Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
Add more filters

Country/Region as subject
Publication year range
1.
JAMA ; 331(8): 632-634, 2024 02 27.
Article in English | MEDLINE | ID: mdl-38294772

ABSTRACT

This Medical News article discusses a recent study that examined the remedies most often recommended on Reddit for diluting psychedelic experiences.


Subject(s)
Hallucinogens , Protective Agents , Social Media , Hallucinogens/adverse effects , Hallucinogens/antagonists & inhibitors , Protective Agents/therapeutic use
2.
Article in English | MEDLINE | ID: mdl-26874330

ABSTRACT

BACKGROUND: Salvinorin-A is a terpene found in the leaves of the plant Salvia divinorum. When administered to humans, salvinorin-A induces an intense but short-lasting modified state of awareness, sharing features with those induced by the classical serotonin-2A receptor agonist psychedelics. However, unlike substances such as psilocybin or mescaline, salvinorin-A shows agonist activity at the kappa-opioid receptor rather than at the serotonin-2A receptor. Here, we assessed the involvement of kappa-opioid receptor and serotonin-2A agonism in the subjective, cardiovascular, and neuroendocrine effects of salvinorin-A in humans. METHODS: We conducted a placebo-controlled, randomized, double-blind study with 2 groups of 12 healthy volunteers with experience with psychedelic drugs. There were 4 experimental sessions. In group 1, participants received the following treatment combinations: placebo+placebo, placebo+salvinorin-A, naltrexone+placebo, and naltrexone+salvinorin-A. Naltrexone, a nonspecific opioid receptor antagonist, was administered at a dose of 50mg orally. In group 2, participants received the treatment combinations: placebo+placebo, placebo+salvinorin-A, ketanserin+placebo, and ketanserin+salvinorin-A. Ketanserin, a selective serotonin-2A antagonist, was administered at a dose of 40mg orally. RESULTS: Inhalation of 1mg of vaporized salvinorin-A led to maximum plasma concentrations at 1 and 2 minutes after dosing. When administered alone, salvinorin-A severely reduced external sensory perception and induced intense visual and auditory modifications, increased systolic blood pressure, and cortisol and prolactin release. These effects were effectively blocked by naltrexone, but not by ketanserin. CONCLUSIONS: Results support kappa opioid receptor agonism as the mechanism of action underlying the subjective and physiological effects of salvinorin-A in humans and rule out the involvement of a serotonin-2A-mediated mechanism.


Subject(s)
Diterpenes, Clerodane/antagonists & inhibitors , Healthy Volunteers/psychology , Ketanserin/pharmacology , Naltrexone/pharmacology , Perception/drug effects , Adult , Blood Pressure/drug effects , Diterpenes, Clerodane/blood , Diterpenes, Clerodane/pharmacology , Double-Blind Method , Drug Interactions , Female , Hallucinogens/antagonists & inhibitors , Hallucinogens/pharmacology , Humans , Hydrocortisone/metabolism , Male , Narcotic Antagonists/pharmacology , Prolactin/metabolism , Serotonin Antagonists/pharmacology , Young Adult
3.
Int J Neuropsychopharmacol ; 17(8): 1269-82, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24650558

ABSTRACT

5-Methoxy-N,N-dimethyltryptamine (5-MeO-DMT) is a natural hallucinogen component of Ayahuasca, an Amazonian beverage traditionally used for ritual, religious and healing purposes that is being increasingly used for recreational purposes in US and Europe. 5MeO-DMT is of potential interest for schizophrenia research owing to its hallucinogenic properties. Two other psychotomimetic agents, phencyclidine and 2,5-dimethoxy-4-iodo-phenylisopropylamine (DOI), markedly disrupt neuronal activity and reduce the power of low frequency cortical oscillations (<4 Hz, LFCO) in rodent medial prefrontal cortex (mPFC). Here we examined the effect of 5-MeO-DMT on cortical function and its potential reversal by antipsychotic drugs. Moreover, regional brain activity was assessed by blood-oxygen level dependent (BOLD) functional magnetic resonance imaging (fMRI). 5-MeO-DMT disrupted mPFC activity, increasing and decreasing the discharge of 51 and 35% of the recorded pyramidal neurons, and reducing (-31%) the power of LFCO. The latter effect depended on 5-HT1A and 5-HT2A receptor activation and was reversed by haloperidol, clozapine, risperidone, and the mGlu2/3 agonist LY379268. Likewise, 5-MeO-DMT decreased BOLD responses in visual cortex (V1) and mPFC. The disruption of cortical activity induced by 5-MeO-DMT resembles that produced by phencyclidine and DOI. This, together with the reversal by antipsychotic drugs, suggests that the observed cortical alterations are related to the psychotomimetic action of 5-MeO-DMT. Overall, the present model may help to understand the neurobiological basis of hallucinations and to identify new targets in antipsychotic drug development.


Subject(s)
Antipsychotic Agents/pharmacology , Banisteriopsis/chemistry , Brain Waves/drug effects , Hallucinogens/pharmacology , Methoxydimethyltryptamines/pharmacology , Prefrontal Cortex/drug effects , Visual Cortex/drug effects , Amino Acids/pharmacology , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Clozapine/pharmacology , Excitatory Amino Acid Agonists/pharmacology , Functional Neuroimaging , Hallucinogens/antagonists & inhibitors , Haloperidol/pharmacology , Magnetic Resonance Imaging , Male , Methoxydimethyltryptamines/antagonists & inhibitors , Prefrontal Cortex/blood supply , Prefrontal Cortex/physiology , Pyramidal Cells/drug effects , Pyramidal Cells/physiology , Rats , Risperidone/pharmacology , Visual Cortex/blood supply , Visual Cortex/physiology
4.
Trends Neurosci ; 32(4): 225-32, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19269047

ABSTRACT

Research on psychedelics such as lysergic acid diethylamide (LSD) and dissociative drugs such as phencyclidine (PCP) and the symptoms, neurochemical abnormalities and treatment of schizophrenia have converged. The effects of hallucinogenic drugs resemble some of the core symptoms of schizophrenia. Some atypical antipsychotic drugs were identified by their high affinity for serotonin 5-HT(2A) receptors, which is also the target of LSD-like drugs. Several effects of PCP-like drugs are strongly affected by both 5-HT(2A) and metabotropic glutamate 2/3 receptor modulation. A serotonin-glutamate receptor complex in cortical pyramidal neurons has been identified that might be the target both of psychedelics and the atypical and glutamate classes of antipsychotic drugs. Recent results on the receptor, signalling and circuit mechanisms underlying the response to psychedelic and antipsychotic drugs might lead to unification of the serotonin and glutamate neurochemical hypotheses of schizophrenia.


Subject(s)
Hallucinogens/therapeutic use , Schizophrenia/drug therapy , Animals , Glutamic Acid/metabolism , Hallucinogens/adverse effects , Hallucinogens/antagonists & inhibitors , Humans , Models, Neurological , Receptor, Serotonin, 5-HT2A/genetics , Receptor, Serotonin, 5-HT2A/metabolism , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, Metabotropic Glutamate/metabolism , Schizophrenia/genetics , Schizophrenia/physiopathology , Serotonin/metabolism , Serotonin 5-HT2 Receptor Agonists
5.
Int J Neuropsychopharmacol ; 13(1): 71-82, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19288975

ABSTRACT

The N-methyl-D-aspartate (NMDA) glutamate receptor antagonist ketamine may have rapid, albeit transient, antidepressant properties. This study in patients with treatment-resistant major depression (TRD) aimed to (1) replicate the acute efficacy of single-dose intravenous (i.v.) ketamine; (2) test the efficacy of the glutamate-modulating agent riluzole in preventing post-ketamine relapse; and (3) examine whether pretreatment with lamotrigine would attenuate ketamine's psychotomimetic effects and enhance its antidepressant activity. Twenty-six medication-free patients received open-label i.v. ketamine (0.5 mg/kg over 40 min). Two hours prior to infusion, patients were randomized to lamotrigine (300 mg) or placebo. Seventeen patients (65%) met response criterion (50% reduction from baseline on the Montgomery-Asberg Depression Rating Scale) 24 h following ketamine. Lamotrigine failed to attenuate the mild, transient side-effects associated with ketamine and did not enhance its antidepressant effects. Fourteen patients (54%) met response criterion 72 h following ketamine and proceeded to participate in a 32-d, randomized, double-blind, placebo-controlled, flexible-dose continuation trial of riluzole (100-200 mg/d). The main outcome measure was time-to-relapse. An interim analysis found no significant differences in time-to-relapse between riluzole and placebo groups [log-rank chi(2) = 0.17, d.f. = 1, p = 0.68], with 80% of patients relapsing on riluzole vs. 50% on placebo. The trial was thus stopped for futility. This pilot study showed that a sub-anaesthetic dose of i.v. ketamine is well-tolerated in TRD, and may have rapid and sustained antidepressant properties. Riluzole did not prevent relapse in the first month following ketamine. Further investigation of relapse prevention strategies post-ketamine is necessary.


Subject(s)
Depression/drug therapy , Depression/prevention & control , Excitatory Amino Acid Antagonists/pharmacology , Ketamine/antagonists & inhibitors , Riluzole/therapeutic use , Triazines/pharmacology , Adult , Aged , Depression/diagnosis , Drug Interactions , Drug Resistance , Excitatory Amino Acid Antagonists/administration & dosage , Female , Hallucinogens/antagonists & inhibitors , Humans , Injections, Intravenous , Ketamine/administration & dosage , Lamotrigine , Male , Middle Aged , Pilot Projects , Placebos , Secondary Prevention , Time Factors , Treatment Outcome , Triazines/therapeutic use
6.
Pharmacol Res ; 61(5): 430-6, 2010 May.
Article in English | MEDLINE | ID: mdl-20045056

ABSTRACT

Neboglamine is a functional modulator of the glycine site on the N-methyl-d-aspartate (NMDA) receptor. Dysfunction of this receptor has been associated with negative and cognitive symptoms in schizophrenia. Thus, we tested the hypothesis that neboglamine behaves as a potential antipsychotic. We compared the effects of neboglamine, D-serine, clozapine, and haloperidol on the expression of Fos-like immunoreactivity (FLI), a marker of neuronal activation, in rat forebrain. We also studied the effects of these agents on phencyclidine (PCP)-induced behaviour in rats, a model predictive of potential antipsychotic activity. Neboglamine, like haloperidol and clozapine, significantly increased the number of FLI-positive cells in the prefrontal cortex, nucleus accumbens, and lateral septal nucleus (3.2-, 4.8-, and 4.5-fold over control, respectively). Haloperidol dramatically increased FLI (390-fold over control) in the dorsolateral striatum, a brain region in which neboglamine and clozapine had no effect. The pattern of FLI induced by neboglamine closely matched that of d-serine, an endogenous agonist at the glycine site of NMDA receptors. Consistent with this finding, neboglamine restored NMDA-mediated neurotransmitter release in frontal cortex punches exposed to the NMDA antagonist PCP. In the behavioural model, all test compounds significantly inhibited PCP-induced hyperlocomotion. Unlike haloperidol and clozapine, neither neboglamine nor D-serine affected the basal levels of locomotor activity. Moreover, oral neboglamine dose-dependently inhibited both the hyperlocomotion and the frequency of rearing behaviour induced by PCP. These results, while confirming that the NMDA glycine site is a feasible target for activating the frontostriatal system, support the clinical evaluation of neboglamine as a treatment for schizophrenia.


Subject(s)
Antipsychotic Agents/pharmacology , Behavior, Animal/drug effects , Pentanoic Acids/pharmacology , Receptors, N-Methyl-D-Aspartate/drug effects , Animals , Clozapine/pharmacology , Dose-Response Relationship, Drug , Hallucinogens/antagonists & inhibitors , Hallucinogens/pharmacology , Haloperidol/pharmacology , Immunohistochemistry , Male , Motor Activity/drug effects , Phencyclidine/antagonists & inhibitors , Phencyclidine/pharmacology , Prosencephalon/drug effects , Prosencephalon/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Wistar , Receptors, Glycine/drug effects , Serine/metabolism
7.
J Neurochem ; 108(3): 755-66, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19187094

ABSTRACT

Phosphodiesterase 5 (PDE5) inhibitors are often used in combination with club drugs such as 3,4-methylenedioxymethamphetamine (MDMA or ecstasy). We investigated the consequences of such combination in the serotonergic system of the rat. Oral administration of sildenafil citrate (1.5 or 8 mg/kg) increased brain cGMP levels and protected in a dose-dependent manner against 5-hydroxytryptamine depletions caused by MDMA (3 x 5 mg/kg, i.p., every 2 h) in the striatum, frontal cortex and hippocampus without altering the acute hyperthermic response to MDMA. Intrastriatal administration of the protein kinase G (PKG) inhibitor, KT5823 [(9S, 10R, 12R)-2,3,9,10,11,12-Hexahydro-10-methoxy-2,9-dimethyl-1-oxo-9,12-epoxy-1H-diindolo[1,2,3-fg:3',2',1'-kl]pyrrolo[3,4-i][1,6]benzodiazocine-10-carboxylic acid, methyl ester)], suppressed sildenafil-mediated protection. By contrast, the cell permeable cGMP analogue, 8-bromoguanosine cyclic 3',5'-monophosphate, mimicked sildenafil effects further suggesting the involvement of the PKG pathway in mediating sildenafil protection. Because mitochondrial ATP-sensitive K(+) channels are a target for PKG, we next administered the specific mitochondrial ATP-sensitive K(+) channel blocker, 5-hydroxydecanoic acid, 30 min before sildenafil. 5-hydroxydecanoic acid completely reversed the protection afforded by sildenafil, thereby implicating the involvement of mitochondrial ATP-sensitive K(+) channels. Sildenafil also increased Akt phosphorylation, and so the possible involvement of the Akt/endothelial nitric oxide synthase (eNOS)/sGC signalling pathway was analysed. Neither the phosphatidylinositol 3-kinase inhibitor, wortmannin, nor the selective eNOS inhibitor, L-N5-(1-iminoethyl)-L-ornithine dihydrochloride, reversed the protection afforded by sildenafil, suggesting that Akt/eNOS/sGC cascade does not participate in the protective mechanisms. Our data also show that the protective effect of sildenafil can be extended to vardenafil, another PDE5 inhibitor. In conclusion, sildenafil protects against MDMA-induced long-term reduction of indoles by a mechanism involving increased production of cGMP and subsequent activation of PKG and mitochondrial ATP-sensitive K(+) channel opening.


Subject(s)
Hallucinogens/antagonists & inhibitors , Hallucinogens/toxicity , N-Methyl-3,4-methylenedioxyamphetamine/antagonists & inhibitors , N-Methyl-3,4-methylenedioxyamphetamine/toxicity , Phosphodiesterase 5 Inhibitors , Phosphodiesterase Inhibitors/pharmacology , Piperazines/pharmacology , Serotonin/deficiency , Sulfones/pharmacology , Animals , Blotting, Western , Body Temperature/drug effects , Brain Chemistry/drug effects , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinases/metabolism , Hydroxyindoleacetic Acid/metabolism , Imidazoles/pharmacology , KATP Channels/drug effects , Male , Microinjections , Mitochondria/drug effects , Mitochondria/metabolism , Neostriatum/drug effects , Purines/pharmacology , Rats , Rats, Wistar , Serotonin Plasma Membrane Transport Proteins/metabolism , Signal Transduction/drug effects , Sildenafil Citrate , Triazines/pharmacology , Vardenafil Dihydrochloride
8.
Pharmacol Biochem Behav ; 88(3): 189-95, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17936884

ABSTRACT

It has been observed that agents with agonist activity at 5-HT2A receptors prevent neurotoxicity induced by the non-competitive NMDA antagonist, dizocilpine (MK-801). Subsequent behavioral studies reported complete antagonism by LSD and DOM of the stimulus effects of the related NMDA antagonist, phencyclidine [PCP]. The present study sought to extend those observations to include other psychoactive drugs. Male F-344 rats were trained in a 2-lever, fixed-ratio 10, food-reinforced task with PCP (3.0 mg/kg; IP; 30 min pretreatment) as a discriminative stimulus. Tests of generalization were then conducted using the training dose of PCP in combination with a range of doses of DOM, LSD, d-amphetamine, MDMA, psilocybin, buspirone, and GHB. All of the drugs tested in combination with PCP produced a statistically significant diminution of PCP-appropriate responding but for none was antagonism complete. These data, obtained using a stimulus control model of the hallucinogenic effects of PCP, fail to support the hypothesis that LSD and DOM completely antagonize stimulus control by PCP. Instead, the data suggest complex interactions between PCP-induced stimulus control and a variety of psychoactive drugs including GHB, an agent with no known affinity for serotonergic receptors.


Subject(s)
Excitatory Amino Acid Antagonists/pharmacology , Hallucinogens/antagonists & inhibitors , Hallucinogens/pharmacology , Phencyclidine/antagonists & inhibitors , Phencyclidine/pharmacology , Psychotropic Drugs/pharmacology , Anesthetics, Intravenous/pharmacology , Animals , Conditioning, Operant/drug effects , Dopamine Agents/pharmacology , Dose-Response Relationship, Drug , Male , Rats , Rats, Inbred F344 , Serotonin Agents/pharmacology , Sodium Oxybate/pharmacology
9.
Biol Psychiatry ; 62(6): 680-6, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17511969

ABSTRACT

BACKGROUND: Although dopamine (DA) has been implicated in the psychostimulant properties of 3,4-methylenedioxymethamphetamine (MDMA), there is no detailed information on its modalities of action on single ventral midbrain dopaminergic neurons. METHODS: We examined the actions of MDMA on intracellularly recorded dopaminergic neurons maintained in slices. RESULTS: At 1 micromol/L, MDMA depolarized and excited the cells; at 3 micromol/L, either excited or inhibited the neurons. Interestingly, higher concentrations (10-30 micromol/L) inhibited firing through membrane hyperpolarization or caused an outward current. Whereas MDMA's excitatory effects were antagonized by pindolol, indicating involvement of 5-HT 1B receptors, the inhibitory effects were counteracted by sulpiride indicating involvement D2 receptors. Treatment of the cells with carbidopa eliminated MDMA-induced firing inhibition and membrane hyperpolarization. MDMA enhanced DA-induced cellular responses but reduced those of amphetamine. Cocaine-induced outward currents were not affected by MDMA. These actions are consistent with inhibition of the DA transporter. Moreover, MDMA depressed the GABA(B) IPSP by activating 5-HT 1B receptors. CONCLUSIONS: Our data demonstrate that 3-30 micromol/L MDMA preferentially inhibits the dopaminergic cells via indirect activation of D2 autoreceptors due to increased extracellular concentration of DA. In contrast, reduction of the GABA(B) IPSP could partially account for excitation caused by 1-3 micromol/L drug.


Subject(s)
Hallucinogens/pharmacology , Mesencephalon/drug effects , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Action Potentials/drug effects , Amphetamine/pharmacology , Animals , Autoreceptors/drug effects , Carbidopa/pharmacology , Cocaine/pharmacology , Dopamine/physiology , Dopamine Plasma Membrane Transport Proteins/pharmacology , Dose-Response Relationship, Drug , Hallucinogens/antagonists & inhibitors , Membrane Potentials/drug effects , Mesencephalon/physiology , N-Methyl-3,4-methylenedioxyamphetamine/antagonists & inhibitors , Neural Inhibition/drug effects , Neurons/drug effects , Neurons/physiology , Pindolol/pharmacology , Receptors, Dopamine D2/drug effects , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/physiology
10.
Neuroscience ; 148(1): 212-20, 2007 Aug 10.
Article in English | MEDLINE | ID: mdl-17629409

ABSTRACT

BACKGROUND: 3,4-Methylenedioxymethamphetamine (MDMA) causes persistent decreases in brain 5-HT content and 5-HT transporter (SERT) binding, with no detectable changes in SERT protein. Such data suggest that MDMA impairs 5-HT transmission but leaves 5-HT nerve terminals intact. To further test this hypothesis, we carried out two types of experiments in rats exposed to high-dose MDMA. First, we examined the effects of MDMA on SERT binding and function using different in vitro assay conditions. Next, we treated rats with the 5-HT precursor, l-5-hydroxytryptophan (5-HTP), in an attempt to restore MDMA-induced depletions of 5-HT. METHODS: Rats received three i.p. injections of saline or MDMA (7.5 mg/kg), one injection every 2 h. Rats in one group were decapitated, and brain tissue was assayed for SERT binding and [(3)H]5-HT uptake under conditions of normal (100 or 126 mM) and low (20 mM) NaCl concentration. Rats from another group received saline or 5-hydroxytryptophan/benserazide (5-HTP-B), each drug at 50 mg/kg i.p., and were killed 2 h later. RESULTS: MDMA reduced SERT binding to 10% of control when assayed in 100 mM NaCl, but this reduction was only 55% of control in 20 mM NaCl. MDMA decreased immunoreactive 5-HT in caudate and hippocampus to about 35% of control. Administration of 5-HTP-B to MDMA-pretreated rats significantly increased the 5-HT signal toward normal levels in caudate (85% of control) and hippocampus (66% of control). CONCLUSION: 1) Following high-dose MDMA treatment sufficient to reduce SERT binding by 90%, a significant number of functionally intact 5-HT nerve terminals survive. 2) The degree of MDMA-induced decreases in SERT binding depends on the in vitro assay conditions. 3) 5-HTP-B restores brain 5-HT depleted by MDMA, suggesting that this approach might be clinically useful in abstinent MDMA users.


Subject(s)
5-Hydroxytryptophan/pharmacology , Brain Chemistry/drug effects , Brain/drug effects , N-Methyl-3,4-methylenedioxyamphetamine/antagonists & inhibitors , Serotonin/deficiency , Animals , Antidepressive Agents, Second-Generation/pharmacology , Binding, Competitive/drug effects , Binding, Competitive/physiology , Brain/metabolism , Brain Chemistry/physiology , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Drug Interactions/physiology , Hallucinogens/antagonists & inhibitors , Hallucinogens/toxicity , Male , N-Methyl-3,4-methylenedioxyamphetamine/toxicity , Nerve Degeneration/chemically induced , Nerve Degeneration/drug therapy , Nerve Degeneration/physiopathology , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Radioligand Assay , Rats , Rats, Sprague-Dawley , Recovery of Function/drug effects , Recovery of Function/physiology , Serotonin Agents/pharmacology , Serotonin Agents/toxicity , Serotonin Plasma Membrane Transport Proteins/drug effects , Serotonin Plasma Membrane Transport Proteins/metabolism , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
11.
Psychopharmacology (Berl) ; 192(1): 9-15, 2007 May.
Article in English | MEDLINE | ID: mdl-17235609

ABSTRACT

RATIONALE: The cognitive and attentional deficits observed in schizophrenic patients are now considered central to the pathophysiology of the disorder. These deficits include an inability to filter sensory input as measured by, e.g., prepulse inhibition (PPI) reflex. Administration of phencyclidine (PCP), a drug that can induce a schizophrenia-like psychosis in humans, disrupts PPI in experimental animals. In rodents, this PCP-induced deficit can be blocked by pretreatment with nitric oxide (NO) synthase inhibitors. This suggests that some of the behavioral effects of PCP are mediated via NO. The substrate for in vivo NO production is L-arginine, and active transport of L-arginine via the cationic amino acid transporter may serve as a regulatory mechanism in NO production. OBJECTIVES: The aim of the present study was to study the effects of L-arginine transport inhibition, using acute and repeated L-lysine treatment, on PCP-induced disruption of PPI in mice. RESULTS: Subchronic, and to some extent acute, pretreatment with L-lysine blocked a PCP-induced deficit in PPI without affecting basal PPI. CONCLUSIONS: L-lysine has been shown to block L-arginine transport in vitro, most likely via a competitive blockade and down regulation of cationic amino acid transporters. However, the importance of L-arginine transport as a regulatory mechanism in NO production in vivo is still not clear. The present results lend further support to the notion that some of the effects of PCP in the central nervous system are mediated via NO and that L-arginine transport may play a role in the regulation of NO production in the brain.


Subject(s)
Amino Acids/pharmacology , Hallucinogens/antagonists & inhibitors , Lysine/pharmacology , Phencyclidine/antagonists & inhibitors , Reflex, Startle/drug effects , Amino Acid Transport Systems, Basic/metabolism , Amino Acids/administration & dosage , Animals , Arginine/metabolism , Behavior, Animal/drug effects , Biological Transport/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Down-Regulation/drug effects , Hallucinogens/pharmacology , Lysine/administration & dosage , Male , Mice , Nitric Oxide/biosynthesis , Nitric Oxide/metabolism , Phencyclidine/pharmacology , Reflex/drug effects , Schizophrenia
12.
Pharmacol Biochem Behav ; 87(4): 472-80, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17688928

ABSTRACT

Although psilocybin has been trained in the rat as a discriminative stimulus, little is known of the pharmacological receptors essential for stimulus control. In the present investigation rats were trained with psilocybin and tests were then conducted employing a series of other hallucinogens and presumed antagonists. An intermediate degree of antagonism of psilocybin was observed following treatment with the 5-HT(2A) receptor antagonist, M100907. In contrast, no significant antagonism was observed following treatment with the 5-HT(1A/7) receptor antagonist, WAY-100635, or the DA D(2) antagonist, remoxipride. Psilocybin generalized fully to DOM, LSD, psilocin, and, in the presence of WAY-100635, DMT while partial generalization was seen to 2C-T-7 and mescaline. LSD and MDMA partially generalized to psilocybin and these effects were completely blocked by M-100907; no generalization of PCP to psilocybin was seen. The present data suggest that psilocybin induces a compound stimulus in which activity at the 5-HT(2A) receptor plays a prominent but incomplete role. In addition, psilocybin differs from closely related hallucinogens such as 5-MeO-DMT in that agonism at 5-HT(1A) receptors appears to play no role in psilocybin-induced stimulus control.


Subject(s)
Discrimination, Psychological/drug effects , Generalization, Stimulus/drug effects , Hallucinogens/pharmacology , Psilocybin/pharmacology , Animals , Conditioning, Operant/drug effects , Data Interpretation, Statistical , Dose-Response Relationship, Drug , Hallucinogens/antagonists & inhibitors , Lysergic Acid Diethylamide/pharmacology , Male , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Phencyclidine/pharmacology , Psilocybin/antagonists & inhibitors , Rats , Rats, Inbred F344 , Reinforcement Schedule
13.
Neuropsychopharmacology ; 30(6): 1056-63, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15668722

ABSTRACT

3,4-Methylenedioxy-N-methamphetamine (MDMA or 'ecstasy') is a psychoactive substance, first described as an appetite suppressant in humans, inducing side effects and even death. MDMA increases serotonin (5-HT) levels, and 5-HT inhibits food intake, but the 5-HT receptors involved in MDMA-induced changes in feeding behavior are unknown. We examined whether a systemic MDMA injection would reduce the physiological drive to eat in starved mice and tested if the inactivation of 5-HT1B or 5-HT2C receptors could restore this response. Our results indicate that in starved mice, MDMA (10 mg/kg) provoked an initial hypophagia for 1 h (-77%) followed by a period of hyperphagia (studied between 1 and 3 h). This biphasic feeding behavior due to MDMA treatment was maintained in 5-HT1B receptor-null mice or in animals treated with the 5-HT1B/1D receptor antagonist GR127935 (3 or 10 mg/kg). In contrast, MDMA-induced hypophagia (for the first 1 h period) was suppressed when combined with the 5-HT2C receptor antagonist RS102221 (2 mg/kg). However, RS102221 did not alter MDMA-induced hyperphagia (for the 1-3 h period) but did exert a stimulant effect, when administered alone, during that period. We have previously shown that MDMA or 5-HT1A/1B receptor agonist RU24969 fails to stimulate locomotor activity in 5-HT1B receptor-null mice. Our present data indicate that the 5-HT2C receptor antagonist RS102221 suppresses MDMA-induced hyperlocomotion. These findings provide the first evidence that the inactivation of 5-HT2C receptors may reduce hypophagia and motor response to MDMA, while a genetic deficit or pharmacological inactivation of 5-HT1B receptors was insufficient to alter the feeding response to MDMA.


Subject(s)
Hallucinogens/pharmacology , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Receptor, Serotonin, 5-HT1B/physiology , Receptor, Serotonin, 5-HT2C/drug effects , Serotonin Antagonists/pharmacology , Spiro Compounds/pharmacology , Sulfonamides/pharmacology , Animals , Body Weight/drug effects , Eating/drug effects , Hallucinogens/antagonists & inhibitors , Kinetics , Male , Mice , Mice, Knockout , N-Methyl-3,4-methylenedioxyamphetamine/antagonists & inhibitors , Oxadiazoles/pharmacology , Piperazines/pharmacology , Receptor, Serotonin, 5-HT1B/genetics , Starvation/psychology
14.
Psychopharmacology (Berl) ; 179(4): 854-62, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15645221

ABSTRACT

RATIONALE: The drug discrimination procedure is the most frequently used in vivo model of hallucinogen activity. Historically, most drug discrimination studies have been conducted in the rat. With the development of genetically modified mice, a powerful new tool has become available for investigating the mechanisms of drug-induced behavior. The current paper is part of an ongoing effort to determine the utility of the drug discrimination technique for evaluating hallucinogenic drugs in mice. OBJECTIVE: To establish the training procedures and characterize the stimulus properties of (+)lysergic acid diethylamide (LSD) in mice. METHODS: Using a two-lever drug discrimination procedure, C57Bl/6J mice were trained to discriminate 0.45 mg/kg LSD vs saline on a VI30 sec schedule of reinforcement, with vanilla-flavored Ensure serving as the reinforcer. RESULTS: As in rats, acquisition was orderly, but the training dose was nearly five-fold higher for mice than rats. LSD lever selection was dose-dependent. Time-course studies revealed a rapid loss of the LSD stimulus effects. The 5-HT(2A/2C) receptor agonist, 2,5-dimethoxy-4-bromoamphetamine [(-)DOB] (1.0 mg/kg), substituted fully for LSD and the 5-HT(1A) receptor agonist, 8-hydroxy-2-(di-n-propylamino)-tetralin (8-OH-DPAT) (1.6 mg/kg), substituted partially for LSD. Pretreatment with the 5-HT(2A) receptor-selective antagonist, MDL 100907, or the 5-HT(1A)-selective antagonist WAY 100635, showed that each antagonist only partially blocked LSD discrimination. Substitution of 1.0 mg/kg (-)DOB for LSD was fully blocked by pretreatment with MDL 100907 but unaltered by WAY 100635 pretreatment. CONCLUSIONS: These data suggest that in mice the stimulus effects of LSD have both a 5-HT(2A) receptor and a 5-HT(1A) receptor component.


Subject(s)
Discrimination, Psychological/drug effects , Hallucinogens/pharmacology , Lysergic Acid Diethylamide/pharmacology , DOM 2,5-Dimethoxy-4-Methylamphetamine/analogs & derivatives , DOM 2,5-Dimethoxy-4-Methylamphetamine/pharmacology , Animals , Cues , Discrimination Learning , Dose-Response Relationship, Drug , Fluorobenzenes/pharmacology , Food , Hallucinogens/antagonists & inhibitors , Lysergic Acid Diethylamide/antagonists & inhibitors , Male , Mice , Mice, Inbred C57BL , Piperidines/pharmacology , Receptor, Serotonin, 5-HT2A/drug effects , Reinforcement Schedule , Reinforcement, Psychology , Serotonin Antagonists/pharmacology
15.
Psychopharmacology (Berl) ; 179(2): 336-48, 2005 May.
Article in English | MEDLINE | ID: mdl-15645224

ABSTRACT

RATIONALE: Phencyclidine (PCP), a glutamate/N-methyl-D-aspartate (NMDA) receptor antagonist, has been shown to induce a range of symptoms similar to those of patients with schizophrenia, while D-amphetamine induces predominantly positive symptoms. Previous studies in our laboratory have shown that PCP can selectively impair the performance of an operant reversal-learning task in the rat. Furthermore, we found that the novel antipsychotic ziprasidone, but not the classical antipsychotic haloperidol, could prevent the PCP-induced deficit. OBJECTIVES: The aim of the present study was to validate the model further using the atypical antipsychotic clozapine and then to investigate the effects of lamotrigine, a broad-spectrum anticonvulsant that is known to reduce glutamate release in vitro and is able to prevent ketamine-induced psychotic symptoms in healthy human volunteers. A further aim was to compare effects of PCP and D-amphetamine in the test and investigate the effects of the typical antipsychotic haloperidol against the latter. METHODS: Female hooded-Lister rats were food deprived and trained to respond for food in a reversal-learning paradigm. RESULTS: PCP at 1.5 mg/kg and 2.0 mg/kg and D-amphetamine at 0.5 mg/kg significantly and selectively impaired performance in the reversal phase of the task. The cognitive deficit induced by 1.5 mg/kg PCP was attenuated by prior administration of lamotrigine (20 mg/kg and 30 mg/kg) or clozapine (5 mg/kg), but not haloperidol (0.05 mg/kg). In direct contrast, haloperidol (0.05 mg/kg), but not lamotrigine (25 mg/kg) or clozapine (5 mg/kg), prevented a similar cognitive impairment produced by D-amphetamine (0.5 mg/kg). CONCLUSIONS: Our findings provide further data to support the use of PCP-induced disruption of reversal learning in rodents to investigate novel antipsychotic drugs. The results also provide evidence for different mechanisms of PCP and D-amphetamine-induced disruption of performance in the test, and their different sensitivities to typical and atypical antipsychotic drugs.


Subject(s)
Antimanic Agents/pharmacology , Antipsychotic Agents/pharmacology , Central Nervous System Stimulants/antagonists & inhibitors , Central Nervous System Stimulants/toxicity , Clozapine/pharmacology , Dextroamphetamine/antagonists & inhibitors , Dextroamphetamine/toxicity , Hallucinogens/antagonists & inhibitors , Hallucinogens/toxicity , Learning Disabilities/chemically induced , Learning Disabilities/prevention & control , Phencyclidine/antagonists & inhibitors , Phencyclidine/toxicity , Reversal Learning/drug effects , Triazines/pharmacology , Animals , Conditioning, Operant/drug effects , Dose-Response Relationship, Drug , Female , Haloperidol/pharmacology , Lamotrigine , Rats
16.
Psychopharmacology (Berl) ; 179(4): 804-12, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15619107

ABSTRACT

RATIONALE: Many behavioral effects of delta-9-tetrahydrocannabinol (THC), including its discriminative-stimulus effects, are modulated by endogenous opioid systems. OBJECTIVE: To investigate opioid receptor subtypes involved in the discriminative effects of THC. METHODS: Rats trained to discriminate 3 mg/kg i.p. of THC from vehicle using a two-lever operant drug-discrimination procedure, were tested with compounds that bind preferentially or selectively to either mu-, delta- or kappa-opioid receptors. RESULTS: The preferential mu-opioid receptor agonist heroin (0.3-1.0 mg/kg, i.p.), the selective delta-opioid receptor agonist SNC-80 (1-10 mg/kg, i.p.) and the selective kappa-opioid receptor agonist U50488 (1-10 mg/kg, i.p.) did not produce generalization to the discriminative effects of THC when given alone. However, heroin, but not SNC-80 or U50488, significantly shifted the dose-response curve for THC discrimination to the left. Also, the preferential mu-opioid receptor antagonist naltrexone (0.1-1 mg/kg, i.p.), the selective delta-opioid receptor antagonist, naltrindole (1-10 mg/kg, i.p.) and the kappa-opioid receptor antagonist nor-binaltorphimine (n-BNI, 5 mg/kg, s.c.), did not significantly reduce the discriminative effects of the training dose of THC. However, naltrexone, but not naltrindole or n-BNI, significantly shifted the dose-response curve for THC discrimination to the right. Finally, naltrexone, but not naltrindole or n-BNI, blocked the leftward shift in the dose-response curve for THC discrimination produced by heroin. CONCLUSIONS: mu- but not delta- or kappa-opioid receptors are involved in the discriminative effects of THC. Given the role that mu-opioid receptors play in THC's rewarding effects, the present findings suggest that discriminative-stimulus effects and rewarding effects of THC involve similar neural mechanisms.


Subject(s)
Discrimination, Psychological/drug effects , Dronabinol/pharmacology , Hallucinogens/pharmacology , Receptors, Opioid, delta/drug effects , Receptors, Opioid, kappa/drug effects , Receptors, Opioid, mu/drug effects , 3,4-Dichloro-N-methyl-N-(2-(1-pyrrolidinyl)-cyclohexyl)-benzeneacetamide, (trans)-Isomer/pharmacology , Analgesics, Non-Narcotic/pharmacology , Animals , Benzamides/pharmacology , Conditioning, Operant/drug effects , Discrimination Learning/drug effects , Dose-Response Relationship, Drug , Dronabinol/antagonists & inhibitors , Food , Hallucinogens/antagonists & inhibitors , Heroin/pharmacology , Male , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Narcotics/pharmacology , Piperazines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/antagonists & inhibitors , Receptors, Opioid, kappa/agonists , Receptors, Opioid, kappa/antagonists & inhibitors , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/antagonists & inhibitors , Reinforcement, Psychology
17.
Pharmacol Biochem Behav ; 82(2): 404-10, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16253319

ABSTRACT

MDMA (N-methyl-3,4-methylenedioxyamphetamine) produces a discriminative stimulus (DS) effect in animals, but attempts to completely block this action with selective neurotransmitter antagonists have not been very successful. Biochemically, MDMA can increase synaptic levels of serotonin, dopamine, and norepinephrine that, conceivably, might interact with multiple populations or subpopulations of neurotransmitter receptors. The present study attempted to antagonize the DS effects of MDMA using the nonselective agents clozapine, cyproheptadine, and pizotyline. An extensive and comparative radioligand binding profile was also obtained for the latter two agents. The purported antagonists were administered in combination with the training dose of MDMA to groups of Sprague-Dawley rats trained to discriminate 1.5 mg/kg of MDMA from saline vehicle in a standard two-lever operant paradigm using a VI-15s schedule of reinforcement. Clozapine was without effect at the doses evaluated, and cyproheptadine only partially attenuated MDMA-appropriate responding. In contrast, pizotyline (AD50=2.5 mg/kg), in combination with the MDMA training dose, resulted in a dose related decrease in percent drug-appropriate responding to saline levels. In a separate group of animals trained to discriminate the structurally-related agent N-methyl-1-(4-methoxyphenyl)-2-aminopropane (PMMA) from vehicle, pretreatment with pizotyline also resulted in a substantial decrease in drug-appropriate responding. The results with cyproheptadine and pizotyline in the binding assays confirmed that these agents display high affinity for multiple subpopulations of serotonergic, dopaminergic, adrenergic, histaminergic, and cholinergic receptors. The overall results of the present investigation indicate that pizotyline, which is clinically available in some countries, might be of clinical utility in the treatment of MDMA overdose.


Subject(s)
Central Nervous System Stimulants/antagonists & inhibitors , Hallucinogens/antagonists & inhibitors , N-Methyl-3,4-methylenedioxyamphetamine/antagonists & inhibitors , Pizotyline/pharmacology , Serotonin Antagonists/pharmacology , Animals , Central Nervous System Stimulants/pharmacology , Cyproheptadine/pharmacology , Discrimination, Psychological/drug effects , Dose-Response Relationship, Drug , Generalization, Psychological , Hallucinogens/pharmacokinetics , Hallucinogens/pharmacology , Male , Methamphetamine/analogs & derivatives , Methamphetamine/pharmacokinetics , Methamphetamine/pharmacology , N-Methyl-3,4-methylenedioxyamphetamine/pharmacokinetics , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Pizotyline/pharmacokinetics , Rats , Rats, Sprague-Dawley , Receptors, Neurotransmitter/drug effects , Receptors, Neurotransmitter/metabolism , Receptors, Serotonin/drug effects , Receptors, Serotonin/metabolism , Serotonin Antagonists/pharmacokinetics
18.
Neurosci Biobehav Rev ; 23(2): 287-93, 1998.
Article in English | MEDLINE | ID: mdl-9884122

ABSTRACT

Phencyclidine (PCP) can induce a model psychosis in humans that mimics the positive and negative symptoms of schizophrenia. In the social interaction test PCP induces stereotyped behaviour and social isolation in rats, and these behaviours can be inhibited by antipsychotic drugs. In order to further evaluate the predictive validity of this model of schizophrenia the anxiolytic diazepam (0.02-17.5 micromol/kg; 0.005-5.0 mg/kg), the antidepressant citalopram (0.62-19.8 micromol/kg; 0.3-4.0 mg/kg), the opioid agonist methadone (0.36-5.8 micromol/kg; 0.13-2.0 mg/kg) and the opioid antagonist naloxone (0.34-22.0 micromol/kg; 0.13-8.0 mg/kg) were tested as examples of drugs without antipsychotic activity. The experiments demonstrated that these compounds did not specifically inhibit the behavioural effects of PCP. So far only antipsychotic drugs have been able to specifically inhibit the PCP-induced behaviours.


Subject(s)
Hallucinogens/pharmacology , Interpersonal Relations , Phencyclidine/pharmacology , Social Isolation/psychology , Stereotyped Behavior/drug effects , Animals , Anti-Anxiety Agents/pharmacology , Citalopram/pharmacology , Diazepam/pharmacology , Drug Interactions , Hallucinogens/antagonists & inhibitors , Male , Methadone/pharmacology , Motor Activity/drug effects , Naloxone/pharmacology , Narcotic Antagonists/pharmacology , Narcotics/pharmacology , Phencyclidine/antagonists & inhibitors , Rats , Rats, Wistar , Selective Serotonin Reuptake Inhibitors/pharmacology
19.
J Cereb Blood Flow Metab ; 17(2): 168-74, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9040496

ABSTRACT

In acute brain injury syndromes, the potent N-methyl-D-aspartate (NMDA) antagonist, MK-801, can prevent neuronal degeneration, and the general anesthetics, isoflurane and propofol, may also provide neuroprotective benefits. An obstacle to the use of NMDA antagonists for neuroprotective purposes is that they can cause a neurotoxic vacuole reaction in cerebrocortical neurons. This study demonstrates the ability of isoflurane and propofol to prevent the neurotoxic vacuole reaction induced by MK-801. Low sedative doses of inhaled isoflurane (1%) or intravenous (i.v.) propofol (7.5 mg/kg/h) were as effective as higher general anesthetic doses. Thus, in the clinical management of acute brain injury conditions such as stroke and brain trauma, administration of one of these anesthetic agents together with an NMDA antagonist may be an excellent formula for obtaining optimal neuroprotection while eliminating serious side effects.


Subject(s)
Anesthetics, Inhalation/therapeutic use , Brain Damage, Chronic/prevention & control , Dizocilpine Maleate/toxicity , Excitatory Amino Acid Antagonists/toxicity , Gyrus Cinguli/drug effects , Hallucinogens/toxicity , Isoflurane/therapeutic use , Neuroprotective Agents/toxicity , Propofol/therapeutic use , Receptors, N-Methyl-D-Aspartate/drug effects , Anesthetics, Inhalation/pharmacology , Animals , Dizocilpine Maleate/antagonists & inhibitors , Female , Gyrus Cinguli/pathology , Hallucinogens/antagonists & inhibitors , Isoflurane/pharmacology , Nerve Degeneration/drug effects , Neuroprotective Agents/antagonists & inhibitors , Propofol/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/physiology
20.
Neuropsychopharmacology ; 18(5): 339-51, 1998 May.
Article in English | MEDLINE | ID: mdl-9536447

ABSTRACT

The 5-HT2A and 5-HT2C antagonists MDL 100,907 and SER-082 were tested with the 5-HT2A/C agonist DOI and the 5-HT1A/2A/2C agonist LSD in the Behavioral Pattern Monitor, which provides multiple measures of locomotor and investigatory activity. Previous investigations have shown that these measures load onto three independent behavioral factors: amount of activity, exploratory behavior, and behavioral organization. Rats pretreated with saline, MDL 100,907 (0.25-2.0 mg/kg), or SER-082 (0.5-1.0 mg/kg) were treated with saline, 0.25 mg/kg DOI, or 60 micrograms/kg LSD. All effects of DOI were blocked by all doses of MDL 100,907, but only by the highest dose of SER-082. While the effects of LSD on activity and exploratory behavior were largely unaffected, either pretreatment antagonized the effects of LSD on behavioral organization. Thus, all of these effects of DOI were attributable to 5-HT2A receptors, whereas the effect of LSD on behavioral organization was influenced by both 5-HT2A and 5-HT2C receptors.


Subject(s)
Hallucinogens/pharmacology , Motor Activity/drug effects , Receptors, Serotonin/drug effects , Amphetamines/antagonists & inhibitors , Amphetamines/pharmacology , Animals , Exploratory Behavior/drug effects , Fluorobenzenes/pharmacology , Hallucinogens/antagonists & inhibitors , Lysergic Acid Diethylamide/pharmacology , Male , Piperidines/pharmacology , Rats , Rats, Sprague-Dawley , Serotonin Antagonists/pharmacology , Serotonin Receptor Agonists/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL