/kg; 64.9 ± 2.8%, 20 µg/kg; 83.1 ± 3.0%, 200 µg/kg). phMRI study revealed significant positive responses to raclopride at 200 µg/kg specifically in the striatum and nucleus accumbens, related to dopaminergic system. Slight fMRI responses were observed at 20 µg/kg in some areas corresponding to the striatum and nucleus accumbens. There were no noticeable fMRI responses at 8 µg/kg raclopride administration. Raclopride at 200 µg/kg significantly increased the cataleptic score, although, at 8 and 20 µg/kg, raclopride had no significant effects. These findings showed that raclopride-induced fMRI responses were observed at doses inducing cataleptic behavior and high D2 receptor occupancy, suggesting that phMRI can be useful for dose selection in clinical trial as an evaluation method of brain activity, which reflects behavioral responses induced by target engagements.


Subject(s)
Corpus Striatum/metabolism , Dopamine Antagonists/pharmacokinetics , Freezing Reaction, Cataleptic/drug effects , Nucleus Accumbens/metabolism , Raclopride/pharmacokinetics , Animals , Corpus Striatum/diagnostic imaging , Corpus Striatum/physiology , Magnetic Resonance Imaging , Male , Nucleus Accumbens/diagnostic imaging , Nucleus Accumbens/physiology , Positron-Emission Tomography , Protein Binding , Rats , Rats, Wistar , Receptors, Dopamine D2/metabolism
5.
J Ethnopharmacol ; 260: 113048, 2020 Oct 05.
Article in English | MEDLINE | ID: mdl-32525067

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Lavender (Lavandula angustifolia) essential oil (EO) has a long history of use in emotional illness, including anxiety disorders. Cognitive mechanisms of learning and memory play a pivotal role in the etiology and maintenance of anxiety since exposure to cues related to aversive situations induces high arousal and anticipatory anxiety. Memory become labile after its reactivation and can be modulated by reconsolidation or extinction. Inhibition of memory reconsolidation or facilitation of memory extinction may be effective in preventing or minimizing the effect of contextual cues on anticipatory anxiety. AIM OF THE STUDY: We investigated the effect of Lavandula angustifolia EO in the memory updating of conditioned contextual fear. MATERIALS AND METHODS: Adult male C57Bl6 mice were submitted to fear conditioning. Two days after conditioning the mice underwent a reactivation session in a hybrid context and were then immediately exposed to vaporized water or essential oil at concentrations of 1%, 2.5% or 5% for 3 h. Two days later, the mice were tested in the original or an altered context and their freezing behavior was measured. In addition, mice were subjected to a fear memory recovery protocol followed by a reinstatement session. RESULTS: In the contextual fear test, 1% essential oil, but not 2.5% or 5%, reduced the freezing behavior response, whereas after a reinstatement session, exposure to 1% essential oil increased the freezing behavior response. CONCLUSIONS: These results suggest that Lavandula angustifolia essential oil enhances memory extinction and, consequently, inhibits memory updating.


Subject(s)
Aromatherapy , Behavior, Animal/drug effects , Conditioning, Psychological/drug effects , Extinction, Psychological/drug effects , Fear/drug effects , Memory/drug effects , Oils, Volatile/administration & dosage , Plant Oils/administration & dosage , Administration, Inhalation , Animals , Cues , Freezing Reaction, Cataleptic/drug effects , Humans , Lavandula , Mice, Inbred C57BL , Time Factors
6.
Brain Struct Funct ; 225(7): 1957-1966, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32594260

ABSTRACT

Electrical stimulation of the dorsolateral periaqueductal gray (dlPAG) in rats has been shown to elicit panic-like behaviour and can be a useful as an unconditioned stimulus for modelling anticipatory fear and agoraphobia in a contextual fear conditioning paradigm. In this study, we further analysed our previous data on the effects of escitalopram (a selective serotonin reuptake inhibitor, SSRI) and buspirone (a 5-HT1A receptor partial agonist) on dlPAG-induced anticipatory fear behaviour in a rat model using freezing as a measure. We then attempted to unravel some of the interactions with dopamine signalling using tyrosine hydroxylase (TH) immunohistochemistry to probe the effects on dopaminergic neurons. We showed that acute treatment of escitalopram, but not buspirone, was effective in reducing anticipatory freezing behaviour, while chronic administrations of both drugs were effective. We found that the dlPAG stimulation induced increase number of dopaminergic neurons in the ventral tegmental area (VTA) which was reversed in both chronic buspirone and escitalopram groups. We further found a strong positive correlation between the number of dopaminergic neurons and freezing in the VTA and showed positive correlations between dopaminergic neurons in the VTA and substantia nigra pars compacta (SNpc) in escitalopram and buspirone groups, respectively. Overall, we showed that chronic treatment with an SSRI and a 5-HT1A agonist reduced anticipatory freezing behaviour which seems to be associated, through correlative studies, with a reversal of dlPAG stimulation induced increase in number of dopaminergic neurons in the VTA and/or SNpc.


Subject(s)
Buspirone/pharmacology , Citalopram/pharmacology , Dopaminergic Neurons/drug effects , Mesencephalon/drug effects , Periaqueductal Gray/physiology , Selective Serotonin Reuptake Inhibitors/pharmacology , Serotonin Receptor Agonists/pharmacology , Animals , Deep Brain Stimulation , Dopaminergic Neurons/metabolism , Electric Stimulation , Fear/drug effects , Fear/physiology , Freezing Reaction, Cataleptic/drug effects , Freezing Reaction, Cataleptic/physiology , Male , Mesencephalon/metabolism , Rats , Rats, Wistar
7.
Horm Behav ; 124: 104777, 2020 08.
Article in English | MEDLINE | ID: mdl-32439347

ABSTRACT

Neuropeptide oxytocin (OT) is involved in the regulation of social and non-social behaviour. The central nucleus of amygdala (CeA), part of the limbic system, plays an important role in learning, memory, anxiety and reinforcing mechanisms. CeA has been shown to be rich in OT receptors in rodents. Our previous findings indicated that OT in the rat CeA has a dose dependent rewarding and anxiolytic effect. The aim of our present study was to examine in the CeA the possible interaction of OT and D2 dopamine (DA) receptor antagonist Sulpiride on reinforcement in place preference test and on anxiety in elevated plus maze test. Wistar rats were microinjected bilaterally with 10 ng OT. In different group of animals 4 µg D2 DA receptor antagonist was applied. Other animals received D2 DA receptor antagonist 15 min before 10 ng OT treatment or vehicle solution into the CeA. Rats receiving 10 ng OT spent significantly longer time in the treatment quadrant during the test session in conditioned place preference test. Prior treatment with D2 DA receptor antagonist blocked the rewarding effects of OT. Antagonist in itself did not influence the time rats spent in the treatment quadrant. In elevated plus maze test, rats receiving 10 ng OT spent significantly longer time on the open arms. Prior treatment with D2 DA receptor antagonist blocked the effects of OT. Our results show that DA system plays a role in positive reinforcing and anxiolytic effects of OT because D2 DA receptor antagonist can block these actions.


Subject(s)
Anti-Anxiety Agents/pharmacology , Oxytocin/pharmacology , Receptors, Dopamine D2/physiology , Spatial Behavior/drug effects , Amygdala/drug effects , Amygdala/metabolism , Animals , Anxiety/drug therapy , Behavior, Animal/drug effects , Conditioning, Classical/drug effects , Dopamine D2 Receptor Antagonists/pharmacology , Fear/drug effects , Freezing Reaction, Cataleptic/drug effects , Male , Maze Learning/drug effects , Rats , Rats, Wistar , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , Receptors, Oxytocin/metabolism , Receptors, Oxytocin/physiology , Reinforcement, Psychology , Reward , Sulpiride/pharmacology
8.
Pharmacol Biochem Behav ; 194: 172938, 2020 07.
Article in English | MEDLINE | ID: mdl-32376258

ABSTRACT

Evidence indicates that periaqueductal gray matter (PAG) plays an important role in defensive responses and pain control. The activation of cannabinoid type-1 (CB1) or mu-opioid (MOR) receptors in the dorsal region of this structure (dPAG) inhibits fear and facilitates antinociception induced by different aversive stimuli. However, it is still unknown whether these two receptors work cooperatively in order to achieve these inhibitory actions. This study investigated the involvement and a likely interplay between CB1 and MOR receptors localized into the dPAG on the regulation of fear-like defensive responses and antinociception (evaluated in tail-flick test) evoked by dPAG chemical stimulation with N-methyl-d-aspartate (NMDA). Before the administration of NMDA, animals were first intra-dPAG injected with the CB1 agonist ACEA (0.5 pmol), or with the MOR agonist DAMGO (0.5 pmol) in combination with the respective antagonists AM251 (CB1 antagonist, 100 pmol) or CTOP (MOR antagonist, 1 nmol). To investigate the interplay between these receptors, microinjection of CTOP was combined with ACEA, or microinjection of AM251 was combined with DAMGO. Our results showed that both the intra-PAG treatments with ACEA or DAMGO inhibited NMDA-induced freezing expression, whereas only the treatment with DAMGO increased antinociception induced with NMDA, which are completely blocked by its respective antagonists. Interestingly, the inhibitory effects of ACEA or DAMGO on freezing was blocked by CTOP and AM251, respectively, indicating a functional interaction between these two receptors in the mediation of defensive behaviors. However, this cooperative interaction was not observed during the NMDA-induced antinociception. Our findings indicate that there is a cooperative action between the MOR and CB1 receptors within the dPAG and it is involved in the mediation of NMDA-induced defensive responses. Additionally, the MORs into the dPAG are involved in the modulation of the antinociceptive effects that follow a fear-like defense-reaction induced by dPAG chemical stimulation with NMDA.


Subject(s)
Fear/drug effects , N-Methylaspartate/pharmacology , Nociception/drug effects , Periaqueductal Gray/metabolism , Receptor, Cannabinoid, CB1/metabolism , Receptors, Opioid, mu/metabolism , Analgesics, Opioid/pharmacology , Animals , Arachidonic Acids/pharmacology , Behavior, Animal/drug effects , Cannabinoid Receptor Agonists/pharmacology , Cannabinoid Receptor Antagonists/pharmacology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Freezing Reaction, Cataleptic/drug effects , Male , Microinjections , Pain/drug therapy , Pain/metabolism , Pain Measurement/drug effects , Piperidines/pharmacology , Pyrazoles/pharmacology , Rats , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/antagonists & inhibitors , Somatostatin/analogs & derivatives , Somatostatin/pharmacology
9.
Molecules ; 25(4)2020 Feb 24.
Article in English | MEDLINE | ID: mdl-32102354

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors with three isoforms (PPARα, PPARß/δ, PPARγ) and can regulate pain, anxiety, and cognition. However, their role in conditioned fear and pain-fear interactions has not yet been investigated. Here, we investigated the effects of systemically administered PPAR antagonists on formalin-evoked nociceptive behaviour, fear-conditioned analgesia (FCA), and conditioned fear in the presence of nociceptive tone in rats. Twenty-three and a half hours following fear conditioning to context, male Sprague-Dawley rats received an intraplantar injection of formalin and intraperitoneal administration of vehicle, PPARα (GW6471), PPARß/δ (GSK0660) or PPARγ (GW9662) antagonists, and 30 min later were re-exposed to the conditioning arena for 15 min. The PPAR antagonists did not alter nociceptive behaviour or fear-conditioned analgesia. The PPARα and PPARß/δ antagonists prolonged context-induced freezing in the presence of nociceptive tone without affecting its initial expression. The PPARγ antagonist potentiated freezing over the entire trial. In conclusion, pharmacological blockade of PPARα and PPARß/δ in the presence of formalin-evoked nociceptive tone, impaired short-term, within-trial fear-extinction in rats without affecting pain response, while blockade of PPARγ potentiated conditioned fear responding. These results suggest that endogenous signalling through these three PPAR isoforms may reduce the expression of conditioned fear in the presence of nociceptive tone.


Subject(s)
Conditioning, Psychological/drug effects , Fear/drug effects , Nociceptive Pain/drug therapy , PPAR alpha/genetics , PPAR delta/genetics , PPAR gamma/genetics , PPAR-beta/genetics , Analgesia/methods , Anilides/pharmacology , Animals , Extinction, Psychological/drug effects , Formaldehyde/administration & dosage , Freezing Reaction, Cataleptic/drug effects , Gene Expression , Male , Nociceptive Pain/chemically induced , Nociceptive Pain/physiopathology , Nociceptive Pain/psychology , Oxazoles/pharmacology , PPAR alpha/antagonists & inhibitors , PPAR alpha/metabolism , PPAR delta/antagonists & inhibitors , PPAR delta/metabolism , PPAR gamma/antagonists & inhibitors , PPAR gamma/metabolism , PPAR-beta/antagonists & inhibitors , PPAR-beta/metabolism , Rats , Rats, Sprague-Dawley , Sulfones/pharmacology , Thiophenes/pharmacology , Tyrosine/analogs & derivatives , Tyrosine/pharmacology
10.
Mol Neurobiol ; 57(5): 2144-2166, 2020 May.
Article in English | MEDLINE | ID: mdl-31960362

ABSTRACT

Frontocortical NMDA receptors are pivotal in regulating cognition and mood, are hypofunctional in schizophrenia, and may contribute to autistic spectrum disorders. Despite extensive interest in agents potentiating activity at the co-agonist glycine modulatory site, few comparative functional studies exist. This study systematically compared the actions of the glycine reuptake inhibitors, sarcosine (40-200 mg/kg) and ORG24598 (0.63-5 mg/kg), the agonists, glycine (40-800 mg/kg), and D-serine (10-160 mg/kg) and the partial agonists, S18841 (2.5 mg/kg s.c.) and D-cycloserine (2.5-40 mg/kg) that all dose-dependently prevented scopolamine disruption of social recognition in adult rats. Over similar dose ranges, they also prevented a delay-induced impairment of novel object recognition (NOR). Glycine reuptake inhibitors specifically elevated glycine but not D-serine levels in rat prefrontal cortical (PFC) microdialysates, while glycine and D-serine markedly increased levels of glycine and D-serine, respectively. D-Cycloserine slightly elevated D-serine levels. Conversely, S18841 exerted no influence on glycine, D-serine, other amino acids, monamines, or acetylcholine. Reversal of NOR deficits by systemic S18841 was prevented by the NMDA receptor antagonist, CPP (20 mg/kg), and the glycine modulatory site antagonist, L701,324 (10 mg/kg). S18841 blocked deficits in NOR following microinjection into the PFC (2.5-10 µg/side) but not the striatum. Finally, in rats socially isolated from weaning (a neurodevelopmental model of schizophrenia), S18841 (2.5 and 10 mg/kg s.c.) reversed impairment of NOR and contextual fear-motivated learning without altering isolation-induced hyperactivity. In conclusion, despite contrasting neurochemical profiles, partial glycine site agonists and glycine reuptake inhibitors exhibit comparable pro-cognitive effects in rats of potential relevance to treatment of schizophrenia and other brain disorders where cognitive performance is impaired.


Subject(s)
Cognition Disorders/drug therapy , Cognition/drug effects , Glycine Agents/pharmacology , Glycine/metabolism , Memory, Short-Term/drug effects , Neurotransmitter Uptake Inhibitors/pharmacology , Nootropic Agents/pharmacology , Amino Acids/analysis , Animals , Autism Spectrum Disorder/drug therapy , Cycloserine/pharmacology , Dose-Response Relationship, Drug , Freezing Reaction, Cataleptic/drug effects , Glycine/agonists , Glycine/analogs & derivatives , Glycine/pharmacology , Male , Motor Activity/drug effects , Prefrontal Cortex/drug effects , Rats , Rats, Wistar , Receptors, Glycine/drug effects , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/physiology , Recognition, Psychology/drug effects , Sarcosine/pharmacology , Schizophrenia/drug therapy , Scopolamine/antagonists & inhibitors , Serine/pharmacology , Social Behavior
11.
Behav Brain Res ; 377: 112236, 2020 01 13.
Article in English | MEDLINE | ID: mdl-31536735

ABSTRACT

CO2 inhalation can provoke panic attacks in humans, and the likelihood is increased in patients with panic disorder. Identifying brain sites involved could provide important mechanistic insight into the illness. In mice, the amygdala has been suggested to promote CO2 responses; however, recent studies in humans with amygdala damage indicate the amygdala is not required for CO2-induced fear and panic and might actually oppose these responses. To clarify the role of the amygdala, we produced lesions in mice paralleling the human lesions, and characterized behavioral responses to CO2. Compared to sham controls, we found that amygdala-lesioned mice froze less to 10% CO2, and unlike shams they also began to jump frenetically. At 20% CO2, controls also exhibited jumping, suggesting it is a normal response to more extreme CO2 concentrations. The effect of amygdala lesions was specific to CO2 as amygdala-lesioned mice did not jump in response to a predator odor or to an auditory conditioned stimulus. In amygdala-lesioned mice, jumping evoked by 10% CO2 was eliminated by co-lesioning the dorsal periaqueductal gray, a structure implicated in panic and escape-related behaviors. Together, these observations suggest a dual role for the amygdala in the CO2 response: promoting CO2-induced freezing, and opposing CO2-induced jumping, which may help explain the exaggerated CO2 responses in humans with amygdala lesions.


Subject(s)
Amygdala/physiology , Behavior, Animal , Carbon Dioxide/pharmacology , Fear/drug effects , Locomotion , Amygdala/pathology , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Carbon Dioxide/administration & dosage , Freezing Reaction, Cataleptic/drug effects , Freezing Reaction, Cataleptic/physiology , Locomotion/drug effects , Locomotion/physiology , Mice , Mice, Inbred C57BL , Periaqueductal Gray/pathology
12.
Dev Psychobiol ; 62(3): 380-385, 2020 04.
Article in English | MEDLINE | ID: mdl-31621064

ABSTRACT

The ontogeny and NMDA-receptor (NMDAR) mechanisms of context conditioning were examined during standard contextual fear conditioning (sCFC) - involving context and context-shock learning in the same trial - as a comparison with our previous reports on the Context Preexposure Facilitation Effect (CPFE), which separates these two types of learning by 24 hr. In Experiment 1, systemic administration of the NMDAR antagonist, MK-801, prior to conditioning disrupted retention but not post-shock freezing during sCFC in PD31 rats. Experiment 2 replicated and extended this effect to PD17 versus PD31 rats. Consistent with Experiment 1, pre-training MK-801 spared post-shock freezing but impaired retention freezing in PD31 rats. In contrast, pre-training MK-801 disrupted post-shock freezing in PD17 rats, which showed no retention freezing regardless of drug. These results reveal developmental differences in the role of NMDAR activity in the acquisition versus retention of a context-shock association during sCFC in pre-weanling and adolescent rats.


Subject(s)
Behavior, Animal/physiology , Conditioning, Classical/physiology , Excitatory Amino Acid Antagonists/pharmacology , Fear/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Retention, Psychology/physiology , Animals , Behavior, Animal/drug effects , Conditioning, Classical/drug effects , Dizocilpine Maleate/pharmacology , Fear/drug effects , Female , Freezing Reaction, Cataleptic/drug effects , Freezing Reaction, Cataleptic/physiology , Male , Rats , Rats, Long-Evans , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Retention, Psychology/drug effects
13.
Horm Behav ; 119: 104651, 2020 03.
Article in English | MEDLINE | ID: mdl-31790664

ABSTRACT

The estrogen receptor (ER) mechanisms by which 17ß-estradiol influences depressive-like behaviour have primarily been investigated acutely and not within an animal model of depression. Therefore, the current study aimed to dissect the contribution of ERα and ERß to the effects of 17ß-estradiol under non-stress and chronic stress conditions. Ovariectomized (OVX) or sham-operated mice were treated chronically (47 days) with 17ß-estradiol (E2), the ERß agonist diarylpropionitrile (DPN), the ERα agonist propylpyrazole-triol (PPT), or vehicle. On day 15 of treatment, mice from each group were assigned to chronic unpredictable stress (CUS; 28 days) or non-CUS conditions. Mice were assessed for anxiety- and depressive-like behaviour and hypothalamic-pituitary-adrenal (HPA) axis function. Cytokine and chemokine levels, and postsynaptic density protein 95 were measured in the hippocampus and frontal cortex, and adult hippocampal neurogenesis was assessed. Overall, the effects of CUS were more robust that those of estrogenic treatments, as seen by increased immobility in the tail suspension test (TST), reduced PSD-95 expression, reduced neurogenesis in the ventral hippocampus, and HPA axis negative feedback dysregulation. However, we also observe CUS-dependent and -independent effects of ovarian status and estrogenic treatments. The effects of CUS on PSD-95 expression, the cytokine milieu, and in TST were largely driven by PPT and DPN, indicating that these treatments were not protective. Independent of CUS, estradiol increased neurogenesis in the dorsal hippocampus, blunted the corticosterone response to an acute stressor, and increased anxiety-like behaviour. These findings provide insights into the complexities of estrogen signaling in modulating depressive-like phenotypes under non-stress and chronic stress conditions.


Subject(s)
Depression/metabolism , Estrogen Receptor alpha/agonists , Estrogen Receptor beta/agonists , Stress, Psychological/metabolism , Animals , Chronic Disease , Corticosterone/metabolism , Depression/etiology , Depression/psychology , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Estrogen Receptor beta/metabolism , Female , Freezing Reaction, Cataleptic/drug effects , Freezing Reaction, Cataleptic/physiology , Hippocampus/drug effects , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , Mice , Mice, Inbred C57BL , Nitriles/pharmacology , Ovariectomy , Phenols/pharmacology , Phenotype , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/metabolism , Propionates/pharmacology , Pyrazoles/pharmacology , Stress, Psychological/complications , Stress, Psychological/pathology , Stress, Psychological/psychology
14.
Behav Brain Res ; 381: 112380, 2020 03 02.
Article in English | MEDLINE | ID: mdl-31765726

ABSTRACT

Buspirone presents a unique profile of action, which involves activation of 5-HT1A receptors and complex effects on D2-like dopaminergic receptors. This medication is studied in terms of potential clinical repositioning to conditions that are associated with dopaminergic dysfunctions including schizophrenia and substance use disorder. Buspirone antagonizes D3 and D4 receptors, however, depending on the dose it differentially interacts with D2 receptors. Previously, we reported that some of D2/D3 dopaminergic agonists attenuate PTSD-like behavioral symptoms in mice. Here we investigated whether buspirone could also affect PTSD-like symptoms. We used the single prolonged stress (mSPS) protocol to induce PTSD-like behavior in adult male CD-1 mice. Buspirone (0.5, 2, or 10 mg/kg, i.p.) was injected for 15 consecutive days. The subjects were repeatedly examined in a variety of behavioral tests measuring conditioned freezing response, antidepressant-like effects, anxiety, and ultrasonic vocal response to the restraint stress. Mouse SPS resulted in prolonged immobility in the forced swim test and freezing in the fear-conditioning test, and produced symptoms of anxiety. Buspirone dose-dependently decreased the exaggerated freezing response in mice, but only at the dose of 2 mg/kg exhibited the anxiolytic-like effect in the elevated plus maze test. Buspirone reduced the number of ultrasonic calls in mSPS-exposed mice but revealed no antidepressant-like effect in the forced swim test. Present data suggest some positive effects of buspirone in the treatment of selected PTSD-like symptoms and prompt for its further clinical evaluation.


Subject(s)
Behavior, Animal/drug effects , Buspirone/pharmacology , Serotonin Receptor Agonists/pharmacology , Stress Disorders, Post-Traumatic/physiopathology , Animals , Conditioning, Psychological/drug effects , Disease Models, Animal , Fear/drug effects , Freezing Reaction, Cataleptic/drug effects , Locomotion/drug effects , Mice , Receptors, Dopamine D2/drug effects , Restraint, Physical , Vocalization, Animal/drug effects
15.
Aquat Toxicol ; 216: 105310, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31580997

ABSTRACT

Pesticide mixtures are increasingly used to fight pest species that developed resistance to pesticides. To assess the pesticide control efficiency and to reduce ecological damage to non-target species, it is important to quantify the effect of these mixtures and compare them with the effect of their single pesticides on pest species, non-target species and their predator-prey interactions. We studied the effects of the chemical pesticide chlorpyrifos (CPF), the biopesticide Bacillus thuringiensis israelensis (Bti) and their mixture both on the direct mortality and on the mortality by predation. We focused on larvae of a CPF-resistant and a non-resistant strain of the vector mosquito Culex quinquefasciatus and its predator, the pygmy backswimmer Plea minutissima. In the CPF-Bti mixture, both pesticides interacted antagonistically for direct mortality. Exposure to the mixture caused equal direct mortality and equal mortality by predation in both strains. As expected, exposure to CPF resulted in less direct mortality and less mortality by predation in the CPF-resistant mosquito strain compared to the non-resistant strain. Notably, Bti caused a higher mortality in the mosquito larvae of the CPF-resistant strain compared to the non-resistant strain. Furthermore, the predator killed more mosquito larvae of the resistant strain compared to the non-resistant strain when exposed before to Bti alone. These observations identify a novel cost of resistance to a chemical pesticide in terms of increased vulnerability to a biopesticide.


Subject(s)
Biological Control Agents/toxicity , Pesticides/toxicity , Predatory Behavior/drug effects , Animals , Bacillus thuringiensis/drug effects , Chlorpyrifos/toxicity , Culex/drug effects , Freezing Reaction, Cataleptic/drug effects , Heteroptera/drug effects , Larva/drug effects , Linear Models , Swimming , Water Pollutants, Chemical/toxicity
16.
Neurobiol Learn Mem ; 166: 107088, 2019 12.
Article in English | MEDLINE | ID: mdl-31513850

ABSTRACT

Female reproductive experience has been shown to alter the hormonal, neurobiological and behavioural features of fear extinction, which is the laboratory basis of exposure therapy. This raises uncertainties as to whether pharmacological agents that enhance fear extinction in reproductively inexperienced females are equally effective in reproductively experienced females. The aim of the current study was therefore to compare the effects of two pharmacological enhancers of fear extinction, d-cycloserine (DCS) and estradiol, between nulliparous (virgin) and primiparous (reproductively experienced) female rats. In Experiment 1, nulliparous and primiparous females received systemic administration of either DCS or saline immediately after extinction training, and were tested for extinction recall the following day. DCS enhanced extinction recall in nulliparous females that showed low levels of freezing at the end of extinction training, but not among those that showed high levels of freezing at the end of extinction training. DCS did not enhance fear extinction in primiparous females, regardless of their level of freezing at the end of extinction training. In Experiment 2, nulliparous and primiparous female rats received systemic administration of either estradiol or vehicle prior to extinction training. Estradiol enhanced extinction recall among nulliparous females, but not primiparous females. Increasing the dose of estradiol administered prior to extinction training did not alter the outcomes in primiparous females (Experiment 3). Together, these findings suggest that reproductive status may be an important individual difference factor associated with the response to pharmacological modulators of extinction in rats. The implications of these findings for the pharmacological augmentation of exposure therapy in clinical populations are discussed.


Subject(s)
Cycloserine/pharmacology , Estradiol/pharmacology , Extinction, Psychological/drug effects , Fear/drug effects , Parity , Animals , Conditioning, Classical/drug effects , Female , Freezing Reaction, Cataleptic/drug effects , Rats , Rats, Sprague-Dawley
17.
Learn Mem ; 26(7): 245-251, 2019 07.
Article in English | MEDLINE | ID: mdl-31209119

ABSTRACT

Vagus nerve stimulation (VNS) enhances extinction of conditioned fear in rats. Previous findings support the hypothesis that VNS effects on extinction are due to enhanced consolidation of extinction memories through promotion of plasticity in extinction-related brain pathways however, alternative explanations are plausible. According to one hypothesis, VNS may produce a hedonic effect and enhance extinction through counter-conditioning. According to another hypothesis, VNS reduces anxiety during exposure and this weakens the association of conditioned stimuli with aversive conditioned responses. The present set of experiments (1) used conditioned place preference (CPP) to identify potential rewarding effects associated with VNS and (2) examined the peripheral effects of VNS on anxiety and extinction enhancement. Male Sprague-Dawley rats were surgically implanted with cuff electrodes around the vagus nerve and subjected to a CPP task in which VNS and sham stimulation were each paired with one of two distinct contexts over the course of 5 d. Following this procedure, rats did not show a place preference, suggesting that VNS is not rewarding or aversive. The role of the peripheral parasympathetic system in the anxiolytic effect of VNS on the elevated plus maze was examined by blocking peripheral muscarinic receptors with intraperitoneal administration of methyl scopolamine prior to VNS. Methyl scopolamine blocked the VNS-induced reduction in anxiety but did not interfere with VNS enhancement of extinction of conditioned fear, indicating that the anxiety-reducing effect of VNS is not necessary for the extinction enhancement.


Subject(s)
Anxiety/physiopathology , Extinction, Psychological/physiology , Fear/physiology , Parasympathetic Nervous System/physiopathology , Vagus Nerve Stimulation , Animals , Anxiety/drug therapy , Conditioning, Classical/physiology , Efferent Pathways/physiology , Electrodes, Implanted , Electroshock , Exploratory Behavior/drug effects , Exploratory Behavior/physiology , Freezing Reaction, Cataleptic/drug effects , Freezing Reaction, Cataleptic/physiology , Male , Maze Learning/physiology , Models, Neurological , Models, Psychological , Muscarinic Antagonists/pharmacology , Muscarinic Antagonists/therapeutic use , N-Methylscopolamine/pharmacology , N-Methylscopolamine/therapeutic use , Rats , Rats, Sprague-Dawley , Receptors, Muscarinic/physiology
18.
Neurochem Int ; 129: 104488, 2019 10.
Article in English | MEDLINE | ID: mdl-31202882

ABSTRACT

Aversive conditions elicit anxiety responses that prepare the organism to an eventual threat. Nonetheless, prolonged anxiety is a pathological condition associated with various neuropsychiatric disorders. Here, we evaluated whether the conspecific alarm substance (CAS), a chemical cue that elicits aversion, influences anxiety-like behaviors and modulates brain oxidative stress-related parameters in wild-type (WT) and leopard (leo) zebrafish following a repeated exposure protocol. CAS exposure was performed for 5 min, once daily for 7 consecutive days. In the 8th day, animals were tested in the light/dark and novel tank tests and their brains were further dissected for biochemical analyses. CAS chronically induced anxiogenic-like states in WT and leo populations when their behaviors were analyzed in the light/dark and novel tank tests. CAS also increased catalase (CAT) and glutathione S-transferase (GST) activities, as well as non-protein thiol (NPSH) content in WT and leo, but only leo had increased thiobarbituric reactive substance (TBARS) levels in the brain. At baseline conditions, leo was more 'anxious' when compared to WT, displaying lower CAT activity and carbonylated protein (CP) levels. Overall, CAS chronically triggers anxiety-like behavior in zebrafish populations, which may be associated with changes in oxidative stress-related parameters. Furthermore, the use of different zebrafish populations may serve as an interesting tool in future research aiming to investigate the neurobehavioral bases of neuropsychiatric disorders in vertebrates.


Subject(s)
Anxiety/physiopathology , Avoidance Learning/physiology , Brain/physiopathology , Exploratory Behavior/physiology , Fear/physiology , Freezing Reaction, Cataleptic/physiology , Oxidative Stress , Zebrafish/physiology , Animals , Anxiety/chemically induced , Anxiety/genetics , Avoidance Learning/drug effects , Brain/metabolism , Catalase/analysis , Exploratory Behavior/drug effects , Fear/drug effects , Female , Freezing Reaction, Cataleptic/drug effects , Glutathione Transferase/analysis , Lipid Peroxidation/drug effects , Male , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Oxidative Stress/drug effects , Pheromones/pharmacology , Protein Carbonylation/drug effects , Sulfhydryl Compounds/analysis , Superoxide Dismutase/analysis , Tissue Extracts/pharmacology , Zebrafish/genetics , Zebrafish Proteins/analysis , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics , Zebrafish Proteins/physiology
19.
Article in English | MEDLINE | ID: mdl-30776402

ABSTRACT

Individuals with fear-associated conditions such as panic disorder (PD) and posttraumatic stress disorder (PTSD) display increased emotional responses to interoceptive triggers, such as CO2 inhalation, that signal a threat to physiological homeostasis. Currently, effector systems and mechanisms underlying homeostatic modulation of fear memory are not well understood. In this regard, the renin angiotensin system (RAS), particularly the angiotensin receptor type 1 (AT1R), a primary homeostatic regulatory target, has gained attention. RAS polymorphisms have been reported in PD and PTSD, and recent studies report AT1R-mediated modulation of fear extinction. However, contribution of AT1Rs in fear evoked by the interoceptive threat of CO2 has not been investigated. Using pharmacological, behavioral, and AT1R/ACE gene transcription analyses, we assessed central AT1R recruitment in CO2-associated fear. CO2 inhalation led to significant AT1R and ACE mRNA upregulation in homeostatic regulatory regions, subfornical organ (SFO) and paraventricular nucleus (PVN), in a temporal manner. Intracerebroventricular infusion of selective AT1R antagonist, losartan, significantly attenuated freezing during CO2 inhalation, and during re-exposure to CO2 context, suggestive of AT1R modulation of contextual fear. Regional Fos mapping in losartan-treated mice post-behavior revealed significantly attenuated labeling in areas regulating defensive behavior, contextual fear, and threat responding; such as, the bed nucleus of stria terminalis, dorsal periaqueductal gray, hypothalamic nuclei, hippocampus, and prefrontal areas such as the prelimbic, infralimbic, and anterior cingulate cortices. Sub-regions of the amygdala did not show CO2-associated AT1R regulation or altered Fos labeling. Collectively, our data suggests central AT1R recruitment in modulation of fear behaviors associated with CO2 inhalation via engagement of neurocircuits regulating homeostasis and defensive behaviors. Our data provides mechanistic insights into the interoceptive regulation of fear, relevant to fear related disorders such as PD and PTSD.


Subject(s)
Carbon Dioxide/metabolism , Fear/physiology , Neural Pathways/physiology , Receptor, Angiotensin, Type 1/physiology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin-Converting Enzyme Inhibitors/metabolism , Animals , Brain/physiology , Freezing Reaction, Cataleptic/drug effects , Homeostasis/physiology , Infusions, Intraventricular , Losartan/pharmacology , Male , Mice , Paraventricular Hypothalamic Nucleus/metabolism , Receptor, Angiotensin, Type 1/metabolism , Subfornical Organ/metabolism , Up-Regulation
20.
Behav Brain Res ; 356: 227-235, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30098408

ABSTRACT

We examined interval timing - time perception in the seconds-to-minutes range - of the fear-inducing stimulus and the role of the amygdala in this phenomenon. Rats were initially trained to perform a temporal bisection task, in which their responses to levers A and B were reinforced following 2-s and 8-s tones, respectively. After acquisition, the rats were also presented with tones of intermediate durations and pressed one of the two levers to indicate whether the tone duration was closer to 2 or 8 s. Subsequently, the rats underwent differential fear conditioning, in which one frequency tone (conditioned stimulus; CS+) was paired with an electric foot shock, whereas another frequency tone (CS-) was presented alone. The rats were then infused with artificial cerebrospinal fluid (aCSF) or the GABAA agonist muscimol into the bilateral basolateral amygdala (BLA) before performing the bisection task with CS+ and CS-. In rats infused with aCSF, the psychophysical function shifted rightward in CS+ relative to that in CS-. Moreover, the point of subjective equality of the CS+ was higher than that of CS-, suggesting that the duration of the fear -CS was perceived as shorter than that of the neutral CS. However, muscimol infusion into the BLA abolished this difference, suggesting that BLA inactivation suppresses the effect of the fear -CS. Our results demonstrate that normal BLA activity is essential for fear-induced underestimation of time.


Subject(s)
Amygdala/drug effects , Basolateral Nuclear Complex/drug effects , Fear/physiology , Animals , Conditioning, Classical/drug effects , Conditioning, Classical/physiology , Freezing Reaction, Cataleptic/drug effects , GABA Agonists/pharmacology , Male , Muscimol/pharmacology , Rats, Wistar , Time Perception/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...