Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 511
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Neurosci ; 36(18): 5160-9, 2016 05 04.
Article in English | MEDLINE | ID: mdl-27147666

ABSTRACT

UNLABELLED: Schizophrenia-related psychosis is associated with disturbances in mesolimbic dopamine (DA) transmission, characterized by hyperdopaminergic activity in the mesolimbic pathway. Currently, the only clinically effective treatment for schizophrenia involves the use of antipsychotic medications that block DA receptor transmission. However, these medications produce serious side effects leading to poor compliance and treatment outcomes. Emerging evidence points to the involvement of a specific phytochemical component of marijuana called cannabidiol (CBD), which possesses promising therapeutic properties for the treatment of schizophrenia-related psychoses. However, the neuronal and molecular mechanisms through which CBD may exert these effects are entirely unknown. We used amphetamine (AMPH)-induced sensitization and sensorimotor gating in rats, two preclinical procedures relevant to schizophrenia-related psychopathology, combined with in vivo single-unit neuronal electrophysiology recordings in the ventral tegmental area, and molecular analyses to characterize the actions of CBD directly in the nucleus accumbens shell (NASh), a brain region that is the current target of most effective antipsychotics. We demonstrate that Intra-NASh CBD attenuates AMPH-induced sensitization, both in terms of DAergic neuronal activity measured in the ventral tegmental area and psychotomimetic behavioral analyses. We further report that CBD controls downstream phosphorylation of the mTOR/p70S6 kinase signaling pathways directly within the NASh. Our findings demonstrate a novel mechanism for the putative antipsychotic-like properties of CBD in the mesolimbic circuitry. We identify the molecular signaling pathways through which CBD may functionally reduce schizophrenia-like neuropsychopathology. SIGNIFICANCE STATEMENT: The cannabis-derived phytochemical, cannabidiol (CBD), has been shown to have pharmacotherapeutic efficacy for the treatment of schizophrenia. However, the mechanisms by which CBD may produce antipsychotic effects are entirely unknown. Using preclinical behavioral procedures combined with molecular analyses and in vivo neuronal electrophysiology, our findings identify a functional role for the nucleus accumbens as a critical brain region whereby CBD can produce effects similar to antipsychotic medications by triggering molecular signaling pathways associated with the effects of classic antipsychotic medications. Specifically, we report that CBD can attenuate both behavioral and dopaminergic neuronal correlates of mesolimbic dopaminergic sensitization, via a direct interaction with mTOR/p70S6 kinase signaling within the mesolimbic pathway.


Subject(s)
Amphetamine/antagonists & inhibitors , Behavior, Animal/drug effects , Cannabidiol/pharmacology , Central Nervous System Stimulants/antagonists & inhibitors , Dopamine Uptake Inhibitors/antagonists & inhibitors , Dopaminergic Neurons/drug effects , Limbic System/physiology , Neural Pathways/drug effects , Neurons/drug effects , Ribosomal Protein S6 Kinases, 70-kDa/drug effects , TOR Serine-Threonine Kinases/drug effects , Amphetamine/pharmacology , Animals , Central Nervous System Stimulants/pharmacology , Dopamine Uptake Inhibitors/pharmacology , Male , Motor Activity/drug effects , Nucleus Accumbens/drug effects , Rats , Rats, Sprague-Dawley , Reflex, Startle/drug effects
2.
J Neurosci ; 34(25): 8499-506, 2014 Jun 18.
Article in English | MEDLINE | ID: mdl-24948805

ABSTRACT

Drug addiction has devastating consequences on social behaviors and can lead to the impairment of social bonding. Accumulating evidence indicates that alterations in oxytocin (OT) and dopamine (DA) neurotransmission within brain reward circuitry may be involved. We investigated this possibility, as well as the therapeutic potential of OT for drug-induced social deficits, using the prairie vole (Microtus ochrogaster)-a socially monogamous rodent that forms enduring pair bonds between adult mates. We demonstrate that repeated exposure to the commonly abused psychostimulant amphetamine (AMPH) inhibits the formation of partner preferences (an index of pair bonding) in female prairie voles. AMPH exposure also altered OT and DA neurotransmission in regions that mediate partner preference formation: it decreased OT and DA D2 receptor immunoreactivity in the medial prefrontal cortex (mPFC) and nucleus accumbens (NAcc), respectively, and increased NAcc DA levels. Administration of OT directly into the mPFC of AMPH-exposed voles restored partner preferences, and altered NAcc DA levels, and this effect was dependent on OT receptor activation. Together, these data suggest that repeated AMPH exposure impairs pair bonding through an OT-mediated mechanism, and that OT and DA systems within brain reward circuitry may interact to mediate the complex relationship between drug abuse and social bonding. Further, these results provide empirical support for the idea that the central OT system may represent an important target for the treatment of social deficits in addiction.


Subject(s)
Amphetamine-Related Disorders/metabolism , Amphetamine/toxicity , Dopamine/metabolism , Nucleus Accumbens/metabolism , Oxytocin/physiology , Pair Bond , Social Behavior , Amphetamine/antagonists & inhibitors , Amphetamine/metabolism , Animals , Arvicolinae , Female , Male , Microdialysis/methods , Oxytocin/administration & dosage
3.
Proc Natl Acad Sci U S A ; 109(30): 12165-70, 2012 Jul 24.
Article in English | MEDLINE | ID: mdl-22689948

ABSTRACT

Neuregulin 1 (NRG1) and ErbB4, critical neurodevelopmental genes, are implicated in schizophrenia, but the mediating mechanisms are unknown. Here we identify a genetically regulated, pharmacologically targetable, risk pathway associated with schizophrenia and with ErbB4 genetic variation involving increased expression of a PI3K-linked ErbB4 receptor (CYT-1) and the phosphoinositide 3-kinase subunit, p110δ (PIK3CD). In human lymphoblasts, NRG1-mediated phosphatidyl-inositol,3,4,5 triphosphate [PI(3,4,5)P3] signaling is predicted by schizophrenia-associated ErbB4 genotype and PIK3CD levels and is impaired in patients with schizophrenia. In human brain, the same ErbB4 genotype again predicts increased PIK3CD expression. Pharmacological inhibition of p110δ using the small molecule inhibitor, IC87114, blocks the effects of amphetamine in a mouse pharmacological model of psychosis and reverses schizophrenia-related phenotypes in a rat neonatal ventral hippocampal lesion model. Consistent with these antipsychotic-like properties, IC87114 increases AKT phosphorylation in brains of treated mice, implicating a mechanism of action. Finally, in two family-based genetic studies, PIK3CD shows evidence of association with schizophrenia. Our data provide insight into a mechanism of ErbB4 association with schizophrenia; reveal a previously unidentified biological and disease link between NRG1-ErbB4, p110δ, and AKT; and suggest that p110δ is a previously undescribed therapeutic target for the treatment of psychiatric disorders.


Subject(s)
Adenine/analogs & derivatives , ErbB Receptors/metabolism , Neuregulin-1/metabolism , Phosphoinositide-3 Kinase Inhibitors , Quinazolines/pharmacology , Schizophrenia/genetics , Schizophrenia/metabolism , Signal Transduction/physiology , Adenine/chemistry , Adenine/pharmacology , Amphetamine/antagonists & inhibitors , Analysis of Variance , Animals , Antipsychotic Agents/pharmacology , B-Lymphocytes , Blotting, Western , Cell Line, Transformed , Class I Phosphatidylinositol 3-Kinases , ErbB Receptors/genetics , Flow Cytometry , Genetic Association Studies , Humans , Mice , Molecular Structure , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Quinazolines/chemistry , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Receptor, ErbB-4 , Reverse Transcriptase Polymerase Chain Reaction , Schizophrenia/drug therapy
4.
J Sleep Res ; 22(5): 581-8, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23879373

ABSTRACT

The objective of the present study was to further elucidate the mechanisms involved in the wake-promoting effects of psychomotor stimulants. Many previous studies have tightly linked the effects of stimulants to dopamine neurotransmission, and some studies indicate that serotonin 2A receptors modulate these effects. However, the role of dopamine in arousal is controversial, most notably because dopamine neurons do not change firing rates across arousal states. In the present study, we examined the wake-promoting effects of the dopamine-releaser amphetamine using non-invasive telemetric monitoring. These effects were evaluated in rhesus monkeys as a laboratory animal model with high translational relevance for human disorders of sleep and arousal. To evaluate the role of dopamine in the wake-promoting effects of amphetamine, we used in vivo microdialysis targeting the caudate nucleus, as this approach provides clearly interpretable measures of presynaptic dopamine release. This is beneficial in the present context because some of the inconsistencies between previous studies examining the role of dopamine in arousal may be related to differences between postsynaptic dopamine receptors. We found that amphetamine significantly and dose-dependently increased arousal at doses that engendered higher extracellular dopamine levels. Moreover, antagonism of serotonin 2A receptors attenuated the effects of amphetamine on both wakefulness and dopamine overflow. These findings further elucidate the role of dopamine and serotonin 2A receptors in arousal, and they suggest that increased dopamine neurotransmission may be necessary for the wake-promoting effects of amphetamine, and possibly other stimulants.


Subject(s)
Amphetamine/antagonists & inhibitors , Amphetamine/pharmacology , Arousal/drug effects , Dopamine/metabolism , Receptor, Serotonin, 5-HT2A/metabolism , Serotonin 5-HT2 Receptor Antagonists/pharmacology , Animals , Arousal/physiology , Caudate Nucleus/drug effects , Caudate Nucleus/metabolism , Central Nervous System Stimulants/pharmacology , Circadian Rhythm/drug effects , Circadian Rhythm/physiology , Dopamine Uptake Inhibitors/pharmacology , Female , Fluorobenzenes/pharmacology , Macaca mulatta , Microdialysis , Piperidines/pharmacology , Receptors, Dopamine/metabolism , Sleep/drug effects , Sleep/physiology , Telemetry , Wakefulness/drug effects , Wakefulness/physiology
5.
Int J Neuropsychopharmacol ; 15(9): 1275-85, 2012 Oct.
Article in English | MEDLINE | ID: mdl-21939588

ABSTRACT

Recent studies indicate that bipolar disorder is associated with mitochondrial dysfunction and oxidative stress. Previous studies in our laboratory have shown that the mood stabilizer lithium inhibits oxidative stress. The α,ß-unsaturated aldehyde 4-hydroxy-2-nonenal (4-HNE), a major product of lipid peroxidation, is able to exert cytotoxicity and disturb cellular function by forming protein adducts. The purpose of this study is to determine whether chronic lithium treatment prevents 4-HNE-protein adduction in an amphetamine-induced hyperactive mania-like model. We found that repeated amphetamine stimulation significantly induced hyperactive behaviour, decreased activities of mitochondrial complexes I and III, and increased 4-HNE-protein adducts in rat frontal cortex, and that chronic lithium treatment inhibited both amphetamine-induced hyperactivity and 4-HNE-protein adduction. Monoamine neurotransmitters are involved in the aetiology and pathology of bipolar disorder and other psychiatric diseases, and also contribute significantly to amphetamine-induced behavioural effects. Vesicular monoamine transporter 2 (VMAT2) is critical in packaging monoamine neurotransmitters. We found that 4-HNE can form protein adducts with VMAT2. Repeated amphetamine stimulation significantly increased 4-HNE-VMAT2 adducts, while chronic lithium treatment reduced amphetamine-increased 4-HNE-VMAT2 adducts in rat frontal cortex. Our findings suggest that chronic lithium treatment may inhibit amphetamine-induced hyperactive mania-like behaviour by preventing 4-HNE-VMAT2 adduction. This finding also indicates that prevention of 4-HNE-VMAT2 adduction may contribute in part to the pharmacological action of lithium for the treatment of bipolar disorder.


Subject(s)
Aldehydes/metabolism , Amphetamine/antagonists & inhibitors , Amphetamine/pharmacology , Antimanic Agents/pharmacology , Central Nervous System Stimulants/antagonists & inhibitors , Central Nervous System Stimulants/pharmacology , Lithium/pharmacology , Nerve Tissue Proteins/metabolism , Prefrontal Cortex/metabolism , Animals , Bipolar Disorder/chemically induced , Bipolar Disorder/metabolism , Blotting, Western , Electron Transport Complex I/metabolism , Electron Transport Complex III/metabolism , Immunohistochemistry , Immunoprecipitation , Male , Motor Activity/drug effects , Prefrontal Cortex/drug effects , Rats , Rats, Sprague-Dawley , Vesicular Monoamine Transport Proteins/metabolism
6.
J Neurosci ; 30(50): 17051-62, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21159975

ABSTRACT

L-type Ca(2+) channel (LTCC)-activated signaling cascades contribute significantly to psychostimulant-induced locomotor sensitization; however, the precise contribution of the two brain-specific subunits Ca(v)1.2 and Ca(v)1.3 remains mostly unknown. In this study, by using amphetamine and cocaine locomotor sensitization in mutant mice expressing dihydropyridine (DHP)-insensitive Ca(v)1.2 LTCCs (Ca(v)1.2DHP(-/-)), we find that, as opposed to a previously identified role of the Ca(v)1.3 subunit of LTCCs in development of sensitization, the Ca(v)1.2 subunit mediates expression of amphetamine and cocaine sensitization when examined after a 14 d drug-free period. Molecular studies to further elucidate the role of Ca(v)1.2 versus Ca(v)1.3 LTCCs in activating signaling pathways in the nucleus accumbens (NAc) of drug-naive versus drug-preexposed mice examined 14 d later revealed that an acute amphetamine and cocaine challenge in drug-naive mice increases Ser133 cAMP response element-binding protein (CREB) phosphorylation in the NAc via Ca(v)1.3 channels and via a dopamine D(1)-dependent mechanism, independent of the extracellular signal-regulated kinase (ERK) pathway, an important mediator of psychostimulant-induced plasticity. In contrast, in amphetamine- and cocaine-preexposed mice, an amphetamine or cocaine challenge no longer activates CREB unless Ca(v)1.2 LTCCs are blocked. This Ca(v)1.2-dependent blunting of CREB activation that underlies expression of locomotor sensitization occurs only after extended drug-free periods and involves recruitment of D(1) receptors and the ERK pathway. Thus, our results demonstrate that specific LTCC subunits are required for the development (Ca(v)1.3) versus expression (Ca(v)1.2) of psychostimulant sensitization and that subunit-specific signaling pathways recruited by psychostimulants underlies long-term drug-induced behavioral responses.


Subject(s)
Amphetamine/pharmacology , Calcium Channels, L-Type/drug effects , Central Nervous System Stimulants/pharmacology , Cocaine/pharmacology , Motor Activity/drug effects , Nifedipine/pharmacology , Nucleus Accumbens/drug effects , Signal Transduction/drug effects , Aminoacetonitrile/analogs & derivatives , Aminoacetonitrile/pharmacology , Amphetamine/antagonists & inhibitors , Animals , Benzazepines/pharmacology , Calcium Channels, L-Type/genetics , Central Nervous System Stimulants/antagonists & inhibitors , Cocaine/antagonists & inhibitors , Cyclic AMP Response Element-Binding Protein/metabolism , Drug Interactions , Male , Mice , Mice, Knockout , Motor Activity/genetics , Nucleus Accumbens/metabolism
7.
Synapse ; 65(6): 505-12, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20936684

ABSTRACT

It has been shown that a single exposure to amphetamine is sufficient to induce long-term behavioral, neurochemical, and neuroendocrine sensitization in rats. Dopaminergic neurotransmission in the nucleus accumbens and the caudate-putamen plays a critical role in the addictive properties of drugs of abuse. Angiotensin (Ang) II receptors are found on the soma and terminals of mesolimbic dopaminergic neurons and it has been shown that Ang II acting through its AT1 receptors facilitates dopamine release. The hypothesis was tested that Ang II AT1 receptors are involved in the neuroadaptative changes induced by a single exposure to amphetamine and that such changes are related to the development of behavioral and neurochemical sensitization. For this purpose, the study examined the expression of amphetamine-enhanced (0.5 mg kg⁻¹ i.p.) locomotor activity in animals pretreated with candesartan, an AT1 blocker, (3 mg kg⁻¹ p.o. x 5 days), 3 weeks after an amphetamine injection (5 mg kg⁻¹ i.p.). Dopaminergic hyperreactivity was tested by measuring the 3H-DA release in vitro from caudate-putamen and nucleus accumbens slices, induced by K+ stimulus. It was confirmed the behavioral sensitization in the two-injection protocol and candesartan pretreatment attenuate this response. It was also found that AT1 blockade pretreatment did not affect the locomotor response to dopamine agonists. In respect to the neurochemical sensitization tested using ex vivo 3H-DA release experiments it was found that AT1 receptor pretreatment blunted the enhanced response induced by K+ stimulus. The results support the idea that the development of neuroadaptive changes induced by amphetamine involves brain AT1 Ang II receptor activation.


Subject(s)
Amphetamine-Related Disorders/metabolism , Amphetamine-Related Disorders/psychology , Amphetamine/toxicity , Angiotensin II Type 1 Receptor Blockers/pharmacology , Behavior, Animal/drug effects , Receptor, Angiotensin, Type 1/metabolism , Amphetamine/antagonists & inhibitors , Amphetamine-Related Disorders/physiopathology , Animals , Behavior, Animal/physiology , Disease Models, Animal , Injections, Intraperitoneal , Male , Organ Culture Techniques , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1/physiology
8.
Neuropharmacology ; 197: 108754, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34389398

ABSTRACT

Alzheimer's disease (AD) is a profoundly debilitating neurodegenerative disorder characterized most notably by progressive cognitive decline, but also agitation and behavioral disturbances that are extremely disruptive to patient and caregiver. Current pharmacological treatments for these symptoms have limited efficacy and significant side effects. We have recently reported the discovery of Compound 24, an M4 positive allosteric modulator (PAM) that is potent, highly selective, and devoid of cholinergic-like side effects in rats. In order to further evaluate the translatability of the effects of compound 24 in primates, here we describe the effect of Compound 24 on three behavioral and cognition assays in rhesus monkeys, the stimulant induced motor activity (SIMA) assay, the object retrieval detour task (ORD), and the visuo-spatial paired-associates learning (vsPAL) task. As far as we know, this is the first such characterization of an M4 PAM in non-human primate. Compound 24 and the clinical standard olanzapine attenuated amphetamine induced hyperactivity to a similar degree. In addition, Compound 24 demonstrated procognitive effects in scopolamine-impaired ORD and vsPAL, and these effects were of similar magnitude to donepezil. These findings suggest that M4 PAMs may be beneficial to diseases such as Alzheimer's disease and schizophrenia, which are marked by behavioral disturbances as well as deficits in cognitive function.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/psychology , Behavior, Animal/drug effects , Cholinergic Agents/pharmacology , Cognition Disorders/drug therapy , Receptor, Muscarinic M4/drug effects , Schizophrenia/drug therapy , Schizophrenic Psychology , Amphetamine/antagonists & inhibitors , Amphetamine/pharmacology , Animals , Association Learning/drug effects , Central Nervous System Stimulants , Cholinergic Agents/pharmacokinetics , Cognition Disorders/psychology , Hyperkinesis/chemically induced , Hyperkinesis/prevention & control , Macaca mulatta , Male , Motor Activity/drug effects , Olanzapine/pharmacology , Orientation/drug effects
10.
Science ; 172(3980): 281-4, 1971 Apr 16.
Article in English | MEDLINE | ID: mdl-5548711

ABSTRACT

Injection of norepinephrine in the lateral ventricles of rats recovering from lateral hypothalamic anorexia caused immediate feeding and, frequently, overeating. Intraventricular administration of the alpha-noradrenergic blocker, phentolamine, suppressed feeding in both normal rats and rats that had recovered from lateral hypothalamic lesions. Feeding is reinforced by ascending medial forebrain bundle fibers that form alpha-noradrenergic synapses in the hypothalamus and forebrain. Damage to these fibers suppresses feeding by reducing noradrenergic transmission and, hence, the rewarding value of food. Recovery of feeding after hypothalamic lesions coincides with the recovery of noradrenergic reward function.


Subject(s)
Appetite/drug effects , Feeding Behavior , Hypothalamus/physiology , Norepinephrine/pharmacology , Amphetamine/antagonists & inhibitors , Animals , Diencephalon/physiology , Isotonic Solutions , Milk , Norepinephrine/physiology , Phentolamine/pharmacology , Propranolol/pharmacology , Rats , Reward , Self Stimulation , Synapses/physiology , Telencephalon/physiology
11.
J Pharmacol Exp Ther ; 326(1): 286-95, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18441249

ABSTRACT

Previous studies identified partial inhibitors and allosteric modulators of 5-hydroxytryptamine ([5-amino-3-(3,4-dichlorophenyl)-1,2-dihydropyrido[3,4-b]pyrazin-7-yl]carbamic acid ethyl ester [SoRI-6238], 4-(2-[bis(4-fluorophenyl)methoxy]ethyl)-1-(2-trifluoromethyl-benzyl)-piperidine [TB-1-099]) and dopamine transporters N-(diphenylmethyl)-2-phenyl-4-quinazolinamine, [SoRI-9804]). We report here the identification of three novel allosteric modulators of the dopamine transporter [N-(2,2-diphenylethyl)-2-phenyl-4-quinazolinamine [SoRI-20040], N-(3,3-diphenylpropyl)-2-phenyl-4-quinazolinamine [SoRI-20041], and [4-amino-6-[(diphenylmethyl)amino]-5-nitro-2-pyridinyl]carbamic acid ethyl ester [SoRI-2827]]. Membranes were prepared from human embryonic kidney cells expressing the cloned human dopamine transporter (hDAT). [(125)I]3beta-(4'-Iodophenyl)tropan-2beta-carboxylic acid methyl ester ([(125)I]RTI-55) binding and other assays followed published procedures. SoRI-20040, SoRI-20041, and SoRI-2827 partially inhibited [(125)I]RTI-55 binding, with EC(50) values ranging from approximately 1.4 to 3 microM and E(max) values decreasing as the [(125)I]RTI-55 concentrations increased. All three compounds decreased the [(125)I]RTI-55 B(max) value and increased the apparent K(d) value in a manner well described by a sigmoid dose-response curve. In dissociation rate experiments, SoRI-20040 (10 microM) and SoRI-20041 (10 microM), but not SoRI-2827 (10 microM), slowed the dissociation of [(125)I]RTI-55 from hDAT by approximately 30%. Using rat brain synaptosomes, all three agents partially inhibited [(3)H]dopamine uptake, with EC(50) values ranging from 1.8 to 3.1 microM and decreased the V(max) value in a dose-dependent manner. SoRI-9804 and SoRI-20040 partially inhibited amphetamine-induced dopamine transporter-mediated release of [(3)H]1-methyl-4-phenylpyridinium ion from rat caudate synaptosomes in a dose-dependent manner. Viewed collectively, we report several compounds that allosterically modulate hDAT binding and function, and we identify novel partial inhibitors of amphetamine-induced dopamine release.


Subject(s)
Amphetamine/antagonists & inhibitors , Amphetamine/pharmacology , Dopamine Antagonists/pharmacology , Dopamine/metabolism , Vesicular Biogenic Amine Transport Proteins/antagonists & inhibitors , Vesicular Biogenic Amine Transport Proteins/metabolism , Animals , Cell Line , Dopamine Antagonists/chemistry , Humans , Male , Protein Binding/drug effects , Protein Binding/physiology , Rats , Rats, Sprague-Dawley
12.
Schizophr Res ; 105(1-3): 224-35, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18710798

ABSTRACT

INTRODUCTION: Amisulpride's high and selective affinity for dopamine D2/3 (Ki 1.3/2.4 nM) receptors, lack of affinity for serotonin receptors, and its unusually high therapeutic doses (400-800 mg/day) makes it unique among atypical antipsychotics and prompted us to compare its actions with other antipsychotics in animal models. METHODS: Amisulpride's effects on amphetamine and phencyclidine induced locomotor activity (AIL/PIL), conditioned avoidance response, catalepsy (CAT), subcortical Fos expression, and plasma prolactin was correlated to its time-course striatal D2/3 and prefrontal 5-HT2 receptor occupancy (D(2/3)/5-HT2RO); in comparison to haloperidol, clozapine, and risperidone. RESULTS: Unlike the atypicals clozapine and risperidone, amisulpride lacked 5-HT2RO and showed a 'delayed' pattern of D2/3RO: 43, 60 and 88% after 1, 2 and 6 h (100 mg/kg), respectively, despite a quick onset (1 h) and decline (6 h) of prolactin elevation. While haloperidol and risperidone were effective at D2RO>60%, clozapine at D2/3RO<50%, amisulpride was effective only when its D2RO exceeded 60% with a delayed latency and lasted longer than other antipsychotics. CAT was observed for haloperidol and risperidone when D2RO exceeded 80%, while in the case of amisulpride, CAT was not observed even when doses exceeded 90% D2/3RO. Amisulpride also displayed functional limbic selectivity in Fos expression like the other atypicals. CONCLUSIONS: Amisulpride's "delayed" functional profile on acute administration and the need for high doses is most likely due to its poor blood-brain-barrier penetration; however, it is distinct from other atypicals in showing low motor side-effects, activity against phencyclidine, and a mesolimbic preference, despite no action on serotonin receptors.


Subject(s)
Antipsychotic Agents/pharmacology , Brain/drug effects , Receptors, Dopamine/drug effects , Receptors, Serotonin/drug effects , Sulpiride/analogs & derivatives , Amisulpride , Amphetamine/antagonists & inhibitors , Amphetamine/pharmacology , Animals , Antipsychotic Agents/metabolism , Basal Ganglia/drug effects , Basal Ganglia/metabolism , Behavior, Animal/drug effects , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain/metabolism , Clozapine/metabolism , Clozapine/pharmacology , Conditioning, Psychological/drug effects , Haloperidol/metabolism , Haloperidol/pharmacology , Male , Models, Animal , Motor Activity/drug effects , Phencyclidine/antagonists & inhibitors , Phencyclidine/pharmacology , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Prolactin/blood , Prolactin/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Dopamine/metabolism , Receptors, Dopamine D2/drug effects , Receptors, Dopamine D2/metabolism , Receptors, Dopamine D3/drug effects , Receptors, Dopamine D3/metabolism , Receptors, Serotonin/metabolism , Risperidone/metabolism , Risperidone/pharmacology , Sulpiride/metabolism , Sulpiride/pharmacology
13.
Alcohol ; 42(2): 123-7, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18358991

ABSTRACT

Most drugs of abuse cause a locomotor stimulation, an effect, at least in part, mediated by increased accumbal dopamine (DA) overflow. Locomotor stimulation has been suggested to be a putative endophenotype for drug addiction. We therefore investigated the effects of aripiprazole, a partial DA D2-receptor agonist, on ethanol as well as amphetamine-induced locomotor stimulation. In the present series of experiments, we found that aripiprazole (1.25 mg/kg, intraperitoneally [i.p.]) antagonized ethanol (1.75 g/kg, i.p.) as well as amphetamine (2 mg/kg, i.p.)induced locomotor stimulation in mice. We suggest that this effect might be related to aripiprazole's ability to alleviate drug-induced hyperdopaminergia without causing hypodopaminergia. Given that altered DA functions in drug dependence have been observed, it may be suggested that aripiprazole could be a new treatment strategy for treatment of drug dependence.


Subject(s)
Amphetamine/antagonists & inhibitors , Antipsychotic Agents/pharmacology , Ethanol/antagonists & inhibitors , Motor Activity/drug effects , Piperazines/pharmacology , Quinolones/pharmacology , Animals , Aripiprazole , Male , Mice , Receptors, Dopamine D2/drug effects , Receptors, Dopamine D2/physiology
14.
Pharmacol Rep ; 60(4): 490-8, 2008.
Article in English | MEDLINE | ID: mdl-18799817

ABSTRACT

New hypotheses regarding affective disorders suggest a critical role for cellular resilience and plasticity. Bcl-2 is a central protein in these processes and is elevated by mood stabilizers and antidepressants. In previous studies, mice with targeted mutations of Bcl-2 showed anxiety-related behavioral changes. The present study further explored the relationship between Bcl-2 and behavior using mice with a targeted mutation but with a different background strain than previously tested. Bcl-2 heterozygous mice (B6;129S2-Bcl-2/J) were tested in models of depression, mania and anxiety. Compared to Wild Type (WT) controls, mutant mice showed behaviors modeling two facets of mania: increased reward seeking and amphetamine sensitization. Moreover, the sensitization was attenuated by chronic pretreatment with lithium. In contrast to previous data, the mutation did not affect measures of anxiety. Although data are still minimal, it supports additional studies of the role of Bcl-2 in affective and anxiety disorders. The importance of background strain in behavioral phenotypes of mutant mice is known and the current lack of effect on anxiety measures may be related to high baseline anxiety of WTanimals. More precise studies of Bcl-2 in affective and anxiety disorders will be possible when specific pharmacological modulators of Bcl-2 become available.


Subject(s)
Amphetamine/antagonists & inhibitors , Antimanic Agents/pharmacology , Disease Models, Animal , Genes, bcl-2/genetics , Lithium Chloride/pharmacology , Mood Disorders/genetics , Motor Activity/drug effects , Proto-Oncogene Proteins c-bcl-2/deficiency , Amphetamine/pharmacology , Animals , Male , Mice
15.
Neuropharmacology ; 140: 246-259, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30005976

ABSTRACT

Group II metabotropic glutamate receptors (mGluR2 and mGluR3) are implicated in a number of psychiatric disorders. They also control sleep-wake architecture and may offer novel therapeutic targets. However, the roles of the mGluR2 versus mGluR3 subtypes are not well understood. Here, we have taken advantage of the recently described mutant strain of Han Wistar rats, which do not express mGluR2 receptors, to investigate behavioural, sleep and EEG responses to mGluR2/3 ligands. The mGluR2/3 agonist, LY354740 (10 mg/kg), reversed amphetamine- and phencyclidine-induced locomotion and rearing behaviours in control Wistar but not in mGluR2 lacking Han Wistar rats. In control Wistar but not in Han Wistar rats the mGluR2/3 agonist LY379268 (3 & 10 mg/kg) induced REM sleep suppression with dose-dependent effects on wake and NREM sleep. By contrast, the mGluR2/3 antagonist LY3020371 (3 & 10 mg/kg) had wake-promoting effects in both rat strains, albeit smaller in the mGluR2-lacking Han Wistar rats, indicating both mGluR2 and mGluR3-mediated effects on wakefulness. LY3020371 enhanced wake cortical oscillations in the theta (4-9 Hz) and gamma (30-80 Hz) range in both Wistar and Han Wistar rat strains, whereas LY379268 reduced theta and gamma oscillations in control Wistar rats, with minimal effects in Han Wistar rats. Together these studies illustrate the significant contribution of mGluR2 to the antipsychotic-like, sleep and EEG effects of drugs acting on group II mGluRs. However, we also provide evidence of a role for mGluR3 activity in the control of sleep and wake cortical theta and gamma oscillations.


Subject(s)
Antipsychotic Agents/pharmacology , Receptors, Metabotropic Glutamate/physiology , Sleep/physiology , Wakefulness/drug effects , Wakefulness/physiology , Amino Acids/pharmacology , Amphetamine/antagonists & inhibitors , Amphetamine/pharmacology , Animals , Bridged Bicyclo Compounds/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cyclohexanes/pharmacology , Dose-Response Relationship, Drug , Excitatory Amino Acid Agonists/pharmacology , Gamma Rhythm/drug effects , Gamma Rhythm/physiology , Locomotion/drug effects , Locomotion/physiology , Male , Motor Activity/drug effects , Motor Activity/physiology , Mutation , Phencyclidine/antagonists & inhibitors , Phencyclidine/pharmacology , Rats , Receptors, Metabotropic Glutamate/deficiency , Receptors, Metabotropic Glutamate/genetics , Sleep/drug effects , Theta Rhythm/drug effects , Theta Rhythm/physiology
16.
Psychopharmacology (Berl) ; 235(7): 1887-1896, 2018 07.
Article in English | MEDLINE | ID: mdl-29572651

ABSTRACT

RATIONALE: We have recently shown that the benzodiazepine diazepam inhibits dopamine release in the NAc and blocks the increased release of dopamine induced by DL-amphetamine. Rewarding stimuli and many drugs of abuse can induce dopamine release in the nucleus accumbens as well as 50-kHz ultrasonic vocalizations (USVs) in rats. OBJECTIVES: In the present study, we tested the hypothesis that diazepam can also block the increase in locomotor activity and USVs elicited by amphetamine. METHODS: Fifty-kilohertz USVs, stereotypy, and locomotor behavior were scored in adult male Wistar rats treated with i.p. injections of saline, 3 mg/kg DL-amphetamine, 2 mg/kg diazepam, 0.2 mg/kg haloperidol, or a combination of these drugs. RESULTS: In agreement with previous studies, amphetamine caused significant increases in the number of USV calls, stereotypies, and locomotor activity. The D2 dopamine receptor antagonist haloperidol blocked the effects of amphetamine on USVs, stereotypy, and locomotor activity. Diazepam blocked the effect of amphetamine on USV and stereotypy, but not on horizontal locomotion. CONCLUSIONS: These results suggest that diazepam blocks the rewarding effect of amphetamine. This finding is promising for basic research regarding treatments of substance use disorders and evaluation of the impact of benzodiazepines on motivation.


Subject(s)
Amphetamine/pharmacology , Diazepam/pharmacology , Locomotion/drug effects , Stereotyped Behavior/drug effects , Ultrasonic Waves , Vocalization, Animal/drug effects , Amphetamine/antagonists & inhibitors , Animals , Dopamine/pharmacology , Dopamine Agents/pharmacology , GABA Modulators/pharmacology , Locomotion/physiology , Male , Motivation/drug effects , Motivation/physiology , Nucleus Accumbens/drug effects , Rats , Rats, Wistar , Receptors, Dopamine D1/antagonists & inhibitors , Stereotyped Behavior/physiology , Vocalization, Animal/physiology
17.
Psychopharmacology (Berl) ; 190(2): 201-11, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17119931

ABSTRACT

RATIONALE: Non-adherence with medication remains the major correctable cause of poor outcome in schizophrenia. However, few treatments have addressed this major determinant of outcome with novel long-term delivery systems. OBJECTIVES: The aim of this study was to provide biological proof of concept for a long-term implantable antipsychotic delivery system in rodents and rabbits. MATERIALS AND METHODS: Implantable formulations of haloperidol were created using biodegradable polymers. Implants were characterized for in vitro release and in vivo behavior using prepulse inhibition of startle in rats and mice, as well as pharmacokinetics in rabbits. RESULTS: Behavioral measures demonstrate the effectiveness of haloperidol implants delivering 1 mg/kg in mice and 0.6 mg/kg in rats to block amphetamine (10 mg/kg) in mice or apomorphine (0.5 mg/kg) in rats. Additionally, we demonstrate the pattern of release from single polymer implants for 1 year in rabbits. CONCLUSIONS: The current study suggests that implantable formulations are a viable approach to providing long-term delivery of antipsychotic medications in vivo using animal models of behavior and pharmacokinetics. In contrast to depot formulations, implantable formulations could last 6 months or longer. Additionally, implants can be removed throughout the delivery interval, offering a degree of reversibility not available with depot formulations.


Subject(s)
Antipsychotic Agents/administration & dosage , Antipsychotic Agents/pharmacokinetics , Arousal/drug effects , Haloperidol/administration & dosage , Haloperidol/pharmacokinetics , Inhibition, Psychological , Reflex, Startle/drug effects , Acoustic Stimulation , Amphetamine/antagonists & inhibitors , Amphetamine/pharmacology , Animals , Apomorphine/antagonists & inhibitors , Apomorphine/pharmacology , Arousal/physiology , Biocompatible Materials , Drug Implants , Lactic Acid , Long-Term Care , Male , Mice , Mice, Inbred C57BL , Polyglycolic Acid , Polylactic Acid-Polyglycolic Acid Copolymer , Polymers , Rabbits , Rats , Rats, Sprague-Dawley , Reflex, Startle/physiology
18.
Regul Pept ; 144(1-3): 6-9, 2007 Dec 04.
Article in English | MEDLINE | ID: mdl-17706801

ABSTRACT

CART peptide has been shown to regulate the actions of psychomotor stimulants. Here we have further investigated the role of the biologically active CART 55-102 peptide in the nucleus accumbens (NAcc) in the expression of behavioral sensitization by amphetamine (AMPH). Rats were pre-exposed 5 times to either saline or AMPH (1 mg/kg, i.p.). After 2 weeks of withdrawal, rats were microinjected into the NAcc with saline or CART 55-102 (1.0, or 2.5 microg/0.5 microl/side) followed by AMPH challenge (1 mg/kg, i.p.). The enhanced increase of locomotion and rearing produced by repeated AMPH pre-exposures was dose-dependently inhibited by microinjection into the NAcc of CART 55-102 peptide. These results indicate that CART 55-102 peptide in the NAcc can play a compensatory inhibitory role in the expression of behavioral sensitization by AMPH and further suggest that CART peptide may be a useful target to control the drug addiction by psychomotor stimulants.


Subject(s)
Amphetamine/antagonists & inhibitors , Behavior, Animal/drug effects , Central Nervous System Stimulants/antagonists & inhibitors , Nerve Tissue Proteins/pharmacology , Nucleus Accumbens/drug effects , Peptide Fragments/pharmacology , Animals , Behavior, Animal/physiology , Locomotion/drug effects , Locomotion/radiation effects , Male , Microinjections , Nerve Tissue Proteins/administration & dosage , Nucleus Accumbens/physiology , Peptide Fragments/administration & dosage , Rats , Rats, Sprague-Dawley
19.
Pharmacol Biochem Behav ; 159: 1-5, 2017 08.
Article in English | MEDLINE | ID: mdl-28642068

ABSTRACT

Drug dependence seems to involve a learning and memory process. Since learning and memory depend on protein synthesis, drug dependence may depend on protein synthesis, too. Drug-induced reward is a crucial effect for the development of drug-dependence. We used chloramphenicol (CAP, a protein synthesis inhibitor), to evaluate its effects on amphetamine (amph)-seeking behavior, on CB1R expression and on protein synthesis in general, in specific areas of the brain. Two groups of Wistar adult male rats were subjected to amph-induced conditioned place preference (CPP). Rats in group 1 received amph and were kept in the chamber for 30min. Once this period elapsed, they received a subcutaneous injection of saline (veh) and were returned to their home-cage. Rats in group 2 were also treated with amph but received CAP (150mg/kgsc) instead of saline. Once CPP was evaluated rats were sacrificed and the prefrontal cortex (PFC), the nucleus accumbens (NAcc) and the hippocampus (Hipp) were isolated and prepared for CB1R Western blot analysis. A vivarium reared group of rats was added as a non-experimentally manipulated control group. Results indicate that group 1 developed CPP while increasing CB1R expression in the NAcc. Group 2 did not develop CPP, had lower CB1R expression in the PFC and lacked the CB1R increase in the NAcc observed in the amph+veh group. These results support the notion that among the underlying mechanisms for amph-seeking reward is an increase in CB1R, further supporting an interaction between dopamine/endocannabinoids in CPP learning.


Subject(s)
Amphetamine/antagonists & inhibitors , Amphetamine/pharmacology , Central Nervous System Stimulants/antagonists & inhibitors , Central Nervous System Stimulants/pharmacology , Chloramphenicol/pharmacology , Conditioning, Operant/drug effects , Nucleus Accumbens/metabolism , Prefrontal Cortex/metabolism , Protein Synthesis Inhibitors/pharmacology , Receptor, Cannabinoid, CB1/biosynthesis , Receptor, Cannabinoid, CB1/drug effects , Animals , Male , Memory/drug effects , Nucleus Accumbens/drug effects , Prefrontal Cortex/drug effects , Psychomotor Performance/drug effects , Rats , Rats, Wistar
20.
Neurochem Int ; 49(1): 28-40, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16490285

ABSTRACT

The present study investigated oxidative damage and neuroprotective effect of the antiparkinsonian drug, L-deprenyl in neuronal death produced by intranigral infusion of a potent mitochondrial complex-I inhibitor, rotenone in rats. Unilateral stereotaxic intranigral infusion of rotenone caused significant decrease of striatal dopamine levels as measured employing HPLC-electrochemistry, and loss of tyrosine hydroxylase immunoreactivity in the perikarya of ipsilateral substantia nigra (SN) neurons and their terminals in the striatum. Rotenone-induced increases in the salicylate hydroxylation products, 2,3- and 2,5-dihydroxybenzoic acid indicators of hydroxyl radials in mitochondrial P2 fraction were dose-dependently attenuated by L-deprenyl. L-deprenyl (0.1-10mg/kg; i.p.) treatment dose-dependently attenuated rotenone-induced reductions in complex-I activity and glutathione (GSH) levels in the SN, tyrosine hydroxylase immunoreactivity in the striatum or SN as well as striatal dopamine. Amphetamine-induced stereotypic rotations in these rats were also significantly inhibited by deprenyl administration. The rotenone-induced elevated activities of cytosolic antioxidant enzymes superoxide dismutase and catalase showed further significant increase following L-deprenyl. Our findings suggest that unilateral intranigral infusion of rotenone reproduces neurochemical, neuropathological and behavioral features of PD in rats and L-deprenyl can rescue the dopaminergic neurons from rotenone-mediated neurodegeneration in them. These results not only establish oxidative stress as one of the major causative factors underlying dopaminergic neurodegeneration as observed in Parkinson's disease, but also support the view that deprenyl is a potent free radical scavenger and an antioxidant.


Subject(s)
Nerve Degeneration/drug therapy , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Parkinsonian Disorders/drug therapy , Selegiline/pharmacology , Substantia Nigra/drug effects , Amphetamine/adverse effects , Amphetamine/antagonists & inhibitors , Animals , Catalase/drug effects , Catalase/metabolism , Disease Models, Animal , Dopamine/metabolism , Dopamine Agents/adverse effects , Dose-Response Relationship, Drug , Electron Transport Complex I/drug effects , Electron Transport Complex I/metabolism , Hydroxyl Radical/metabolism , Male , Nerve Degeneration/chemically induced , Nerve Degeneration/physiopathology , Neurons/drug effects , Neurons/metabolism , Neuroprotective Agents/therapeutic use , Oxidative Stress/physiology , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/physiopathology , Rats , Rats, Sprague-Dawley , Rotenone/antagonists & inhibitors , Rotenone/toxicity , Selegiline/therapeutic use , Substantia Nigra/metabolism , Substantia Nigra/physiopathology , Superoxide Dismutase/drug effects , Superoxide Dismutase/metabolism , Tyrosine 3-Monooxygenase/drug effects , Tyrosine 3-Monooxygenase/metabolism , Uncoupling Agents/antagonists & inhibitors , Uncoupling Agents/toxicity , Up-Regulation/drug effects , Up-Regulation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL