Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters











Publication year range
1.
Drug Alcohol Depend Rep ; 7: 100148, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37033159

ABSTRACT

Background: Studies of nonhuman primates with exposures of up to 100 days of cocaine self-administration (SA) have provided evidence that the central effects of cocaine progress over time. These durations of cocaine exposure, however, may be insufficient to capture the extent of the neurobiological alterations observed in cocaine users, many of whom use the drug for years. The goal of the present study was to determine whether 1.5 years of cocaine SA would result in further progression of alterations in functional brain activity. Methods: Adult male rhesus monkeys were exposed to 300 sessions of high-dose cocaine SA over 1.5 years. Following the final session rates of local cerebral glucose utilization (LCGU) were assessed with the 2-[14C]-deoxyglucose method and compared to rates of LCGU in control monkeys who responded for food reinforcement. In addition, LCGU in these animals was compared to a previously published group of monkeys that had self-administered cocaine or food for 100 sessions over a 4-5 month period. Results: Compared to 100 days of exposure, 300 days of cocaine SA further reduced LCGU in the post-commissural striatum and produced reductions in areas unaffected by the shorter duration of exposure, such as the hypothalamus, all of the amygdala, and large expanses of cortex. Conclusions: These findings demonstrate a clear progression of the impact of cocaine on functional activity with increasing durations of drug experience and have important implications for the development of potential strategies for the treatment of cocaine use disorder.

2.
Neuropsychopharmacology ; 48(2): 290-298, 2023 01.
Article in English | MEDLINE | ID: mdl-34385608

ABSTRACT

Previous studies in humans and in animals have shown dramatic effects of cocaine on measures of brain function that persist into abstinence. The purpose of this study was to examine the neurobiological consequences of abstinence from cocaine, using a model that removes the potential confound of cocaine cues. Adult male rhesus monkeys self-administered cocaine (0.3 mg/kg/injection; N = 8) during daily sessions or served as food-reinforcement controls (N = 4). Two times per week, monkeys were placed in a neutral environment and presented with a cartoon video for ~30 min, sometimes pre- and sometimes post-operant session, but no reinforcement was presented during the video. After ~100 sessions and when the cocaine groups had self-administered 900 mg/kg cocaine, the final experimental condition was a terminal 2-[14C]-deoxyglucose procedure, which occurred in the neutral (cartoon video) environment; for half of the monkeys in each group, this occurred after 1 day of abstinence and for the others after 30 days of abstinence. Rates of local cerebral glucose metabolism were measured in 57 brain regions. Global rates of cerebral metabolism were significantly lower in animals 1 day and 30 days post-cocaine self-administration when compared to those of food-reinforced controls. Effects were larger in 30- vs. 1-day cocaine abstinence, especially in prefrontal, parietal and cingulate cortex, as well as dorsal striatum and thalamus. Because these measures were obtained from monkeys while in a neutral environment, the deficits in glucose utilization can be attributed to the consequences of cocaine exposure and not to effects of conditioned stimuli associated with cocaine.


Subject(s)
Cocaine-Related Disorders , Cocaine , Animals , Humans , Male , Macaca mulatta , Self Administration , Cocaine-Related Disorders/metabolism , Brain , Dose-Response Relationship, Drug
5.
Brain Res ; 1761: 147387, 2021 Feb 23.
Article in English | MEDLINE | ID: mdl-33631209

ABSTRACT

Phenmetrazine (PHEN) is a putative treatment for cocaine and psychostimulant recidivism; however, neurochemical changes underlying its activity have not been fully elucidated. We sought to characterize brain homeostatic adaptations to chronic PHEN, specifically on functional brain activity (local cerebral glucose utilization), G-Protein Coupled Receptor-stimulated G-protein activation, and phosphorylation of ERK1/2Thr202/Tyr204, GSK3ßTyr216, and DARPP-32Thr34. Male Sprague-Dawley rats were implanted with sub-cutaneous minipumps delivering either saline (vehicle), acute (2-day) or chronic (14-day) low dose (25 mg/kg/day) or high dose (50 mg/kg/day) PHEN. Acute administration of high dose PHEN increased local cerebral glucose utilization measured by 2-[14C]-deoxyglucose uptake in basal ganglia and motor-related regions of the rat brain. However, chronically treated animals developed tolerance to these effects. To identify the neurochemical changes associated with PHEN's activity, we performed [35S]GTPγS binding assays on unfixed and immunohistochemistry on fixed coronal brain sections. Chronic PHEN treatment dose-dependently attenuated D2 dopamine and α2-adrenergic, but not 5-HT1A, receptor-mediated G-protein activation. Two distinct patterns of effects on pERK1/2 and pDARPP-32 were observed: 1) chronic low dose PHEN decreased pERK1/2, and also significantly increased pDARPP-32 levels in some regions; 2) acute and chronic PHEN increased pERK1/2, but chronic high dose PHEN treatment tended to decrease pDARPP-32. Chronic low dose, but not high dose, PHEN significantly reduced pGSK3ß levels in several regions. Our study provides definitive evidence that extended length PHEN dosage schedules elicit distinct modes of neuronal acclimatization in cellular signaling. These pharmacodynamic modifications should be considered in drug development for chronic use.

7.
Brain Res ; 1727: 146552, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31726041

ABSTRACT

The mesocorticolimbic system is comprised of dopaminergic neurons in the ventral tegmental area (VTA) and their projection targets in the ventral striatum, amygdala, prefrontal cortex, and hippocampus, among others. Regulation of dopamine transmission within this system is achieved in part through a negative feedback mechanism via dopamine D2 autoreceptors located on somatodendrites and terminals of VTA dopaminergic neurons. Dysregulation of this mechanism has been implicated in addiction and other psychiatric disorders, although the biological bases for these associations are unclear. In order to elucidate the functional consequences of VTA D2 receptor dysregulation, this study investigated alterations in local cerebral glucose utilization throughout the brain following Drd2 knockdown in the VTA. Male Sprague-Dawley rats received bilateral injections of lentivirus encoding shRNAs against the rat dopamine D2 receptor, scrambled shRNA or phosphate buffered saline. The autoradiographic 2-[14C]deoxyglucose metabolic mapping procedure was conducted 22 days post-infection. Brains were sectioned for autoradiography and glucose utilization was measured across distinct regions throughout the brain. Local cerebral glucose utilization was found to be elevated in the Drd2 knockdown group as compared to control groups. These greater levels of metabolic activity following Drd2 knockdown in the VTA were observed not only in the mesocorticolimbic system and associated dopamine pathways, but also in a global pattern that included many areas with far less concentrated VTA dopamine inputs. This suggests that even a partial Drd2 deletion in the VTA can have widespread consequences and impact information flow in diverse networks that process sensory, cognitive, motor and emotional information.


Subject(s)
Receptors, Dopamine D2/physiology , Ventral Tegmental Area/physiology , Amygdala/physiology , Animals , Gene Knockdown Techniques , Glucose/metabolism , Hippocampus/physiology , Lentivirus , Male , Prefrontal Cortex/physiology , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D2/genetics , Ventral Striatum/physiology
8.
Brain Struct Funct ; 224(4): 1417-1428, 2019 May.
Article in English | MEDLINE | ID: mdl-30747315

ABSTRACT

It has been shown that exposure to cocaine can result in neuroinflammatory responses. Microglia, the resident CNS immune cells, undergo a transition to an activated state when challenged. In rodents, and possibly humans, cocaine exposure activates microglia. The goal of this study was to assess the extent and magnitude of microglial activation in rhesus monkeys with an extensive history of cocaine self-administration. Male rhesus monkeys (N = 4/group) were trained to respond on a fixed-interval 3-min schedule of food or 0.3 mg/kg/injection cocaine presentation (30 reinforcers/session) for 300 sessions. At the end of the final session, monkeys were administered 2-[14C]deoxyglucose intravenously and 45 min later euthanized. Brain sections were used for autoradiographic assessments of glucose utilization and for microglia activation with [3H]PK11195, a marker for the microglial 18-kDa translocator protein. There were no group differences in gray matter [3H]PK11195 binding, while binding was significantly greater in cocaine self-administration animals as compared to food controls in 8 of the 11 white matter tracts measured at the striatal level. Binding did not differ from control at other levels. There were also significant increases in white matter local cerebral glucose utilization at the striatal level, which were positively correlated with [3H]PK11195 binding. The present findings demonstrate an elevation in [3H]PK11195 binding in forebrain white matter tracts of nonhuman primates with a prolonged history of cocaine self-administration. These elevations were also associated with greater cerebral metabolic rates. These data suggest that white matter deficits may contribute to behavioral, motivational, and cognitive impairments observed in cocaine abusers.


Subject(s)
Cocaine/administration & dosage , Frontal Lobe/drug effects , Glucose/metabolism , Microglia/drug effects , White Matter/drug effects , Animals , Drug-Seeking Behavior , Frontal Lobe/metabolism , Gray Matter/drug effects , Gray Matter/metabolism , Macaca mulatta , Male , Microglia/metabolism , Reinforcement Schedule , White Matter/metabolism
9.
Biol Psychiatry ; 80(9): 702-710, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27059874

ABSTRACT

BACKGROUND: A major goal of treatments for cocaine addiction is to reduce relapse-associated cravings, which are typically induced by environmental stimuli associated with cocaine use and related to changes in dopamine neurotransmission. METHODS: The present study used an animal model of cocaine seeking to determine functional consequences of cue exposure using fluorodeoxyglucose positron emission tomography and to relate findings to juvenile levels of dopamine transporter and D2-like receptor availabilities determined before any drug exposure. Adult male rhesus monkeys (N = 11) self-administered cocaine (0.2 mg/kg per injection) under a second-order schedule of reinforcement, in which responding was maintained by conditioned reinforcers. Positron emission tomography scans assessing glucose utilization, a marker of functional activation, were conducted during cocaine-cue responding and food-reinforced responding in a context where cocaine was never available. RESULTS: Compared with the noncocaine condition, we found significant functional activation in the medial prefrontal cortex, anterior cingulate, precuneus region of the parietal cortex, and striatum-findings similar to those reported in humans who abuse cocaine. Furthermore, these functional activations in the prefrontal, cingulate, and parietal cortex measured during cocaine-cue responding were significantly correlated with juvenile measures of dopamine transporter availability, whereas no significant relationship with prior D2-like receptor availability was observed in any brain region. CONCLUSIONS: The similarity between the present findings and findings in humans who use cocaine supports the use of this model for examination of factors that affect the development and intensity of cue-induced drug seeking and provides evidence for potential biomarkers for the evaluation of potential treatments (behavioral and pharmacologic) for cocaine abuse.


Subject(s)
Brain/drug effects , Brain/metabolism , Cocaine/administration & dosage , Cues , Dopamine Plasma Membrane Transport Proteins/metabolism , Drug-Seeking Behavior/physiology , Reinforcement Schedule , Animals , Brain/diagnostic imaging , Craving/drug effects , Craving/physiology , Glucose/metabolism , Macaca mulatta , Male , Positron-Emission Tomography , Receptors, Dopamine D2/metabolism , Self Administration
10.
Addict Biol ; 21(3): 519-29, 2016 May.
Article in English | MEDLINE | ID: mdl-25684556

ABSTRACT

Exposure to stimuli and environments associated with drug use is considered one of the most important contributors to relapse among substance abusers. Neuroimaging studies have identified neural circuits underlying these responses in cocaine-dependent subjects. But these studies are often difficult to interpret because of the heterogeneity of the participants, substances abused, and differences in drug histories and social variables. Therefore, the goal of this study was to assess the functional effects of exposure to cocaine-associated stimuli in a non-human primate model of cocaine self-administration, providing precise control over these variables, with the 2-[(14) C]deoxyglucose method. Rhesus monkeys self-administered 0.3 mg/kg/injection cocaine (n = 4) under a fixed-interval 3-minute (FI 3-min) schedule of reinforcement (30 injections/session) for 100 sessions. Control animals (n = 4) underwent identical schedules of food reinforcement. Sessions were then discontinued for 30 days, after which time, monkeys were exposed to cocaine- or food-paired cues, and the 2-[(14) C]deoxyglucose experiment was conducted. The presentation of the cocaine-paired cues resulted in significant increases in functional activity within highly restricted circuits that included portions of the pre-commissural striatum, medial prefrontal cortex, rostral temporal cortex and limbic thalamus when compared with control animals presented with the food-paired cues. The presentation of cocaine-associated cues increased brain functional activity in contrast to the decreases observed after cocaine consumption. Furthermore, the topography of brain circuits engaged by the expectation of cocaine is similar to the distribution of effects during the earliest phases of cocaine self-administration, prior to the onset of neuroadaptations that accompany chronic cocaine exposure.


Subject(s)
Behavior, Animal , Brain/metabolism , Cocaine-Related Disorders/metabolism , Cocaine/administration & dosage , Cues , Dopamine Uptake Inhibitors/administration & dosage , Animals , Autoradiography , Carbon Radioisotopes , Deoxyglucose , Disease Models, Animal , Macaca mulatta , Male , Neostriatum/metabolism , Prefrontal Cortex/metabolism , Reinforcement Schedule , Reinforcement, Psychology , Self Administration , Spectrophotometry , Temporal Lobe/metabolism , Thalamus/metabolism
11.
Brain Struct Funct ; 221(5): 2703-15, 2016 06.
Article in English | MEDLINE | ID: mdl-26013302

ABSTRACT

Repeated exposure to cocaine is known to dysregulate the norepinephrine system, and norepinephrine has also been implicated as having a role in abstinence and withdrawal. The goal of this study was to determine the effects of exposure to cocaine self-administration and subsequent abstinence on regulatory elements of the norepinephrine system in the nonhuman primate brain. Rhesus monkeys self-administered cocaine (0.3 mg/kg/injection, 30 reinforcers/session) under a fixed-interval 3-min schedule of reinforcement for 100 sessions. Animals in the abstinence group then underwent a 30-day period during which no operant responding was conducted, followed by a final session of operant responding. Control animals underwent identical schedules of food reinforcement and abstinence. This duration of cocaine self-administration has been shown previously to increase levels of norepinephrine transporters (NET) in the ventral noradrenergic bundle terminal fields. In contrast, in the current study, abstinence from chronic cocaine self-administration resulted in elevated levels of [(3)H]nisoxetine binding to the NET primarily in dorsal noradrenergic bundle terminal field structures. As compared to food reinforcement, chronic cocaine self-administration resulted in decreased binding of [(3)H]RX821002 to α2-adrenoceptors primarily in limbic-related structures innervated by both dorsal and ventral bundles, as well as elevated binding in the striatum. However, following abstinence from responding for cocaine binding to α2-adrenoceptors was not different than in control animals. These data demonstrate the dynamic nature of the regulation of norepinephrine during cocaine use and abstinence, and provide further evidence that the norepinephrine system should not be overlooked in the search for effective pharmacotherapies for cocaine dependence.


Subject(s)
Brain/metabolism , Cocaine-Related Disorders/metabolism , Cocaine/administration & dosage , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Adrenergic alpha-2 Receptor Antagonists/administration & dosage , Animals , Autoradiography , Brain/diagnostic imaging , Fluoxetine/administration & dosage , Fluoxetine/analogs & derivatives , Idazoxan/administration & dosage , Idazoxan/analogs & derivatives , Macaca mulatta , Male , Neural Pathways/metabolism , Reinforcement Schedule , Self Administration
12.
Neuropharmacology ; 85: 528-37, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24953829

ABSTRACT

Cocaine users exhibit a wide range of behavioral impairments accompanied by brain structural, neurochemical and functional abnormalities. Metabolic mapping studies in cocaine users and animal models have shown extensive functional alterations throughout the striatum, limbic system, and cortex. Few studies, however, have evaluated the persistence of these effects following cessation of cocaine availability. The purpose of this study, therefore, was to assess the functional effects of re-exposure to cocaine in nonhuman primates after the discontinuation of cocaine self-administration for 30 or 90 days, using the quantitative autoradiographic 2-[14C]deoxyglucose (2DG) method. Rhesus monkeys self-administered cocaine (fixed interval 3-min schedule, 30 infusions per session, 0.3 mg/kg/infusion) for 100 sessions followed by 30 (n=4) or 90 days (n=3) during which experimental sessions were not conducted. Food-reinforced control animals (n=5) underwent identical schedules of reinforcement. Animals were then re-exposed to cocaine or food for one final session and the 2DG method applied immediately after session completion. Compared to controls, re-exposure to cocaine after 30 or 90 day drug-free periods resulted in lower rates of glucose utilization in ventral and dorsal striatum, prefrontal and temporal cortex, limbic system, thalamus, and midbrain. These data demonstrate that vulnerability to the effects of cocaine persists for as long as 90 days after cessation of drug use. While there was some evidence for recovery (fewer brain areas were affected by cocaine re-exposure at 90 days as compared to 30 days), this was not uniform across regions, thus suggesting that recovery occurs at different rates in different brain systems.


Subject(s)
Brain/drug effects , Brain/metabolism , Cocaine-Related Disorders/metabolism , Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Animals , Autoradiography , Carbon Radioisotopes , Deoxyglucose , Food , Glucose/metabolism , Macaca mulatta , Male , Random Allocation , Reinforcement, Psychology , Self Administration , Substance Withdrawal Syndrome , Time Factors
13.
Drug Alcohol Depend ; 137: 143-7, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24529965

ABSTRACT

BACKGROUND: Neuroimaging studies of cocaine users have demonstrated white matter abnormalities associated with behavioral measures of impulsivity and decision-making deficits. The underlying bases for this dysregulation in white matter structure and function have yet to be determined. The aim of the present studies was to investigate the influence of prolonged cocaine self-administration on the levels of myelin-associated proteins and mRNAs in nonhuman primate white matter. METHODS: Rhesus monkeys (N=4) self-administered cocaine (0.3mg/kg/inj, 30 reinforcers per session) for 300 sessions. Control animals (N=4) responded for food. Following the final session monkeys were euthanized and white matter tissue at three brain levels was processed for immunoblotting analysis of proteolipid protein (PLP) and myelin basic protein (MBP), as well as for in situ hybridization histochemical analysis of PLP and MBP mRNAs. RESULTS: Both MBP and PLP immunoreactivities in white matter at the level of the precommissural striatum were significantly lower in tissue from monkeys self-administering cocaine as compared to controls. No significant differences were seen for either protein at the levels of the prefrontal cortex or postcommissural striatum. In addition, no differences were observed in expression of mRNA for either protein. CONCLUSIONS: These preliminary findings, in a nonhuman model of prolonged cocaine self-administration, provide further evidence that compromised myelin may underlie the deficits in white matter integrity described in studies of human cocaine users.


Subject(s)
Brain/drug effects , Cocaine/administration & dosage , Myelin Proteins , Nerve Fibers, Myelinated/drug effects , Animals , Brain/metabolism , Macaca mulatta , Male , Myelin Proteins/metabolism , Nerve Fibers, Myelinated/metabolism , Random Allocation , Self Administration , Time Factors
14.
Brain Res ; 1527: 67-78, 2013 Aug 21.
Article in English | MEDLINE | ID: mdl-23806775

ABSTRACT

Rearing young rodents in socially isolated or environmentally enriched conditions has been shown to affect numerous components of the dopamine system as well as behavior. Methylphenidate (MPH), a commonly used dopaminergic agent, may affect animals differently based on rearing environment. Here we examined the interaction between environment and chronic MPH treatment at clinically relevant doses, administered via osmotic minipump. Young Sprague Dawley rats (PND 21) were assigned to environmentally enriched, pair-housed, or socially isolated rearing conditions, and treated with either 0, 2, 4, or 8 mg/kg/day MPH for 3 weeks. At the end of the treatment period, animals were tested for locomotor activity and anxiety-like behavior. The densities of D1-like and D2-like receptors were measured in the striatum using in vitro receptor autoradiography. Locomotor activity and anxiety-like behavior were increased in isolated animals compared to pair-housed and enriched animals. The density of D1-like receptors was greater in isolated animals, but there were no differences between groups in D2-like receptor density. Finally, there were no effects of MPH administration on any reported measure. This study provides evidence for an effect of early rearing environment on the dopamine system and behavior, and also suggests that MPH administration may not have long-term consequences.


Subject(s)
Behavior, Animal/drug effects , Brain/drug effects , Central Nervous System Stimulants/pharmacology , Methylphenidate/pharmacology , Receptors, Dopamine/biosynthesis , Social Isolation , Aging , Animals , Autoradiography , Brain/metabolism , Housing, Animal , Male , Motor Activity/drug effects , Rats , Rats, Sprague-Dawley , Social Isolation/psychology
15.
Neuropsychopharmacology ; 34(5): 1162-71, 2009 Apr.
Article in English | MEDLINE | ID: mdl-18769473

ABSTRACT

Although dysregulation within the dopamine (DA) system is a hallmark feature of chronic cocaine exposure, the question of whether these alterations persist into abstinence remains largely unanswered. Nonhuman primates represent an ideal model in which to assess the effects of abstinence on the DA system following chronic cocaine exposure. In this study, male rhesus monkeys self-administered cocaine (0.3 mg/kg per injection, 30 reinforcers per session) under a fixed-interval 3-min schedule for 100 days followed by either 30 or 90 days abstinence. This duration of cocaine self-administration has been previously shown to decrease DA D2-like receptor densities and increase levels of D1-like receptors and DA transporters (DAT). Responding by control monkeys was maintained by food presentation under an identical protocol and the same abstinence periods. [(3)H]SCH 23390 binding to DA D1 receptors following 30 days of abstinence was significantly higher in all portions of the striatum, compared to control animals, whereas [(3)H]raclopride binding to DA D2 receptors was not different between groups. [(3)H]WIN 35 428 binding to DAT was also significantly higher throughout virtually all portions of the dorsal and ventral striatum following 30 days of abstinence. Following 90 days of abstinence, however, levels of DA D1 receptors and DAT were not different from control values. Although these results indicate that there is eventual recovery of the separate elements of the DA system, they also highlight the dynamic nature of these components during the initial phases of abstinence from chronic cocaine self-administration.


Subject(s)
Cocaine-Related Disorders/physiopathology , Cocaine/administration & dosage , Corpus Striatum/physiopathology , Dopamine/metabolism , Animals , Benzazepines/pharmacology , Central Nervous System Stimulants/administration & dosage , Central Nervous System Stimulants/toxicity , Cocaine/analogs & derivatives , Cocaine/pharmacology , Cocaine/toxicity , Corpus Striatum/drug effects , Dopamine Antagonists/pharmacology , Dopamine D2 Receptor Antagonists , Dopamine Plasma Membrane Transport Proteins/metabolism , Dopamine Uptake Inhibitors/pharmacology , Macaca mulatta , Male , Raclopride/pharmacology , Receptors, Dopamine D1/antagonists & inhibitors , Self Administration , Tritium
16.
Brain Struct Funct ; 213(1-2): 73-91, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18283492

ABSTRACT

The monoamines in the amygdala modulate multiple aspects of emotional processing in the mammalian brain, and organic or pharmacological dysregulation of these systems can result in affective pathologies. Knowledge of the normal distribution of these neurotransmitters, therefore, is central to our understanding of both the normal processes regulated by the amygdala and the pathological conditions associated with monoaminergic dysregulation. The monoaminergic transporters have proven to be accurate and reliable markers of the distributions of their substrates. The purpose of this review was twofold: First, to briefly recount the functional relevance of dopamine, serotonin, and norepinephrine transmission in the amygdala, and second, to describe and compare the distributions of the monoamine transporters in the rodent, monkey, and human brain. The transporters were found to be heterogeneously distributed in the amygdala. The dopamine transporter (DAT) is consistently found to be extremely sparsely distributed, however the various accounts of its subregional topography are inconsistent, making any cross-species comparisons difficult. The serotonin transporter (SERT) had the greatest overall degree of labeling of the three markers, and was characterized by substantial inter-species variability in its relative distribution. The norepinephrine transporter (NET) was shown to possess an intermediate level of labeling, and like the SERT, its distribution is not consistent across the three species. The results of these comparisons indicate that caution should be exercised when using animal models to investigate the complex processes modulated by the monoamines in the amygdala, as their relative contributions to these functions may differ across species.


Subject(s)
Amygdala/metabolism , Neurons/metabolism , Plasma Membrane Neurotransmitter Transport Proteins/metabolism , Animals , Biogenic Monoamines/metabolism , Haplorhini , Humans , Neurons/cytology , Rodentia , Serotonin/metabolism , Species Specificity
17.
Prog Neuropsychopharmacol Biol Psychiatry ; 31(8): 1593-600, 2007 Nov 15.
Article in English | MEDLINE | ID: mdl-17900777

ABSTRACT

Repeated exposure to psychostimulant drugs such as cocaine has been shown in numerous studies to produce significant neuroadaptations in both structure and function throughout the brain. Nonhuman primate models provide a way to systematically evaluate these adaptations engendered by cocaine self-administration and simulate the progressive nature of cocaine addiction in humans. Functional activity, measured using the 2-[14C]deoxyglucose method, was evaluated at selected critical time points over the course of chronic cocaine self-administration in rhesus monkeys. The effects of cocaine exposure in the initial stages of self-administration resulted in changes in functional activity in a highly restricted network of interconnected brain regions when compared to activity in food-reinforced controls. This pattern of changes was confined mainly to ventromedial prefrontal cortex and ventral striatum. Following chronic exposure to cocaine self-administration, however, the spatial extent and intensity of significant alterations in functional activity expanded considerably. The shift in topography of these changes was orderly, originating ventromedially in the prefrontal cortical-ventral striatal network and expanding dorsally to encompass the dorsal striatum. A strikingly similar progression occurred within the cortical areas that project to each of these striatal regions. Preliminary studies suggest that this pattern is maintained despite periods of abstinence from cocaine. The shifting patterns of cerebral metabolic function that accompany longer durations of cocaine self-administration may underlie many of the characteristics of chronic drug exposure, and may provide transitional mechanisms to more compulsive cocaine use.


Subject(s)
Cocaine-Related Disorders/psychology , Cocaine/pharmacology , Animals , Brain/pathology , Brain Chemistry/drug effects , Cocaine-Related Disorders/pathology , Cocaine-Related Disorders/physiopathology , Disease Models, Animal , Humans , Substance Withdrawal Syndrome/psychology
18.
Eur J Neurosci ; 23(11): 3109-18, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16820001

ABSTRACT

Previous studies utilizing a nonhuman primate model have shown that cocaine self-administration in its initial stages is accompanied by alterations in functional activity largely within the prefrontal cortex and ventral striatum. Continued cocaine exposure may considerably change this response. The purpose of the present investigation was to characterize the effects of reinforcing doses of cocaine on cerebral metabolism in a nonhuman primate model of cocaine self-administration, following an extended history of cocaine exposure, using the quantitative 2-[(14)C]deoxyglucose (2-DG) method. Rhesus monkeys were trained to self-administer 0.03 mg/kg/injection (n = 4) or 0.3 mg/kg/injection (n = 4) cocaine and compared to monkeys trained to respond under an identical schedule of food reinforcement (n = 6). Monkeys received 30 reinforcers per session for a total of 100 sessions. Metabolic mapping was conducted at the end of the final session. After this extended history, cocaine self-administration dose-dependently reduced glucose utilization throughout the striatum and prefrontal cortex similarly to the initial stages of self-administration. However, glucose utilization was also decreased in a dose-independent manner in large portions of the temporal lobe including the amygdala, hippocampus and surrounding neocortex. The recruitment of temporal structures indicates that the pattern of changes in functional activity has undergone significant expansion beyond limbic regions into association areas that mediate higher order cognitive and emotional processing. These data strongly contribute to converging evidence from human studies demonstrating structural and functional abnormalities in temporal and prefrontal areas of cocaine abusers, and suggest that substance abusers may undergo progressive cognitive decline with continued exposure to cocaine.


Subject(s)
Cocaine/administration & dosage , Deoxyglucose/metabolism , Dopamine Uptake Inhibitors/administration & dosage , Reinforcement, Psychology , Temporal Lobe/drug effects , Analysis of Variance , Animals , Autoradiography , Behavior, Animal , Brain Mapping , Carbon Isotopes/metabolism , Dose-Response Relationship, Drug , Glucose/metabolism , Macaca mulatta , Male , Reinforcement Schedule , Self Administration/methods , Temporal Lobe/metabolism
19.
Psychopharmacology (Berl) ; 180(4): 781-8, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15739079

ABSTRACT

RATIONALE: While cocaine blocks dopamine and serotonin transporters, considerably less emphasis has been placed on its effects following blockade of the norepinephrine transporter (NET). To date, no studies have made a systematic investigation of the effects of chronic cocaine on primate NET density. OBJECTIVE: We previously reported increases in NET density in portions of the monkey bed nucleus of stria terminalis after 100 days of cocaine self-administration. In the present study we extend these findings and assess the changes in [3H]nisoxetine binding in additional brain regions of rhesus monkeys chronically self-administrating cocaine. RESULTS: [3H]Nisoxetine binding sites in the A1 noradrenergic cell group were significantly higher after 5 days of cocaine exposure. One hundred days of self-administration also induced a higher density of NET binding within the A1 cell group; however, in addition, the effects extended to the nucleus prepositus, as well as forebrain regions such as hypothalamic nuclei, basolateral amygdala, parasubiculum, and entorhinal cortex. CONCLUSIONS: These data demonstrate that cocaine self-administration alters the noradrenergic system of nonhuman primates. Although cocaine affected NET binding sites in the forebrain projections of both the ventral (VNAB) and dorsal (DNAB) noradrenergic bundles, the alteration in the A1 cell group at the early time-point suggests that the VNAB appears to be more sensitive than the DNAB to the effects of cocaine. Given the role of norepinephrine in arousal and attention, as well as mediating responses to stress, long-term exposure to cocaine is likely to result in significant changes in the way in which information is perceived and processed by the central nervous system of long-term cocaine users.


Subject(s)
Brain/drug effects , Cocaine/administration & dosage , Dopamine Uptake Inhibitors/administration & dosage , Norepinephrine Plasma Membrane Transport Proteins/metabolism , Animals , Autoradiography/methods , Behavior, Animal , Brain/anatomy & histology , Brain/metabolism , Brain Mapping , Carbon Isotopes/pharmacokinetics , Deoxyglucose/pharmacokinetics , Drug Administration Schedule , Fluoxetine/analogs & derivatives , Fluoxetine/pharmacokinetics , Macaca mulatta , Male , Protein Binding/drug effects , Self Administration , Tritium/pharmacokinetics
20.
Neurosci Lett ; 370(2-3): 201-5, 2004 Nov 11.
Article in English | MEDLINE | ID: mdl-15488323

ABSTRACT

Cocaine abuse is associated with autonomic dysregulation, such as altered blood pressure and heart rate. Both central and peripheral mechanisms have been implicated in mediating these changes, however, to date, no study has examined functional changes in activity within central autonomic-associated brain regions in response to cocaine in non-human primates. The aim of the present study was to measure local cerebral glucose utilization, in selected autonomic brain regions, in rhesus monkeys that had self-administered cocaine (0.3 mg/kg/infusion) for 5 days (initial) or 100 days (chronic). Measurements were compared with control monkeys, in which responding was maintained by food reinforcement. In general, decreased rates of glucose utilization were observed in targeted areas following both 5 and 100 days of cocaine self-administration compared to control values. However, after initial stages of cocaine exposure, significant reductions in the forebrain were restricted to the bed nucleus of stria terminalis and in the brainstem to the nucleus tractus solitarius and dorsomotor nucleus of the vagus nerve. The pattern of significantly altered functional activity induced by chronic 100-day cocaine self-administration extended within the forebrain to include the paraventricular hypothalamic nucleus, and in the brainstem to include additional autonomic-related nuclei, the nucleus ambiguus and locus coeruleus. These results suggest that even at the initial stages of cocaine self-administration, functional changes in activity occur in autonomic and reward-related brain regions. These alterations progress with prolonged cocaine exposure, and therefore may be involved in mediating changes in central autonomic control and the neuroadaptations reported to result from chronic drug abuse.


Subject(s)
Brain , Cardiovascular Physiological Phenomena/drug effects , Cocaine/administration & dosage , Vasoconstrictor Agents/administration & dosage , Animals , Behavior, Animal , Brain/anatomy & histology , Brain/drug effects , Brain/physiology , Carbon Isotopes/pharmacokinetics , Densitometry , Deoxyglucose/pharmacokinetics , Drug Administration Schedule , Macaca mulatta , Male , Reinforcement, Psychology , Self Administration/methods
SELECTION OF CITATIONS
SEARCH DETAIL