Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
Article in English | MEDLINE | ID: mdl-38671147

ABSTRACT

Stress affects brain serotonin (5HT) and dopamine (DA) function, and the effectiveness of 5HT and DA to regulate stress and emotional responses. However, our understanding of the long-term impact of early life adversity (ELA) on primate brain monoaminergic systems during adolescence is scarce and inconsistent. Filling this gap in the literature is critical, given that the emergence of psychopathology during adolescence has been related to deficits in these systems. Here, we use a translational nonhuman primate (NHP) model of ELA (infant maltreatment by the mother) to examine the long-term impact of ELA on adolescent 5HT1A, 5HT2A and D2 receptor systems. These receptor systems were chosen based on their involvement in stress/emotional control, as well as reward and reinforcement. Rates of maternal abuse, rejection, and infant's vocalizations were obtained during the first three postnatal months, and hair cortisol concentrations obtained at 6 months postnatal were examined as early predictors of binding potential (BP) values obtained during adolescence using positron emission tomography (PET) imaging. Maltreated animals demonstrated significantly lower 5HT1A receptor BP in prefrontal cortical areas as well as the amygdala and hippocampus, and lower 5HT2A receptor BP in striatal and prefrontal cortical areas. Maltreated animals also demonstrated significantly lower D2 BP in the amygdala. None of the behavioral and neuroendocrine measurements obtained early in life predicted any changes in BP data. Our findings suggest that early caregiving experiences regulate the development of brain 5HT and DA systems in primates, resulting in long-term effects evident during adolescence.

2.
J Am Assoc Lab Anim Sci ; 61(2): 165-172, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35012705

ABSTRACT

Rhesus monkeys are naturally social animals, and behavioral management strategies have focused on promoting pairhousing in laboratory settings as an alternative to individual or group housing. In humans, co-sleeping can have a major impact on bed partners' sleep, raising the possibility that pair-housing also may influence sleep parameters in monkeys. In the present study, we investigated if pair-housing would impact home-cage partner's sleep in female rhesus monkeys, and if nighttime separation using socialization panels would alter this pattern. Sleep parameters of 10 experimentally naïve adult female rhesus monkeys (5 pairs) were evaluated for 7 consecutive days using actigraphy monitors attached to primate collars. Paired animals then were separated by socialization panels during the night, and sleep-associated measures were evaluated for 7 consecutive days. The data showed that sleep efficiency was significantly lower when monkeys were pairhoused as compared with when they were separated. On the nights when subjects were pair-housed, a positive correlation was detected for sleep measures (both sleep latency and efficiency) of both members of a pair (R2's = 0.16-0.5), suggesting that pair-housing influences sleep quality. On nights when subjects were separated, no correlations were observed for sleep measures between members of the pairs (R2's = 0.004-0.01), suggesting that when separated, the home-cage partner's sleep no longer influenced the partner's sleep. Our results indicate that pair-housing has a strong impact on the home-cage partner's sleep, and that this pattern can be prevented by nighttime separation using socialization panels. Studies evaluating sleep in pair-housed monkeys should consider the effects that the partner's sleep may have on the subject's sleep. Sleep is a biologic phenomenon and experimental outcome that affects physical and behavioral health and altered sleep due to pair-housing may affect a range of research outcomes.


Subject(s)
Actigraphy , Sleep , Actigraphy/methods , Animals , Female , Housing, Animal , Humans , Macaca mulatta
3.
Psychopharmacology (Berl) ; 237(12): 3583-3589, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32821985

ABSTRACT

RATIONALE: It is critical to identify potential risk factors, such as a history of early life stress (ELS), that may confer specific vulnerabilities to increased drug intake. OBJECTIVE: In this study, we examined whether male and female rhesus monkeys with a history of ELS (infant maltreatment; MALT) demonstrated significantly greater cocaine intake compared with controls. METHODS: Monkeys were trained to self-administer cocaine during 4-h sessions at a peak dose (0.003-0.1 mg/kg/infusion; extended access, "EA peak") and a dose of 0.1 mg/kg/infusion (EA 0.1) of cocaine. These data were compared with data obtained previously in monkeys trained during 1-h limited access (LA) sessions at the same peak dose of cocaine used here (Wakeford et al. Psychopharmacology, 236:2785-2796, 2019). RESULTS: Monkeys significantly increased total number of infusions earned in EA compared with LA, but total session response rates significantly decreased in EA compared with LA. There was no evidence of escalation in drug intake when we compared response rates to obtain the first 20 cocaine infusions between LA and EA peak conditions. Moreover, there was no evidence of escalation in drug intake during an additional 7 weeks of self-administration at 0.1 mg/kg/injection. CONCLUSIONS: The current study expands on previous reports demonstrating that rhesus macaques did not escalate cocaine intake under the experimental conditions employed and extended these findings by using a unique population of nonhuman primates with a history of infant MALT to test the hypothesis that ELS is a risk factor for escalation of cocaine intake in nonhuman primates. There was no clear evidence of escalation in cocaine intake as a consequence of ELS.


Subject(s)
Aging , Cocaine-Related Disorders , Cocaine , Stress, Psychological , Animals , Female , Male , Aging/psychology , Cocaine/administration & dosage , Cocaine-Related Disorders/psychology , Dose-Response Relationship, Drug , Macaca mulatta , Self Administration , Stress, Psychological/psychology
4.
Psychopharmacology (Berl) ; 236(9): 2785-2796, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31115612

ABSTRACT

RATIONALE: Early life stress (ELS), including childhood maltreatment, is a predictive factor for the emergence of cocaine use disorders (CUDs) in adolescence. OBJECTIVE: Accordingly, we examined whether post-pubertal male and female rhesus macaques that experienced infant maltreatment (maltreated, n = 7) showed greater vulnerability to cocaine self-administration in comparison with controls (controls, n = 7). METHODS: Infant emotional reactivity was measured to assess differences in behavioral distress between maltreated and control animals as a result of early life caregiving. Animals were then surgically implanted with indwelling intravenous catheters and trained to self-administer cocaine (0.001-0.3 mg/kg/infusion) under fixed-ratio schedules of reinforcement. Days to acquisition, and sensitivity to (measured by the EDMax dose of cocaine) and magnitude (measured by response rates) of the reinforcing effects of cocaine were examined in both groups. RESULTS: Maltreated animals demonstrated significantly higher rates of distress (e.g., screams) in comparison with control animals. When given access to cocaine, control males required significantly more days to progress through terminal performance criteria compared with females and acquired cocaine self-administration slower than the other three experimental groups. The dose that resulted in peak response rates did not differ between groups or sex. Under 5-week, limited-access conditions, males from both groups had significantly higher rates of responding compared with females. CONCLUSIONS: In control monkeys, these data support sex differences in cocaine self-administration, with females being more sensitive than males. These findings also suggest that ELS may confer enhanced sensitivity to the reinforcing effects of cocaine, especially in males.


Subject(s)
Cocaine/administration & dosage , Dopamine Uptake Inhibitors/administration & dosage , Reinforcement Schedule , Sex Characteristics , Sexual Maturation/drug effects , Stress, Psychological/psychology , Animals , Cocaine-Related Disorders/psychology , Dose-Response Relationship, Drug , Female , Macaca mulatta , Male , Random Allocation , Reaction Time/drug effects , Reaction Time/physiology , Reinforcement, Psychology , Self Administration , Sexual Maturation/physiology
5.
Neuropharmacology ; 151: 13-20, 2019 06.
Article in English | MEDLINE | ID: mdl-30922893

ABSTRACT

The recreational drug 3,4-methylenedioxymethamphetamine (MDMA) has well documented prosocial effects and is currently under clinical investigation as a treatment for patients with PTSD, autism, and other conditions. Early clinical trials have found that MDMA-assisted therapy may have robust long-lasting therapeutic effects, yet the mechanism by which acute treatments produce these long-term effects is unclear. Sensitization to certain behavioral drug effects is a common rodent model used to assess long-lasting neurobiological adaptations induced by acute drug treatments. Nine independent experiments were undertaken to investigate if and how mice sensitize to the prosocial effects of MDMA. When treated with 7.8 mg/kg MDMA and paired every other day for a week, MDMA-induced social interaction increased precipitously across treatment sessions. This previously unreported phenomenon was investigated and found to be heavily influenced by a social context and 5-HT2AR activation. Social sensitization did not appear to develop if mice were administered MDMA in isolation, and pretreatment with MDL100907, a selective 5-HT2AR antagonist, inhibited the development of social sensitization. However, when MDL100907 was administered to mice that had already been sensitized, it did not attenuate social interaction, suggesting that 5-HT2AR activity may be necessary for the development of social sensitization but not the expression of MDMA-induced social behavior. Additional investigation is warranted to further explore the phenomenon of social sensitization and to determine the underlying neurobiological mechanisms.


Subject(s)
Behavior, Animal/drug effects , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Serotonin Agents/pharmacology , Social Behavior , Animals , Fluorobenzenes/pharmacology , Male , Mice , Motor Activity/drug effects , Piperidines/pharmacology , Serotonin Antagonists/pharmacology
6.
Psychopharmacology (Berl) ; 236(7): 2105-2118, 2019 Jul.
Article in English | MEDLINE | ID: mdl-30879118

ABSTRACT

RATIONALE: Substance use disorders are characterized by a loss of executive control over reward-based decision-making, and disruption of fronto-striatal connectivity has been implicated in this process. Sub-anesthetic ketamine has recently been shown to bolster fronto-striatal connectivity in drug-naïve subjects. OBJECTIVES: The influence of ketamine treatment was examined on the disruptive effects of cocaine on functional connectivity (FC) and on cocaine-seeking behavior in female rhesus monkeys. METHODS: Three female rhesus were trained for unanesthetized MRI scanning. Each received three drug-naïve/abstinent pharmacological MRI scans with acute injections of saline, cocaine (0.3 mg/kg i.v.), and cocaine (0.3 mg/kg i.v.) 48-h after a ketamine treatment (low dose = 0.345 mg/kg bolus + 0.256 mg/kg/h for 1 h; i.v.), and a fourth scan with saline injection following 2 months of daily cocaine self-administration. A separate cohort of five rhesus (4 female), all with extensive histories of cocaine exposure, underwent reinstatement testing 48 h after ketamine (or vehicle) treatment. Two sub-anesthetic doses were tested: low dose and high dose = 0.69 mg/kg + 0.512 mg/kg/h for 1 h. RESULTS: Ketamine treatment attenuated the effects of cocaine on both global and fronto-striatal FC in drug-naïve/abstinent subjects. Two months of daily cocaine self-administration led to prolonged disruption of both global and fronto-striatal FC. Cocaine-seeking behavior during reinstatement was reduced following ketamine treatment at the low dose, but not high dose. CONCLUSION: These findings illustrate the disruptive effects of cocaine on functional connectivity and provide evidence for the potential efficacy of ketamine as a treatment for stimulant use disorder.


Subject(s)
Cocaine-Related Disorders/drug therapy , Cocaine/administration & dosage , Corpus Striatum/drug effects , Frontal Lobe/drug effects , Ketamine/therapeutic use , Nerve Net/drug effects , Anesthetics, Dissociative/pharmacology , Anesthetics, Dissociative/therapeutic use , Animals , Cocaine-Related Disorders/diagnostic imaging , Corpus Striatum/diagnostic imaging , Extinction, Psychological/drug effects , Female , Frontal Lobe/diagnostic imaging , Ketamine/pharmacology , Macaca mulatta , Magnetic Resonance Imaging/methods , Nerve Net/diagnostic imaging , Reward , Self Administration , Treatment Outcome
7.
Neuropsychopharmacology ; 44(3): 478-486, 2019 02.
Article in English | MEDLINE | ID: mdl-30188516

ABSTRACT

Perseverative behavior has been highly implicated in addiction. Activation of serotonin 2C receptors (5-HT2CRs) attenuates cocaine and high caloric food intake, but whether a 5-HT2CR agonist can reduce high caloric diet (HCD) or methamphetamine (METH) intake and response perseveration remains unknown. Clarifying the role of 5-HT2CRs in these behaviors will improve knowledge of neurochemical processes that regulate flexible decision-making and whether improvements in decision-making are accompanied by decreases in HCD or METH intake. This study evaluated the effects of long-term HCD and METH intake on reversal learning in female rhesus monkeys. The effects of the 5-HT2CR agonist WAY163909 on reversal learning before and after extended HCD or METH intake, and on food intake, was also tested. Moreover, we examined whether the 5-HT2CR is necessary for the effects of WAY163909. WAY163909 was given prior to reversal learning at baseline and after extended HCD or METH intake, and prior to measures of food intake. Extended intake of METH or the HCD increased perseverative errors during reversal. WAY163909 increased correct responses and decreased perseverative errors, both before and after extended HCD or METH intake. Similarly, WAY163909 decreased consumption of a HCD, but not a low caloric diet. The effects of WAY163909 on all these measures were blocked by co-administration with a 5-HT2CR antagonist. These data indicate that long-term HCD or METH intake disrupts flexible decision-making. Further, the results suggest that reductions in food intake produced by WAY163909 are associated with parallel improvements in decision-making strategies, underscoring the role of the 5-HT2CR for these behavioral effects.


Subject(s)
Behavior, Animal/drug effects , Central Nervous System Stimulants/pharmacology , Decision Making/drug effects , Dietary Fats/pharmacology , Feeding Behavior/drug effects , Methamphetamine/pharmacology , Receptor, Serotonin, 5-HT2C/physiology , Reversal Learning/drug effects , Serotonin 5-HT2 Receptor Agonists/pharmacology , Serotonin 5-HT2 Receptor Antagonists/pharmacology , Animals , Azepines/pharmacology , Female , Indoles/pharmacology , Macaca mulatta
8.
Drug Alcohol Depend ; 194: 252-256, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30469095

ABSTRACT

BACKGROUND: Serotonin 5-HT2A receptor antagonists and 5-HT2C receptor agonists have been proposed as important candidates for the development of pharmacotherapies for psychostimulant abuse, with evidence suggesting that those receptors may act together to control behavior. However, the role of 5-HT2A receptors on the reinforcing effects of psychostimulant drugs has not been fully elucidated. METHODS: In the present study, we investigated the effects of the selective 5HT2A receptor antagonist M100907 alone or in combination with the selective 5HT2C agonist WAY 163909 on intravenous methamphetamine self-administration in rhesus macaques (N = 3). Methamphetamine self-administration (0.01-0.03 mg/kg/inf) was evaluated under a fixed-ratio 20-schedule of reinforcement, and acute pretreatments were conducted 1 h (M100907) or 45 min (WAY 163,909) prior to the beginning of self-administration sessions at the EDMax dose of methamphetamine once stability criteria were met. RESULTS: Pretreatment with M100907 (0.03-0.3 mg/kg, i.m.) dose-dependently attenuated methamphetamine self-administration, with the highest dose significantly decreasing response rates compared to vehicle. Combined administration of ineffective doses of M100907 and WAY 163,909 had no effects on methamphetamine self-administration. CONCLUSIONS: Our study indicates that acute selective 5-HT2A receptor blockade decreases peak methamphetamine intake in nonhuman primates. Combination approaches with sub-threshold doses of 5-HT2A receptor antagonists and 5-HT2C receptor agonists, on the other hand, do not seem to be effective in decreasing methamphetamine reinforcement. Further studies are needed in order to investigate the effects of chronic treatments with M100 on complete METH SA dose-response curves.


Subject(s)
Azepines/administration & dosage , Central Nervous System Stimulants/administration & dosage , Fluorobenzenes/administration & dosage , Indoles/administration & dosage , Methamphetamine/administration & dosage , Piperidines/administration & dosage , Serotonin 5-HT2 Receptor Agonists/administration & dosage , Serotonin 5-HT2 Receptor Antagonists/administration & dosage , Animals , Behavior, Addictive/drug therapy , Behavior, Addictive/psychology , Dose-Response Relationship, Drug , Drug Therapy, Combination , Female , Macaca mulatta , Male , Reinforcement, Psychology , Self Administration , Serotonin Antagonists/administration & dosage , Treatment Outcome
9.
Neurobiol Stress ; 9: 188-198, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30450384

ABSTRACT

Adolescence represents a developmental stage in which initiation of drug use typically occurs and is marked by dynamic neurobiological changes. These changes present a sensitive window during which perturbations to normative development lead to alterations in brain circuits critical for stress and emotional regulation as well as reward processing, potentially resulting in an increased susceptibility to psychopathologies. The occurrence of early life stress (ELS) is related to a greater risk for the development of substance use disorders (SUD) during adolescence. Studies using nonhuman primates (NHP) are ideally suited to examine how ELS may alter the development of neurobiological systems modulating the reinforcing effects of drugs, given their remarkable neurobiological, behavioral, and developmental homologies to humans. This review examines NHP models of ELS that have been used to characterize its effects on sensitivity to drug reinforcement, and proposes future directions using NHP models of ELS and drug abuse in an effort to develop more targeted intervention and prevention strategies for at risk clinical populations.

10.
Comp Med ; 68(6): 496-502, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30486918

ABSTRACT

An adult rhesus macaque developed seizures after the induction of ischemic stroke. Initially, on the day of surgery, a focal ischemic lesion was present exclusively in the right caudate nucleus. By 48 h after stroke induction, the lesion had extended into the putamen, when a seizure was observed. Our report highlights the temporal changes in infarction of unilateral basal ganglia after acute stroke and the accompanying clinical symptoms. This unusual case may provide additional information regarding the involvement of the basal ganglia in seizures, given that prior case reports and studies usually have not described the temporal and spatial evolution of the lesion before clinical symptoms emerge.


Subject(s)
Basal Ganglia/diagnostic imaging , Macaca mulatta , Monkey Diseases/diagnostic imaging , Seizures/veterinary , Stroke/veterinary , Animals , Magnetic Resonance Imaging , Seizures/diagnostic imaging , Stroke/diagnostic imaging
11.
Exp Clin Psychopharmacol ; 26(4): 410-420, 2018 08.
Article in English | MEDLINE | ID: mdl-29939048

ABSTRACT

3,4-Methylenedioxymethamphetamine (MDMA) affects monoaminergic pathways that play a critical role in sleep-wake cycles. Dopaminergic mechanisms are thought to mediate the sleep-disrupting effects of stimulant drugs. However, the mechanisms underlying the effects of MDMA on sleep-wake cycles and the effects of R(-) MDMA, a stereoisomer that lacks dopaminergic activity, on sleep remain unknown. The aim of the present study was to investigate the effects of racemic MDMA and R(-) MDMA on daytime activity and sleep-like parameters evaluated with actigraphy in adult rhesus macaques (Macaca mulatta, n = 6). Actiwatch monitors were attached to the monkeys' collars and actigraphy recording was conducted during baseline conditions and after the administration of acute intramuscular injections of saline (vehicle), racemic MDMA (0.3, 1.0, or 1.7 mg/kg), or R(-) MDMA (0.3, 1.0, or 1.7 mg/kg) at 9 or 16 h (3 h before "lights off"). Morning treatments had no effects on sleep-like parameters. Racemic MDMA decreased general daytime activity during the first hour after injection and increased daytime activity at 3 hr posttreatment. Although afternoon administration of racemic MDMA increased sleep latency, it improved other sleep parameters, decreasing wake time after sleep onset (WASO) and increasing sleep efficiency for subjects with low baseline sleep efficiency. Afternoon treatment with R(-) MDMA improved sleep measures, increasing sleep efficiency and decreasing sleep latency and WASO, while having no effects on daytime activity. The stimulant and sleep-disrupting effects of racemic MDMA are likely mediated by dopaminergic and noradrenergic mechanisms, while serotonergic pathways appear to be involved in the sleep-promoting effects of MDMA. (PsycINFO Database Record


Subject(s)
Actigraphy/methods , Motor Activity/drug effects , N-Methyl-3,4-methylenedioxyamphetamine/administration & dosage , N-Methyl-3,4-methylenedioxyamphetamine/chemistry , Sleep/drug effects , Wakefulness/drug effects , Animals , Central Nervous System Stimulants/pharmacology , Female , Hallucinogens/chemistry , Macaca mulatta , Male , Motor Activity/physiology , Sleep/physiology , Wakefulness/physiology
12.
Psychoneuroendocrinology ; 94: 143-151, 2018 08.
Article in English | MEDLINE | ID: mdl-29783162

ABSTRACT

Alterations in peripheral immune markers are observed in individuals with post-traumatic stress disorder (PTSD). PTSD is characterized in part by impaired extinction of fear memory for a traumatic experience. We hypothesized that fear memory extinction is regulated by immune signaling stimulated when fear memory is retrieved. The relationship between fear memory and the peripheral immune response was tested using auditory Pavlovian fear conditioning in mice. Memory for the association was quantified by the amount of conditioned freezing exhibited in response to the conditioned stimulus (CS), extinction and time-dependent changes in circulating inflammatory cytokines. Brief extinction training with 12 CS rapidly and acutely increased circulating levels of the cytokine interleukin-6 (IL-6), downstream IL-6 signaling, other IL-6 related pro-inflammatory cytokines. Transgenic manipulations or neutralizing antibodies that inhibit IL-6 activity did not affect conditioned freezing during the acquisition of fear conditioning or extinction but significantly reduced conditioned freezing 24 h after extinction training with 12 CS. Conversely, conditioned freezing after extinction training was unchanged by IL-6 inhibition when 40 CS were used during the extinction training session. In addition to effectively diminishing conditioned freezing, extinction training with 40 CS also diminished the subsequent IL-6 response to the CS. These data demonstrate that IL-6 released following fear memory retrieval contributes to the maintenance of that fear memory and that this effect is extinction dependent. These findings extend the current understanding for the role of the immune system in PTSD and suggest that IL-6 and other IL-6 related pro-inflammatory cytokines may contribute to the persistence of fear memory in PTSD where fear memory extinction is impaired.


Subject(s)
Fear/physiology , Memory/physiology , Stress, Psychological/immunology , Animals , Conditioning, Classical/physiology , Disease Models, Animal , Extinction, Psychological/physiology , Interleukin-6/analysis , Interleukin-6/metabolism , Male , Mental Recall/physiology , Mice , Mice, Inbred C57BL , Prefrontal Cortex/physiology , Stress Disorders, Post-Traumatic/immunology , Stress Disorders, Post-Traumatic/psychology , Wounds and Injuries/immunology , Wounds and Injuries/psychology
13.
Neuropharmacology ; 128: 196-206, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28993129

ABSTRACT

S,R(+/-)-3,4-methylenedioxymethamphetamine (SR-MDMA) is an amphetamine derivative with prosocial and putative therapeutic effects. Ongoing clinical trials are investigating it as a treatment for post-traumatic stress disorder (PTSD) and other conditions. However, its potential for adverse effects such as hyperthermia and neurotoxicity may limit its clinical viability. We investigated the hypothesis that one of the two enantiomers of SR-MDMA, R-MDMA, would retain the prosocial and therapeutic effects but with fewer adverse effects. Using male Swiss Webster and C57BL/6 mice, the prosocial effects of R-MDMA were measured using a social interaction test, and the therapeutic-like effects were assessed using a Pavlovian fear conditioning and extinction paradigm relevant to PTSD. Locomotor activity and body temperature were tracked after administration, and neurotoxicity was evaluated post-mortem. R-MDMA significantly increased murine social interaction and facilitated extinction of conditioned freezing. Yet, unlike racemic MDMA, it did not increase locomotor activity, produce signs of neurotoxicity, or increase body temperature. A key pharmacological difference between R-MDMA and racemic MDMA is that R-MDMA has much lower potency as a dopamine releaser. Pretreatment with a selective dopamine D1 receptor antagonist prevented SR-MDMA-induced hyperthermia, suggesting that differential dopamine signaling may explain some of the observed differences between the treatments. Together, these results indicate that the prosocial and therapeutic effects of SR-MDMA may be separable from the stimulant, thermogenic, and potential neurotoxic effects. To what extent these findings translate to humans will require further investigation, but these data suggest that R-MDMA could be a more viable therapeutic option for the treatment of PTSD and other disorders for which SR-MDMA is currently being investigated.


Subject(s)
Hallucinogens/pharmacology , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Racemases and Epimerases/pharmacology , Animals , Body Temperature/drug effects , Brain/drug effects , Brain/metabolism , Conditioning, Classical/drug effects , Dopamine/metabolism , Dopamine Plasma Membrane Transport Proteins/metabolism , Extinction, Psychological/drug effects , Fear/drug effects , Glial Fibrillary Acidic Protein/metabolism , Interpersonal Relations , Locomotion/drug effects , Male , Mice , Mice, Inbred C57BL , Neurotoxicity Syndromes/prevention & control , Serotonin/metabolism , Stereoisomerism
14.
Psychopharmacology (Berl) ; 235(2): 377-392, 2018 02.
Article in English | MEDLINE | ID: mdl-29248945

ABSTRACT

The use of (±)-3,4-methylenedioxymethamphetamine ((±)-MDMA) as an adjunct to psychotherapy in the treatment of psychiatric and behavioral disorders dates back over 50 years. Only in recent years have controlled and peer-reviewed preclinical and clinical studies lent support to (±)-MDMA's hypothesized clinical utility. However, the clinical utility of (±)-MDMA is potentially mitigated by a range of demonstrated adverse effects. One potential solution could lie in the individual S(+) and R(-) enantiomers that comprise (±)-MDMA. Individual enantiomers of racemic compounds have been employed in psychiatry to improve a drug's therapeutic index. Although no research has explored the individual effects of either S(+)-MDMA or R(-)-MDMA in humans in a controlled manner, preclinical research has examined similarities and differences between the two molecules and the racemic compound. This review addresses information related to the pharmacodynamics, neurotoxicity, physiological effects, and behavioral effects of S(+)-MDMA and R(-)-MDMA that might guide preclinical and clinical research. The current preclinical evidence suggests that R(-)-MDMA may provide an improved therapeutic index, maintaining the therapeutic effects of (±)-MDMA with a reduced side effect profile, and that future investigations should investigate the therapeutic potential of R(-)-MDMA.


Subject(s)
Hallucinogens/chemistry , Hallucinogens/therapeutic use , Mental Disorders/drug therapy , N-Methyl-3,4-methylenedioxyamphetamine/chemistry , N-Methyl-3,4-methylenedioxyamphetamine/therapeutic use , Animals , Humans , Mental Disorders/psychology , Stereoisomerism , Stress, Psychological/drug therapy , Stress, Psychological/psychology
15.
Behav Pharmacol ; 28(8): 610-622, 2017 12.
Article in English | MEDLINE | ID: mdl-29049083

ABSTRACT

Ketamine has been used as a pharmacological model for schizophrenia as subanesthetic infusions have been shown to produce temporary schizophrenia-like symptoms in healthy humans. More recently, ketamine has emerged as a potential treatment for multiple psychiatric disorders, including treatment-resistant depression and suicidal ideation. However, the mechanisms underlying both the psychotomimetic and the therapeutic effects of ketamine remain poorly understood. This review provides an overview of what is known of the neural mechanisms underlying the effects of ketamine and details what functional MRI studies have yielded at a systems level focused on brain circuitry. Multiple analytic approaches show that ketamine exerts robust and consistent effects at the whole-brain level. These effects are highly conserved across human and nonhuman primates, validating the use of nonhuman primate models for further investigations with ketamine. Regional analysis of brain functional connectivity suggests that the therapeutic potential of ketamine may be derived from a strengthening of executive control circuitry, making it an intriguing candidate for the treatment of drug abuse. There are still important questions about the mechanism of action and the therapeutic potential of ketamine that can be addressed using appropriate functional neuroimaging techniques.


Subject(s)
Brain/drug effects , Brain/physiology , Excitatory Amino Acid Antagonists/pharmacology , Ketamine/pharmacology , Magnetic Resonance Imaging , Psychotropic Drugs/pharmacology , Animals , Brain/diagnostic imaging , Humans , Magnetic Resonance Imaging/methods , Neural Pathways/diagnostic imaging , Neural Pathways/drug effects , Neural Pathways/physiology
16.
Psychopharmacology (Berl) ; 234(19): 2883-2895, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28741031

ABSTRACT

RATIONALE: 3,4-Methylenedioxymethamphetamine (MDMA) persistently improves symptoms of post-traumatic stress disorder (PTSD) when combined with psychotherapy. Studies in rodents suggest that these effects can be attributed to enhancement of fear memory extinction. Therefore, MDMA may improve the effects of exposure-based therapy for PTSD, particularly in treatment-resistant patients. However, given MDMA's broad pharmacological profile, further investigation is warranted before moving to a complex clinical population. OBJECTIVES: We aimed to inform clinical research by providing a translational model of MDMA's effect, and elucidating monoaminergic mechanisms through which MDMA enhances fear extinction. METHODS: We explored the importance of monoamine transporters targeted by MDMA to fear memory extinction, as measured by reductions in conditioned freezing and fear-potentiated startle (FPS) in mice. Mice were treated with selective inhibitors of individual monoamine transporters prior to combined MDMA treatment and fear extinction training. RESULTS: MDMA enhanced the lasting extinction of FPS. Acute and chronic treatment with a 5-HT transporter (5-HTT) inhibitor blocked MDMA's effect on fear memory extinction. Acute inhibition of dopamine (DA) and norepinephrine (NE) transporters had no effect. 5-HT release alone did not enhance extinction. Blockade of MDMA's effect by 5-HTT inhibition also downregulated 5-HT2A-mediated behavior, and 5-HT2A antagonism disrupted MDMA's effect on extinction. CONCLUSIONS: We validate enhancement of fear memory extinction by MDMA in a translational behavioral model, and reveal the importance of 5-HTT and 5-HT2A receptors to this effect. These observations support future clinical research of MDMA as an adjunct to exposure therapy, and provide important pharmacological considerations for clinical use in a population frequently treated with 5-HTT inhibitors.


Subject(s)
Extinction, Psychological/drug effects , Fear/drug effects , Memory/drug effects , N-Methyl-3,4-methylenedioxyamphetamine/toxicity , Selective Serotonin Reuptake Inhibitors/pharmacology , Serotonin Plasma Membrane Transport Proteins , Animals , Dose-Response Relationship, Drug , Extinction, Psychological/physiology , Fear/physiology , Fear/psychology , Male , Memory/physiology , Mice , Mice, Inbred C57BL , N-Methyl-3,4-methylenedioxyamphetamine/therapeutic use , Serotonin Plasma Membrane Transport Proteins/metabolism , Selective Serotonin Reuptake Inhibitors/therapeutic use , Stress Disorders, Post-Traumatic/chemically induced , Stress Disorders, Post-Traumatic/drug therapy , Stress Disorders, Post-Traumatic/psychology
17.
Psychopharmacology (Berl) ; 234(15): 2277-2287, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28589263

ABSTRACT

RATIONALE: Methamphetamine is one of the most largely consumed illicit drugs, and its use is associated with abuse liability and several adverse health effects, such as sleep impairment. Importantly, sleep quality can influence addiction treatment outcomes. Evidence suggests that tolerance can develop to the sleep-disrupting effects of stimulant drugs. OBJECTIVE: The aim of the present study was to investigate the development of tolerance to the actigraphy-based sleep-disrupting and stimulant effects of methamphetamine self-administration in rhesus monkeys. METHODS: Methamphetamine (0.03 mg/kg/inf, i.v.) self-administration was carried out following three different protocols: 14 consecutive days of self-administration, 5 days/week for 3 weeks, with a 2-day interval between 5-day blocks of self-administration, and 3 days/week for 3 weeks, with a 4-day interval between 3-day blocks of self-administration. Daytime activity and activity-based sleep measures were evaluated with Actiwatch monitors a week before (baseline parameters) and throughout each protocol. RESULTS: Methamphetamine self-administration markedly disrupted sleep-like measures and increased daytime activity. Tolerance developed to those effects with repeated methamphetamine intake exceeding five consecutive days. Inclusion of washout periods (2 or 4 days) between blocks of methamphetamine self-administration attenuated the development of tolerance, with longer breaks from methamphetamine intake being more effective in maintaining the sleep-disrupting and stimulant effects of methamphetamine. CONCLUSIONS: Tolerance can develop to the stimulant and sleep-disrupting effects of methamphetamine self-administration. Interruption of drug intake extends the effects of methamphetamine on sleep-like measures and daytime activity.


Subject(s)
Actigraphy/methods , Drug Tolerance/physiology , Methamphetamine/administration & dosage , Sleep Wake Disorders/chemically induced , Sleep/drug effects , Sleep/physiology , Animals , Central Nervous System Stimulants/administration & dosage , Female , Humans , Locomotion/drug effects , Locomotion/physiology , Macaca mulatta , Male , Self Administration , Sleep Wake Disorders/physiopathology , Sleep, REM/drug effects , Sleep, REM/physiology
18.
Psychopharmacology (Berl) ; 234(17): 2607-2617, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28584928

ABSTRACT

RATIONALE: Accumulating evidence shows that the serotonergic system plays a major role in psychostimulant abuse through its interactions with the dopaminergic system. Studies indicate that serotonin 5-HT2C receptors are one of the main classes of receptors involved in mediating the influence of serotonin in drug abuse. OBJECTIVE: The aim of the present study was to evaluate the effects of the selective serotonin 5-HT2C receptor agonist WAY163909 on the behavioral neuropharmacology of cocaine and methamphetamine in adult rhesus macaques. METHODS: Cocaine or methamphetamine self-administration and reinstatement were evaluated under second-order and fixed-ratio schedules of reinforcement, respectively. Cocaine- and methamphetamine-induced increases in dopamine were assessed through in vivo microdialysis targeting the nucleus accumbens. RESULTS: Pretreatment with WAY163909 dose-dependently attenuated cocaine and methamphetamine self-administration and drug-induced reinstatement of extinguished behavior previously maintained by cocaine or methamphetamine delivery. In an additional experiment, WAY163909 induced a dose-dependent attenuation of cocaine- or methamphetamine-induced dopamine overflow in the nucleus accumbens. CONCLUSIONS: Our data indicate that selective 5-HT2C receptor activation decreases drug intake and drug-seeking behavior in nonhuman primate models of psychostimulant abuse through neurochemical mechanisms involved in the modulation of mesolimbic dopamine.


Subject(s)
Azepines/pharmacology , Cocaine/administration & dosage , Dopamine/metabolism , Drug-Seeking Behavior/drug effects , Indoles/pharmacology , Methamphetamine/administration & dosage , Nucleus Accumbens/drug effects , Serotonin 5-HT2 Receptor Agonists/pharmacology , Amphetamine-Related Disorders/drug therapy , Amphetamine-Related Disorders/metabolism , Animals , Azepines/therapeutic use , Central Nervous System Stimulants/administration & dosage , Cocaine-Related Disorders/drug therapy , Cocaine-Related Disorders/metabolism , Female , Indoles/therapeutic use , Macaca mulatta , Male , Microdialysis , Nucleus Accumbens/metabolism , Self Administration , Serotonin 5-HT2 Receptor Agonists/therapeutic use
19.
Neuropharmacology ; 123: 299-309, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-28495376

ABSTRACT

GABAA receptor positive allosteric modulators (GABAA receptor modulators) are commonly used for the treatment of insomnia. Nevertheless, the effects of these compounds on psychostimulant-induced sleep impairment are poorly understood. Because GABAA receptor modulators have been shown to decrease the abuse-related effects of psychostimulants, the aim of the present study was to evaluate the effects of temazepam (0.3, 1.0 or 3.0 mg/kg) and eszopiclone (0.3, 1.0 or 3.0 mg/kg), two GABAA receptor modulators, on the behavioral neuropharmacology of methamphetamine in adult rhesus macaques (n = 5). Sleep-like measures and general daytime activity were evaluated with Actiwatch monitors. Methamphetamine self-administration (0.03 mg/kg/inf) was evaluated during morning sessions. Methamphetamine-induced dopamine overflow was assessed through in vivo microdialysis targeting the nucleus accumbens. Nighttime treatment with either temazepam or eszopiclone was ineffective in improving sleep-like measures disrupted by methamphetamine self-administration. Acute pretreatment with a low dose of temazepam before self-administration sessions increased methamphetamine self-administration without affecting normal daytime home-cage activity. At a high dose, acute temazepam pretreatment decreased methamphetamine self-administration and attenuated methamphetamine-induced increases in dopamine in the nucleus accumbens, without decreasing general daytime activity. Acute eszopiclone treatment exerted no effects on methamphetamine intake or drug-induced increases in dopamine. Our study suggests that treatments based on GABAA receptor modulators are not effective for the treatment of sleep disruption in the context of psychostimulant use. In addition, distinct GABAA receptor modulators differentially modulated the abuse-related effects of methamphetamine, with acute treatment with the high efficacy GABAA receptor modulator temazepam decreasing the behavioral and neurochemical effects of methamphetamine.


Subject(s)
Amphetamine-Related Disorders/drug therapy , Dopamine/metabolism , Eszopiclone/pharmacology , GABA-A Receptor Agonists/pharmacology , Sleep/drug effects , Temazepam/pharmacology , Amphetamine-Related Disorders/metabolism , Animals , Central Nervous System Stimulants/administration & dosage , Dose-Response Relationship, Drug , Female , Macaca mulatta , Male , Methamphetamine/administration & dosage , Receptors, GABA-A/metabolism , Self Administration
20.
Neuropsychopharmacology ; 42(10): 1962-1971, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28425496

ABSTRACT

3,4-Methylenedioxymethamphetamine (MDMA) increases sociality in humans and animals. Release of serotonin (5-HT) is thought to have an important role in the increase in social behaviors, but the mechanisms underlying these effects are poorly understood. Despite the advantages of nonhuman primate models, no studies have examined the mechanisms of the social effects of MDMA in nonhuman primates. The behavior and vocalizations of four group-housed squirrel monkeys were examined following administration of MDMA, its enantiomers, and methamphetamine. 5-HT receptor antagonists and agonists were given as drug pretreatments. Data were analyzed using linear mixed-effects models. MDMA and its enantiomers increased affiliative social behaviors and vocalizations, whereas methamphetamine had only modest effects on affiliative behaviors. Pretreatment with a 5-HT2A receptor antagonist and a 5-HT2C receptor agonist attenuated the MDMA-induced increase in social behaviors, while a 5-HT1A receptor antagonist did not alter affiliative vocalizations and increased MDMA-induced social contact. Nonhuman primates show MDMA-specific increases in affiliative social behaviors following MDMA administration, in concordance with human and rodent studies. MDMA-induced increases in social behaviors are 5-HT2A, but not 5-HT1A, receptor dependent. Understanding the neurochemical mechanisms mediating the prosocial effects of MDMA could help in the development of novel therapeutics with the unique social effects of MDMA but fewer of its limitations.


Subject(s)
N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Psychotropic Drugs/pharmacology , Receptor, Serotonin, 5-HT2A/metabolism , Social Behavior , Animals , Dose-Response Relationship, Drug , Linear Models , Male , Methamphetamine/pharmacology , Receptor, Serotonin, 5-HT1A/metabolism , Saimiri , Serotonin Agents/pharmacology , Vocalization, Animal/drug effects , Vocalization, Animal/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...