Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Eur J Neurosci ; 44(7): 2446-2454, 2016 10.
Article in English | MEDLINE | ID: mdl-27453431

ABSTRACT

The key problem in treating cocaine addiction is the maintenance of a drug-free state as negative emotional symptoms during abstinence often trigger relapse. The mechanisms underpinning the emotional dysregulation during abstinence are currently not well-understood. There is evidence suggesting a role of the neuropeptide oxytocin in the modulation of drug addiction processes. However, its involvement during long-term abstinence from cocaine use remains unclear. In this study, we aimed to behaviourally characterize a mouse model of long-term cocaine withdrawal and assess the effect of chronic cocaine administration and long-term cocaine abstinence on the central oxytocinergic system and the hypothalamic-pituitary-adrenal axis. Fourteen-day escalating-dose cocaine administration (3 × 15-30 mg/kg/day) and 14-day withdrawal increased plasma corticosterone levels and oxytocin receptor (OTR) binding in piriform cortex, lateral septum and amygdala. A specific cocaine withdrawal-induced increase in OTR binding was observed in the medial septum. These biochemical alterations occurred concomitantly with the emergence of memory impairment, contextual psychomotor sensitization and an anhedonic and anxiogenic phenotype during withdrawal. Our study established a clear relationship between cocaine abstinence and emotional impairment in a novel translationally relevant model of cocaine withdrawal and demonstrated for the first time brain region-specific neuroadaptations of the oxytocin system, which may contribute to abstinence-induced negative emotional state.


Subject(s)
Cocaine-Related Disorders/metabolism , Cocaine/pharmacology , Emotions/drug effects , Hypothalamo-Hypophyseal System/drug effects , Receptors, Oxytocin/metabolism , Animals , Behavior, Animal , Corticosterone/blood , Hypothalamo-Hypophyseal System/metabolism , Male , Mice , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/metabolism , Protein Binding , Substance Withdrawal Syndrome/drug therapy , Substance Withdrawal Syndrome/metabolism , Up-Regulation
2.
Article in English | MEDLINE | ID: mdl-26861145

ABSTRACT

BACKGROUND: A difficult problem in treating opioid addicts is the maintenance of a drug-free state because of the negative emotional symptoms associated with withdrawal, which may trigger relapse. Several lines of evidence suggest a role for the metabotropic glutamate receptor 5 in opioid addiction; however, its involvement during opioid withdrawal is not clear. METHODS: Mice were treated with a 7-day escalating-dose morphine administration paradigm. Following withdrawal, the development of affective behaviors was assessed using the 3-chambered box, open-field, elevated plus-maze and forced-swim tests. Metabotropic glutamate receptor 5 autoradiographic binding was performed in mouse brains undergoing chronic morphine treatment and 7 days withdrawal. Moreover, since there is evidence showing direct effects of opioid drugs on the metabotropic glutamate receptor 5 system, the presence of an metabotropic glutamate receptor 5/µ-opioid receptor interaction was assessed by performing metabotropic glutamate receptor 5 autoradiographic binding in brains of mice lacking the µ-opioid receptor gene. RESULTS: Withdrawal from chronic morphine administration induced anxiety-like, depressive-like, and impaired sociability behaviors concomitant with a marked upregulation of metabotropic glutamate receptor 5 binding. Administration of the metabotropic glutamate receptor 5 antagonist, 3-((2-Methyl-4-thiazolyl)ethynyl)pyridine, reversed morphine abstinence-induced depressive-like behaviors. A brain region-specific increase in metabotropic glutamate receptor 5 binding was observed in the nucleus accumbens shell, thalamus, hypothalamus, and amygdala of µ-opioid receptor knockout mice compared with controls. CONCLUSIONS: These results suggest an association between metabotropic glutamate receptor 5 alterations and the emergence of opioid withdrawal-related affective behaviors. This study supports metabotropic glutamate receptor 5 system as a target for the development of pharmacotherapies for the treatment of opioid addiction. Moreover, our data show direct effects of µ-opioid receptor system manipulation on metabotropic glutamate receptor 5 binding in the brain.


Subject(s)
Emotions/drug effects , Morphine/adverse effects , Receptor, Metabotropic Glutamate 5/metabolism , Receptors, Opioid, mu/genetics , Substance Withdrawal Syndrome/metabolism , Animals , Behavior, Animal/drug effects , Brain/metabolism , Male , Mice , Mice, Knockout , Morphine/pharmacology , Up-Regulation/drug effects
3.
Neuropharmacology ; 105: 520-532, 2016 06.
Article in English | MEDLINE | ID: mdl-26896754

ABSTRACT

The major challenge in treating methamphetamine addicts is the maintenance of a drug free-state since they experience negative emotional symptoms during abstinence, which may trigger relapse. The neuronal mechanisms underlying long-term withdrawal and relapse are currently not well-understood. There is evidence suggesting a role of the oxytocin (OTR), µ-opioid receptor (MOPr), dopamine D2 receptor (D2R), corticotropin-releasing factor (CRF) systems and the hypothalamic-pituitary-adrenal (HPA)-axis in the different stages of methamphetamine addiction. In this study, we aimed to characterize the behavioral effects of methamphetamine withdrawal in mice and to assess the modulation of the OTR, MOPr, D2R, CRF and HPA-axis following chronic methamphetamine administration and withdrawal. Ten-day methamphetamine administration (2 mg/kg) increased OTR binding in the amygdala, whilst 7 days of withdrawal induced an upregulation of this receptor in the lateral septum. Chronic methamphetamine treatment increased plasma OT levels that returned to control levels following withdrawal. In addition, methamphetamine administration and withdrawal increased striatal MOPr binding, as well as c-Fos(+)/CRF(+) neuronal expression in the amygdala, whereas an increase in plasma corticosterone levels was observed following METH administration, but not withdrawal. No differences were observed in the D2R binding following METH administration and withdrawal. The alterations in the OTR, MOPr and CRF systems occurred concomitantly with the emergence of anxiety-related symptoms and the development of psychomotor sensitization during withdrawal. Collectively, our findings indicate that chronic methamphetamine use and abstinence can induce brain-region specific neuroadaptations of the OTR, MOPr and CRF systems, which may, at least, partly explain the withdrawal-related anxiogenic effects.


Subject(s)
Amphetamine-Related Disorders/metabolism , Corticotropin-Releasing Hormone/metabolism , Receptors, Dopamine D2/metabolism , Receptors, Opioid, mu/metabolism , Receptors, Oxytocin/metabolism , Substance Withdrawal Syndrome/metabolism , Amphetamine-Related Disorders/complications , Amphetamine-Related Disorders/pathology , Amygdala/drug effects , Amygdala/metabolism , Amygdala/pathology , Animals , Anxiety/etiology , Anxiety/metabolism , Anxiety/pathology , Central Nervous System Stimulants/administration & dosage , Central Nervous System Stimulants/adverse effects , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/pathology , Corticosterone/blood , Disease Models, Animal , Male , Methamphetamine/administration & dosage , Methamphetamine/adverse effects , Mice, Inbred C57BL , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Proto-Oncogene Proteins c-fos/metabolism , Random Allocation , Septum of Brain/drug effects , Septum of Brain/metabolism , Septum of Brain/pathology , Substance Withdrawal Syndrome/pathology , Substance Withdrawal Syndrome/psychology
4.
Addict Biol ; 21(4): 811-25, 2016 07.
Article in English | MEDLINE | ID: mdl-25975203

ABSTRACT

Addiction to psychostimulants is a major public health problem with no available treatment. Adenosine A2A receptors (A2A R) co-localize with metabotropic glutamate 5 receptors (mGlu5 R) in the striatum and functionally interact to modulate behaviours induced by addictive substances, such as alcohol. Using genetic and pharmacological antagonism of A2A R in mice, we investigated whether A2A R-mGlu5 R interaction can regulate the locomotor, stereotypic and drug-seeking effect of methamphetamine and cocaine, two drugs that exhibit distinct mechanism of action. Genetic deletion of A2A R, as well as combined administration of sub-threshold doses of the selective A2A R antagonist (SCH 58261, 0.01 mg/kg, i.p.) with the mGlu5 R antagonist, 3-((2-methyl-4-thiazolyl)ethynyl)pyridine (0.01 mg/kg, i.p.), prevented methamphetamine- but not cocaine-induced hyperactivity and stereotypic rearing behaviour. This drug combination also prevented methamphetamine-rewarding effects in a conditioned-place preference paradigm. Moreover, mGlu5 R binding was reduced in the nucleus accumbens core of A2A R knockout (KO) mice supporting an interaction between these receptors in a brain region crucial in mediating addiction processes. Chronic methamphetamine, but not cocaine administration, resulted in a significant increase in striatal mGlu5 R binding in wild-type mice, which was absent in the A2A R KO mice. These data are in support of a critical role of striatal A2A R-mGlu5 R functional interaction in mediating the ambulatory, stereotypic and reinforcing effects of methamphetamine but not cocaine-induced hyperlocomotion or stereotypy. The present study highlights a distinct and selective mechanistic role for this receptor interaction in regulating methamphetamine-induced behaviours and suggests that combined antagonism of A2A R and mGlu5 R may represent a novel therapy for methamphetamine addiction.


Subject(s)
Corpus Striatum/drug effects , Drug-Seeking Behavior/drug effects , Methamphetamine/pharmacokinetics , Psychomotor Performance/drug effects , Receptor, Adenosine A2A/drug effects , Receptor, Metabotropic Glutamate 5/drug effects , Animals , Behavior, Animal/drug effects , Central Nervous System Stimulants/pharmacology , Disease Models, Animal , Male , Mice , Mice, Knockout
5.
Eur Neuropsychopharmacol ; 25(12): 2459-64, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26475574

ABSTRACT

Relapse to illicit drug-seeking following abstinence is a major challenge for the treatment of addiction as no effective pharmacotherapy is available. We have recently shown that activating the central oxytocinergic system prevents emotional impairment and stress-induced reinstatement associated with opioid withdrawal. Here, we investigated whether the oxytocin analogue carbetocin (CBT) is able to reverse morphine-primed reinstatement of conditioned-place preference (CPP) in mice. The mechanism underlining the behavioural effect of CBT was investigated by assessing the involvement of the striatal noradrenergic and dopaminergic systems in CBT reversal of priming- and stress-induced reinstatement of opioid CPP. In addition, given recent evidence suggesting the presence of oxytocin receptor (OTR)-µ-opioid receptor (MOPr) interactions in the brain, we further explored these interactions by carrying out OTR autoradiographic binding in brain of mice lacking MOPr. CBT administration prevented priming-induced reinstatement of morphine CPP. While an acute effect of CBT in enhancing dopamine turnover was observed following stress- and priming-induced reinstatement, CBT significantly decreased striatal noradrenaline turnover only following priming-induced reinstatement. Moreover, a significant brain region- specific increase in OTR binding was observed in MOPr knockout mice, indicating the presence of a possible OTR-MOPr interaction, which may be involved in the modulation of relapse. These results support the oxytocinergic system as a promising target for the prevention of relapse to opioid use and highlight the differential involvement of monoaminergic systems on the effects of OTR stimulation in preventing stress- and priming-induced reinstatement of opioid CPP behaviour.


Subject(s)
Carbenicillin/pharmacology , Dopamine/metabolism , Drug-Seeking Behavior/drug effects , Morphine/administration & dosage , Norepinephrine/metabolism , Receptors, Opioid, mu/metabolism , Analysis of Variance , Animals , Conditioning, Operant/drug effects , Corticosterone/blood , Locomotion/drug effects , Male , Mice , Mice, Inbred C57BL , Protein Binding/drug effects , Receptors, Oxytocin/metabolism , Regression Analysis
6.
Addict Biol ; 20(5): 902-12, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25522112

ABSTRACT

The key problem for the treatment of drug addiction is relapse to drug use after abstinence that can be triggered by drug-associated cues, re-exposure to the drug itself and stress. Understanding the neurobiological mechanisms underlying relapse is essential in order to develop effective pharmacotherapies for its prevention. Given the evidence implicating the metabotropic glutamate receptor 5 (mGlu5 R), µ-opioid receptor (MOPr), κ-opioid receptor (ΚOPr) and oxytocin receptor (OTR) systems in cocaine addiction and relapse, our aim was to assess the modulation of these receptors using a mouse model of cue- and priming-induced reinstatement of cocaine seeking. Male mice were trained to self-administer cocaine (1 mg/kg/infusion, i.v.) and were randomized into different groups: (1) cocaine self-administration; (2) cocaine extinction; (3) cocaine-primed (10 mg/kg i.p.); or (4) cue-induced reinstatement of cocaine seeking. Mice undergoing the same protocols but receiving saline instead of cocaine were used as controls. Quantitative autoradiography of mGlu5 R, MOPr, KOPr and OTR showed a persistent cocaine-induced upregulation of the mGlu5 R and OTR in the lateral septum and central amygdala, respectively. Moreover, a downregulation of mGlu5 R and MOPr was observed in the basolateral amygdala and striatum, respectively. Further, we showed that priming- but not cue-induced reinstatement upregulates mGlu5 R and MOPr binding in the nucleus accumbens core and basolateral amygdala, respectively, while cue- but not priming-induced reinstatement downregulates MOPr binding in caudate putamen and nucleus accumbens core. This is the first study to provide direct evidence of reinstatement-induced receptor alterations that are likely to contribute to the neurobiological mechanisms underpinning relapse to cocaine seeking.


Subject(s)
Behavior, Animal/drug effects , Brain/metabolism , Cocaine-Related Disorders/metabolism , Cues , Receptor, Metabotropic Glutamate 5/metabolism , Receptors, Opioid, mu/metabolism , Animals , Autoradiography , Brain/drug effects , Conditioning, Operant , Disease Models, Animal , Male , Mice , Receptor, Metabotropic Glutamate 5/drug effects , Receptors, Opioid, mu/drug effects , Recurrence , Self Administration , Up-Regulation/physiology
7.
PLoS One ; 9(6): e99711, 2014.
Article in English | MEDLINE | ID: mdl-24978193

ABSTRACT

Nucleotides have effects on immune cells which are complex but generally proinflammatory, and have been suggested to play a role in smoking-related lung diseases. However, there have been no studies directly measuring functional responses to nucleotides in human lungs taken from smokers. We used fragments of post mortem human lung from smokers and non-smokers, incubated them with a range of nucleotides (4-1000 µM) in the presence of lipopolysaccharide (LPS; 10 µg/ml) for 24 hours and measured cytokines (IL-1ß, IFNγ, IL-17, TNFα, IL-6, IL-8, IL-2 and IL-10) in the supernatants using multiplex immunoassays. Although the basal cytokine levels in the smokers were generally higher in the smokers than the non-smokers, there were no significant differences in either the basal release or the LPS-stimulated release of any of the cytokines when lungs from smokers and non-smokers were compared. There were no significant effects of ATP, ADP, AMP, UTP, α,ß-methylene-ATP, P1, P4-diATP, 2-methylthio-ATP or Bz-ATP on the release of cytokines from the lungs. However, the stable ATP analogue ATPγS increased the release of IL-1ß and IFNγ, and the effect was greatly increased in lungs from smokers. In non-smokers but not in smokers ATPγS increased the release of IL-17. Overall these results clearly demonstrate for the first time that in normal human lung a stable ATP analogue can enhance LPS-induced pro-inflammatory cytokine release, and that these effects are greatly altered by a prior history of smoking. This provides strong support for the suggestion that nucleotides are involved in the pathogenesis of smoking-related diseases.


Subject(s)
Cytokines/metabolism , Lung/drug effects , Nucleotides/pharmacology , Smoking/adverse effects , Case-Control Studies , Cytokines/genetics , Humans , Lipopolysaccharides/toxicity , Lung/immunology , Lung/metabolism
8.
Pharmacol Biochem Behav ; 119: 72-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23680573

ABSTRACT

There is mounting evidence that the neuropeptide oxytocin is a possible candidate for the treatment of drug addiction. Oxytocin was shown to reduce methamphetamine self-administration, conditioned place-preference, hyperactivity and reinstatement in rodents, highlighting its potential for the management of methamphetamine addiction. Thus, we hypothesised that the central endogenous oxytocinergic system is dysregulated following chronic methamphetamine administration. We tested this hypothesis by examining the effect of chronic methamphetamine administration on oxytocin receptor density in mice brains with the use of quantitative receptor autoradiographic binding. Saline (4ml/kg/day, i.p.) or methamphetamine (1mg/kg/day, i.p.) was administered daily for 10 days to male, CD1 mice. Quantitative autoradiographic mapping of oxytocin receptors was carried out with the use of [(125)I]-vasotocin in brain sections of these animals. Chronic methamphetamine administration induced a region specific upregulation of oxytocin receptor density in the amygdala and hypothalamus, but not in the nucleus accumbens and caudate putamen. As there is evidence suggesting an involvement of central adenosine A2A receptors on central endogenous oxytocinergic function, we investigated whether these methamphetamine-induced oxytocinergic neuroadaptations are mediated via an A2A receptor-dependent mechanism. To test this hypothesis, autoradiographic oxytocin receptor binding was carried out in brain sections of male CD1 mice lacking A2A receptors which were chronically treated with methamphetamine (1mg/kg/day, i.p. for 10 days) or saline. Similar to wild-type animals, chronic methamphetamine administration induced a region-specific upregulation of oxytocin receptor binding in the amygdala and hypothalamus of A2A receptor knockout mice and no genotype effect was observed. These results indicate that chronic methamphetamine use can induce profound neuroadaptations of the oxytocinergic receptor system in brain regions associated with stress, emotionality and social bonding and that these neuroadaptations are independent on the presence of A2A receptors. These results may at least partly explain some of the behavioural consequences of chronic methamphetamine use.


Subject(s)
Amygdala/drug effects , Hypothalamus/drug effects , Methamphetamine/pharmacology , Receptor, Adenosine A2A/metabolism , Receptors, Oxytocin/drug effects , Up-Regulation/drug effects , Amygdala/metabolism , Animals , Female , Hypothalamus/metabolism , Male , Methamphetamine/administration & dosage , Mice , Mice, Knockout , Receptors, Oxytocin/metabolism
9.
Neuropsychopharmacology ; 39(4): 855-65, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24129263

ABSTRACT

The main challenge in treating opioid addicts is to maintain abstinence due to the affective consequences associated with withdrawal which may trigger relapse. Emerging evidence suggests a role of the neurohypophysial peptide oxytocin (OT) in the modulation of mood disorders as well as drug addiction. However, its involvement in the emotional consequences of drug abstinence remains unclear. We investigated the effect of 7-day opioid abstinence on the oxytocinergic system and assessed the effect of the OT analogue carbetocin (CBT) on the emotional consequences of opioid abstinence, as well as relapse. Male C57BL/6J mice were treated with a chronic escalating-dose morphine regimen (20-100 mg/kg/day, i.p.). Seven days withdrawal from this administration paradigm induced a decrease of hypothalamic OT levels and a concomitant increase of oxytocin receptor (OTR) binding in the lateral septum and amygdala. Although no physical withdrawal symptoms or alterations in the plasma corticosterone levels were observed after 7 days of abstinence, mice exhibited increased anxiety-like and depressive-like behaviors and impaired sociability. CBT (6.4 mg/kg, i.p.) attenuated the observed negative emotional consequences of opioid withdrawal. Furthermore, in the conditioned place preference paradigm with 10 mg/kg morphine conditioning, CBT (6.4 mg/kg, i.p.) was able to prevent the stress-induced reinstatement to morphine-seeking following extinction. Overall, our results suggest that alterations of the oxytocinergic system contribute to the mechanisms underlying anxiety, depression, and social deficits observed during opioid abstinence. This study also highlights the oxytocinergic system as a target for developing pharmacotherapy for the treatment of emotional impairment associated with abstinence and thereby prevention of relapse.


Subject(s)
Affective Symptoms/etiology , Affective Symptoms/prevention & control , Morphine Dependence/psychology , Oxytocin/analogs & derivatives , Stress, Psychological/prevention & control , Substance Withdrawal Syndrome/complications , Analysis of Variance , Animals , Brain/drug effects , Brain/metabolism , Corticosterone/blood , Dose-Response Relationship, Drug , Drug-Seeking Behavior/drug effects , Male , Mice , Mice, Inbred C57BL , Morphine/adverse effects , Oxytocin/metabolism , Oxytocin/therapeutic use , Reinforcement, Psychology , Stress, Psychological/blood , Time Factors
10.
Neuropharmacology ; 71: 228-36, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23583933

ABSTRACT

Considerable evidence indicates that adenosine A(2A) receptors (A(2A)Rs) modulate cholinergic neurotransmission, nicotinic acetylcholine receptor (nAChR) function, and nicotine-induced behavioural effects. To explore the interaction between A(2A) and nAChRs, we examined if the complete genetic deletion of adenosine A(2A)Rs in mice induces compensatory alterations in the binding of different nAChR subtypes, and whether the long-term effects of nicotine on nAChR regulation are altered in the absence of the A(2A)R gene. Quantitative autoradiography was used to measure cytisine-sensitive [¹²5I]epibatidine and [¹²5I]α-bungarotoxin binding to α4ß2* and α7 nAChRs, respectively, in brain sections of drug-naïve (n = 6) or nicotine treated (n = 5-7), wild-type and adenosine A(2A)R knockout mice. Saline or nicotine (7.8 mg/kg/day; free-base weight) were administered to male CD1 mice via subcutaneous osmotic minipumps for a period of 14 days. Blood plasma levels of nicotine and cotinine were measured at the end of treatment. There were no compensatory developmental alterations in nAChR subtype distribution or density in drug-naïve A(2A)R knockout mice. In nicotine treated wild-type mice, both α4ß2* and α7 nAChR binding sites were increased compared with saline treated controls. The genetic ablation of adenosine A(2A)Rs prevented nicotine-induced upregulation of α7 nAChRs, without affecting α4ß2* receptor upregulation. This selective effect was observed at plasma levels of nicotine that were within the range reported for smokers (10-50 ng ml⁻¹). Our data highlight the involvement of adenosine A(2A)Rs in the mechanisms of nicotine-induced α7 nAChR upregulation, and identify A(2A)Rs as novel pharmacological targets for modulating the long-term effects of nicotine on α7 receptors.


Subject(s)
Brain/drug effects , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Receptor, Adenosine A2A/metabolism , Receptors, Nicotinic/metabolism , Up-Regulation/drug effects , alpha7 Nicotinic Acetylcholine Receptor/biosynthesis , Animals , Brain/metabolism , Brain/pathology , Bridged Bicyclo Compounds, Heterocyclic/metabolism , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Bungarotoxins/metabolism , Bungarotoxins/pharmacology , Cotinine/blood , Male , Mice , Mice, Inbred Strains , Mice, Knockout , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Nicotine/blood , Nicotine/pharmacokinetics , Nicotinic Agonists/blood , Nicotinic Agonists/metabolism , Nicotinic Agonists/pharmacokinetics , Protein Subunits/agonists , Protein Subunits/metabolism , Pyridines/metabolism , Pyridines/pharmacology , Receptor, Adenosine A2A/genetics , Receptors, Nicotinic/chemistry , Tobacco Use Disorder/blood , Tobacco Use Disorder/metabolism , Tobacco Use Disorder/pathology , alpha7 Nicotinic Acetylcholine Receptor/agonists , alpha7 Nicotinic Acetylcholine Receptor/metabolism
11.
Synapse ; 66(5): 383-90, 2012 May.
Article in English | MEDLINE | ID: mdl-22213208

ABSTRACT

Adenosine, acting on adenosine A(2A) receptors (A2ARs), regulates addictive processes induced by drugs of abuse. This study investigates the role of A(2A) adenosine receptors in neurochemical and behavioral responses to an acute cocaine challenge. Changes in the extracellular levels of dopamine (DA) in the nucleus accumbens (NAc) of mice lacking A(2A) adenosine receptors and wild type (WT) littermates after an acute cocaine (20 mg/kg) administration were evaluated by in vivo microdialysis studies. Locomotor effects induced by cocaine were measured during the microdialysis procedure. Cocaine-evoked increases in extracellular DA were not sustained in mice lacking A(2A) Rs in comparison with wild-type mice (P < 0.05). Cocaine administration significantly increased ambulatory activity in both genotypes. However, overall locomotor activity was further increased, whereas rest and small local movement measures were significantly attenuated in the A(2A) R knockout mice compared with WT littermates (P < 0.05). Our findings support an important role for adenosine A(2A) R in modulating the acute effects of cocaine, as demonstrated by the decrease in cocaine-evoked dopaminergic transmission in the NAc. Furthermore, the results support an important antagonistic role of A(2A) R in vivo in regulating psychostimulant-induced hyperlocomotion.


Subject(s)
Cocaine/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Dopamine/metabolism , Locomotion/drug effects , Motor Activity/drug effects , Nucleus Accumbens/drug effects , Receptor, Adenosine A2A/genetics , Animals , Behavior, Animal/drug effects , Male , Mice , Mice, Knockout , Microdialysis , Receptor, Adenosine A2A/physiology
12.
J Biol Chem ; 285(51): 40104-13, 2010 Dec 17.
Article in English | MEDLINE | ID: mdl-20940302

ABSTRACT

Endothelial cells (ECs) express a Nox2 enzyme, which, by generating reactive oxygen species (ROS), contributes to EC redox signaling and angiotensin II (AngII)-induced endothelial dysfunction. ECs also express abundantly an adenosine A(2A) receptor (A(2A)R), but its role in EC ROS production remains unknown. In this study, we investigated the role of A(2A)R in the regulation of Nox2 activity and signaling in ECs with or without acute AngII stimulation. In cultured ECs (SVEC4-10), AngII (100 nm, 30 min) significantly increased Nox2 membrane translocation and association with A(2A)R. These were accompanied by p47(phox), ERK1/2, p38 MAPK, and Akt phosphorylation and an increased ROS production (169 ± 0.04%). These AngII effects were inhibited back to the control levels by a specific A(2A)R antagonist (SCH58261), or adenosine deaminase, or by knockdown of A(2A)R or Nox2 using specific siRNAs. Knockdown of A(2A)R, as determined by Western blotting, decreased Nox2 and p47(phox) expression. In wild-type mouse aorta, SCH58261 significantly reduced acute AngII-induced ROS production and preserved endothelium-dependent vessel relaxation to acetylcholine. These results were further confirmed by using aortas from A(2A)R knock-out mice. In conclusion, A(2A)R is involved in the regulation of EC ROS production by Nox2. Inhibition or blockade of A(2A)R protects ECs from acute AngII-induced oxidative stress, MAPK activation, and endothelium dysfunction.


Subject(s)
Angiotensin II/pharmacology , Endothelial Cells/metabolism , Membrane Glycoproteins/metabolism , NADPH Oxidases/metabolism , Reactive Oxygen Species/metabolism , Receptor, Adenosine A2A/metabolism , Signal Transduction/drug effects , Adenosine A2 Receptor Antagonists/pharmacology , Angiotensin II/metabolism , Animals , Cells, Cultured , Enzyme Activation/drug effects , Enzyme Activation/genetics , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/genetics , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , NADPH Oxidase 2 , NADPH Oxidases/biosynthesis , NADPH Oxidases/genetics , Phosphorylation/drug effects , Phosphorylation/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Pyrimidines/pharmacology , Receptor, Adenosine A2A/genetics , Signal Transduction/physiology , Triazoles/pharmacology , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
13.
Free Radic Biol Med ; 44(7): 1433-42, 2008 Apr 01.
Article in English | MEDLINE | ID: mdl-18206127

ABSTRACT

Cardiac tissues express constitutively an NADPH oxidase, which generates reactive oxygen species (ROS) and is involved in redox signaling. Myocardial metabolism generates abundant adenosine, which binds to its receptors and plays important roles in cardiac function. The adenosine A2A receptor (A2AR) has been found to be expressed in cardiac myocytes and coronary endothelial cells. However, the role of the A2AR in the regulation of cardiac ROS production remains unknown. We found that knockout of A2AR significantly decreased (39+/-8%) NADPH-dependent O2- production in mouse hearts compared to age (10 weeks)-matched wild-type controls. This was accompanied by a significant decrease in Nox2 (a catalytic subunit of NADPH oxidase) protein expression, and down-regulation of ERK1/2, p38MAPK, and JNK phosphorylation (all P<0.05). In wild-type mice, intraperitoneal injection of the selective A2AR antagonist SCH58261 (3-10 mg/kg body weight for 90 min) inhibited phosphorylation of p47phox (a regulatory subunit of Nox2), which was accompanied by a down-regulated cardiac ROS production (48+/-8%), and decreased JNK and ERK1/2 activation by 54+/-28% (all P<0.05). In conclusion, A2AR through MAPK signaling regulates p47phox phosphorylation and cardiac ROS production by NADPH oxidase. Modulation of A2AR activity may have potential therapeutic applications in controlling ROS production by NADPH oxidase in the heart.


Subject(s)
Gene Expression Regulation, Enzymologic , Myocardium/enzymology , NADPH Oxidases/metabolism , Receptor, Adenosine A2A/metabolism , Adenosine/metabolism , Animals , MAP Kinase Signaling System , Mice , Mice, Knockout , Microscopy, Fluorescence , Models, Biological , Myocardium/metabolism , Phosphorylation , Reactive Oxygen Species , Signal Transduction , Time Factors
14.
Eur J Pharmacol ; 562(1-2): 68-71, 2007 May 07.
Article in English | MEDLINE | ID: mdl-17331495

ABSTRACT

Paracetamol is an effective analgesic but its mechanism of action is unclear. We investigated the effect of paracetamol and the analgesic adjuvant caffeine on the activity of NO synthase in mouse spinal cord and cerebellar slices in vitro, by measuring the conversion of [(3)H]arginine to [(3)H]citrulline. Paracetamol (100 microM) had no effect on NO synthase activity in cerebellum, but in the spinal cord both paracetamol (100 microM) and caffeine (30 microM) attenuated glutamate (5 mM)-induced [(3)H]citrulline production and in combination they abolished it. In conclusion paracetamol inhibits spinal cord NO synthesis and this may be related to its analgesic effects.


Subject(s)
Acetaminophen/pharmacology , Nitric Oxide/biosynthesis , Spinal Cord/drug effects , Analgesics, Non-Narcotic/pharmacology , Animals , Arginine/pharmacology , Caffeine/pharmacology , Central Nervous System Stimulants/pharmacology , Cerebellum/drug effects , Cerebellum/metabolism , Citrulline/biosynthesis , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Glutamic Acid/pharmacology , In Vitro Techniques , Male , Mice , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Spinal Cord/metabolism
15.
Pain ; 129(3): 287-294, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17134834

ABSTRACT

Adenosine is a neuromodulator with complex effects on pain pathways. Mice lacking the adenosine A2A receptor are hypoalgesic, and have altered analgesic responses to receptor-selective opioid agonists. These and other findings suggest a role for the adenosine A2A receptor in sensitizing afferent fibres projecting to the spinal cord. To test this hypothesis formalin (20 microl, 5%) was injected into the paw and nociceptive responses were measured in wildtype and adenosine A2A receptor knockout mice. There was a significant reduction in nociception associated with sensory nerve activation in the knockout mice as measured by time spent biting/licking the formalin-injected paw and number of flinches seen during the first phase, but only the number of flinches was reduced during the second inflammatory phase. In addition, the selective adenosine A2A antagonist SCH58261 (3 and 10 mg/kg) also antagonised both phases of the formalin test. We also labelled NMDA glutamate and NK1 receptors in spinal cord sections as an indirect measure of nociceptive transmission from peripheral sites to the spinal cord. [3H]-Substance P binding to NK1 receptors was unaltered but there was a substantial reduction in binding of [3H]-MK801 to NMDA glutamate receptors in all regions of the spinal cord from knockout mice. The decrease in NMDA glutamate receptor binding may reflect reduced peripheral sensory input to the spinal cord during development and could relate to the hypoalgesia in this genotype. These results support a key role for the adenosine A2A receptor in peripheral nociceptive pathways.


Subject(s)
Formaldehyde , Hyperalgesia/chemically induced , Hyperalgesia/physiopathology , Receptors, Adenosine A2/metabolism , Receptors, Glutamate/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Spinal Cord/physiopathology , Animals , Male , Mice , Mice, Knockout , Pain Measurement/drug effects , Pain Threshold , Protein Binding , Receptors, Adenosine A2/genetics , Signal Transduction , Spinal Cord/drug effects
16.
Eur J Pharmacol ; 531(1-3): 80-6, 2006 Feb 15.
Article in English | MEDLINE | ID: mdl-16448647

ABSTRACT

This study investigated the involvement of adenosine receptors in the interaction between paracetamol and caffeine in mice, using the adenosine A2A receptor antagonist 5-amino-7-(2-phenylethyl)-2-(2-furyl)pyrazolo[4,3-e]-1,2,4-triazolo[1,5-c]pyrimidine (SCH58261) and the adenosine A2B receptor antagonist 1-propyl-8-p-sulfophenylxanthine (PSB1115), in the tail immersion and hot-plate tests. Paracetamol (10-200 mg/kg) was antinociceptive in both tests, but, in contrast to previous studies, caffeine (10 mg/kg) was pronociceptive in the tail immersion test, and reduced the effects of paracetamol in both tests. SCH58261 (3 mg/kg) was antinociceptive in both tests and in its presence paracetamol (50 mg/kg) had no further effect. PSB1115 (10 mg/kg) had little effect alone but potentiated the effect of paracetamol (50 mg/kg) in the hot-plate test and abolished it in the tail immersion test. These results suggest that adenosine A2B receptors may be involved in the action of paracetamol in a pathway-dependent manner, and also support the existence of pronociceptive adenosine A2A receptors.


Subject(s)
Acetaminophen/pharmacology , Adenosine A2 Receptor Antagonists , Caffeine/pharmacology , Nociceptors/drug effects , Pyrimidines/pharmacology , Triazoles/pharmacology , Analgesics, Non-Narcotic/pharmacology , Animals , Central Nervous System Stimulants/pharmacology , Dose-Response Relationship, Drug , Female , Male , Mice , Mice, Knockout , Neuroprotective Agents/pharmacology , Nociceptors/physiopathology , Pain/physiopathology , Pain/prevention & control , Pain Measurement/methods , Receptor, Adenosine A2A/genetics , Receptor, Adenosine A2A/physiology , Receptor, Adenosine A2B/physiology , Sulfonic Acids/pharmacology , Time Factors , Xanthines/pharmacology
17.
Int J Exp Pathol ; 86(4): 247-55, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16045547

ABSTRACT

The objective was to test the hypothesis that dietary copper inhibits atherosclerosis by inducing superoxide dismutase (SOD) and potentiating nitric oxide (NO). New Zealand White rabbits were fed either a cholesterol diet (n = 8) or a cholesterol diet containing 0.02% copper acetate (n = 8) for 13 weeks. We found that the intimal area was significantly smaller in the animals supplemented with copper (P < 0.005), although integrated plasma cholesterol levels were not significantly different. This was associated with a significant increase in aortic copper content (P < 0.05), SOD activity (P < 0.05) and Cu/Zn SOD mRNA (P < 0.05) and a significant decrease in nitrotyrosine content (P < 0.05). Furthermore, there was a positive correlation between aortic copper content and SOD activity (P < 0.005, R(2) = 0.83) and a negative correlation between aortic superoxide dimutase activity and nitrotyrosine content (P < 0.005, R(2) = 0.93). In organ bath experiments, the relaxation of precontracted carotid artery rings to calcium ionophore was greater in animals supplemented with copper. No difference in response to sodium nitroprusside was observed. These data suggest that in the cholesterol-fed rabbit, copper supplements inhibit the progression of atherosclerosis by increasing SOD expression, thereby reducing the interaction of NO with superoxide, and hence potentiating NO-mediated pathways that may protect against atherosclerosis.


Subject(s)
Aorta, Thoracic/metabolism , Arteriosclerosis/diet therapy , Copper/administration & dosage , Dietary Supplements , Nitric Oxide/metabolism , Superoxide Dismutase/metabolism , Animals , Aorta, Thoracic/enzymology , Arteriosclerosis/enzymology , Arteriosclerosis/metabolism , Calcimycin/pharmacology , Carotid Arteries/drug effects , Cholesterol/blood , Copper/analysis , Ionophores/pharmacology , Muscle, Smooth/drug effects , Oxidative Stress/drug effects , Rabbits , Tyrosine/analogs & derivatives , Tyrosine/analysis
18.
Eur J Pharmacol ; 508(1-3): 99-106, 2005 Jan 31.
Article in English | MEDLINE | ID: mdl-15680259

ABSTRACT

Quantitative autoradiography has been used to assess whether [3H]paracetamol (3 microM) binds specifically to any area of the murine brain and spinal cord and to investigate whether paracetamol (1-100 microM) competes for binding to the nociceptin opioid peptide (NOP) receptor or to the nitrobenzylthioinosine (NBTI)-sensitive adenosine transporter in the brains of mice. [3H]paracetamol binding was homogenous and, although there was some indication of specific binding overall, this binding in most individual regions failed to reach statistical significance. However, thoracic segments of the spinal cord were found to have significantly higher specific binding than cervical and lumbar regions. Paracetamol did not significantly compete for binding to the NOP receptor or to the NBTI-sensitive adenosine transporter, showing that it does not mediate its effect via these sites. Although paracetamol did bind specifically to the murine brain and spinal cord, the binding was not region-specific, suggesting binding is not related to any particular neurotransmitter system.


Subject(s)
Acetaminophen/metabolism , Brain/metabolism , Spinal Cord/metabolism , Thioinosine/analogs & derivatives , Acetaminophen/pharmacology , Analysis of Variance , Animals , Autoradiography/methods , Binding Sites , Binding, Competitive/drug effects , Male , Membrane Transport Proteins/metabolism , Mice , Nucleoside Transport Proteins , Opioid Peptides/metabolism , Receptors, Opioid/metabolism , Thioinosine/metabolism , Thioinosine/pharmacology , Tritium , Nociceptin Receptor , Nociceptin
19.
Eur J Pharmacol ; 504(1-2): 55-9, 2004 Nov 03.
Article in English | MEDLINE | ID: mdl-15507221

ABSTRACT

The binding of the adenosine A(2A) receptor antagonist [3H] 4-(2-[7-amino-2-(2-furyl)[1,2,4]-triazolo[2,3-a][1,3,5]triazin-5-ylamino]ethyl)phenol ([3H]ZM 241385) to mouse brain and spinal cord was investigated. In brain homogenates, single-site binding was observed with a Bmax of 299+/-28 fmol mg(-1) protein and a Kd of 0.75+/-0.08 nM. In autoradiographic studies, there was a high density of specific binding of [3H]ZM 241385 in the striatum, with a very low density in the cortex and no binding elsewhere in the brain or in the spinal cord. All specific binding of [3H]ZM 241385 was lost in genetically modified mice lacking the adenosine A(2A) receptor, confirming the selectivity of this radioligand.


Subject(s)
Receptor, Adenosine A2A/deficiency , Receptor, Adenosine A2A/metabolism , Triazines/metabolism , Triazoles/metabolism , Animals , Binding Sites/drug effects , Brain/metabolism , Dose-Response Relationship, Drug , Male , Mice , Mice, Knockout , Protein Binding/physiology , Receptor, Adenosine A2A/genetics , Tritium
20.
Brain Res ; 1025(1-2): 1-9, 2004 Oct 29.
Article in English | MEDLINE | ID: mdl-15464738

ABSTRACT

There is a large body of evidence indicating important interactions between the adenosine and the opioid systems in regulating pain, opioid dependence and withdrawal. Mice lacking the proenkephalin gene and therefore lacking the endogenous enkephalin peptides have been successfully developed and exhibit decreased locomotor activity, are hyperalgesic and show enhanced anxiety and aggression. In addition, an upregulation of mu and delta receptors was also observed in the brains of knockout mice. To investigate if there are any compensatory alterations in adenosine systems in the brains of mutant mice, we have carried out quantitative autoradiographic mapping of A(1) and A(2A) adenosine receptors and nitrobenzylthioinosine (NBTI)-sensitive adenosine transporters in the brains of wild-type and homozygous enkephalin knockout mice. Adjacent coronal brain sections were cut from brains of +/+ and -/- mice for the determination of binding of [(3)H]1,3-dipropyl-8-cyclopentylxanthine ([(3)H]DPCPX), [(3)H]2-[p-(2-carbonylethyl)phenylethylamino]-5'-N-ethylcarboxamidoadenosine ([(3)H]CGS21680) or [(3)H]NBTI to A(1) and A(2A) adenosine receptors and NBTI-sensitive adenosine transporters, respectively. A small but significant increase in [(3)H]DPCPX and [(3)H]NBTI binding but no significant change in [(3)H]CGS21680 binding was detected in enkephalin knockout brains. The results provide further evidence of functional interactions in the brain between opioid receptors and A(1) adenosine receptors as well as NBTI-sensitive adenosine transporters but not A(2A) receptors.


Subject(s)
Brain/metabolism , Enkephalins/deficiency , Membrane Transport Proteins/metabolism , Protein Precursors/deficiency , Receptors, Purinergic P1/metabolism , Thioinosine/analogs & derivatives , Thioinosine/pharmacology , Animals , Autoradiography , Brain/drug effects , Enkephalins/biosynthesis , Enkephalins/genetics , Male , Membrane Transport Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Nucleoside Transport Proteins , Protein Binding , Protein Precursors/biosynthesis , Protein Precursors/genetics , Receptors, Purinergic P1/genetics , Xanthines/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...