Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 100
1.
Transl Psychiatry ; 3: e310, 2013 Oct 08.
Article En | MEDLINE | ID: mdl-24105441

The dynorphin opioid peptides control glutamate neurotransmission in the hippocampus. Alcohol-induced dysregulation of this circuit may lead to impairments in spatial learning and memory. This study examines whether changes in the hippocampal dynorphin and glutamate systems are related, and contribute to impairment of spatial learning and memory in a rat model of cognitive deficit associated with alcohol binge drinking. Hippocampal dynorphins (radioimmunoassay) and glutamate (in vivo microdialysis) were analyzed in Wistar rats exposed to repeated moderate-dose ethanol bouts that impair spatial learning and memory in the Water Maze Task (WMT). The highly selective, long-acting κ-opioid receptor (KOR) antagonist nor-binaltorphimine (nor-BNI) was administered systemically or into the hippocampal CA3 region to test a role of dynorphins in alcohol-induced dysregulations in glutamate neurotransmission and behavior in the WMT. The ethanol treatment impaired learning and memory, upregulated dynorphins and increased glutamate overflow in the CA3 region. Administration of nor-BNI after cessation of ethanol exposure reversed ethanol-induced changes in glutamate neurotransmission in animals exposed to ethanol and normalized their performance in the WMT. The findings suggest that impairments of spatial learning and memory by binge-like ethanol exposure are mediated through the KOR activation by upregulated dynorphins resulting in elevation in glutamate levels. Selective KOR antagonists may correct alcohol-induced pathological processes, thus representing a novel pharmacotherapy for treating of ethanol-related cognitive deficits.


CA3 Region, Hippocampal/drug effects , Central Nervous System Depressants/pharmacology , Dynorphins/drug effects , Ethanol/pharmacology , Glutamic Acid/drug effects , Memory/drug effects , Animals , CA3 Region, Hippocampal/metabolism , Dynorphins/metabolism , Glutamic Acid/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Learning/drug effects , Learning/physiology , Maze Learning , Memory/physiology , Microdialysis , Naltrexone/analogs & derivatives , Narcotic Antagonists , Radioimmunoassay , Rats , Rats, Wistar , Receptors, Opioid, kappa/antagonists & inhibitors , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , Up-Regulation/drug effects , Up-Regulation/physiology
2.
Cell Mol Life Sci ; 69(6): 857-96, 2012 Mar.
Article En | MEDLINE | ID: mdl-22002579

The dynorphin/κ-opioid receptor system has been implicated in the pathogenesis and pathophysiology of several psychiatric disorders. In the present review, we present evidence indicating a key role for this system in modulating neurotransmission in brain circuits that subserve mood, motivation, and cognitive function. We overview the pharmacology, signaling, post-translational, post-transcriptional, transcriptional, epigenetic and cis regulation of the dynorphin/κ-opioid receptor system, and critically review functional neuroanatomical, neurochemical, and pharmacological evidence, suggesting that alterations in this system may contribute to affective disorders, drug addiction, and schizophrenia. We also overview the dynorphin/κ-opioid receptor system in the genetics of psychiatric disorders and discuss implications of the reviewed material for therapeutics development.


Dynorphins/physiology , Mental Disorders/etiology , Receptors, Opioid, kappa/physiology , Brain/physiology , Central Nervous System Stimulants/pharmacology , Cyclic AMP Response Element-Binding Protein/physiology , Dynorphins/genetics , Enkephalins/genetics , Epigenesis, Genetic , Gene Expression Regulation , Humans , Mental Disorders/drug therapy , Models, Animal , Protein Precursors/genetics , Receptors, Opioid, kappa/agonists , Receptors, Opioid, kappa/genetics , Self Stimulation , Signal Transduction
3.
Eur J Neurosci ; 30(2): 272-8, 2009 Jul.
Article En | MEDLINE | ID: mdl-19614973

Mu opioid receptor (MOR) regulation of somatodendritic dopamine neurotransmission in the ventral tegmental area (VTA) was investigated using conventional microdialysis in freely moving rats and mice. Reverse dialysis of the MOR agonist DAMGO (50 and 100 microm) into the VTA of rats produced a concentration-dependent increase in dialysate dopamine concentrations. Basal dopamine overflow in the VTA was unaltered in mice lacking the MOR gene. However, basal gamma-aminobutyric acid (GABA) overflow in these animals was significantly increased, whereas glutamate overflow was decreased. Intra-VTA perfusion of DAMGO into wild-type (WT) mice increased dopamine overflow. GABA concentrations were decreased, whereas glutamate concentrations in the VTA were unaltered. Consistent with the loss of MOR, no effect of DAMGO was observed in MOR knockout (KO) mice. These data provide the first direct demonstration of tonically active MOR systems in the VTA that regulate basal glutamatergic and GABAergic neurotransmission in this region. We hypothesize that increased GABAergic neurotransmission following constitutive deletion of MOR is due to the elimination of a tonic inhibitory influence of MOR on GABAergic neurons in the VTA, whereas decreased glutamatergic neurotransmission in MOR KO mice is a consequence of intensified GABA tone on glutamatergic neurons and/or terminals. As a consequence, somatodendritic dopamine release is unaltered. Furthermore, MOR KO mice do not exhibit the positive correlation between basal dopamine levels and the glutamate/GABA ratio observed in WT mice. Together, our findings indicate a critical role of VTA MOR in maintaining an intricate balance between excitatory and inhibitory inputs to dopaminergic neurons.


Dendrites/physiology , Dopamine/metabolism , Glutamic Acid/physiology , Receptors, Opioid, mu/physiology , gamma-Aminobutyric Acid/physiology , Animals , Dendrites/drug effects , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats , Rats, Sprague-Dawley , Receptors, Opioid, mu/agonists , Somatosensory Cortex/drug effects , Somatosensory Cortex/physiology , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/physiology
4.
Neuropsychopharmacology ; 34(4): 887-98, 2009 Mar.
Article En | MEDLINE | ID: mdl-18704097

Studies in experimental animals have shown that individuals exhibiting enhanced sensitivity to the locomotor-activating and rewarding properties of drugs of abuse are at increased risk for the development of compulsive drug-seeking behavior. The purpose of the present study was to assess the effect of constitutive deletion of delta-opioid receptors (DOPr) on the rewarding properties of morphine as well as on the development of sensitization and tolerance to the locomotor-activating effects of morphine. Locomotor activity testing revealed that mice lacking DOPr exhibit an augmentation of context-dependent sensitization following repeated, alternate injections of morphine (20 mg/kg; s.c.; 5 days). In contrast, the development of tolerance to the locomotor-activating effects of morphine following chronic morphine administration (morphine pellet: 25 mg: 3 days) is reduced relative to WT mice. The conditioned rewarding effects of morphine were reduced significantly in DOPrKO mice as compared to WT controls. Similar findings were obtained in response to pharmacological inactivation of DOPr in WT mice, indicating that observed effects are not due to developmental adaptations that occur as a consequence of constitutive deletion of DOPr. Together, these findings indicate that the endogenous DOPr system is recruited in response to both repeated and chronic morphine administration and that this recruitment serves an essential function in the development of tolerance, behavioral sensitization, and the conditioning of opiate reward. Importantly, they demonstrate that DOPr has a distinct role in the development of each of these drug-induced adaptations. The anti-rewarding and tolerance-reducing properties of DOPr antagonists may offer new opportunities for the treatment and prevention of opioid dependence as well as for the development of effective analgesics with reduced abuse liability.


Drug Tolerance/physiology , Morphine/pharmacology , Motor Activity/drug effects , Receptors, Opioid, delta/metabolism , Reward , Animals , Conditioning, Psychological/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Receptors, Opioid, delta/antagonists & inhibitors , Spatial Behavior/drug effects
5.
CNS Neurol Disord Drug Targets ; 7(5): 442-53, 2008 Nov.
Article En | MEDLINE | ID: mdl-19128202

Drug addiction is a chronic, relapsing disorder that is characterized by a compulsion to take drug regardless of the adverse consequences that may ensue. Although the involvement of mesoaccumbal dopamine neurons in the initiation of drug abuse is well-established, neuroadaptations within the limbic cortical- striatopallidal circuit that occur as a consequence of repeated drug use are thought to lead to the behavioral dysregulation that characterizes addiction. Opioid receptors and their endogenous ligands are enriched in brain regions comprising this system and are, thus, strategically located to modulate neurotransmission therein. This article will review data suggesting an important role of mu-opioid receptor (MOPr) and delta opioid receptor (DOPr) systems in mediating the rewarding effects of several classes of abused drugs and that aberrant activity of these opioid systems may not only contribute to the behavioral dysregulation that characterizes addiction but to individual differences in addiction vulnerability.


Brain/metabolism , Opioid Peptides/metabolism , Receptors, Opioid, delta/metabolism , Receptors, Opioid, mu/metabolism , Substance-Related Disorders/metabolism , Animals , Brain/drug effects , Brain/physiopathology , Humans , Limbic System/drug effects , Limbic System/metabolism , Limbic System/physiopathology , Narcotics/pharmacology , Neural Pathways/drug effects , Neural Pathways/metabolism , Neural Pathways/physiopathology , Receptors, Opioid, delta/drug effects , Receptors, Opioid, mu/drug effects , Reward , Substance-Related Disorders/drug therapy , Substance-Related Disorders/physiopathology
6.
Pharmacol Ther ; 116(2): 306-21, 2007 Nov.
Article En | MEDLINE | ID: mdl-17868902

Drug addiction is a chronic relapsing disease in which drug administration becomes the primary stimulus that drives behavior regardless of the adverse consequence that may ensue. As drug use becomes more compulsive, motivation for natural rewards that normally drive behavior decreases. The discontinuation of drug use is associated with somatic signs of withdrawal, dysphoria, anxiety, and anhedonia. These consequences of drug use are thought to contribute to the maintenance of drug use and to the reinstatement of compulsive drug use that occurs during the early phase of abstinence. Even, however, after prolonged periods of abstinence, 80-90% of human addicts relapse to addiction, suggesting that repeated drug use produces enduring changes in brain circuits that subserve incentive motivation and stimulus-response (habit) learning. A major goal of addiction research is the identification of the neural mechanisms by which drugs of abuse produce these effects. This article will review data showing that the dynorphin/kappa-opioid receptor (KOPr) system serves an essential function in opposing alterations in behavior and brain neurochemistry that occur as a consequence of repeated drug use and that aberrant activity of this system may not only contribute to the dysregulation of behavior that characterizes addiction but to individual differences in vulnerability to the pharmacological actions of cocaine and alcohol. We will provide evidence that the repeated administration of cocaine and alcohol up-regulates the dynorphin/KOPr system and that pharmacological treatments that target this system may prove effective in the treatment of drug addiction.


Dynorphins/metabolism , Receptors, Opioid, kappa/metabolism , Substance-Related Disorders/physiopathology , Alcoholism/physiopathology , Animals , Behavior, Addictive/physiopathology , Brain/metabolism , Cocaine/pharmacology , Cocaine/toxicity , Cocaine-Related Disorders/physiopathology , Ethanol/pharmacology , Ethanol/toxicity , Humans , Up-Regulation
7.
J Neurochem ; 99(3): 892-9, 2006 Nov.
Article En | MEDLINE | ID: mdl-16925594

A deficit in the serotonin 5-HT(1A) receptor has been found in panic and post-traumatic stress disorders, and genetic inactivation of the receptor results in an anxiety-like phenotype in mice on both the C57Bl6 and Swiss-Webster genetic backgrounds. Anxiety is associated with increased neuronal activity in the prefrontal cortex and here we describe changes in glutamate and GABA uptake of C57Bl6 receptor null mice. Although these alterations were not present in Swiss-Webster null mice, we have previously reported reductions in GABA(A) receptor expression in these but not in C57Bl6 null mice. This demonstrates that inactivation of the 5-HT(1A) receptor elicits different and genetic background-dependent perturbations in the prefrontal cortex GABA/glutamate system. These perturbations can result in a change in the balance between excitation and inhibition, and indeed both C57Bl6 and Swiss-Webster null mice show signs of increased neuronal excitability. Because neuronal activity in the prefrontal cortex controls the extent of response to anxiogenic stimuli, the genetic background-specific perturbations in glutamate and GABA neurotransmission in C57Bl6 and Swiss-Webster 5-HT(1A) receptor null mice may contribute to their shared anxiety phenotype. Our study shows that multiple strains of genetically altered mice could help us to understand the common and individual features of anxiety.


Anxiety/genetics , Anxiety/psychology , Glutamic Acid/physiology , Prefrontal Cortex/metabolism , Receptor, Serotonin, 5-HT1A/genetics , Receptor, Serotonin, 5-HT1A/physiology , gamma-Aminobutyric Acid/physiology , Animals , Behavior, Animal/physiology , Blotting, Western , Chromatography, High Pressure Liquid , Excitatory Amino Acid Transporter 3/metabolism , Genes, fos/genetics , Immunohistochemistry , In Vitro Techniques , Mice , Mice, Inbred C57BL , Mice, Knockout , Microdialysis , Phenotype , Potassium Chloride/pharmacology , Sodium/physiology , Stress, Psychological/metabolism , Stress, Psychological/psychology , Synaptosomes/drug effects , Synaptosomes/metabolism , gamma-Aminobutyric Acid/metabolism
8.
Eur J Neurosci ; 23(1): 229-38, 2006 Jan.
Article En | MEDLINE | ID: mdl-16420432

Quantitative and conventional microdialysis were used to investigate the effects of constitutive deletion of the prodynorphin gene on basal dopamine (DA) dynamics in the nucleus accumbens (NAc) and the responsiveness of DA neurons to an acute cocaine challenge. Saline- and cocaine-evoked locomotor activity were also assessed. Quantitative microdialysis revealed that basal extracellular DA levels were decreased, while the DA extraction fraction, an indirect measure of DA uptake, was unchanged in dynorphin (DYN) knockout (KO) mice. The ability of cocaine to increase NAc DA levels was reduced in KO. Similarly, cocaine-evoked locomotor activity was decreased in KO. The selective kappa opioid receptor agonist U-69593 decreased NAc dialysate DA levels in wildtype mice and this effect was enhanced in KO. Administration of the selective kappa opioid receptor (KOPr) antagonist nor-binaltorphimine to KO mice attenuated the decrease in cocaine-induced DA levels. However, it was ineffective in altering the decreased locomotor response to cocaine. These studies demonstrate that constitutive deletion of prodynorphin is associated with a reduction of extracellular NAc DA levels and a decreased responsiveness to acute cocaine. Data regarding the effects of U-69593 and nor-binaltorphimine in KO suggest that the kappa opioid receptor is up-regulated as a consequence of prodynorphin gene deletion and that this adaptation underlies the decrease in basal DA dynamics and cocaine-evoked DA levels observed in DYN KO mice. These findings suggest that the phenotype of DYN KO mice is not solely due to loss of endogenous opioid peptide but also reflects developmental compensations that occur at the level of the opioid receptor.


Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Dopamine/metabolism , Dynorphins/deficiency , Nucleus Accumbens/drug effects , Analgesics/pharmacology , Animals , Area Under Curve , Behavior, Animal , Benzeneacetamides/pharmacology , Chromatography, High Pressure Liquid/methods , Dose-Response Relationship, Drug , Drug Interactions , Electrochemistry/methods , Mice , Mice, Inbred C57BL , Mice, Knockout , Microdialysis/methods , Motor Activity/drug effects , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Nucleus Accumbens/physiology , Pyrrolidines/pharmacology
9.
Neuroscience ; 127(2): 497-503, 2004.
Article En | MEDLINE | ID: mdl-15262338

Conventional brain microdialysis was used to assess basal and cocaine-induced dopamine (DA) levels in the nucleus accumbens of wildtype (WT) C57BL/6J mice and mice with constitutive deletion of ether mu- or delta-opioid receptors (MOR or DOR knockout [KO], respectively). Locomotor activity was assessed in these same animals. Basal locomotor activity of DOR KO was elevated relative to MOR KO, but did not differ from that of WT mice. DOR mice, but not WT or MOR KO, exhibited a significant increase in activity in response to an injection of saline. The acute administration of cocaine produced a dose-related increase in locomotor activity in the three genotypes. The locomotor activating effects of a low dose (10 mg/kg) of cocaine were enhanced in DOR KO mice whereas the locomotor activating effects of both a low and higher (20 mg/kg) dose of cocaine were reduced in MOR KO animals. Microdialysis studies revealed no difference between genotypes in basal DA levels. Acute administration of cocaine, but not saline, increased DA levels in WT and KO animals. Paradoxically, however, the magnitude of this effect was smaller in DOR KO as compared with that in either WT or MOR KO. These data indicate that constitutive deletion of either MOR or DOR results in contrasting effects upon responsiveness to cocaine, which is consistent with the distinct phenotypes previously described for these mutants.


Brain Chemistry/drug effects , Brain/drug effects , Cocaine-Related Disorders/metabolism , Cocaine/pharmacology , Receptors, Opioid, delta/physiology , Receptors, Opioid, mu/physiology , Animals , Brain/metabolism , Brain/physiopathology , Brain Chemistry/physiology , Cocaine-Related Disorders/genetics , Cocaine-Related Disorders/physiopathology , Dopamine/metabolism , Dose-Response Relationship, Drug , Extracellular Fluid/drug effects , Extracellular Fluid/metabolism , Genotype , Mice , Mice, Knockout , Microdialysis , Motor Activity/drug effects , Motor Activity/physiology , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Opioid Peptides/metabolism , Receptors, Opioid, delta/deficiency , Receptors, Opioid, delta/genetics , Receptors, Opioid, mu/deficiency , Receptors, Opioid, mu/genetics
10.
Brain Res ; 975(1-2): 244-7, 2003 Jun 13.
Article En | MEDLINE | ID: mdl-12763614

Behavioral immobility during forced swim is usually considered a consequence of inescapable stress, and is used to screen antidepressant drugs. However, immobility in this test may also result from inhibition of neural functions because of brain hypothermia due to body cooling. To explore this possibility, we measured brain temperature dynamics during a 10-min forced swim in cold (25 degrees C) and warm (37 degrees C) water and correlated brain temperatures with behavioral changes. Cold water forced swim resulted in significant brain hypothermia (-6-7 degrees C) and immobility, while no immobility was observed during warm water forced swim, when brain temperature transiently increased (0.5 degrees C) then decreased below baseline in the post-swim period. These data suggest that immobility, which rapidly develops during forced swim in cold water, may result from dramatic inhibition of neural functions because of severe brain hypothermia.


Behavior, Animal/physiology , Brain/physiopathology , Hypothermia/psychology , Motor Activity/physiology , Swimming/physiology , Swimming/psychology , Animals , Brain Chemistry/physiology , Calcium Channels/physiology , Cold Temperature , Dopamine/metabolism , Hypothermia/physiopathology , Male , Mice , Neostriatum/metabolism , Neostriatum/physiology , Sodium Channels/physiology
11.
J Neurochem ; 81(5): 1035-42, 2002 Jun.
Article En | MEDLINE | ID: mdl-12065616

An involvement of the D(3) dopamine receptor in the regulation of extracellular dopamine has been suggested. However, the mechanisms mediating this effect are unclear. We have used the technique of no net flux microdialysis under transient conditions to examine the influence of the D(3) -preferring agonist (+)-PD128907 upon extracellular dopamine levels in the nucleus accumbens of the mouse. (+)-PD 128907 (0.1 mg/kg intraperitoneally) significantly decreased extracellular dopamine. This decrease was associated with a marked increase in the extraction fraction, which suggests an increase in dopamine clearance. The ability of D(3) -preferring compounds to modulate dopamine uptake was investigated in vitro using rotating disk electrode voltammetry. (+)-PD 128907 (10 nm) significantly increased the initial clearance rate of 3 microm dopamine in rat nucleus accumbens tissue suspensions. Kinetic analysis revealed no change in the apparent K (m) of uptake but it showed a 33% increase in V (max). In contrast, the D(3) antagonist GR 103691 (10 nm) significantly decreased dopamine uptake. Consistent with the low levels of D(3) receptors in the dorsal striatum, neither compound affected uptake in tissue suspensions from this brain region. These data indicate that D(3) receptor activation increases dopamine uptake in the nucleus accumbens and suggest that this receptor subtype can regulate extracellular dopamine by modulating the DA transporter activity.


Dopamine/metabolism , Extracellular Space/metabolism , Nucleus Accumbens/metabolism , Receptors, Dopamine D2/metabolism , Animals , Benzopyrans/pharmacology , Biphenyl Compounds/pharmacology , Dopamine/analysis , Dopamine/pharmacokinetics , Dopamine Agonists/pharmacology , Dopamine Antagonists/pharmacology , Dopamine D2 Receptor Antagonists , Dose-Response Relationship, Drug , Electrochemistry , Extracellular Space/chemistry , In Vitro Techniques , Injections, Intraperitoneal , Ligands , Male , Mice , Microdialysis , Nucleus Accumbens/drug effects , Oxazines/pharmacology , Piperazines/pharmacology , Receptors, Dopamine D2/agonists , Receptors, Dopamine D3
12.
Neuroscience ; 112(4): 907-19, 2002.
Article En | MEDLINE | ID: mdl-12088750

Despite an abundance of studies on mechanisms of behavioral sensitization, considerable uncertainty exists as to whether alterations in dopamine neurotransmission underlie the exacerbated behavioral effects of cocaine observed during the early stages of abstinence. One of the factors contributing to the uncertainty and controversy may be the limitations in utilized measurement techniques (mostly conventional microdialysis). The techniques of quantitative microdialysis under transient conditions and rotating disk electrode voltammetry were used to characterize basal dopamine dynamics as well as time-related changes in extracellular dopamine concentrations and dopamine uptake that occur in response to an acute drug challenge in control animals and animals with previous history of cocaine. Basal extracellular dopamine concentrations were unaltered on abstinence day 3 from repeated cocaine administration (5 days, 20 mg/kg, i.p.). The extraction fraction of dopamine, an indirect measure of dopamine uptake, was significantly lower in cocaine-sensitized animals relative to controls. These two facts, taken together, suggest that basal dopamine release is depressed in cocaine-sensitized animals on abstinence day 3. At the same time, a cocaine challenge decreased the extraction fraction and increased the extracellular dopamine concentration in both experimental groups. The magnitude of the increase in extracellular dopamine concentration was greater in cocaine-sensitized animals, while the ability of cocaine to decrease the extraction fraction was unaltered, suggesting that the increase in extracellular dopamine concentration reflects an increase in drug-evoked dopamine release. Moreover, cocaine-pretreated rats demonstrated greater depolarization-induced dopamine release and the ability of dopamine D(2) receptor agonist, quinpirole, to inhibit release was decreased in these animals. These data demonstrate that a cocaine treatment regimen resulting in behavioral sensitization is associated with a reduction in basal dopamine release, an enhancement in both cocaine and K(+)-evoked dopamine release, and a subsensitivity of dopamine D(2) autoreceptors that regulate dopamine release in the nucleus accumbens.


Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Dopamine/metabolism , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Substance Withdrawal Syndrome/metabolism , Animals , Cocaine/metabolism , Dopamine Uptake Inhibitors/metabolism , Extracellular Space/metabolism , Illicit Drugs/pharmacology , Male , Microdialysis , Rats , Rats, Sprague-Dawley , Time Factors
13.
Neuropharmacology ; 41(3): 351-9, 2001 Sep.
Article En | MEDLINE | ID: mdl-11522326

An involvement of the D3 dopamine receptor in the modulation of extracellular dopamine concentrations is suggested by pharmacological studies. However, recent studies using D3 receptor knock out mice indicated that several functions previously attributed to the D3 receptor are mediated by other receptor types. In the present study, we used the no-net flux microdialysis technique to characterize: (i) basal dopamine dynamics in the ventral striatum of D3 knock out and wild type mice and (ii) the effects of the putative D3-receptor selective agonist (+)-PD 128907. Neither the extracellular dopamine concentration nor the in vivo extraction fraction, an indirect measure of basal dopamine uptake, differed between D3 knock out and wild type mice. Moreover, no differences in potassium (60 mM) or cocaine (5 or 20 mg/kg i.p.) evoked dopamine concentrations were detected between the two genotypes. However, intra-striatal or systemic administration of doses of (+)-PD 128907 that failed to modify dopamine concentrations in knock out mice significantly decreased dialysate dopamine concentrations in the wild type. Comparison of the concentration-response curve for (+)-PD 128907 revealed IC(25) values of 61 and 1327 nM in wild type and knock out mice, respectively, after intra-striatal infusions. Similar differences were obtained after systemic administration of the D3 preferring agonist (IC(25) 0.05 and 0.44 mg/kg i.p. in wild type and knock out mice, respectively). We conclude that the activation of the D3 receptor decreases extracellular dopamine levels and that, at sufficiently low doses, the effects of (+)-PD 128907 on extracellular dopamine are selectively mediated by the D3 receptor.


Benzopyrans/pharmacology , Dopamine Agonists/pharmacology , Dopamine/metabolism , Oxazines/pharmacology , Receptors, Dopamine D2/agonists , Animals , Brain/anatomy & histology , Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Kinetics , Male , Mice , Mice, Knockout , Microdialysis , Potassium/pharmacology , Receptors, Dopamine D2/genetics , Receptors, Dopamine D3
14.
Ann N Y Acad Sci ; 937: 50-73, 2001 Jun.
Article En | MEDLINE | ID: mdl-11458540

The repeated, intermittent use of cocaine and other drugs of abuse produces profound and often long-lasting alterations in behavior and brain chemistry. It has been suggested that these consequences of drug use play a critical role in drug craving and relapse to addiction. This article reviews the effects of psychostimulant administration on dopaminergic and excitatory amino acid neurotransmission in brain regions comprising the brain's motive circuit and provides evidence that the activation of endogenous kappa-opioid receptor systems in these regions opposes the behavioral and neurochemical consequences of repeated drug use. The role of this opioid system in mediating alterations in mood and affect that occur during abstinence from repeated psychostimulant use are also discussed.


Amphetamine/pharmacology , Cocaine-Related Disorders/physiopathology , Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Receptors, Opioid, kappa/drug effects , Receptors, Opioid, kappa/physiology , Adaptation, Psychological , Affect , Animals , Basal Ganglia/drug effects , Basal Ganglia/physiology , Brain Chemistry/drug effects , Humans , Ligands , Receptors, Dopamine/drug effects , Receptors, Dopamine/physiology , Thalamic Nuclei/drug effects , Thalamic Nuclei/physiology
15.
Psychopharmacology (Berl) ; 155(1): 35-42, 2001 Apr.
Article En | MEDLINE | ID: mdl-11374334

RATIONALE: Since cues that predict aversive outcomes can elicit both avoidance and 20 kHz ultrasonic vocalizations (USVs) in adult rats, 20 kHz USVs may also index the conditioned aversive effects of drugs. OBJECTIVE: We evaluated whether exposure to compartments associated with drugs with aversive effects would selectively increase 20 but not 50 kHz USVs in rats. METHOD: Rats were injected with naloxone (NAL) or lithium chloride (LiCl) and placed in one compartment or with saline (VEH) and placed in another compartment for three 50-min conditioning sessions. 20 kHz USVs, 50 kHz USVs, and time spent in each chamber were recorded during subsequent 15-min testing sessions during which rats had access to both compartments (expt 1) or were confined to the drug- or VEH-paired compartment (expt 2). RESULTS: In expt 1, animals conditioned either with NAL (0.3 and 3.0 mg/kg) or LiCl (10 and 30 mg/kg) emitted increased 20 kHz USVs in the drug-paired compartment, relative to VEH-conditioned controls. Conditioning with high doses of both drugs also increased conditioned place aversion and decreased emission of 50 kHz USVs. In expt 2, restriction of animals to the compartment paired with high doses of NAL and LiCl also increased emission of 20 kHz USVs and decreased 50 kHz USVs, relative to VEH-conditioned controls. CONCLUSIONS: In rats, cues associated with drugs with aversive effects increase 20 kHz USVs and decrease 50 kHz USVs, suggesting that USVs may provide a useful model for predicting the conditioned aversive effects of drugs.


Antimanic Agents/pharmacology , Conditioning, Psychological/drug effects , Lithium Chloride/pharmacology , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Ultrasonics , Vocalization, Animal/drug effects , Analgesics, Opioid/pharmacology , Animals , Conditioning, Psychological/physiology , Cues , Male , Morphine/pharmacology , Rats , Rats, Sprague-Dawley , Vocalization, Animal/physiology
16.
Brain Res ; 902(1): 11-7, 2001 May 25.
Article En | MEDLINE | ID: mdl-11376590

An involvement of serotonin (5-HT) 1A receptors in the etiology of psychiatric disorders has been suggested. Hypo-responsiveness of the 5-HT1A receptor is linked to anxiety and constitutive deletion of the 5-HT1A receptor produces anxiety-like behaviors in the mouse. Evidence that 5-HT1A receptor inactivation increases the therapeutic effects of antidepressants has also been presented. The present studies used in vivo microdialysis and homologous recombination techniques to examine the contribution of 5-HT1A autoreceptors to these effects. Basal and fluoxetine-evoked extracellular concentrations of 5-HT were quantified in the striatum, a projection area of dorsal raphe neurons (DRN), of wild-type (WT) and 5-HT1A receptor knock out (KO) mice. The density of 5-HT transporters was also determined. Basal 5-HT concentrations did not differ in WT and KO mice. Fluoxetine (10 mg/kg) increased 5-HT concentrations in both genotypes. This increase was, however, 2-fold greater in KO mice. In contrast, no differences in K(+)-evoked 5-HT concentrations were seen. Similarly, neither basal nor stimulation-evoked DA differed across genotype. Autoradiography revealed no differences between genotype in the density of 5-HT transporters or post-synaptic 5-HT2A receptors, an index of 5-HT neuronal activity. These experiments demonstrate that, under basal and KCl stimulated conditions, adaptive mechanisms in the 5-HT system compensate for the lack of 5-HT1A autoreceptor regulation of DRN. Furthermore, they suggest that the absence of release-regulating 5-HT1A autoreceptors in the DRN can not account for the anxiety phenotype of KO mice. The enhanced response to fluoxetine in KO mice is consistent with pharmacological studies and suggests that adaptive mechanisms that occur in response to 5-HT1A receptor deletion are insufficient to oppose increases in 5-HT concentrations produced by acute inhibition of the 5-HT transporter.


Brain/drug effects , Fluoxetine/pharmacology , Membrane Transport Proteins , Nerve Tissue Proteins/physiology , Receptors, Presynaptic/physiology , Receptors, Serotonin/physiology , Selective Serotonin Reuptake Inhibitors/pharmacology , Animals , Brain/metabolism , Carrier Proteins/metabolism , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Dopamine/metabolism , Haloperidol/pharmacology , Male , Membrane Glycoproteins/metabolism , Mice , Mice, Knockout , Microdialysis , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Receptors, Presynaptic/deficiency , Receptors, Presynaptic/genetics , Receptors, Serotonin/deficiency , Receptors, Serotonin/genetics , Receptors, Serotonin, 5-HT1 , Serotonin Plasma Membrane Transport Proteins , Spiperone/pharmacology
17.
Neuropsychopharmacology ; 24(4): 441-50, 2001 Apr.
Article En | MEDLINE | ID: mdl-11182539

Previous studies showed that prior administration of kappa-opioid agonists decreased the development of sensitization to some of the behavioral effects of cocaine. The present study sought to determine whether the development of sensitization to cocaine's reinforcing effects was also sensitive to antagonism by kappa-opioid agonists. During a pretreatment phase, the kappa-opioid agonist, U69593 (0.0 or 0.32 mg/kg) was administered prior to (1) 2 daily injections of cocaine (0.0 or 20.0 mg/kg), or (2) cocaine or saline administered via a yoking procedure. Cocaine pretreatment decreased the latency to acquisition of cocaine self-administration. However, prior administration of U69593 during the pretreatment phase failed to attenuate the development of this sensitized response to cocaine's reinforcing effect. In other groups, the effect of acute U69593 pretreatment on the maintenance of cocaine self-administration was examined during a 10 hr session. During training and testing, a stimulus was associated with each self-administered cocaine infusion for one group whereas responding of another group was reinforced by a cocaine infusion alone. On the test day, pretreatment with U69593 (0.32 mg/kg) decreased responding during each hour of the 10 hr session for the group that was reinforced with cocaine plus the cocaine-associated stimulus. U69593 failed to produce a long-lasting disruption of cocaine self-administration for rats that were trained and tested without the cocaine-associated stimulus. These data suggest that the acquisition and maintenance of cocaine self-administration are differentially sensitive to manipulations of kappa-opioid systems. Further, the disruption of cocaine self-administration by U69593 may be due to interactions with mechanisms that underlie facilitative effects of stimuli that have been associated with self-administered cocaine infusions.


Behavior, Addictive/metabolism , Benzeneacetamides , Cocaine-Related Disorders/metabolism , Cocaine/administration & dosage , Pyrrolidines/administration & dosage , Receptors, Opioid, kappa/agonists , Animals , Behavior, Animal/drug effects , Conditioning, Psychological/drug effects , Disease Models, Animal , Drug Administration Schedule , Injections, Intravenous , Male , Photic Stimulation , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Reinforcement, Psychology , Self Administration
18.
Synapse ; 39(4): 343-50, 2001 Mar 15.
Article En | MEDLINE | ID: mdl-11169785

The repeated administration of selective kappa-opioid receptor agonists prevents the locomotor activation produced by acute cocaine administration and the development of cocaine-induced behavioral sensitization. Previous studies have shown that dopamine (DA) D2 autoreceptors modulate the synthesis and release of DA in the striatum. Evidence that kappa agonist treatment downregulates DA D2 receptors in this same brain region has recently been obtained. Accordingly, the present studies were undertaken to examine the influence of repeated kappa-opioid receptor agonist administration on pre- and postsynaptic DA D2 receptor function in the dorsal striatum using pre- and postsynaptic receptor-selective doses of quinpirole. Rats were injected once daily with the selective kappa-opioid receptor agonist U69593 (0.16-0.32 mg/kg s.c.) or vehicle for 3 days. Microdialysis studies assessing basal and quinpirole-evoked (0.05 mg/kg s.c.) DA levels were conducted 2 days later. Basal and quinpirole-stimulated locomotor activity were assessed in a parallel group of animals. The no-net flux method of quantitative microdialysis revealed no effect of U69593 on basal DA dynamics, in that extracellular DA concentration and extraction fraction did not differ in control and U69593-treated animals. Acute administration of quinpirole significantly decreased striatal DA levels in control animals, but in animals treated with U69593, the inhibitory effects of quinpirole were significantly reduced. Quinpirole produced a dose-related increase in locomotor activity in control animals, and this effect was significantly attenuated in U69593-treated animals. These data reveal that prior repeated administration of a selective kappa-opioid receptor agonist attenuates quinpirole-induced alterations in DA neurotransmission and locomotor activity. These results suggest that both pre- and postsynaptic striatal DA D2 receptors may be downregulated following repeated kappa-opioid receptor agonist administration. Synapse 39:343-350, 2001. Published 2001 Wiley-Liss, Inc.


Benzeneacetamides , Dopamine D2 Receptor Antagonists , Pyrrolidines/pharmacology , Receptors, Opioid, kappa/agonists , Animals , Autoreceptors/antagonists & inhibitors , Dopamine/metabolism , Dopamine Agonists/administration & dosage , Dopamine Agonists/pharmacology , Down-Regulation , Male , Microdialysis , Motor Activity/drug effects , Neostriatum/drug effects , Neostriatum/metabolism , Pyrrolidines/administration & dosage , Quinpirole/administration & dosage , Quinpirole/pharmacology , Rats , Rats, Sprague-Dawley , Synaptic Transmission/drug effects
19.
Curr Protoc Neurosci ; Chapter 7: Unit7.1, 2001 May.
Article En | MEDLINE | ID: mdl-18428520

The technique of microdialysis enables the monitoring of neurotransmitters and other molecules in the extracellular environment. This method has undergone several modifications and is now widely used for sampling and quantitating neurotransmitters, neuropeptides, and hormones in the brain and periphery. This unit describes the principles of conventional and quantitative microdialysis as well as strategies in designing a dialysis experiment. It establishes the groundwork for the basic techniques of preparation, conduct, and analysis of dialysis experiments in rodents and subhuman primates. Although the methods described are those used for monitoring CNS function, they can be easily applied with minor modification to other organ systems.


Microdialysis , Animal Structures/chemistry , Animals , Columbidae , Diffusion , Equipment Design , Humans , Inorganic Chemicals/analysis , Mammals , Mathematics , Microdialysis/instrumentation , Microdialysis/methods , Oncorhynchus mykiss , Organ Specificity , Organic Chemicals/analysis , Research Design
20.
Curr Protoc Neurosci ; Chapter 7: Unit7.2, 2001 May.
Article En | MEDLINE | ID: mdl-18428530

Microdialysis is an established sampling technique for the in vivo measurement of a variety of substances in both blood and tissue. These include neurotransmitters and neuropeptides, enzymes, and electrolytes, as well as various hormones and pharmaceutical agents. More recently, microdialysis has been used to infuse exogenous as well as endogenous substances into the brain and spinal cord. In microdialysis, a semipermeable dialysis membrane is introduced into the fluid or tissue compartment to be sampled and perfused with physiological fluid. As a result of passive diffusion, molecules migrate across the membrane along their concentration gradient. Molecules found in high concentrations within the tissue compartment migrate across the membrane into the dialysis tubing where they can be collected for subsequent quantification, whereas molecules found in high concentrations within the membrane diffuse outward into the surrounding tissue compartment. This unit describes construction of concentric and side-by-side microdialysis probes, which differ with respect to size and inflow/outflow tube compositions, as well as a modification of a horizontal probe. This unit also covers methods for surgically implanting microdialysis probes in rats and mice and for conducting microdialysis in vitro and in vivo in rodents. Finally, procedures are provided for carrying out quantitative microdialysis techniques.


Microdialysis/methods , Rats/metabolism , Animals , Brain Chemistry , Catheterization , Equipment Design , Implants, Experimental , Mice , Microdialysis/instrumentation , Stereotaxic Techniques
...