Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Psychopharmacology (Berl) ; 233(7): 1215-25, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26758284

ABSTRACT

RATIONALE: Methoxetamine (MXE) is a ketamine analog sold online that has been subject to widespread abuse for its dissociative and hallucinogenic effects. Previous studies have shown that MXE has high affinity for the phencyclidine (PCP) binding site located within the channel pore of the NMDA receptor (NMDAR), but little is known about its behavioral effects. Dissociative anesthetics such as ketamine and PCP produce a characteristic behavioral profile in rats that includes locomotor hyperactivity and disruption of prepulse inhibition (PPI) of acoustic startle. METHODS: The goal of the present investigation was to determine whether MXE produces PCP-like effects in Sprague-Dawley rats using the PPI paradigm and the behavioral pattern monitor (BPM), which enables analyses of patterns of locomotor activity and investigatory behavior. PPI studies were conducted with several other uncompetitive NMDAR antagonists that produce dissociative effects in humans, including PCP, the S-(+) and R-(-) isomers of ketamine, and N-allylnormetazocine (NANM; SKF-10,047). RESULTS: MXE disrupted PPI when administered at 3 and 10 mg/kg SC. The rank order of potency of MXE and the other test compounds in the PPI paradigm (PCP > MXE > S-(+)-ketamine > NANM > R-(-)-ketamine) parallels their affinities for the PCP binding site reported in the literature. When tested in the BPM, 10 mg/kg MXE induced locomotor hyperactivity, reduced the number of rearings, increased the roughness of locomotor paths, and produced perseverative patterns of locomotion. Administration of PCP (2.25 and 6.75 mg/kg, SC) produced a similar profile of effects in the BPM. CONCLUSIONS: These results indicate that MXE produces a behavioral profile similar to that of other psychotomimetic uncompetitive NMDAR antagonists. Our findings support the classification of MXE as a dissociative drug and suggest that it likely has effects and abuse potential similar to that of PCP and ketamine.


Subject(s)
Cyclohexanones/pharmacology , Cyclohexylamines/pharmacology , Exploratory Behavior/drug effects , Motor Activity/drug effects , Prepulse Inhibition/drug effects , Reflex, Startle/drug effects , Acoustic Stimulation , Animals , Male , Rats , Rats, Sprague-Dawley , Substance-Related Disorders
2.
Int J Neuropsychopharmacol ; 18(11): pyv063, 2015 Jun 02.
Article in English | MEDLINE | ID: mdl-26037489

ABSTRACT

BACKGROUND: Ketamine produces schizophrenia-like behavioral phenotypes in healthy people. Prolonged ketamine effects and exacerbation of symptoms after the administration of ketamine have been observed in patients with schizophrenia. More recently, ketamine has been used as a potent antidepressant to treat patients with major depression. The genes and neurons that regulate behavioral responses to ketamine, however, remain poorly understood. Sp4 is a transcription factor for which gene expression is restricted to neuronal cells in the brain. Our previous studies demonstrated that Sp4 hypomorphic mice display several behavioral phenotypes relevant to psychiatric disorders, consistent with human SP4 gene associations with schizophrenia, bipolar disorder, and major depression. Among those behavioral phenotypes, hypersensitivity to ketamine-induced hyperlocomotion has been observed in Sp4 hypomorphic mice. METHODS: In the present study, we used the Cre-LoxP system to restore Sp4 gene expression, specifically in either forebrain excitatory or GABAergic inhibitory neurons in Sp4 hypomorphic mice. Mouse behavioral phenotypes related to psychiatric disorders were examined in these distinct rescue mice. RESULTS: Restoration of Sp4 in forebrain excitatory neurons did not rescue deficient sensorimotor gating nor ketamine-induced hyperlocomotion. Restoration of Sp4 in forebrain GABAergic neurons, however, rescued ketamine-induced hyperlocomotion, but did not rescue deficient sensorimotor gating. CONCLUSIONS: Our studies suggest that the Sp4 gene in forebrain GABAergic neurons regulates ketamine-induced hyperlocomotion.


Subject(s)
Akathisia, Drug-Induced/physiopathology , Excitatory Amino Acid Antagonists/toxicity , GABAergic Neurons/physiology , Ketamine/toxicity , Prosencephalon/physiopathology , Sp4 Transcription Factor/metabolism , Animals , Cohort Studies , Female , GABAergic Neurons/drug effects , Male , Mice, 129 Strain , Mice, Transgenic , Prosencephalon/drug effects , Sensory Gating/physiology , Sp4 Transcription Factor/genetics
3.
Dev Neurosci ; 37(3): 253-62, 2015.
Article in English | MEDLINE | ID: mdl-26022788

ABSTRACT

Cognitive impairments appear early in the progression of schizophrenia, often preceding the symptoms of psychosis. Thus, the systems subserving these functions may be more vulnerable to, and mechanistically linked with, the initial pathology. Understanding the trajectory of behavioral and anatomical abnormalities relevant to the schizophrenia prodrome and their sensitivity to interventions in relevant models will be critical to identifying early therapeutic strategies. Isolation rearing of rats is an environmental perturbation that deprives rodents of social contact from weaning through adulthood and produces behavioral and neuronal abnormalities that mirror some pathophysiology associated with schizophrenia, e.g. frontal cortex abnormalities and prepulse inhibition (PPI) of startle deficits. Previously, we showed that PPI deficits in isolation-reared rats emerge in mid-adolescence (4 weeks after weaning; approx. postnatal day 52) but are not present when tested at 2 weeks after weaning (approx. postnatal day 38). Because cognitive deficits are reported during early adolescence, are relevant to the prodrome, and are linked to functional outcome, we examined the putative time course of reversal learning deficits in isolation-reared rats. Separate groups of male Sprague Dawley rats were tested in a two-choice discrimination task at 2 and 8 weeks after weaning, on postnatal day 38 and 80, respectively. The isolation-reared rats displayed impaired reversal learning at both time points. Isolation rearing was also associated with deficits in PPI at 4 and 10 weeks after weaning. The reversal learning deficits in the isolated rats were accompanied by reductions in parvalbumin immunoreactivity, a marker for specific subpopulations of GABAergic neurons, in the hippocampus. Hence, isolation rearing of rats may offer a unique model to examine the ontogeny of behavioral and neurobiological alterations that may be relevant to preclinical models of prodromal psychosis. © 2015 S. Karger AG, Basel.


Subject(s)
Behavior, Animal/physiology , Cognition Disorders/physiopathology , Prepulse Inhibition/physiology , Reversal Learning/physiology , Social Isolation , Age Factors , Animals , Cognition Disorders/etiology , Disease Models, Animal , Female , Male , Prodromal Symptoms , Psychotic Disorders/etiology , Rats , Rats, Sprague-Dawley , Schizophrenia/etiology , gamma-Aminobutyric Acid
4.
Behav Pharmacol ; 25(1): 12-22, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24281153

ABSTRACT

HIV infection is frequently comorbid with methamphetamine (METH) dependence. Both factors are associated with impairment in inhibitory function that continues even after abstinence from the drug. Deficits in prepulse inhibition (PPI), a measure of sensorimotor gating, are induced by acute stimulant administration, but the combined effect of HIV and chronic METH exposure on PPI is not well characterized. We quantified baseline acoustic startle and PPI in mice expressing the HIV-1 gp120 envelope protein (gp120tg) and in wild-type (WT) littermates; thereafter, we administered a chronic regimen of METH or vehicle and tested startle and PPI after 7 days of drug withdrawal. We hypothesized that METH-treated gp120tg mice would exhibit PPI deficits compared with vehicle-treated WT or gp120tg animals. Before METH administration, drug-naive female gp120tg mice exhibited decreased PPI compared with female WT mice, whereas male gp120tg mice exhibited increased startle compared with other groups. After drug withdrawal, no consistent genotype effect was observed, but METH-treated mice exhibited increased PPI compared with vehicle, in contrast to previous reports of acute METH-induced PPI deficits. In summary, PPI impairment in HIV could depend on factors such as sex, whereas changes in PPI following METH withdrawal may depend on the quantity and duration of drug exposure.


Subject(s)
Central Nervous System Stimulants/adverse effects , Lameness, Animal/etiology , Methamphetamine/adverse effects , Neural Inhibition/physiology , Reflex, Startle/physiology , Substance Withdrawal Syndrome/complications , Acoustic Stimulation , Analysis of Variance , Animals , Drug Administration Schedule , Female , HIV Envelope Protein gp120/genetics , Humans , Lameness, Animal/virology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Inhibition/drug effects , Reflex, Startle/drug effects , Sex Factors , Time Factors
5.
Behav Brain Res ; 236(1): 210-220, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-22960458

ABSTRACT

Methamphetamine (METH) dependence is frequently comorbid with HIV infection. Both factors are independently characterized by inhibitory deficits, which may manifest as increased motor activity, inappropriate perseverative behavior, and elevated exploratory responses to novel stimuli, but the effect of combined METH exposure and HIV is not well understood. In this study, we administered a chronic escalation/binge regimen of METH or vehicle treatment to wildtype (WT) or transgenic (tg) mice expressing the HIV-1 gp120 envelope protein and quantified disinhibition during the 7 days following drug withdrawal. We hypothesized that gp120tg mice administered chronic METH would exhibit more pronounced inhibitory deficits compared to vehicle-treated WT or gp120tg animals. Our results showed that METH treatment alone increased novel object interaction while female METH-treated gp120tg mice exhibited the highest level of exploration (holepoking) compared to other female mice. Transgenic mice exhibited fewer rears relative to WT, slightly less locomotion, and also demonstrated a trend toward more perseverative motor patterns. In summary, both METH treatment and gp120 expression may modify inhibition, but such effects are selective and dependent upon variations in age and sex that could impact dopamine and frontostriatal function. These findings illustrate the need to improve our knowledge about the combined effects of HIV and substance use and facilitate improved treatment methods for comorbid disease and drug dependence.


Subject(s)
Behavior, Animal/drug effects , Central Nervous System Stimulants/pharmacology , HIV Envelope Protein gp120/genetics , Methamphetamine/pharmacology , Aging/psychology , Amphetamine-Related Disorders/psychology , Animals , Body Temperature/drug effects , Body Weight/drug effects , Darkness , Dose-Response Relationship, Drug , Exploratory Behavior/drug effects , Female , HIV Infections/psychology , Light , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/drug effects , Motor Activity/physiology , Sex Characteristics
6.
Pharmacol Biochem Behav ; 102(1): 88-94, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22487771

ABSTRACT

The indirect serotonin (5-HT) agonist 3,4-methylenedioxymethamphetamine (MDMA) produces a distinct behavioral profile in rats consisting of locomotor hyperactivity, thigmotaxis, and decreased exploration. The indirect 5-HT agonist α-ethyltryptamine (AET) produces a similar behavioral profile. Using the Behavioral Pattern Monitor (BPM), the present investigation examined whether the effects of MDMA and AET are dependent on the novelty of the testing environment. These experiments were conducted in Sprague-Dawley rats housed on a reversed light cycle and tested during the dark phase of the light/dark cycle. We found that racemic MDMA (RS-MDMA; 3 mg/kg, SC) increased locomotor activity in rats tested in novel BPM chambers, but had no effect on locomotor activity in rats habituated to the BPM chambers immediately prior to testing. Likewise, AET (5 mg/kg, SC) increased locomotor activity in non-habituated animals but not in animals habituated to the test chambers. These results were unexpected because previous reports indicate that MDMA has robust locomotor-activating effects in habituated animals. To further examine the influence of habituation on MDMA-induced locomotor activity, we conducted parametric studies with S-(+)-MDMA (the more active enantiomer) in habituated and non-habituated rats housed on a standard or reversed light cycle. Light cycle was included as a variable due to reported differences in sensitivity to serotonergic ligands during the dark and light phases. In confirmation of our initial studies, rats tested during the dark phase and habituated to the BPM did not show an S-(+)-MDMA (3 mg/kg, SC)-induced increase in locomotor activity, whereas non-habituated rats did. By contrast, in rats tested during the light phase, S-(+)-MDMA increased locomotor activity in both non-habituated and habituated rats, although the response in habituated animals was attenuated. The finding that habituation and light cycle interact to influence MDMA- and AET-induced hyperactivity demonstrates that there are previously unrecognized complexities associated with the behavioral effects of these drugs.


Subject(s)
Habituation, Psychophysiologic/drug effects , Motor Activity/drug effects , N-Methyl-3,4-methylenedioxyamphetamine/pharmacology , Serotonin Receptor Agonists/pharmacology , Animals , Habituation, Psychophysiologic/physiology , Male , Motor Activity/physiology , Random Allocation , Rats , Rats, Sprague-Dawley
7.
PLoS One ; 6(7): e19286, 2011.
Article in English | MEDLINE | ID: mdl-21818251

ABSTRACT

Deficits in sensorimotor gating measured by prepulse inhibition (PPI) of the startle have been known as characteristics of patients with schizophrenia and related neuropsychiatric disorders. PPI disruption is thought to rely on the activity of the mesocorticolimbic dopaminergic system and is inhibited by most antipsychotic drugs. These drugs however act also at the nigrostriatal dopaminergic pathway and exert adverse locomotor responses. Finding a way to inhibit the mesocorticolimbic- without affecting the nigrostriatal-dopaminergic pathway may thus be beneficial to antipsychotic therapies. The melanin-concentrating hormone (MCH) system has been shown to modulate dopamine-related responses. Its receptor (MCH1R) is expressed at high levels in the mesocorticolimbic and not in the nigrostriatal dopaminergic pathways. Interestingly a genomic linkage study revealed significant associations between schizophrenia and markers located in the MCH1R gene locus. We hypothesize that the MCH system can selectively modulate the behavior associated with the mesocorticolimbic dopamine pathway. Using mice, we found that central administration of MCH potentiates apomorphine-induced PPI deficits. Using congenic rat lines that differ in their responses to PPI, we found that the rats that are susceptible to apomorphine (APO-SUS rats) and exhibit PPI deficits display higher MCH mRNA expression in the lateral hypothalamic region and that blocking the MCH system reverses their PPI deficits. On the other hand, in mice and rats, activation or inactivation of the MCH system does not affect stereotyped behaviors, dopamine-related responses that depend on the activity of the nigrostriatal pathway. Furthermore MCH does not affect dizocilpine-induced PPI deficit, a glutamate related response. Thus, our data present the MCH system as a regulator of sensorimotor gating, and provide a new rationale to understand the etiologies of schizophrenia and related psychiatric disorders.


Subject(s)
Behavior, Animal/drug effects , Hypothalamic Hormones/pharmacology , Melanins/pharmacology , Mental Disorders/physiopathology , Pituitary Hormones/pharmacology , Animals , Apomorphine/pharmacology , Dizocilpine Maleate/pharmacology , Hypothalamic Hormones/administration & dosage , Injections , Male , Melanins/administration & dosage , Mice , Mice, Inbred C57BL , Neural Inhibition/drug effects , Pituitary Hormones/administration & dosage , Rats , Reflex, Startle/drug effects , Stereotyped Behavior/drug effects
8.
Behav Pharmacol ; 21(2): 135-43, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20215963

ABSTRACT

Pramipexole (PRA) is a preferential D3R agonist that, in rats and humans, modifies prepulse inhibition (PPI) of the acoustic startle reflex, an operational measure of sensorimotor gating. The ability to use similar PPI measures across species, and the relative ease of genetic manipulations in mice, suggests that molecular studies of the D3R regulation of sensorimotor gating might be best pursued in mice. Here, we evaluate the effects of PRA on PPI and locomotion in C57BL/6J mice, the background strain for many gene knockout mouse models. Male C57BL/6J mice were tested for PPI and locomotor activity after injection of PRA. No significant effects of PRA on PPI were observed at any dose (0.1-10.0 mg/kg), but a significant reduction in startle magnitude was observed after 10 mg/kg PRA. In contrast, the D1/2 agonist, apomorphine (5 mg/kg) significantly reduced PPI in these mice. At doses of PRA that did not alter startle magnitude (0.3, 1, 3 mg/kg), significant decreases in the amount of locomotor and investigatory behavior were observed. Distinct from findings in rats and humans, it seems that either: (i) PRA does not activate D3Rs in C57BL/6J mice, or (ii) D3R agonists are not sufficient to alter PPI in this mouse strain.


Subject(s)
Benzothiazoles/pharmacology , Dopamine Agonists/pharmacology , Inhibition, Psychological , Motor Activity/drug effects , Reflex, Startle/drug effects , Animals , Apomorphine/pharmacology , Dose-Response Relationship, Drug , Exploratory Behavior/drug effects , Male , Mice , Mice, Inbred C57BL , Pramipexole , Sensory Gating/drug effects
9.
Hum Mol Genet ; 18(20): 3914-25, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19617637

ABSTRACT

Duplication of human chromosome 22q11.2 is associated with elevated rates of mental retardation, autism and many other behavioral phenotypes. However, because duplications cover 1.5-6 Mb, the precise manner in which segments of 22q11.2 causally affect behavior is not known in humans. We have now determined the developmental impact of over-expression of an approximately 190 kb segment of human 22q11.2, which includes the genes TXNRD2, COMT and ARVCF, on behaviors in bacterial artificial chromosome (BAC) transgenic (TG) mice. BAC TG mice and wild-type (WT) mice were tested for their cognitive capacities, affect- and stress-related behaviors and motor activity at 1 and 2 months of age. An enzymatic assay determined the impact of BAC over-expression on the activity level of COMT. BAC TG mice approached a rewarded goal faster (i.e. incentive learning), but were impaired in delayed rewarded alternation during development. In contrast, BAC TG and WT mice were indistinguishable in rewarded alternation without delays, spontaneous alternation, prepulse inhibition, social interaction, anxiety-, stress- and fear-related behaviors and motor activity. Compared with WT mice, BAC TG mice had an approximately 2-fold higher level of COMT activity in the prefrontal cortex, striatum and hippocampus. These data suggest that over-expression of this 22q11.2 segment enhances incentive learning and impairs the prolonged maintenance of working memory, but has no apparent effect on working memory per se, affect- and stress-related behaviors or motor capacity. High copy numbers of this 22q11.2 segment might contribute to a highly selective set of phenotypes in learning and cognition during development.


Subject(s)
Armadillo Domain Proteins/genetics , Catechol O-Methyltransferase/genetics , Cell Adhesion Molecules/genetics , Chromosomes, Human, Pair 22/genetics , Gene Expression Regulation, Developmental , Learning , Memory, Short-Term , Phosphoproteins/genetics , Thioredoxin Reductase 2/genetics , Animals , Armadillo Domain Proteins/metabolism , Autistic Disorder/genetics , Autistic Disorder/metabolism , Autistic Disorder/psychology , Catechol O-Methyltransferase/metabolism , Cell Adhesion Molecules/metabolism , Chromosomes, Human, Pair 22/metabolism , Disease Models, Animal , Female , Humans , Intellectual Disability/genetics , Intellectual Disability/metabolism , Intellectual Disability/psychology , Male , Mice , Mice, Transgenic , Phosphoproteins/metabolism , Thioredoxin Reductase 2/metabolism
10.
Hum Mol Genet ; 18(9): 1652-60, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19240081

ABSTRACT

Deletion or duplication of the human chromosome 22q11.2 is associated with many behavioral traits and neuropsychiatric disorders, including autism spectrum disorders and schizophrenia. However, why phenotypes vary widely among individuals with identical deletions or duplications of 22q11.2 and which specific 22q11.2 genes contribute to these phenotypes are still poorly understood. Previous studies have identified a approximately 200 kb 22q11.2 region that contributes to behavioral phenotypes in mice. We tested the role of Septin 5 (Sept5), a gene encoded in the approximately 200 kb region, in affective behaviors, cognitive capacities and motor activity. To evaluate the impact of genetic backgrounds on behavioral phenotypes of Sept5 deficiency, we used mice on two genetic backgrounds. Our data show that Sept5 deficiency decreased affiliative active social interaction, but this phenotypic expression was influenced by genetic backgrounds. In contrast, Sept5 deficiency decreased anxiety-related behavior, increased prepulse inhibition and delayed acquisition of rewarded goal approach, independent of genetic background. These data suggest that Sept5 deficiency exerts pleiotropic effects on a select set of affective behaviors and cognitive processes and that genetic backgrounds could provide an epistatic influence on phenotypic expression.


Subject(s)
Behavior, Animal , Cell Cycle Proteins/genetics , Gene Silencing , Motor Activity , Animals , Cell Cycle Proteins/metabolism , Female , Male , Mice , Mice, Knockout , Septins
11.
Psychopharmacology (Berl) ; 201(1): 55-66, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18604652

ABSTRACT

RATIONALE: The hallucinogenic tea known as ayahuasca is made from a combination of psychoactive plants that contribute the active components N,N-dimethyltryptamine (DMT) and 5-methoxy-DMT (5-MeO-DMT), as well as the monoamine oxidase (MAO) inhibitors (MAOIs) harmine and harmaline for oral activity. OBJECTIVE: The present study examined the effects of 5-MeO-DMT in combination with MAOIs in rats using the behavioral pattern monitor, which enables analyses of patterns of locomotor activity and exploration. Interaction studies using the serotonin (5-HT)(1A) antagonist WAY-100635 (1.0 mg/kg) and the 5-HT(2A) antagonist MDL 11,939 (1.0 mg/kg) were also performed to assess the respective contributions of these receptors to the behavioral effects of 5-MeO-DMT in MAOI-treated animals. RESULTS: 5-MeO-DMT (0.01, 0.1, and 1.0 mg/kg) decreased locomotor activity and investigatory behavior. In rats pretreated with a behaviorally inactive dose of harmaline (0.1 mg/kg), 1.0 mg/kg 5-MeO-DMT had biphasic effects on locomotor activity, initially reducing locomotion and then increasing activity as time progressed. The ability of harmaline to shift 5-MeO-DMT to a biphasic locomotor pattern was shared by the selective MAO(A) inhibitor clorgyline, whereas the selective MAO(B) inhibitor (-)-deprenyl was ineffective. The late hyperactivity induced by the combination of 1.0 mg/kg 5-MeO-DMT and 0.3 mg/kg clorgyline was blocked by pretreatment with MDL 11,939. Pretreatment with WAY-100635 failed to attenuate either the early hypoactivity or the late hyperactivity. CONCLUSIONS: The ability of harmaline to modify the behavioral effects of 5-MeO-DMT is mediated by the inhibition of MAO(A). Furthermore, 5-HT(2A) receptors are responsible for the late hyperactivity induced by 5-MeO-DMT in the presence of MAO(A) inhibitors.


Subject(s)
Exploratory Behavior/drug effects , Methoxydimethyltryptamines/pharmacology , Monoamine Oxidase Inhibitors/pharmacology , Psychotropic Drugs/pharmacology , DOM 2,5-Dimethoxy-4-Methylamphetamine/pharmacology , Animals , Behavior, Animal , Behavioral Research/instrumentation , Clorgyline/pharmacology , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Interactions , Drug Therapy, Combination , Harmaline/pharmacology , Hyperkinesis/chemically induced , Male , Methoxydimethyltryptamines/chemistry , Motor Activity/drug effects , Pattern Recognition, Automated , Pharmaceutical Vehicles/administration & dosage , Pharmaceutical Vehicles/chemistry , Piperazines/pharmacology , Piperidines/pharmacology , Psychotropic Drugs/chemistry , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT2A/drug effects , Receptor, Serotonin, 5-HT2A/physiology , Serotonin 5-HT1 Receptor Antagonists , Time Factors
12.
Psychopharmacology (Berl) ; 189(3): 319-29, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17013638

ABSTRACT

RATIONALE: The hallucinogen 5-methoxy-N,N-dimethyltryptamine (5-MeO-DMT) is structurally similar to other indoleamine hallucinogens such as LSD. The present study examined the effects of 5-MeO-DMT in rats using the Behavioral Pattern Monitor (BPM), which enables analyses of patterns of locomotor activity and exploration, and the prepulse inhibition of startle (PPI) paradigm. OBJECTIVES: A series of interaction studies using the serotonin (5-HT)(1A) antagonist WAY-100635 (1.0 mg/kg), the 5-HT(2A) antagonist M100907 (1.0 mg/kg), and the 5-HT(2C) antagonist SER-082 (0.5 mg/kg) were performed to assess the respective contributions of these receptors to the behavioral effects of 5-MeO-DMT (0.01, 0.1, and 1.0 mg/kg) in the BPM and PPI paradigms. RESULTS: 5-MeO-DMT decreased locomotor activity, investigatory behavior, the time spent in the center of the BPM chamber, and disrupted PPI. All of these effects were antagonized by WAY-100635 pretreatment. M100907 pretreatment failed to attenuate any of these effects, while SER-082 pretreatment only antagonized the PPI disruption produced by 5-MeO-DMT. CONCLUSIONS: While the prevailing view was that the activation of 5-HT(2) receptors is solely responsible for hallucinogenic drug effects, these results support a role for 5-HT(1A) receptors in the effects of the indoleamine hallucinogen 5-MeO-DMT on locomotor activity and PPI in rats.


Subject(s)
Methoxydimethyltryptamines/pharmacology , Motor Activity/drug effects , Neural Inhibition/drug effects , Receptor, Serotonin, 5-HT1A/physiology , Receptor, Serotonin, 5-HT2A/physiology , Animals , Lysergic Acid Diethylamide/pharmacology , Male , Neural Inhibition/physiology , Rats , Rats, Sprague-Dawley
13.
Behav Brain Res ; 169(1): 162-7, 2006 Apr 25.
Article in English | MEDLINE | ID: mdl-16406103

ABSTRACT

Male 129T2 and C57BL/6J mice were housed either in groups of three (socials) or singly (isolates) at weaning. Six and seven weeks later, prepulse inhibition (PPI), startle reactivity, and locomotor activity (LMA) were measured. Isolation-reared mice of both strains exhibited PPI deficits compared to socially reared controls in at least one of the two PPI test sessions. Isolation rearing had no effect on startle reactivity or habituation and only 129T2 isolates exhibited increased LMA. Isolation rearing induced locomotor hyperactivity and PPI deficits in mice and may be an effective developmental manipulation to use in combination with studies of genetically altered mice.


Subject(s)
Motor Activity/physiology , Neural Inhibition/physiology , Reflex, Startle/physiology , Social Isolation , Acoustic Stimulation , Analysis of Variance , Animals , Ion Channel Gating/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Models, Animal , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...