Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.850
Filter
1.
Article in English | MEDLINE | ID: mdl-38692472

ABSTRACT

Stress exposure can lead to post-traumatic stress disorder (PTSD) in male and female rats. Social-Single Prolonged Stress (SPS) protocol has been considered a potential PTSD model. This study aimed to pharmacologically validate the Social-SPS as a PTSD model in male and female rats. Male and female Wistar rats (60-day-old) were exposed to Social-SPS protocol and treated with fluoxetine (10 mg/Kg) or saline solution intraperitoneally 24 h before euthanasia. Two cohorts of animals were used; for cohort 1, male and female rats were still undisturbed until day 7 post-Social-SPS exposure, underwent locomotor and conditioned fear behaviors, and were euthanized on day 9. Animals of cohort 2 were subjected to the same protocol but were re-exposed to contextual fear behavior on day 14. Results showed that fluoxetine-treated rats gained less body weight than control and Social-SPS in both sexes. Social-SPS effectively increased the freezing time in male and female rats on day eight but not on day fourteen. Fluoxetine blocked the increase of freezing in male and female rats on day 8. Different mechanisms for fear behavior were observed in males, such as Social-SPS increased levels of glucocorticoid receptors and Beclin-1 in the amygdala. Social-SPS was shown to increase the levels of NMDA2A, GluR-1, PSD-95, and CAMKII in the amygdala of female rats. No alterations were observed in the amygdala of rats on day fourteen. The study revealed that Social-SPS is a potential PTSD protocol applicable to both male and female rats.


Subject(s)
Amygdala , Fear , Fluoxetine , Rats, Wistar , Stress, Psychological , Animals , Male , Female , Fear/drug effects , Fear/physiology , Fluoxetine/pharmacology , Amygdala/drug effects , Amygdala/metabolism , Stress, Psychological/metabolism , Rats , Disease Models, Animal , Stress Disorders, Post-Traumatic/metabolism , Stress Disorders, Post-Traumatic/psychology , Conditioning, Classical/drug effects , Conditioning, Classical/physiology , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Selective Serotonin Reuptake Inhibitors/pharmacology , Disks Large Homolog 4 Protein , Receptors, AMPA
2.
Psychopharmacology (Berl) ; 241(6): 1191-1203, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38383904

ABSTRACT

RATIONALE: Preclinical studies report attenuated ethanol-induced conditioned taste aversion (CTA) following chronic ethanol exposure, suggesting that tolerance develops to the aversive properties of ethanol. However, these studies are confounded by pre-exposure to the unconditioned stimulus (US; ethanol), which is well known to hinder conditioning. OBJECTIVES: This study was designed to determine whether chronic ethanol exposure produces tolerance to the aversive properties of ethanol in the absence of a US pre-exposure confound. METHODS: CTA was performed in adult male and female Long-Evans rats by pairing 0.1% ingested saccharin with an intraperitoneal injection of ethanol (1.5 or 2.0 g/kg) or saline. Rats were then rendered ethanol dependent using chronic intermittent ethanol (CIE) vapor exposure. Controls were exposed to room air (AIR). The effect of chronic ethanol on CTA expression and reconditioning were examined following vapor exposure. RESULTS: Prior to vapor exposure, both sexes developed CTA to a comparable degree with 2.0 g/kg producing greater CTA than 1.5 g/kg ethanol. Following vapor exposure, AIR controls exhibited an increase in CTA magnitude compared to pre-vapor levels. This effect was largely absent in CIE-exposed rats. Re-conditioning after vapor exposure facilitated increased CTA magnitude to a similar degree in AIR- and CIE-exposed males. In contrast, CTA magnitude was unchanged by re-conditioning in females. CONCLUSIONS: These data suggest that chronic ethanol does not facilitate tolerance to the aversive properties of ethanol but rather attenuates incubation of ethanol-induced CTA. Loss of CTA incubation suggests that CIE exposure disrupts circuits encoding aversion.


Subject(s)
Avoidance Learning , Ethanol , Rats, Long-Evans , Saccharin , Taste , Animals , Male , Ethanol/administration & dosage , Ethanol/pharmacology , Female , Rats , Avoidance Learning/drug effects , Avoidance Learning/physiology , Taste/drug effects , Saccharin/administration & dosage , Disease Models, Animal , Alcoholism/physiopathology , Dose-Response Relationship, Drug , Conditioning, Classical/drug effects , Conditioning, Psychological/drug effects
3.
Int J Mol Sci ; 23(3)2022 Feb 08.
Article in English | MEDLINE | ID: mdl-35163844

ABSTRACT

Although women and men are equally likely to receive ketamine following traumatic injury, little is known regarding sex-related differences in the impact of ketamine on traumatic memory. We previously reported that subanesthetic doses of an intravenous (IV) ketamine infusion following fear conditioning impaired fear extinction and altered regional brain glucose metabolism (BGluM) in male rats. Here, we investigated the effects of IV ketamine infusion on fear memory, stress hormone levels, and BGluM in female rats. Adult female Sprague-Dawley rats received a single IV ketamine infusion (0, 2, 10, or 20 mg/kg, over a 2-h period) following auditory fear conditioning (three pairings of tone and footshock). Levels of plasma stress hormones, corticosterone (CORT) and progesterone, were measured after the ketamine infusion. Two days after ketamine infusion, fear memory retrieval, extinction, and renewal were tested over a three-day period. The effects of IV ketamine infusion on BGluM were determined using 18F-fluoro-deoxyglucose positron emission tomography (18F-FDG-PET) and computed tomography (CT). The 2 and 10 mg/kg ketamine infusions reduced locomotor activity, while 20 mg/kg infusion produced reduction (first hour) followed by stimulation (second hour) of activity. The 10 and 20 mg/kg ketamine infusions significantly elevated plasma CORT and progesterone levels. All three doses enhanced fear memory retrieval, impaired fear extinction, and enhanced cued fear renewal in female rats. Ketamine infusion produced dose-dependent effects on BGluM in fear- and stress-sensitive brain regions of female rats. The current findings indicate that subanesthetic doses of IV ketamine produce robust effects on the hypothalamic-pituitary-adrenal (HPA) axis and brain energy utilization that may contribute to enhanced fear memory observed in female rats.


Subject(s)
Anesthetics, Dissociative/administration & dosage , Brain/diagnostic imaging , Conditioning, Psychological/drug effects , Fear/drug effects , Glucose/metabolism , Ketamine/administration & dosage , Anesthetics, Dissociative/adverse effects , Animals , Brain/drug effects , Brain/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Fluorodeoxyglucose F18/administration & dosage , Fluorodeoxyglucose F18/metabolism , Infusions, Intravenous , Ketamine/adverse effects , Positron-Emission Tomography , Rats , Rats, Sprague-Dawley , Tomography, X-Ray Computed
4.
Pharmacol Biochem Behav ; 213: 173338, 2022 02.
Article in English | MEDLINE | ID: mdl-35038444

ABSTRACT

Nicotine-induced rewarding and mood altering effects contribute to the continued use of nicotine and the subsequent development of nicotine dependence. The goal of this study was to assess the role of two specific regulators of G-protein signaling (RGS) proteins namely RGS2 and RGS4 in the above described effects of nicotine. Male and female mice lacking either RGS2 (RGS2 KO) or RGS4 (RGS4 KO), and their respective wildtype (WT) littermates were used in this study. The rewarding effects of nicotine (0.5 mg/kg, base; s.c.) were assessed using the conditioned place preference model. Nicotine-induced anxiolytic-like (0.1 mg/kg, base; i.p.) and antidepressant-like (1 mg/kg, base; i.p.) effects were assessed using the elevated plus maze and tail suspension test, respectively. We also assessed effects of nicotine (0, 0.05, 0.1 & 0.5 mg/kg, base; s.c.) on spontaneous locomotor activity. Nicotine-induced rewarding and antidepressant-like effects were observed in both male and female RGS2 WT mice, but not in mice lacking RGS2 compared to respective controls. In contrast, nicotine-induced rewarding and antidepressant-like effects were observed in both male and female mice lacking RGS4 and their WT littermates. Interestingly, deletion of RGS4 facilitated antidepressant-like effect of nicotine in male, but not female mice compared to respective WT littermates. Nicotine-induced anxiolytic-like effect was not influenced by deletion of either RGS2 or RGS4, irrespective of sex. Nicotine (0.5 mg/kg) decreased locomotor activity in both WT and KO mice compared to respective saline, irrespective of genotype and sex. Taken together, these data provide evidence that RGS2, but not RGS4, plays a role in mediating the rewarding and antidepressant-like effects of nicotine. Further research is required to explore the role of RGS2 after chronic exposure to nicotine.


Subject(s)
Antidepressive Agents/pharmacology , Depression/drug therapy , Nicotine/pharmacology , RGS Proteins/metabolism , Reward , Animals , Anti-Anxiety Agents/pharmacology , Anxiety/drug therapy , Anxiety/metabolism , Conditioning, Psychological/drug effects , Depression/metabolism , Elevated Plus Maze Test , Female , Locomotion/drug effects , Male , Mice
5.
Pharmacol Biochem Behav ; 213: 173319, 2022 02.
Article in English | MEDLINE | ID: mdl-34990706

ABSTRACT

We aimed to determine whether REM sleep deprivation (RSD) affects extinction and reinstatement of methamphetamine (METH) reward memory in male rats and also to evaluate the possible role of dopamine D1-like and D2-like dopamine (DA) receptors in these processes. Male rats were trained to acquire METH-induced place preference (2 mg/kg, i.p.). METH reward memory was then reinstated following a 10-day extinction period. The animals underwent a 72-hour sleep deprivation episode by multiple platforms method (in separate groups), either before the extraction or before the reinstatement of METH reward memory. The animals received SCH 23390 (0.01 or 0.05 mg/kg, i.p.) or sulpiride (20 or 60 mg/kg, i.p.) as antagonists of D1-like and D2-like DA receptors, respectively, either immediately following each daily extinction session or before the reinstatement of METH-seeking behavior. The RSD episode postponed extinction and facilitated reinstatement of METH reward memory. Administration of SCH 23390, but not sulpiride, facilitated METH extinction and decreased reinstatement of the extinguished METH-seeking behavior. Moreover, locomotor activity was not affected by METH and/or the RSD paradigm. The results would seem to suggest that the D1-like, but not the D2-like, DA receptors may be involved in the extinction and reinstatement of the extinguished METH reward memory in RSD animals. Nonetheless, more investigations are needed to elucidate the exact mechanisms involved.


Subject(s)
Drug-Seeking Behavior/drug effects , Methamphetamine/pharmacology , REM Sleep Behavior Disorder/metabolism , Receptors, Dopamine D1/metabolism , Sleep Deprivation/metabolism , Animals , Benzazepines/pharmacology , Conditioning, Psychological/drug effects , Dopamine Agents/pharmacology , Extinction, Psychological/drug effects , Locomotion/drug effects , Male , Memory/drug effects , Rats , Rats, Wistar , Receptors, Dopamine D2/metabolism , Reward , Sleep, REM , Sulpiride/pharmacology
6.
Naunyn Schmiedebergs Arch Pharmacol ; 395(3): 365-376, 2022 03.
Article in English | MEDLINE | ID: mdl-34997272

ABSTRACT

Agmatine (AG), idazoxan (IDZ), and efaroxan (EFR) are imidazoline receptor ligands with beneficial effects in central nervous system disorders. The present study aimed to evaluate the interaction between AG, IDZ, and EFR with an opiate, tramadol (TR), in a conditioned place preference (CPP) paradigm. In the experiment, we used five groups with 8 adult male Wistar rats each. During the condition session, on days 2, 4, 6, and 8, the rats received the drugs (saline, or TR, or IDZ and TR, or EFR and TR, or AG and TR) and were placed in their least preferred compartment. On days 1, 3, 5, and 7, the rats received saline in the preferred compartment. In the preconditioning, the preferred compartment was determined. In the postconditioning, the preference for one of the compartments was reevaluated. TR increased the time spent in the non-preferred compartment. AG decreased time spent in the TR-paired compartment. EFR, more than IDZ, reduced the time spent in the TR-paired compartment, but without statistical significance. AG reversed the TR-induced CPP, while EFR and IDZ only decreased the time spent in the TR-paired compartment, without statistical significance.


Subject(s)
Agmatine/pharmacology , Benzofurans/pharmacology , Idazoxan/pharmacology , Imidazoles/pharmacology , Tramadol/pharmacology , Analgesics, Opioid/pharmacology , Animals , Conditioning, Psychological/drug effects , Drug Interactions , Imidazoline Receptors/drug effects , Imidazoline Receptors/metabolism , Male , Rats , Rats, Wistar , Time Factors
7.
Behav Brain Res ; 417: 113611, 2022 01 24.
Article in English | MEDLINE | ID: mdl-34592376

ABSTRACT

Fear extinction (FExt) is used to treat patients with posttraumatic stress disorder (PTSD). However, fear related to traumatic events can be persistent and return even after successful extinction. The neurochemical control of extinction seems to be performed by several neurotransmitters, including dopamine (DA), through D1 and D2 receptors. Recently, we showed that intranasally applied DA (IN-DA) facilitated the FExt, but the mechanisms by which it promoted this effect are still unknown. This study focused on investigating whether these effects are mediated by the action of DA on D2-like receptors since these receptors seem to be related to neurochemical and molecular changes underlying extinction. Also, we investigated whether IN-DA treatment would affect conditioned fear-induced antinociception (Fear-IA). Rats treated with IN-DA (1 mg/kg) twenty-five minutes after sulpiride (SUL; 40 mg/kg, i.p., D2-antagonist) were subjected to the extinction of contextual fear. IN-DA applied before the extinction session induced the FExt and prevented Fear-IA. These effects were impaired by pre-treatment with SUL, suggesting that the IN-DA effects are mediated by DA on D2-like receptors. SUL per se also facilitated the FExt but did not affect Fear-IA. These data suggest IN-DA as a promising pharmacological tool to supplement the psychotherapy of patients suffering from PTSD.


Subject(s)
Conditioning, Psychological/physiology , Dopamine D2 Receptor Antagonists/pharmacology , Dopamine/pharmacology , Extinction, Psychological/physiology , Fear/physiology , Receptors, Dopamine D2/physiology , Sulpiride/pharmacology , Administration, Intranasal , Animals , Conditioning, Psychological/drug effects , Dopamine Agents/pharmacology , Extinction, Psychological/drug effects , Male , Rats , Sulpiride/antagonists & inhibitors
8.
Behav Brain Res ; 418: 113644, 2022 02 10.
Article in English | MEDLINE | ID: mdl-34757001

ABSTRACT

Epigenetic mechanisms play important roles in the neurobiology of substance use disorder. In particular, bromodomain and extra-terminal domain (BET) proteins, a class of histone acetylation readers, have been found to regulate cocaine conditioned behaviors, but their role in the behavioral response to other drugs of abuse remains unclear. To address this knowledge gap, we examined the effects of the BET inhibitor, JQ1, on nicotine, amphetamine, morphine, and oxycodone conditioned place preference (CPP). Similar to previous cocaine studies, systemic administration of JQ1 caused a dose-dependent reduction in the acquisition of amphetamine and nicotine CPP in male mice. However, in opioid studies, JQ1 did not alter morphine or oxycodone CPP. Investigating the effects of JQ1 on other types of learning and memory, we found that JQ1 did not alter the acquisition of contextual fear conditioning. Together, these results indicate that BET proteins play an important role in the acquisition of psychostimulant-induced CPP but not the acquisition of opioid-induced CPP nor contextual fear conditioning.


Subject(s)
Anesthetics, Local/pharmacology , Azepines/administration & dosage , Behavior, Animal/drug effects , Central Nervous System Stimulants/pharmacology , Conditioning, Psychological/drug effects , Dose-Response Relationship, Drug , Triazoles/administration & dosage , Amphetamine/pharmacology , Animals , Cocaine/pharmacology , Epigenomics , Learning/drug effects , Male , Memory/drug effects , Mice , Morphine/pharmacology , Nicotine/pharmacology
9.
PLoS One ; 16(12): e0260577, 2021.
Article in English | MEDLINE | ID: mdl-34898621

ABSTRACT

Females are more affected by psychiatric illnesses including eating disorders, depression, and post-traumatic stress disorder than males. However, the neural mechanisms mediating these sex differences are poorly understood. Animal models can be useful in exploring such neural mechanisms. Conditioned taste aversion (CTA) is a behavioral task that assesses how animals process the competition between associated reinforcing and aversive stimuli in subsequent task performance, a process critical to healthy behavior in many domains. The purpose of the present study was to identify sex differences in this behavior and associated neural responses. We hypothesized that females would value the rewarding stimulus (Boost®) relative to the aversive stimulus (LiCl) more than males in performing CTA. We evaluated behavior (Boost® intake, LiCl-induced behaviors, ultrasonic vocalizations (USVs), CTA performance) and Fos activation in relevant brain regions after the acute stimuli [acute Boost® (AB), acute LiCl (AL)] and the context-only task control (COT), Boost® only task (BOT) and Boost®-LiCl task (BLT). Acutely, females drank more Boost® than males but showed similar aversive behaviors after LiCl. Females and males performed CTA similarly. Both sexes produced 55 kHz USVs anticipating BOT and inhibited these calls in the BLT. However, more females emitted both 22 kHz and 55 kHz USVs in the BLT than males: the latter correlated with less CTA. Estrous cycle stage also influenced 55 kHz USVs. Fos responses were similar in males and females after AB or AL. Females engaged the gustatory cortex and ventral tegmental area (VTA) more than males during the BOT and males engaged the amygdala more than females in both the BOT and BLT. Network analysis of correlated Fos responses across brain regions identified two unique networks characterizing the BOT and BLT, in both of which the VTA played a central role. In situ hybridization with RNAscope identified a population of D1-receptor expressing cells in the CeA that responded to Boost® and D2 receptor-expressing cells that responded to LiCl. The present study suggests that males and females differentially process the affective valence of a stimulus to produce the same goal-directed behavior.


Subject(s)
Conditioning, Psychological , Proto-Oncogene Proteins c-fos/metabolism , Acoustic Stimulation , Amygdala/drug effects , Amygdala/metabolism , Animals , Conditioning, Psychological/drug effects , Conditioning, Psychological/radiation effects , Female , Lithium Chloride/pharmacology , Male , Rats , Sex Characteristics , Ultrasonics
10.
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
11.
Neurosci Lett ; 764: 136235, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34508846

ABSTRACT

The mechanisms involved in the maintenance of cigarette smoking and nicotine reward remain unclear. Immune response might play an important role in this context. Nicotine may induce both central and systemic inflammatory responses as well as changes in the regulation of brain-derived neurotrophic factor (BDNF). The conditioned place preference (CPP) is a method used for the evaluation of nicotine-induced reward, reproducing nicotine-seeking behavior in humans. So far, there are no studies investigating the relationship between neuroinflammation and nicotine-induced CPP. This study aimed to evaluate the levels of inflammatory mediators and neurotrophic factors in key areas of the central nervous system (CNS) of mice subject to nicotine-induced CPP. CPP was induced with an intraperitoneal administration of 0.5 mg/kg of nicotine in male Swiss mice, using an unbiased protocol. Control group received vehicle by the same route. The levels of cytokines, chemokines, and neurotrophic factors were measured using Enzyme-Linked Immunosorbent Assay (ELISA) in the brain after CPP test. As expected, nicotine induced place preference behavior. In parallel, we observed increased peripheral levels of IL-6 and IL-10 alongside increased hippocampal levels of NGF but decreased GDNF in mice treated with nicotine compared to controls. In the striatum, nicotine promoted decrease of IL-1ß, IL-10 and GDNF levels, while the levels of all the mediators were similar between groups in the pre-frontal cortex. Our results provide evidence on the role of cytokines and neurotrophic factors in nicotine-induced CPP in mice.


Subject(s)
Conditioning, Psychological/drug effects , Neuroinflammatory Diseases/psychology , Nicotine/administration & dosage , Reward , Tobacco Use Disorder/psychology , Animals , Brain-Derived Neurotrophic Factor/analysis , Brain-Derived Neurotrophic Factor/metabolism , Corpus Striatum/immunology , Corpus Striatum/pathology , Disease Models, Animal , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Hippocampus/immunology , Hippocampus/pathology , Humans , Injections, Intraperitoneal , Interleukin-10/analysis , Interleukin-10/metabolism , Interleukin-1beta/analysis , Interleukin-1beta/metabolism , Male , Mice , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/pathology , Nicotine/adverse effects , Prefrontal Cortex/immunology , Prefrontal Cortex/pathology , Tobacco Use Disorder/immunology , Tobacco Use Disorder/pathology
12.
Int Immunopharmacol ; 100: 108123, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34560511

ABSTRACT

The slightly available data about the pathogenesis process of mild repetitive traumatic brain injury (mRTBI) indicates to the necessity of further exploration of mRTBI consequences. Several cellular changes are believed to contribute to the cognitive disabilities, and neurodegenerative changes observed later in persons subjected to mRTBI. We investigated glial fibrillary acidic protein (GFAP), the important severity related biomarker, where it showed further increase after multiple trauma compared to single one. To authenticate our aim, Morin (10 mg/kg loading dose, then twice daily 5 mg/kg for 7 days), MK-801 (1 mg/kg; i.p) and their combination were used. The results obtained has shown that all the chosen regimens opposed the upregulated dementia markers (Aß1-40,p(Thr231)Tau) and inflammatory protein contents/expression of p(Ser53s6)NF-κBp65, TNF-α, IL-6,and IL-1ß and the elevated GFAP in immune stained cortex sections. Additionally, they exerted anti-apoptotic activity by decreasing caspase-3 activity and increasing Bcl-2 contents. Saving brain tissues was evident after these therapeutic agents via upregulating the non-canonical Wnt-1/PKC-α cue and IL-10/p(Tyr(1007/1008))JAK-2/p(Tyr705)STAT-3 signaling pathway to confirm enhancement of survival pathways on the molecular level. Such results were imitated by correcting the injury dependent deviated behavior, where Morin alone or in combination enhanced behavior outcome. On one side, our study refers to the implication of two survival signaling pathways; viz.,the non-canonical Wnt-1/PKC-α and p(Tyr(1007/1008))JAK-2/p(Tyr705)STAT-3 in single and repetitive mRTBI along with distorted dementia markers, inflammation and apoptotic process that finally disrupted behavior. On the other side, intervention through affecting all these targets by Morin alone or with MK-801 affords a promising neuroprotective effect.


Subject(s)
Brain Injuries, Traumatic/drug therapy , Flavonoids/pharmacology , Janus Kinase 2/metabolism , Oligopeptides/metabolism , Phosphopeptides/metabolism , Protein Kinase C-alpha/metabolism , STAT3 Transcription Factor/metabolism , Amyloid beta-Peptides/metabolism , Animals , Apoptosis/drug effects , Brain Injuries, Traumatic/metabolism , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Conditioning, Psychological/drug effects , Dizocilpine Maleate/pharmacology , Fear/drug effects , Inflammation/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , Wnt1 Protein , tau Proteins/metabolism
13.
Int J Mol Sci ; 22(17)2021 Aug 31.
Article in English | MEDLINE | ID: mdl-34502393

ABSTRACT

Beta-phenylethylamine (ß-PEA) is a well-known and widespread endogenous neuroactive trace amine found throughout the central nervous system in humans. In this study, we demonstrated the effects of ß-PEA on psychomotor, rewarding, and reinforcing behaviors and affective state using the open-field test, conditioned place preference (CPP), self-administration, and ultrasonic vocalizations (USVs) paradigms. We also investigated the role of the dopamine (DA) D1 receptor in the behavioral effects of ß-PEA in rodents. Using enzyme-linked immunosorbent assay (ELISA) and Western immunoblotting, we also determined the DA concentration and the DA-related protein levels in the dorsal striatum of mice administered with acute ß-PEA. The results showed that acute ß-PEA increased stereotypic behaviors such as circling and head-twitching responses in mice. In the CPP experiment, ß-PEA increased place preference in mice. In the self-administration test, ß-PEA significantly enhanced self-administration during a 2 h session under fixed ratio (FR) schedules (FR1 and FR3) and produced a higher breakpoint during a 6 h session under progressive ratio schedules of reinforcement in rats. In addition, acute ß-PEA increased 50-kHz USV calls in rats. Furthermore, acute ß-PEA administration increased DA concentration and p-DAT and TH expression in the dorsal striatum of mice. Finally, pretreatment with SCH23390, a DA D1 receptor antagonist, attenuated ß-PEA-induced circling behavior and ß-PEA-taking behavior in rodents. Taken together, these findings suggest that ß-PEA has rewarding and reinforcing effects and psychoactive properties, which induce psychomotor behaviors and a positive affective state by activating the DA D1 receptor in the dorsal striatum.


Subject(s)
Phenethylamines/pharmacology , Receptors, Dopamine D1/metabolism , Affect/drug effects , Affect/physiology , Animals , Behavior, Animal/drug effects , Behavior, Animal/physiology , Conditioning, Operant/drug effects , Conditioning, Operant/physiology , Conditioning, Psychological/drug effects , Dopamine/metabolism , Male , Mice , Mice, Inbred C57BL , Phenethylamines/metabolism , Psychomotor Performance/drug effects , Receptors, Dopamine D1/drug effects , Reinforcement, Psychology , Reward , Self Administration
14.
Drug Des Devel Ther ; 15: 3685-3696, 2021.
Article in English | MEDLINE | ID: mdl-34465980

ABSTRACT

INTRODUCTION: Oleanolic acid (OA) has been shown to be useful for the treatment of mental disorders. METHODS: In this study, we investigated the effects of OA in animal models of spontaneous withdrawal and naloxone-precipitated withdrawal and evaluated the effects of OA on the acquisition, extinction, and reinstatement of morphine-induced conditioned place preference (CPP). RESULTS: OA significantly improved symptoms of withdrawal, and significantly reduced the acquisition and reinstatement of morphine-induced conditioned place preference. Moreover, OA significantly reduced the serum content of 5-hydroxy tryptamine (5-HT) and dopamine (DA) in a dose-dependent manner, and reduced norepinephrine (NE) and 5-HT content in the frontal cortex (PFC), while significantly increasing endorphin content in rats. OA also significantly reduced serum DA content in mice. CONCLUSION: These results indicate that OA can improve the withdrawal symptoms of rats and mice by regulating the DA system and suggest that OA may be useful in treatment of morphine addiction.


Subject(s)
Morphine Dependence/drug therapy , Morphine/pharmacology , Oleanolic Acid/pharmacology , Substance Withdrawal Syndrome/drug therapy , Animals , Conditioning, Psychological/drug effects , Dopamine/metabolism , Dose-Response Relationship, Drug , Male , Mice , Morphine/administration & dosage , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Oleanolic Acid/administration & dosage , Rats , Rats, Sprague-Dawley , Serotonin/metabolism
15.
Int J Mol Sci ; 22(15)2021 Jul 26.
Article in English | MEDLINE | ID: mdl-34360704

ABSTRACT

The activation of the endocannabinoid system controls the release of many neurotransmitters involved in the brain reward pathways, including glutamate. Both endocannabinoid and glutamate systems are crucial for alcohol relapse. In the present study, we hypothesize that N-methyl-D-aspartate (NMDA) glutamate receptors regulate the ability of a priming dose of WIN 55,212-2 to cross-reinstate ethanol-induced conditioned place preference (CPP). To test this hypothesis, ethanol-induced (1.0 g/kg, 10% w/v, i.p.) CPP (unbiased method) was established using male adult Wistar rats. After CPP extinction, one group of animals received WIN 55,212-2 (1.0 and 2.0 mg/kg, i.p.), the cannabinoid receptor 1 (CB1) agonist, or ethanol, and the other group received memantine (3.0 or 10 mg/kg, i.p.), the NMDA antagonist and WIN 55,212-2 on the reinstatement day. Our results showed that a priming injection of WIN 55,212-2 (2.0 mg/kg, i.p.) reinstated (cross-reinstated) ethanol-induced CPP with similar efficacy to ethanol. Memantine (3.0 or 10 mg/kg, i.p.) pretreatment blocked this WIN 55,212-2 effect. Furthermore, our experiments indicated that ethanol withdrawal (7 days withdrawal after 10 days ethanol administration) down-regulated the CNR1 (encoding CB1), GRIN1/2A (encoding GluN1 and GluN2A subunit of the NMDA receptor) genes expression in the prefrontal cortex and dorsal striatum, but up-regulated these in the hippocampus, confirming the involvement of these receptors in ethanol rewarding effects. Thus, our results show that the endocannabinoid system is involved in the motivational properties of ethanol, and glutamate may control cannabinoid induced relapse into ethanol seeking behavior.


Subject(s)
Benzoxazines/pharmacology , Conditioning, Psychological/drug effects , Ethanol/pharmacology , Memantine/pharmacology , Morpholines/pharmacology , Motivation/drug effects , Naphthalenes/pharmacology , Animals , Male , Rats , Rats, Wistar
16.
Biosci Rep ; 41(8)2021 08 27.
Article in English | MEDLINE | ID: mdl-34355745

ABSTRACT

Traditional Chinese medicine detoxification prescription Chaihu-jia-Longgu-Muli decoction (CLMD) relieves depressive symptoms in patients withdrawing from methamphetamine. In the present study, we assessed the effects of CLMD on methamphetamine withdrawal in rats. A methamphetamine-intoxicated rat model was established. Rats were randomly divided into the control, model, high-dosage, medium-dosage, and low-dosage groups, receiving high, medium, and low doses of CLMD, respectively. Weekly body weight measurements revealed that rats treated with methamphetamine had the lowest body weight. The conditioned place preference (CPP) experiment revealed that methamphetamine-intoxicated rats stayed significantly longer in the drug-paired chamber than the control rats. However, after administering high-dosage CLMD, the amount of time the rats spent in the drug-paired chamber was significantly less than that of the model rats. Our open-field test revealed that the model group had lower crossing and rearing scores than the control group. Additionally, rats that received CLMD treatment exhibited higher crossing and rearing scores than the model rats. Striatal dopamine (DA), 5-hydroxytryptamine (5-HT), and endorphins (ß-EP) and serum interleukin (IL)-1α and IL-2 concentrations were estimated. Rats in the model group had lower striatal DA, 5-HT, and ß-EP and higher serum IL-1α and IL-2 concentrations than those in the control group. High-dosage CLMD administration significantly changed the concentrations of these molecules, such that they approached normal concentrations. In general, CLMD could prevent the development of methamphetamine-induced withdrawal symptoms in rats by increasing the DA, 5-HT, and ß-EP and lowering the IL-1α and IL-2 concentrations.


Subject(s)
Behavior, Animal/drug effects , Central Nervous System Stimulants , Conditioning, Psychological/drug effects , Corpus Striatum/drug effects , Drugs, Chinese Herbal/pharmacology , Methamphetamine , Substance Withdrawal Syndrome/drug therapy , Animals , Corpus Striatum/metabolism , Disease Models, Animal , Dopamine/metabolism , Interleukin-1alpha/blood , Interleukin-2/blood , Male , Open Field Test/drug effects , Rats, Sprague-Dawley , Serotonin/metabolism , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/psychology , beta-Endorphin/metabolism
17.
Int J Neuropsychopharmacol ; 24(11): 907-919, 2021 11 12.
Article in English | MEDLINE | ID: mdl-34363482

ABSTRACT

BACKGROUND: Enhancement of N-methyl-D-aspartate (NMDA) receptor function using glycine-site agonist D-cycloserine is known to facilitate fear extinction, providing a means to augment cognitive behavioral therapy in anxiety disorders. A novel class of glycine-site agonists has recently been identified, and we have found that the prototype, AICP, is more effective than D-cycloserine in modulating neuronal function. METHODS: Using novel glycine-site agonist AICP, local infusion studies, and genetic models, we elucidated the role of GluN2C-containing receptors in fear extinction. RESULTS: We tested the effect of intracerebroventricular injection of AICP on fear extinction and found a robust facilitation of fear extinction. This effect was dependent on GluN2C subunit, consistent with superagonist action of AICP at GluN2C-containing receptors. Local infusion studies in wild-type and GluN2C knockout mice suggested that AICP produces its effect via GluN2C-containing receptors in the basolateral amygdala (BLA). Furthermore, consistent with astrocytic expression of GluN2C subunit in the amygdala, we found that AICP did not facilitate fear extinction in mice with conditional deletion of obligatory GluN1 subunit from astrocytes. Importantly, chemogenetic activation of astrocytes in the basolateral amygdala facilitated fear extinction. Acutely, AICP was found to facilitate excitatory neurotransmission in the BLA via presynaptic GluN2C-dependent mechanism. Immunohistochemical studies suggest that AICP-mediated facilitation of fear extinction involves synaptic insertion of α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptor GluA1 subunit. CONCLUSION: These results identify a unique role of astrocytic NMDA receptors composed of GluN2C subunit in extinction of conditioned fear memory and demonstrate that further development of recently identified superagonists of GluN2C-containing receptors may have utility for anxiety disorders.


Subject(s)
Amygdala/drug effects , Astrocytes/metabolism , Extinction, Psychological/drug effects , Fear/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Basolateral Nuclear Complex/metabolism , Conditioning, Psychological/drug effects , Cycloserine/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Mice , Receptors, AMPA/metabolism , Synaptic Transmission/drug effects , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/metabolism
18.
Neurochem Int ; 150: 105157, 2021 11.
Article in English | MEDLINE | ID: mdl-34390773

ABSTRACT

Besides their clinical application, chronic misuse of opioids has often been associated to drug addiction due to their addictive properties, underlying neuroadaptations of AMPA glutamate-receptor-dependent synaptic plasticity. Topiramate (TPM), an AMPAR antagonist, has been used to treat psychostimulants addiction, despite its harmful effects on memory. This study aimed to evaluate the effects of a novel topiramate nanosystem on molecular changes related to morphine reinstatement. Rats were previously exposed to morphine in conditioned place preference (CPP) paradigm and treated with topiramate-chitosan nanoparticles (TPM-CS-NP) or non-encapsulated topiramate in solution (S-TPM) during CPP extinction; following memory performance evaluation, they were re-exposed to morphine reinstatement. While morphine-CPP extinction was comparable among all experimental groups, TPM-CS-NP treatment prevented morphine reinstatement, preserving memory performance, which was impaired by both morphine-conditioning and S-TPM treatment. In the NAc, morphine increased D1R, D2R, D3R, DAT, GluA1 and MOR immunoreactivity. It also increased D1R, DAT, GluA1 and MOR in the dorsal hippocampus. TPM-CS-NP treatment decreased D1R, D3R and GluA1 and increased DAT in the NAc, decreasing GluA1 and increasing D2 and DAT in the dorsal hippocampus. Taken together, we may infer that TPM-CS-NP treatment was able to prevent the morphine reinstatement without memory impairment. Therefore, TPM-CS-NP may be considered an innovative therapeutic tool due to its property to prevent opioid reinstatement because it acts modifying both dopaminergic and glutamatergic neurotransmission, which are commonly related to morphine addiction.


Subject(s)
Chitosan/administration & dosage , Dopamine/metabolism , Glutamic Acid/metabolism , Morphine Dependence/metabolism , Nanoparticles/administration & dosage , Topiramate/administration & dosage , Analgesics, Opioid/pharmacology , Animals , Conditioning, Psychological/drug effects , Conditioning, Psychological/physiology , Drug Therapy, Combination , Extinction, Psychological/drug effects , Extinction, Psychological/physiology , Male , Memory/drug effects , Memory/physiology , Morphine/pharmacology , Morphine Dependence/prevention & control , Rats , Rats, Wistar , Receptors, AMPA/metabolism , Receptors, Dopamine/metabolism
19.
Bull Exp Biol Med ; 171(3): 293-296, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34297286

ABSTRACT

The brain mechanisms underlying conditioned aversion learning in birds were studied using experimental model in young chicks. The learning consisted of a conditioning stimulus presentation followed by a delayed sickness-inducing treatment reinforcement. Intraventricular administration of an NMDA receptor antagonist MK-801, a protein synthesis inhibitor anisomycin, or an inhibitor of glycoprotein fucosylation 2-deoxygalactose just before presentation of the conditioning stimulus prevented aversion learning. Injections of the same chemicals before reinforcement did not affect learning. The obtained results show that the investigated mechanisms underlying aversion learning were critical at the early stage of memory formation. Later processes of association of the conditioning stimulus with the reinforcement appear to be independent of the NMDA receptors and protein synthesis/glycosylation, or alternatively to be located in other brain areas.


Subject(s)
Avoidance Learning/drug effects , Conditioning, Psychological/drug effects , Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Memory, Long-Term/drug effects , Receptors, N-Methyl-D-Aspartate/genetics , Animals , Animals, Newborn , Anisomycin/pharmacology , Avoidance Learning/physiology , Brain/drug effects , Brain/metabolism , Chickens , Conditioning, Psychological/physiology , Fucose/pharmacology , Gene Expression , Glycosylation/drug effects , Injections, Intraventricular , Lithium Chloride/pharmacology , Memory, Long-Term/physiology , Protein Biosynthesis/drug effects , Protein Synthesis Inhibitors/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Reinforcement, Psychology
20.
Pharmacol Biochem Behav ; 208: 173229, 2021 09.
Article in English | MEDLINE | ID: mdl-34246729

ABSTRACT

The present studies investigated the effects of withdrawal from a single binge-like dose of ethanol (hangover) on fear conditioning in male and female Sprague Dawley rats. In Experiment 1, males and females were given 0 or 3.5 g/kg ethanol intraperitoneally (i.p.) and then conditioned to contextual fear 24 h post injection. Withdrawal from acute ethanol enhanced expression of the conditioned freezing response in males, but not in females. Experiment 2 demonstrated that in males, withdrawal from acute ethanol administered 24 h prior to conditioning enhanced contextual fear conditioning, but not auditory-cued fear conditioning. In Experiment 3, male and female rats were given 3.5 g/kg ethanol, and blood ethanol concentrations (BECs) were assessed at various time points for determination of ethanol clearance. Female rats cleared ethanol at a higher rate than males, with 10 h required for females and 14 for males to eliminate ethanol from their systems. Because females cleared ethanol faster than males, in Experiment 4, females were conditioned 18 h after ethanol administration to keep the interval between ethanol clearance and fear conditioning similar to that of males. Withdrawal from acute ethanol given 18 h prior to conditioning did not affect both contextual and auditory-cued fear conditioning in females. In summary, these results highlight sex differences in the impact of withdrawal from acute ethanol (hangover) on fear learning; suggesting that males are more sensitive to hangover-associated enhancement of negative affect than females.


Subject(s)
Conditioning, Psychological/drug effects , Ethanol/pharmacology , Fear/drug effects , Learning/drug effects , Substance Withdrawal Syndrome/psychology , Alcoholic Intoxication/psychology , Animals , Behavior, Animal/drug effects , Central Nervous System Depressants/pharmacology , Cues , Female , Locomotion/drug effects , Male , Rats , Rats, Sprague-Dawley , Sex Characteristics , Substance Withdrawal Syndrome/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...