Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Pharmacol Exp Ther ; 344(2): 501-10, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23197772

ABSTRACT

Dopamine D(3) receptors have eluded definitive linkage to neurologic and psychiatric disorders since their cloning over 20 years ago. We report a new method that does not employ a radiolabel for simultaneously defining in vivo receptor occupancy of D(3) and D(2) receptors in rat brain after systemic dosing using the tracer epidepride (N-[[(2S)-1-ethylpyrrolidin-2-yl]methyl]-5-iodo-2,3-dimethoxybenzamide). Decreases in epidepride binding in lobule 9 of cerebellum (rich in D(3) receptors) were compared with nonspecific binding in the lateral cerebellum. The in vivo occupancy of the dopamine D(3) receptors was dose dependently increased by SB-277011A (trans-N-[4-[2-(6-cyano-1,2,3,4-tetrahydroisoquinolin-2-yl)ethyl]cyclohexyl]-4-quinolinecarboxamide) and U99194 (2,3-dihydro-5,6-dimethoxy- N,N-dipropyl-1H-inden-2-amine). Both antagonists increased extracellular levels of acetylcholine (ACh) in the medial prefrontal cortex of rats and modified brain-tissue levels of ACh and choline. Consistent with these findings, the D(3) receptor antagonists enhanced the acquisition of learning of rats either alone or in the presence of the norepinephrine uptake blocker reboxetine as with the attention-deficit-hyperactivity disorder (ADHD) drug methylphenidate. Like reboxetine, the D(3) receptor antagonists also prevented deficits induced by scopolamine in object recognition memory of rats. Mice in which the dopamine transporter (DAT) has been deleted exhibit hyperactivity that is normalized by compounds that are effective in the treatment of ADHD. Both D(3) receptor antagonists decreased the hyperactivity of DAT(-/-) mice without affecting the activity of wild type controls. The present findings indicate that dopamine D(3) receptor antagonists engender cognition-enhancing and hyperactivity-dampening effects. Thus, D(3) receptor blockade could be considered as a novel treatment approach for cognitive deficits and hyperactivity syndromes, including those observed in ADHD.


Subject(s)
Attention Deficit Disorder with Hyperactivity/drug therapy , Behavior, Animal/drug effects , Brain Chemistry/drug effects , Brain/drug effects , Dopamine Antagonists/pharmacology , Receptors, Dopamine D3/antagonists & inhibitors , Animals , Attention Deficit Disorder with Hyperactivity/metabolism , Brain/metabolism , Dopamine Antagonists/chemistry , Dopamine Antagonists/pharmacokinetics , Dopamine Antagonists/therapeutic use , Dopamine D2 Receptor Antagonists , Indans/chemistry , Indans/pharmacokinetics , Indans/pharmacology , Indans/therapeutic use , Male , Microdialysis , Molecular Structure , Nitriles/chemistry , Nitriles/pharmacokinetics , Nitriles/pharmacology , Nitriles/therapeutic use , Pattern Recognition, Visual/drug effects , Protein Binding , Rats , Rats, Sprague-Dawley , Rats, Wistar , Tetrahydroisoquinolines/chemistry , Tetrahydroisoquinolines/pharmacokinetics , Tetrahydroisoquinolines/pharmacology , Tetrahydroisoquinolines/therapeutic use
2.
Neuroscience ; 144(4): 1470-6, 2007 Feb 23.
Article in English | MEDLINE | ID: mdl-17207580

ABSTRACT

Calcineurin (PP2B) is a Ca(2+)-dependent protein phosphatase enriched in the brain that takes part in intracellular signaling pathways regulating synaptic plasticity and neuronal functions. Calcineurin-dependent pathways are important for complex brain functions such as learning and memory. More recently, they have been suggested to play a role in the processing of emotional information. The aim of this study was to investigate whether calcineurin may be involved in the effect of antidepressants. We first found that chronic antidepressant treatment in mice leads to an increase of calcineurin levels in the hippocampus. We then studied the behavioral and molecular responses to fluoxetine of mice with a genetic overactivation of calcineurin in the hippocampus (constitutively-activated calcineurin transgenic mouse line #98, CN98 mice). We observed that CN98 mice are more sensitive to the behavioral effect of fluoxetine and desipramine tested in the tail suspension test. Moreover, the basal expression of growth factor brain-derived neurotrophic factor and subunit 1 of AMPA glutamate receptor, GluR1, both of which are modified after chronic antidepressant administration, are altered in the hippocampus of CN98 mice. These results suggest that calcineurin-dependent dephosphorylation plays an important role in the mechanisms of action of antidepressants, providing a new starting point for developing improved therapeutic treatments for depression.


Subject(s)
Antidepressive Agents/pharmacology , Calcineurin/metabolism , Depressive Disorder/drug therapy , Depressive Disorder/enzymology , Hippocampus/drug effects , Hippocampus/enzymology , Animals , Antidepressive Agents, Tricyclic/pharmacology , Brain-Derived Neurotrophic Factor/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Calcineurin/genetics , Depressive Disorder/physiopathology , Desipramine/pharmacology , Female , Fluoxetine/pharmacology , Hippocampus/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation/drug effects , Receptors, AMPA/drug effects , Receptors, AMPA/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology , Up-Regulation/drug effects , Up-Regulation/physiology
4.
Mol Psychiatry ; 11(2): 187-95, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16231039

ABSTRACT

Atomoxetine has been approved by the FDA as the first new drug in 30 years for the treatment of attention deficit/hyperactivity disorder (ADHD). As a selective norepinephrine uptake inhibitor and a nonstimulant, atomoxetine has a different mechanism of action from the stimulant drugs used up to now for the treatment of ADHD. Since brain acetylcholine (ACh) has been associated with memory, attention and motivation, processes dysregulated in ADHD, we investigated the effects of atomoxetine on cholinergic neurotransmission. We showed here that, in rats, atomoxetine (0.3-3 mg/kg, i.p.),--increases in vivo extracellular levels of ACh in cortical but not subcortical brain regions. The marked increase of cortical ACh induced by atomoxetine was dependent upon norepinephrine alpha-1 and/or dopamine D1 receptor activation. We observed similar increases in cortical and hippocampal ACh release with methylphenidate (1 and 3 mg/kg, i.p.)--currently the most commonly prescribed medication for the treatment of ADHD--and with the norepinephrine uptake inhibitor reboxetine (3-30 mg/kg, i.p.). Since drugs that increase cholinergic neurotransmission are used in the treatment of cognitive dysfunction and dementias, we also investigated the effects of atomoxetine on memory tasks. We showed that, consistent with its cortical procholinergic and catecholamine-enhancing profile, atomoxetine (1-3 mg/kg, p.o.) significantly ameliorated performance in the object recognition test and the radial arm-maze test.


Subject(s)
Adrenergic Uptake Inhibitors/pharmacology , Cholinergic Fibers/drug effects , Maze Learning/drug effects , Pattern Recognition, Visual/drug effects , Propylamines/pharmacology , Acetylcholine/metabolism , Animals , Atomoxetine Hydrochloride , Central Nervous System Stimulants/pharmacology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cholinergic Fibers/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Male , Methylphenidate/pharmacology , Microdialysis , Morpholines/pharmacology , Norepinephrine/metabolism , Rats , Rats, Sprague-Dawley , Rats, Wistar , Reboxetine , Receptors, Dopamine/drug effects , Receptors, Dopamine/metabolism
5.
Behav Pharmacol ; 16(5-6): 315-31, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16148437

ABSTRACT

Mood and anxiety disorders, the most prevalent of the psychiatric disorders, cause immeasurable suffering worldwide. Despite impressive advances in pharmacological therapies, improvements in efficacy and side-effect profiles are needed. The present literature review examines the role that the endocannabinoid system may play in these disorders and the potential value of targeting this system in the search for novel and improved medications. Cannabis and its major psychoactive component (-)-trans-delta9-tetrahydrocannabinol, have profound effects on mood and can modulate anxiety and mood states. Cannabinoid receptors and other protein targets in the central nervous system (CNS) that modulate endocannabinoid function have been described. The discovery of selective modulators of some of these sites that increase or decrease endocannabinoid neurotransmission, primarily through the most prominent of the cannabinoid receptors in the CNS, the CB1 receptors, combined with transgenic mouse technology, has enabled detailed investigations into the role of these CNS sites in the regulation of mood and anxiety states. Although data point to the involvement of the endocannabinoid system in anxiety states, the pharmacological evidence seems contradictory: both anxiolytic- and anxiogenic-like effects have been reported with both endocannabinoid neurotransmission enhancers and blockers. Due to advances in the development of selective compounds directed at the CB1 receptors, significant progress has been made on this target. Recent biochemical and behavioural findings have demonstrated that blockade of CB1 receptors engenders antidepressant-like neurochemical changes (increases in extracellular levels of monoamines in cortical but not subcortical brain regions) and behavioural effects consistent with antidepressant/antistress activity in rodents.


Subject(s)
Affect/physiology , Anxiety Disorders/physiopathology , Receptor, Cannabinoid, CB1/physiology , Affect/drug effects , Animals , Anxiety Disorders/prevention & control , Cannabinoid Receptor Modulators/physiology , Dronabinol/pharmacology , Humans , Models, Animal , Piperidines/pharmacology , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/antagonists & inhibitors
6.
Mol Psychiatry ; 8(7): 673-9, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12874603

ABSTRACT

Among the five different muscarinic receptors that have been cloned and characterized, M2 and M4 receptors are localized both post- and presynaptically and are believed to have a pronounced autoreceptor role. The functional importance of these receptors in the regulation of acetylcholine release in the hippocampus and in cognitive processes was investigated by using M2 and M4 receptor single knockout (KO) as well as M2/M4 receptor double KO mice. We found profound alterations in acetylcholine homeostasis in the hippocampus of both M2- and M4-KO mice as well as of the combined M2/M4-KOs, as assessed by in vivo microdialysis. Basal acetylcholine efflux in the hippocampus was significantly increased in M4-KO and was elevated further in M2/M4-KOs. The increase in hippocampal acetylcholine induced by local administration of scopolamine was markedly reduced in M2-KO and completely abolished in M2/M4-KOs. In M2-KO and much more in M2/M4-KOs, the increase in hippocampal acetylcholine triggered by exposure to a novel environment was more pronounced both in amplitude and duration, with a similar trend observed for M4-KOs. Dysregulation of cholinergic function in the hippocampus, as it could result from perturbed autoreceptor function, may be associated with cognitive deficits. Importantly, M2- and M2/M4-KO, but not M4-KO, animals showed an impaired performance in the passive avoidance test. Together these results suggest a crucial role for muscarinic M2 and M4 receptors in the tonic and phasic regulation of acetylcholine efflux in the hippocampus as well as in cognitive processes.


Subject(s)
Acetylcholine/metabolism , Hippocampus/metabolism , Receptor, Muscarinic M2/physiology , Receptor, Muscarinic M4/physiology , Animals , Avoidance Learning/drug effects , Avoidance Learning/physiology , Cognition/physiology , Crosses, Genetic , Environment , Habituation, Psychophysiologic/physiology , Hippocampus/drug effects , Hippocampus/physiology , Homeostasis , Male , Mice , Mice, Knockout , Microdialysis , Muscarinic Antagonists/pharmacology , Receptor, Muscarinic M2/deficiency , Receptor, Muscarinic M2/drug effects , Receptor, Muscarinic M2/genetics , Receptor, Muscarinic M4/deficiency , Receptor, Muscarinic M4/drug effects , Receptor, Muscarinic M4/genetics , Scopolamine/pharmacology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
7.
Eur J Pharmacol ; 426(3): R3-4, 2001 Aug 31.
Article in English | MEDLINE | ID: mdl-11527547

ABSTRACT

The effects of the selective cannabinoid CB(1) receptor antagonist N-(piperidin-1-yl)-5-(4-chlorophenyl)-1-(2,4-dichlorophenyl)-4-methyl-1H-pyrazole-3-carboxamide-hydrochloride (SR141716A) on extracellular concentrations of norepinephrine and 5-hydroxytryptamine (5-HT) were assessed by in vivo microdialysis in the anterior hypothalamus of freely moving rats. SR14716A (0.3, 1, 3 mg/kg, i.p.) dose-dependently increased norepinephrine efflux to about 300% of baseline, without affecting 5-HT levels. This increase in norepinephrine outflow could play an important role in the pharmacological and potentially therapeutic actions of SR141716A.


Subject(s)
Hypothalamus/drug effects , Norepinephrine/metabolism , Piperidines/pharmacology , Pyrazoles/pharmacology , Receptors, Drug/antagonists & inhibitors , 3,4-Dihydroxyphenylacetic Acid/metabolism , Animals , Dose-Response Relationship, Drug , Homovanillic Acid/metabolism , Hydroxyindoleacetic Acid/metabolism , Hypothalamus/metabolism , Male , Microdialysis , Rats , Rats, Wistar , Receptors, Cannabinoid , Rimonabant , Serotonin/metabolism
8.
Neuroreport ; 12(7): 1367-70, 2001 May 25.
Article in English | MEDLINE | ID: mdl-11388413

ABSTRACT

Krox-24 is an immediate early gene encoding a zinc-finger transcription factor implicated in several adaptive responses, and its induction by cannabinoids has been reported. We used mice targeted in the Krox-24 gene to specifically dissect the role of this protein in the acute and chronic central actions of cannabinoids. We report here on the ability of cannabinoids to activate G-proteins and to inhibit adenylyl cyclase, and to elicit behavioral responses in wild-type and mutant mice. The behavioral parameters and the biochemical correlates of abstinence after delta9-THC withdrawal were evaluated. We show that Krox-24 is not involved in the acute analgesic effects of delta9-THC and in the SR precipitated delta9-THC withdrawal syndrome.


Subject(s)
Adenylyl Cyclases/drug effects , Behavior, Animal/drug effects , Cannabinoids/pharmacology , Cyclic AMP/genetics , DNA-Binding Proteins/drug effects , GTP-Binding Proteins/drug effects , Immediate-Early Proteins , Mice, Knockout/metabolism , Transcription Factors/drug effects , Adenylyl Cyclases/metabolism , Animals , Behavior, Animal/physiology , Binding, Competitive/drug effects , Binding, Competitive/physiology , Cerebellum/drug effects , Cerebellum/metabolism , Cyclic AMP/metabolism , Cyclic AMP Response Element-Binding Protein/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Early Growth Response Protein 1 , GTP-Binding Proteins/metabolism , Male , Marijuana Abuse/metabolism , Marijuana Abuse/physiopathology , Mice , Mice, Knockout/genetics , Phosphorylation/drug effects , Receptors, Cannabinoid , Receptors, Drug/drug effects , Receptors, Drug/metabolism , Spleen/drug effects , Spleen/metabolism , Substance Withdrawal Syndrome/drug therapy , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/physiopathology , Transcription Factors/genetics , Transcription Factors/metabolism
9.
Br J Pharmacol ; 132(8): 1809-16, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11309253

ABSTRACT

Several compounds, mainly opioid agonists such as methadone, are currently used for long term medication of heroin addicts. Nevertheless, these maintenance treatments have the disadvantage to induce a dependence to another opiate. As interactions between opioid and cannabinoid systems have been demonstrated, the ability of the CB(1) antagonist, SR141716A to reduce morphine-induced addiction was investigated. The effects of SR141716A on the rewarding responses of morphine were evaluated in the place conditioning paradigm. No significant conditioned preference or aversion were observed after repeated treatment with the CB(1) antagonist alone. However, SR141716A was able to antagonize the acquisition of morphine-induced conditioned place preference. SR141716A was co-administered with morphine for 5 days, and the withdrawal syndrome was precipitated by naloxone administration. A reduction in the incidence of two main signs of abstinence: wet dog shakes and jumping was observed while the other were not significantly modified. In contrast, an acute injection of the CB(1) antagonist just before naloxone administration was unable to modify the incidence of the behavioural manifestations of the withdrawal, suggesting that only chronic blockade of CB(1) receptors is able to reduce morphine-induced physical dependence. Several biochemical mechanisms could explain the reduction of opioid dependence by CB(1) antagonists. Whatever the hypotheses, this study supports the reported interaction between the endogenous cannabinoid and opioid systems, and suggests that SR 141716A warrants further investigations for a possible use in opioid addiction.


Subject(s)
Opioid-Related Disorders/drug therapy , Piperidines/therapeutic use , Pyrazoles/therapeutic use , Receptors, Drug/antagonists & inhibitors , Animals , Brain Chemistry/drug effects , Cannabinoids/antagonists & inhibitors , Cannabinoids/metabolism , Conditioning, Operant/drug effects , Dynorphins/metabolism , Male , Mice , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Opioid-Related Disorders/psychology , Radioimmunoassay , Receptors, Cannabinoid , Receptors, Opioid, kappa/metabolism , Rimonabant , Substance Withdrawal Syndrome/metabolism , Synapses/drug effects , Synapses/metabolism
10.
Eur J Neurosci ; 12(3): 1038-46, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10762335

ABSTRACT

Region-specific up-regulation of the cyclic AMP pathway is considered an important molecular mechanism in the origin of the somatic manifestations of the withdrawal syndrome to known drugs of abuse. Nevertheless, the existence of a withdrawal syndrome after prolonged cannabinoid administration has long been a controversial issue. Recent studies, in different species, have shown that withdrawal to prolonged cannabinoid exposure precipitated by the cannabinoid antagonist SR141716A is characterized by physical signs underlying impairment of motor coordination. Interestingly, cannabinoid withdrawal is accompanied by an increase of adenylyl cyclase activity in the cerebellum. Here, we investigate the functional role of the cyclic AMP pathway in the cerebellum in the establishment of cannabinoid withdrawal. We show that after SR141716A precipitation of cannabinoid withdrawal, basal and calcium-calmodulin-stimulated adenylyl cyclase activities as well as active PKA in the cerebellum increase in a transient manner with a temporal profile which matches that of the somatic expression of abstinence. Selectively blocking the up-regulation of the cyclic AMP pathway in the cerebellum, by microinfusing the cyclic AMP blocker Rp-8Br-cAMPS in this region, markedly reduced both PKA activation and the somatic expression of cannabinoid withdrawal. Our results (i) directly link the behavioural manifestations of cannabinoid withdrawal with the up-regulation of the cyclic AMP pathway in the cerebellum, pointing towards common molecular adaptive mechanisms for dependence and withdrawal to most drugs of abuse; (ii) suggest a particular role for the cerebellum as a major neurobiological substrate for cannabinoid withdrawal.


Subject(s)
Cerebellum/enzymology , Cyclic AMP-Dependent Protein Kinases/metabolism , Dronabinol/adverse effects , Hallucinogens/adverse effects , Substance Withdrawal Syndrome/enzymology , 8-Bromo Cyclic Adenosine Monophosphate/administration & dosage , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Adenylyl Cyclases/metabolism , Animals , Behavior, Animal/drug effects , Cyclic AMP/antagonists & inhibitors , Cyclic AMP/physiology , Dronabinol/antagonists & inhibitors , Enzyme Activation/physiology , Hallucinogens/antagonists & inhibitors , Injections , Injections, Intraventricular , Male , Mice , Piperidines/pharmacology , Pyrazoles/pharmacology , Receptors, Drug/antagonists & inhibitors , Rimonabant , Stereotaxic Techniques , Substance Withdrawal Syndrome/prevention & control , Substance Withdrawal Syndrome/psychology , Up-Regulation/drug effects
11.
Biochem Biophys Res Commun ; 250(3): 720-5, 1998 Sep 29.
Article in English | MEDLINE | ID: mdl-9784412

ABSTRACT

The recently discovered endogenous mu receptor selective endomorphin 2 was prepared in tritiated form by a catalytic dehalogenation method resulting in a specific radioactivity of 1.98 TBq/mmol (53.4 Ci/mmol), and used for in vitro labelling of rat brain membranes. The binding was saturable, stereospecific and of high affinity (Kd: 0.97 and 1.12 nM based on kinetic and equilibrium binding studies, respectively). The maximal number of binding sites (Bmax) was found to be 114.8 fmol/mg protein. [3H]Endomorphin 2 was displaced by mu-receptor selective specific peptides and heterocyclic compounds with high affinity, whereas kappa and delta receptor specific ligands were much less potent. The Ki values of endomorphin 1 and 2 in inhibiting [3H]naloxone binding increased by 15-fold in the presence of 100 mM NaCl which indicates the agonist property of these peptides. Endomorphins stimulated [35S]GTPgammaS binding and inhibited adenylyl cyclase activity which also provides evidence for the agonist character of endomorphins.


Subject(s)
Analgesics, Opioid/pharmacology , Brain/metabolism , Cell Membrane/metabolism , Oligopeptides/pharmacology , Receptors, Opioid, mu/agonists , Signal Transduction/drug effects , Animals , Radioligand Assay , Rats , Rats, Wistar
12.
Br J Pharmacol ; 125(7): 1567-77, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9884086

ABSTRACT

Tolerance and dependence induced by chronic delta-9-tetrahydrocannabinol (THC) administration were investigated in mice. The effects on body weight, analgesia and hypothermia were measured during 6 days of treatment (10 or 20 mg kg(-1) THC twice daily). A rapid tolerance to the acute effects was observed from the second THC administration. The selective CB-1 receptor antagonist SR 141716A (10 mg kg(-1)) was administered at the end of the treatment, and somatic and vegetative manifestations of abstinence were evaluated. SR 141716A administration precipitated several somatic signs that included wet dog shakes, frontpaw tremor, ataxia, hunched posture, tremor, ptosis, piloerection, decreased locomotor activity and mastication, which can be interpreted as being part of a withdrawal syndrome. Brains were removed immediately after the behavioural measures and assayed for adenylyl cyclase activity. An increase in basal, forskolin and calcium/calmodulin stimulated adenylyl cyclase activities was specifically observed in the cerebellum of these mice. The motivational effects of THC administration and withdrawal were evaluated by using the place conditioning paradigm. No conditioned change in preference to withdrawal associated environment was observed. In contrast, a conditioned place aversion was produced by the repeated pairing of THC (20 mg kg(-1)), without observing place preference at any of the doses used. This study constitutes a clear behavioural and biochemical model of physical THC withdrawal with no motivational aversive consequences. This model permits an easy quantification of THC abstinence in mice and can be useful for the elucidation of the molecular mechanisms involved in cannabinoid dependence.


Subject(s)
Dronabinol/pharmacology , Hallucinogens/pharmacology , Substance Withdrawal Syndrome/physiopathology , Analysis of Variance , Animals , Body Temperature/drug effects , Body Weight/drug effects , Conditioning, Psychological/drug effects , Cyclic AMP/metabolism , Drug Interactions , Hypothermia/chemically induced , In Vitro Techniques , Male , Mice , Motivation , Narcotics/pharmacology , Pain Measurement/drug effects , Piperidines/pharmacology , Pyrazoles/pharmacology , Rimonabant
14.
Eur J Neurosci ; 8(12): 2671-8, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8996817

ABSTRACT

The specific participation of protein kinases in the expression of the somatic signs of morphine withdrawal has been previously demonstrated, suggesting that changes in intracellular signalling systems are involved in opioid addiction. In the present study, the involvement of protein kinases in the aversive/dysphoric effects of morphine abstinence has been investigated in the nucleus accumbens, because of the critical role played by the mesolimbic system in the rewarding effects of opioids. Rats were chronically treated with morphine, twice a day for 5 days, with doses progressively increased from 5 to 30 mg/kg (i.p.). In addition, microinjections into the nucleus accumbens of the serine-threonine kinase inhibitors H7 or H8 (1 or 10 nmol per side) or saline once daily were also given, both in control and in morphine-treated animals. After these chronic treatments, withdrawal syndrome was induced by naloxone administration (0.1 mg/kg, s.c.), and the motivational component of morphine abstinence was studied using the place aversion paradigm. When administered at the highest dose (10 nmol), H7 and H8 strongly reduced the place aversion induced by naloxone in morphine dependent animals. Protein kinase inhibitors did not induce significant behavioural responses in non-dependent animals. Chronic morphine treatment induced a selective up-regulation of adenylate cyclase activity in the amygdala, without affecting other brain regions. The morphine-increased adenylate cyclase activity in amygdala was reversed by the chronic intra-accumbens microinjections of H7 and H8. These results suggest that serine-threonine kinases in the nucleus accumbens play an important role in the emotional/dysphoric properties which characterize opiate withdrawal.


Subject(s)
Morphine/adverse effects , Nucleus Accumbens/enzymology , Protein Kinases/physiology , Substance Withdrawal Syndrome/physiopathology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Adenylyl Cyclases/metabolism , Animals , Avoidance Learning/drug effects , Behavior, Animal/drug effects , Conditioning, Psychological/drug effects , Enzyme Inhibitors/pharmacology , Injections , Isoquinolines/pharmacology , Male , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Nucleus Accumbens/physiology , Protein Kinase Inhibitors , Rats , Rats, Wistar , Substance Withdrawal Syndrome/psychology
15.
Proc Natl Acad Sci U S A ; 93(20): 11208-12, 1996 Oct 01.
Article in English | MEDLINE | ID: mdl-8855334

ABSTRACT

Nocturnal melatonin production in the pineal gland is under the control of norepinephrine released from superior cervical ganglia afferents in a rhythmic manner, and of cyclic AMP. Cyclic AMP increases the expression of serotonin N-acetyltransferase and of inducible cAMP early repressor that undergo circadian oscillations crucial for the maintenance and regulation of the biological clock. In the present study, we demonstrate a circadian pattern of expression of the calcium/calmodulin activated adenylyl cyclase type 1 (AC1) mRNA in the rat pineal gland. In situ hybridization revealed that maximal AC1 mRNA expression occurred at midday (12:00-15:00), with a very low signal at night (0:00-3:00). We established that this rhythmic pattern was controlled by the noradrenergic innervation of the pineal gland and by the environmental light conditions. Finally, we observed a circadian responsiveness of the pineal AC activity to calcium/calmodulin, with a lag due to the processing of the protein. At midday, AC activity was inhibited by calcium (40%) either in the presence or absence of calmodulin, while at night the enzyme was markedly (3-fold) activated by the calcium-calmodulin complex. These findings suggest (i) the involvement of AC1 acting as the center of a gating mechanism, between cyclic AMP and calcium signals, important for the fine tuning of the pineal circadian rhythm; and (ii) a possible regulation of cyclic AMP on the expression of AC1 in the rat pineal gland.


Subject(s)
Adenylyl Cyclases/metabolism , Circadian Rhythm , Membrane Proteins , Nerve Tissue Proteins , Pineal Gland/enzymology , Animals , Calcium/physiology , Calmodulin/physiology , Gene Expression Regulation, Enzymologic/radiation effects , In Situ Hybridization , Light , Male , Norepinephrine/physiology , RNA, Messenger/genetics , Rats , Rats, Wistar
16.
Nature ; 383(6603): 819-23, 1996 Oct 31.
Article in English | MEDLINE | ID: mdl-8893006

ABSTRACT

Despite tremendous efforts in the search for safe, efficacious and non-addictive opioids for pain treatment, morphine remains the most valuable painkiller in contemporary medicine. Opioids exert their pharmacological actions through three opioid-receptor classes, mu, delta and kappa, whose genes have been cloned. Genetic approaches are now available to delineate the contribution of each receptor in opioid function in vivo. Here we disrupt the mu-opioid-receptor gene in mice by homologous recombination and find that there are no overt behavioural abnormalities or major compensatory changes within the opioid system in these animals. Investigation of the behavioural effects of morphine reveals that a lack of mu receptors abolishes the analgesic effect of morphine, as well as place-preference activity and physical dependence. We observed no behavioural responses related to delta- or kappa-receptor activation with morphine, although these receptors are present and bind opioid ligands. We conclude that the mu-opioid-receptor gene product is the molecular target of morphine in vivo and that it is a mandatory component of the opioid system for morphine action.


Subject(s)
Analgesics/pharmacology , Morphine/pharmacology , Narcotics/pharmacology , Receptors, Opioid, mu/metabolism , Analgesics/adverse effects , Analgesics/metabolism , Animals , Behavior, Animal , Cell Line , Enkephalin, Ala(2)-MePhe(4)-Gly(5)- , Enkephalins/metabolism , Gene Deletion , Mice , Mice, Inbred C57BL , Mice, Knockout , Morphine/adverse effects , Morphine/metabolism , Morphine Dependence/metabolism , Narcotics/adverse effects , Narcotics/metabolism , Receptors, Opioid, delta/metabolism , Receptors, Opioid, kappa/metabolism , Receptors, Opioid, mu/genetics , Reward , Substance Withdrawal Syndrome/metabolism
17.
Science ; 273(5275): 657-9, 1996 Aug 02.
Article in English | MEDLINE | ID: mdl-8662559

ABSTRACT

Chronic morphine administration induces an up-regulation of several components of the cyclic adenosine 5'-monophosphate (cAMP) signal transduction cascade. The behavioral and biochemical consequences of opiate withdrawal were investigated in mice with a genetic disruption of the alpha and Delta isoforms of the cAMP-responsive element-binding protein (CREB). In CREBalphadelta mutant mice the main symptoms of morphine withdrawal were strongly attenuated. No change in opioid binding sites or in morphine-induced analgesia was observed in these mutant mice, and the increase of adenylyl cyclase activity and immediate early gene expression after morphine withdrawal was normal. Thus, CREB-dependent gene transcription is a factor in the onset of behavioral manifestations of opiate dependence.


Subject(s)
Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/physiology , Morphine Dependence/etiology , Morphine/administration & dosage , Substance Withdrawal Syndrome/physiopathology , Adenylyl Cyclases/metabolism , Analgesia , Animals , Behavior, Animal , Cerebral Cortex/enzymology , Drug Tolerance , Gene Expression Regulation , Gene Targeting , Genes, Immediate-Early , Locus Coeruleus/metabolism , Mice , Morphine/adverse effects , Morphine/pharmacology , Morphine Dependence/metabolism , Mutation , Naloxone/pharmacology , Receptors, Opioid/metabolism , Signal Transduction
18.
Neurochem Res ; 20(1): 17-22, 1995 Jan.
Article in English | MEDLINE | ID: mdl-7739754

ABSTRACT

Rat hippocampal slices preloaded with D-[3H]aspartate, a non metabolizable analogue of L-glutamate, were superfused with artificial CSF. Depolarization was induced by 53.5 mM K+, in the presence of Ca2+ (1.3 mM) or Mg2+ (5 mM) to determine the Ca2+ dependent release. Haloperidol added in the superfusion medium at 100 microM reduced by about 60% the Ca2+ dependent release of D-[3H]aspartate. This drug at 20 microM or 100 microM inhibited the non-activated glutamate dehydrogenase (GDH) but had no effect on GDH activated by ADP (2 mM) or leucine (5 mM). In addition no effect was observed on phosphate activated glutaminase (PAG) in the presence either of 20 mM or 5 mM phosphate. These results indicate that the effect of haloperidol is exerted on presynaptic mechanisms regulating neurotransmitter release.


Subject(s)
Aspartic Acid/metabolism , Calcium/pharmacology , Haloperidol/pharmacology , Hippocampus/drug effects , Potassium/pharmacology , Animals , Glutamic Acid/analogs & derivatives , Glutamic Acid/metabolism , Hippocampus/metabolism , In Vitro Techniques , Male , Rats , Rats, Wistar , Synaptic Transmission/drug effects , Tritium
SELECTION OF CITATIONS
SEARCH DETAIL
...