Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Psychopharmacology (Berl) ; 240(10): 2173-2185, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37615683

ABSTRACT

RATIONALE: Long-acting antipsychotics such as haloperidol decanoate are becoming more commonly used. Long-acting depot formulations have several advantages, but secondary negative effects of prolonged delivery, including motivational dysfunctions, could have debilitating effects. Assessing the behavioral changes that emerge during chronic antipsychotic administration in rats could provide insight regarding the development of motivational dysfunctions and drug tolerance. OBJECTIVES: Acute administration of dopamine D2 antagonists such as haloperidol induce motivational deficits in rats, as marked by a shift towards a low-effort bias during effort-based choice tasks. In the present studies, programmable subcutaneous infusion pumps provided continuous and controlled drug delivery of haloperidol. Animals were assessed using a fixed ratio (FR) 5 lever pressing schedule and the FR5/chow feeding test of effort-based choice. The adenosine A2A antagonist istradefylline was studied for its ability to reverse the effects of chronic haloperidol. RESULTS: Continuous chronic infusions of haloperidol produced significant reductions in FR5 performance and a shift from lever pressing to chow intake in rats tested on FR5/chow feeding choice, with no evidence of tolerance over the 4-week infusion period. Behavior returned to baseline during the vehicle-infusion washout period. Istradefylline significantly reversed the effects of haloperidol, increasing lever pressing and decreasing chow intake in haloperidol-treated rats. CONCLUSIONS: These studies provide an important behavioral characterization of the effects of chronically infused haloperidol, and demonstrate that A2A antagonism reverses the effects of chronic haloperidol. This research could contribute to the understanding and treatment of motivational dysfunctions seen in schizophrenia, Parkinson's disease, and other disorders involving dopamine.


Subject(s)
Antipsychotic Agents , Haloperidol , Animals , Rats , Haloperidol/pharmacology , Antipsychotic Agents/pharmacology , Purines , Adenosine
2.
Neuron ; 111(18): 2863-2880.e6, 2023 09 20.
Article in English | MEDLINE | ID: mdl-37451263

ABSTRACT

Changes in the function of inhibitory interneurons (INs) during cortical development could contribute to the pathophysiology of neurodevelopmental disorders. Using all-optical in vivo approaches, we find that parvalbumin (PV) INs and their immature precursors are hypoactive and transiently decoupled from excitatory neurons in postnatal mouse somatosensory cortex (S1) of Fmr1 KO mice, a model of fragile X syndrome (FXS). This leads to a loss of parvalbumin INs (PV-INs) in both mice and humans with FXS. Increasing the activity of future PV-INs in neonatal Fmr1 KO mice restores PV-IN density and ameliorates transcriptional dysregulation in S1, but not circuit dysfunction. Critically, administering an allosteric modulator of Kv3.1 channels after the S1 critical period does rescue circuit dynamics and tactile defensiveness. Symptoms in FXS and related disorders could be mitigated by targeting PV-INs.


Subject(s)
Fragile X Syndrome , Parvalbumins , Humans , Mice , Animals , Parvalbumins/genetics , Parvalbumins/metabolism , Fragile X Mental Retardation Protein/genetics , Interneurons/physiology , Neurons/metabolism , Touch , Fragile X Syndrome/genetics , Mice, Knockout , Disease Models, Animal
3.
Mol Psychiatry ; 26(7): 3018-3033, 2021 07.
Article in English | MEDLINE | ID: mdl-32814812

ABSTRACT

A cardinal feature of post-traumatic stress disorder (PTSD) is a long-lasting paradoxical alteration of memory with hypermnesia for salient traumatic cues and amnesia for peri-traumatic contextual cues. So far, pharmacological therapeutic approach of this stress-related disorder is poorly developed mainly because of the lack of animal model for this paradoxical memory alteration. Using a model that precisely recapitulates the two memory components of PTSD in mice, we tested if brexpiprazole, a new antipsychotic drug with pro-cognitive effects in rodents, may persistently prevent the expression of PTSD-like memory induced by injection of corticosterone immediately after fear conditioning. Acute administration of brexpiprazole (0.3 mg/kg) 7 days' post-trauma first blocks the expression of the maladaptive fear memory for a salient but irrelevant trauma-related cue. In addition, it enhances (with superior efficacy when compared to diazepam, prazosin, and escitalopram) memory for the traumatic context, correct predictor of the threat. This beneficial effect of brexpiprazole is overall maintained 1 week after treatment. In contrast brexpiprazole fully spares normal/adaptive cued fear memory, showing that the effect of this drug is specific to an abnormal/maladaptive (PTSD-like) fear memory of a salient cue. Finally, this treatment not only promotes the switch from PTSD-like to normal fear memory, but also normalizes most of the alterations in the hippocampal-amygdalar network activation associated with PTSD-like memory, as measured by C-Fos expression. Altogether, these preclinical data indicate that brexpiprazole could represent a new pharmacological treatment of PTSD promoting the normalization of traumatic memory.


Subject(s)
Quinolones , Stress Disorders, Post-Traumatic , Animals , Disease Models, Animal , Escitalopram , Fear , Mice , Quinolones/pharmacology , Stress Disorders, Post-Traumatic/drug therapy , Thiophenes
4.
Front Pharmacol ; 11: 427, 2020.
Article in English | MEDLINE | ID: mdl-32390829

ABSTRACT

Amphetamine (AMP), methylphenidate (MPH), and atomoxetine (ATX) are approved treatments for ADHD, and together with nicotine (NIC), represent pharmacological agents widely studied on cognitive domains including attention and impulsive action in humans. These agents thus represent opportunities for clinical observation to be reinvestigated in the preclinical setting, i.e., reverse translation. The present study investigated each drug in male, Long Evans rats trained to perform either (1) the five-choice serial reaction time task (5-CSRTT), (2) Go/NoGo task, or (3) a progressive ratio (PR) task, for the purpose of studying each drug on attention, impulsive action and motivation. Specific challenges were adopted in the 5-CSRTT designed to tax attention and impulsivity, i.e., high frequency of stimulus presentation (sITI), variable reduction in stimulus duration (sSD), and extended delay to stimulus presentation (10-s ITI). Initially, performance of a large (> 80) cohort of rats in each task variant was conducted to examine performance stability over repeated challenge sessions, and to identify subgroups of "high" and "low" attentive rats (sITI and sSD schedules), and "high" and "low" impulsives (10-s ITI). Using an adaptive sequential study design, the effects of AMP, MPH, ATX, and NIC were examined and contrasting profiles noted across the tests. Both AMP (0.03-0.3 mg/kg) and MPH (1-6 mg/kg) improved attentional performance in the sITI but not sSD or 10-s ITI condition, NIC (0.05-0.2 mg/kg) improved accuracy across all conditions. ATX (0.1-1 mg/kg) detrimentally affected performance in the sITI and sSD condition, notably in "high" performers. In tests of impulsive action, ATX reduced premature responses notably in the 10-s ITI condition, and also reduced false alarms in Go/NoGo. Both AMP and NIC increased premature responses in all task variants, although AMP reduced false alarms highlighting differences between these two measures of impulsive action. The effect of MPH was mixed and appeared baseline dependent. ATX reduced break point for food reinforcement suggesting a detrimental effect on motivation for primary reward. Taken together these studies highlight differences between AMP, MPH, and ATX which may translate to their clinical profiles. NIC had the most reliable effect on attentional accuracy, whereas ATX was reliably effective against all tests of impulsive action.

5.
J Clin Invest ; 129(5): 2145-2162, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30985297

ABSTRACT

Vacuolar H+-ATPase-dependent (V-ATPase-dependent) functions are critical for neural proteostasis and are involved in neurodegeneration and brain tumorigenesis. We identified a patient with fulminant neurodegeneration of the developing brain carrying a de novo splice site variant in ATP6AP2 encoding an accessory protein of the V-ATPase. Functional studies of induced pluripotent stem cell-derived (iPSC-derived) neurons from this patient revealed reduced spontaneous activity and severe deficiency in lysosomal acidification and protein degradation leading to neuronal cell death. These deficiencies could be rescued by expression of full-length ATP6AP2. Conditional deletion of Atp6ap2 in developing mouse brain impaired V-ATPase-dependent functions, causing impaired neural stem cell self-renewal, premature neuronal differentiation, and apoptosis resulting in degeneration of nearly the entire cortex. In vitro studies revealed that ATP6AP2 deficiency decreases V-ATPase membrane assembly and increases endosomal-lysosomal fusion. We conclude that ATP6AP2 is a key mediator of V-ATPase-dependent signaling and protein degradation in the developing human central nervous system.


Subject(s)
Central Nervous System/physiopathology , Neurodegenerative Diseases/diagnostic imaging , Neurodegenerative Diseases/genetics , Pluripotent Stem Cells/metabolism , Receptors, Cell Surface/genetics , Vacuolar Proton-Translocating ATPases/genetics , Adolescent , Alternative Splicing , Animals , Apoptosis , Brain/diagnostic imaging , Cell Death , Cell Differentiation , Cell Survival , Child, Preschool , Gene Deletion , Genetic Variation , HEK293 Cells , HeLa Cells , Humans , Lysosomes/metabolism , Male , Mice , Mice, Inbred C57BL , Neural Stem Cells/metabolism , Neurons/metabolism , Proton-Translocating ATPases/genetics , Proton-Translocating ATPases/physiology , Receptors, Cell Surface/physiology , Vacuolar Proton-Translocating ATPases/physiology
6.
Hum Mol Genet ; 28(5): 701-717, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30357341

ABSTRACT

Genetic disruptions of the forkhead box transcription factor FOXP2 in humans cause an autosomal-dominant speech and language disorder. While FOXP2 expression pattern are highly conserved, its role in specific brain areas for mammalian social behaviors remains largely unknown. Here we studied mice carrying a homozygous cortical Foxp2 deletion. The postnatal development and gross morphological architecture of mutant mice was indistinguishable from wildtype (WT) littermates. Unbiased behavioral profiling of adult mice revealed abnormalities in approach behavior towards conspecifics as well as in the reciprocal responses of WT interaction partners. Furthermore mutant mice showed alterations in acoustical parameters of ultrasonic vocalizations, which also differed in function of the social context. Cell type-specific gene expression profiling of cortical pyramidal neurons revealed aberrant regulation of genes involved in social behavior. In particular Foxp2 mutants showed the downregulation of Mint2 (Apba2), a gene involved in approach behavior in mice and autism spectrum disorder in humans. Taken together these data demonstrate that cortical Foxp2 is required for normal social behaviors in mice.


Subject(s)
Behavior, Animal , Cerebral Cortex/metabolism , Cerebral Cortex/physiopathology , Forkhead Transcription Factors/deficiency , Gene Deletion , Repressor Proteins/deficiency , Social Behavior , Animals , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/pathology , Homozygote , Mice , Mice, Knockout , Neurons/metabolism
7.
Pharmacol Biochem Behav ; 153: 141-146, 2017 02.
Article in English | MEDLINE | ID: mdl-28057524

ABSTRACT

Previous studies have shown that partial and full 5-HT1A receptor agonists reduce antipsychotic-induced catalepsy. Consequently, some antipsychotics combining balanced efficacy between dopamine (DA) D2 antagonism or partial agonism and 5-HT1A receptor agonism have a low propensity to induce extrapyramidal side effects (EPS), as reflected by low cataleptogenic activity in rodents. In the present experiments, we attempted to explore the importance of pre- and postsynaptic 5-HT1A agonistic properties of brexpiprazole and aripiprazole in the context of neurological side-effect liabilities. Additional measures of prefrontal cortical serotonin (5-HT) and DA levels using microdialysis were used to support that brexpiprazole has a preferential agonist effect on presynaptic 5-HT1A receptors. Brexpiprazole (3.0 and 10mg/kg, p.o.) as well as aripiprazole (8.0 and 30mg/kg, p.o.) failed to induce catalepsy in rats. Brexpiprazole (10mg/kg, p.o.) significantly reduced the cataleptic response induced by haloperidol (0.63mg/kg, s.c.), while aripiprazole (1.0-100mg/kg, p.o.) failed to reverse the effect of haloperidol and only showed a numeric decrease at 10mg/kg, (p.o.). When 5-HT1A receptors were blocked by the selective antagonist, WAY100635 (1.0mg/kg, s.c.), cataleptogenic properties of brexpiprazole (10mg/kg; p.o), but not aripiprazole (8.0 and 30mg/kg, p.o.) were unmasked. The ("biased") 5-HT1A receptor agonists F15599 (postsynaptic preference) and F13714 (presynaptic preference) had differential effects on haloperidol-induced catalepsy: F13714 (0.16mg/kg, s.c.) counteracted catalepsy, whereas F15599 (0.040mg/kg, s.c.) had no significant effect at regionally-selective doses. These data support a role of presynaptic 5-HT1A receptors in the anticataleptic effect of brexpiprazole. The selective 5-HT2A antagonist M100907 (0.10mg/kg, s.c.) had no effect on haloperidol-induced catalepsy, arguing against a major role of 5-HT2A receptors in the cataleptogenic profile of brexpiprazole. The findings with brexpiprazole were supported using microdialysis studies: Brexpiprazole (3.0 and 10mg/kg, p.o.) decreased extracellular 5-HT levels in the medial prefrontal cortex (mPFC), while it failed to affect extracellular DA in the same samples, suggesting that the 5-HT1A agonist properties of brexpiprazole may be preferentially presynaptic. In conclusion, these results confirm that brexpiprazole and aripiprazole have low propensities to induce EPS. However, the low EPS risk of brexpiprazole is more likely dependent on its agonist properties on presynaptic 5-HT1A receptors, while that of aripiprazole is less sensitive to 5-HT1A receptor antagonism.


Subject(s)
Basal Ganglia Diseases/chemically induced , Quinolones/toxicity , Receptor, Serotonin, 5-HT1A/physiology , Thiophenes/toxicity , Animals , Aripiprazole/toxicity , Catalepsy/chemically induced , Dopamine/analysis , Haloperidol/pharmacology , Male , Piperazines/pharmacology , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Serotonin/analysis
8.
Cell Rep ; 7(6): 1779-88, 2014 Jun 26.
Article in English | MEDLINE | ID: mdl-24931612

ABSTRACT

Over the course of cortical neurogenesis, the transition of progenitors from proliferation to differentiation requires a precise regulation of involved gene networks under varying environmental conditions. In order to identify such regulatory mechanisms, we analyzed microRNA (miRNA) target networks in progenitors during early and late stages of neurogenesis. We found that cyclin D1 is a network hub whose expression is miRNA-dosage sensitive. Experimental validation revealed a feedback regulation between cyclin D1 and its regulating miRNAs miR-20a, miR-20b, and miR-23a. Cyclin D1 induces expression of miR-20a and miR-20b, whereas it represses miR-23a. Inhibition of any of these miRNAs increases the developmental stage-specific mean and dynamic expression range (variance) of cyclin D1 protein in progenitors, leading to reduced neuronal differentiation. Thus, miRNAs establish robustness and stage-specific adaptability to a critical dosage-sensitive gene network during cortical neurogenesis. Understanding such network regulatory mechanisms for key developmental events can provide insights into individual susceptibilities for genetically complex neuropsychiatric disorders.


Subject(s)
Gene Regulatory Networks , MicroRNAs/genetics , Neurogenesis/genetics , Stem Cells/cytology , Animals , Cell Differentiation/genetics , Humans , Mice , Mice, Transgenic
9.
Science ; 339(6117): 332-5, 2013 Jan 18.
Article in English | MEDLINE | ID: mdl-23329050

ABSTRACT

Repeated traumatic events induce long-lasting behavioral changes that are key to organism adaptation and that affect cognitive, emotional, and social behaviors. Rodents subjected to repeated instances of aggression develop enduring social aversion and increased anxiety. Such repeated aggressions trigger a stress response, resulting in glucocorticoid release and activation of the ascending dopamine (DA) system. We bred mice with selective inactivation of the gene encoding the glucocorticoid receptor (GR) along the DA pathway, and exposed them to repeated aggressions. GR in dopaminoceptive but not DA-releasing neurons specifically promoted social aversion as well as dopaminergic neurochemical and electrophysiological neuroadaptations. Anxiety and fear memories remained unaffected. Acute inhibition of the activity of DA-releasing neurons fully restored social interaction in socially defeated wild-type mice. Our data suggest a GR-dependent neuronal dichotomy for the regulation of emotional and social behaviors, and clearly implicate GR as a link between stress resiliency and dopaminergic tone.


Subject(s)
Anxiety/metabolism , Dopamine/metabolism , Dopaminergic Neurons/metabolism , Receptors, Glucocorticoid/metabolism , Social Alienation , Social Isolation , Stress, Psychological/metabolism , Animals , Fear , Mice , Mice, Mutant Strains , Receptors, Dopamine/metabolism , Receptors, Glucocorticoid/genetics
10.
PLoS Genet ; 7(7): e1002145, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21765815

ABSTRACT

Forkhead-box protein P2 is a transcription factor that has been associated with intriguing aspects of cognitive function in humans, non-human mammals, and song-learning birds. Heterozygous mutations of the human FOXP2 gene cause a monogenic speech and language disorder. Reduced functional dosage of the mouse version (Foxp2) causes deficient cortico-striatal synaptic plasticity and impairs motor-skill learning. Moreover, the songbird orthologue appears critically important for vocal learning. Across diverse vertebrate species, this well-conserved transcription factor is highly expressed in the developing and adult central nervous system. Very little is known about the mechanisms regulated by Foxp2 during brain development. We used an integrated functional genomics strategy to robustly define Foxp2-dependent pathways, both direct and indirect targets, in the embryonic brain. Specifically, we performed genome-wide in vivo ChIP-chip screens for Foxp2-binding and thereby identified a set of 264 high-confidence neural targets under strict, empirically derived significance thresholds. The findings, coupled to expression profiling and in situ hybridization of brain tissue from wild-type and mutant mouse embryos, strongly highlighted gene networks linked to neurite development. We followed up our genomics data with functional experiments, showing that Foxp2 impacts on neurite outgrowth in primary neurons and in neuronal cell models. Our data indicate that Foxp2 modulates neuronal network formation, by directly and indirectly regulating mRNAs involved in the development and plasticity of neuronal connections.


Subject(s)
Brain/embryology , Forkhead Transcription Factors/genetics , Gene Regulatory Networks , Neurites/metabolism , Repressor Proteins/genetics , Animals , Cell Line, Tumor , Chromatin Immunoprecipitation , Corpus Striatum/growth & development , Gene Expression Profiling , Gene Expression Regulation, Developmental , Mice , Mice, Inbred C57BL , Models, Biological , Mutation , Oligonucleotide Array Sequence Analysis/methods , Primary Cell Culture , RNA, Messenger/genetics , RNA, Messenger/metabolism
11.
Neuropharmacology ; 59(6): 468-73, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20624407

ABSTRACT

Serotonin 2C receptors (5-HT(2C)R) have been shown to undergo post-transcriptional RNA editing. This modification affects the affinity, coupling and constitutive activity of the receptor. In vivo, manipulations such as stress or antidepressant administration dramatically modify the pattern of 5-HT(2C)R mRNA editing, suggesting that this phenomenon might be involved in the pathophysiology of stress-related disorders. Indeed, alterations of 5-HT(2C)R mRNA editing have been observed in depressed patients. Thus, the recent development of mice expressing either the non-edited (5-HT(2C)R-INI) or the fully-edited form of 5-HT(2C)R (5-HT(2C)R-VGV) provides a novel opportunity to investigate the relevance of this phenomenon in the context of stress-related disorders. We observed that both 5-HT(2C)R-INI and 5-HT(2C)R-VGV mice exhibit exaggerated anxiety-like behaviors in the elevated-plus maze paradigm. This phenotype was observed when the INI or VGV mutations were present in mice on a BALB/c background, as well as non-significant trends in the same direction in mice on a C57BL/6J background. In animal models of antidepressant-like activity, the absence of editing of 5-HT(2C)R mRNA (5-HT(2C)R-INI) induced an increase in the time spent immobile in the forced-swim test (FST) and tail suspension test (TST). Complete editing of 5-HT(2C) receptor mRNA (5-HT(2C)R-VGV) induced antidepressant-like behavior in the FST and TST, as reflected by a significant decrease in time spent immobile. These phenotypes were unrelated to alterations in locomotor activity in both 5-HT(2C)R-INI and 5-HT(2C)R-VGV. In the TST, these phenotypes were accompanied by a decrease and an increase in response to desipramine in 5-HT(2C)R-INI and 5-HT(2C)R-VGV, respectively. These data constitute the first in vivo demonstration of a role for 5-HT(2C)R mRNA editing in anxiety- and depression-related behaviors.


Subject(s)
Anxiety/genetics , Behavior, Animal/physiology , Depression/genetics , RNA Editing/genetics , Receptor, Serotonin, 5-HT2C/genetics , Analysis of Variance , Animals , Female , Hindlimb Suspension , Male , Mice , Motor Activity/genetics , Receptor, Serotonin, 5-HT2C/metabolism , Swimming , Tail
12.
Int J Neuropsychopharmacol ; 13(3): 321-34, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20003619

ABSTRACT

Clinically, SSRIs are widely prescribed in the treatment of several anxiety disorders, although very few pre-clinical studies have observed a beneficial effect of this class of drugs in animal models of anxiety. Furthermore, the biphasic pattern observed clinically, an exacerbation of anxiety followed by beneficial effects, is rarely observed in animal studies. In the present study we document this clinical phenomenon in several behavioural paradigms. While a single injection of citalopram induced anxiogenic effects, three administrations of citalopram were sufficient to elicit anxiolytic effects. Congruent with these data, we observed that short-term repeated administration of citalopram was accompanied by increased activation of cAMP response element-binding protein (CREB) in the hippocampus and desensitization of 5-HT1A receptors, two phenomena well associated with chronic rather than acute actions of antidepressants. Moreover, effects of citalopram were abolished in CREBalphaDelta mutant animals in the elevated zero maze (EZM) and tail suspension test (TST), but not in novelty-induced hypophagia (NIH). Further, the desensitization of 5-HT1A receptors elicited by citalopram was not affected by CREB deficiency. The significance of the EZM and TST paradigms in predicting therapeutic efficacy is well known while effects in NIH and 5-HT1A sensitization are less well-established. These data demonstrate that behavioural responses to citalopram are dependent on the frequency of its administration, and that these responses are differentially dependent on CREB function.


Subject(s)
Antidepressive Agents, Second-Generation/pharmacology , Anxiety , Citalopram/pharmacology , Depression , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Animals , Anxiety/drug therapy , CREB-Binding Protein/genetics , Chlordiazepoxide/pharmacology , Depression/drug therapy , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Schedule , Gene Expression Regulation/drug effects , Hindlimb Suspension/methods , Hypothermia/chemically induced , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Statistics, Nonparametric
13.
J Neurosci ; 28(48): 12834-44, 2008 Nov 26.
Article in English | MEDLINE | ID: mdl-19036977

ABSTRACT

RNA editing that converts adenosine to inosine replaces the gene-encoded Ile, Asn, and Ile (INI) of serotonin [5-hydroxytryptamine (5-HT)] receptor 2C (5-HT(2C)R) with Val, Gly, and Val (VGV). Up to 24 different 5-HT(2C)R isoforms are detected in different brain regions (Burns et al., 1997; Fitzgerald et al., 1999; Wang et al., 2000). To elucidate the physiological significance of 5-HT(2C)R mRNA editing, we derived mutant mouse lines harboring a knock-in INI or VGV allele, resulting in sole expression of one of two extremely different editing isoforms 5-HT(2C)R-INI (editing blocked) or -VGV (fully edited). Although INI mice grew normally, VGV mice had a severely reduced fat mass, despite compensatory hyperphagia, as a result of constitutive activation of the sympathetic nervous system and increased energy expenditure. Furthermore, serotonergic neurotransmission was oversensitized in VGV mice, most likely because of the increased cell surface expression of VGV receptors. Melanocortin 4 receptor (MC4R) regulates energy homeostasis (Balthasar et al., 2005; Heisler et al., 2006; Lam et al., 2008), and Mc4r(-/-) mice are obese because of hyperphagia and reduced energy expenditure (Huszar et al., 1997). However, the elevated energy expenditure of VGV mice could not be rescued in the Mc4r(-/-) background, indicating the presence of a distinct signaling pathway mediated via 5-HT(2C)R-VGV that dominates the MC4R-dependent pathway in control of energy expenditure. Our results highlight the importance of regulated 5-HT(2C)R mRNA editing, because dysregulation could result in the pathological consequences such as growth retardation seen in VGV mice.


Subject(s)
Adipose Tissue/metabolism , Energy Metabolism/genetics , RNA Editing/genetics , RNA, Messenger/genetics , Receptor, Serotonin, 5-HT2C/genetics , Amino Acid Sequence/genetics , Animals , Female , Gene Knock-In Techniques , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Protein Isoforms/genetics , Receptor, Melanocortin, Type 4/genetics , Receptor, Melanocortin, Type 4/metabolism , Serotonin/metabolism , Signal Transduction/genetics , Sympathetic Nervous System/physiopathology , Synaptic Transmission/genetics
14.
J Pharmacol Exp Ther ; 321(1): 172-7, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17215447

ABSTRACT

There is an increasing demand for a novel non-nicotinic, nondopaminergic therapeutic approach to nicotine addiction. GABAergic mechanisms have been implicated in drug dependence. Recently, a novel GABAB receptor allosteric-positive modulator, GS39783, was characterized. There are no investigations to date on the effects of GABAB receptor-positive modulators in animal models of nicotine reinforcement. Conditioned place preference (CPP) paradigms are based on the principle that animals, like humans, would learn to seek environmental stimuli that have been previously associated with rewarding events. Here we show that nicotine (0.06 mg/kg s.c.) induced a robust CPP response. Furthermore, GS39783 (30-100 mg/kg p.o.) during the conditioning phase blocked the rewarding effects of nicotine in the CPP paradigm in rats. However, GS39783 did not significantly alter the CPP effects of nicotine when given only immediately before the CPP test. A growing body of evidence suggests that repeated administration of drugs of abuse induced long-term molecular changes in brain plasticity, most notably an accumulation of DeltaFosB, in the striatal complex that contribute to the manifestation of dependence. There was a significant accumulation of DeltaFosB in the nucleus accumbens, but not in the dorsal striatum, of rats treated daily for 5 days with nicotine (0.06 mg/kg i.p.). GS39783 completely (30-100 mg/kg p.o.) counteracted these nicotine-induced molecular adaptations when given before the CPP acquisition phase but not when administered immediately before the test phase. Taken together, the behavioral and molecular changes induced by nicotine occur in concert and are concomitantly amenable to reversal by GABAB receptor-positive modulators.


Subject(s)
Cyclopentanes/pharmacology , GABA Modulators/pharmacology , Nicotine/antagonists & inhibitors , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Nucleus Accumbens/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Pyrimidines/pharmacology , Receptors, GABA-B/physiology , Animals , Blotting, Western , Conditioning, Operant/drug effects , Nucleus Accumbens/drug effects , Rats , Rats, Wistar , Reinforcement, Psychology , Reward
15.
Neuropsychopharmacology ; 32(2): 388-98, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16710312

ABSTRACT

Exposure to cocaine induces selective behavioral and molecular adaptations. In rodents, acute cocaine induces increased locomotor activity, whereas prolonged drug exposure results in behavioral locomotor sensitization, which is thought to be a consequence of drug-induced neuroadaptive changes. Recent attention has been given to compounds activating GABA(B) receptors as potential antiaddictive therapies. In particular, the principle of allosteric positive GABA(B) receptor modulators is very promising in this respect, as positive modulators lack the sedative and muscle relaxant properties of full GABA(B) receptor agonists such as baclofen. Here, we investigated the effects of systemic application of the GABA(B) receptor-positive modulator GS39783 (N,N'-dicyclopentyl-2-methylsulfanyl-5-nitro-pyrimidine-4, 6-diamine) in animals treated with acute and chronic cocaine administration. Both GS39783 and baclofen dose dependently attenuated acute cocaine-induced hyperlocomotion. Furthermore, both compounds also efficiently blocked cocaine-induced Fos induction in the striatal complex. In chronic studies, GS39783 induced a modest attenuation of cocaine-induced locomotor sensitization. Chronic cocaine induces the accumulation of the transcription factor deltaFosB and upregulates cAMP-response-element-binding protein (CREB) and dopamine- and cAMP-regulated phosphoprotein of 32 kDa (DARPP-32). GS39783 blocked the induction/activation of DARPP-32 and CREB in the nucleus accumbens and dorsal striatum and partially inhibited deltaFosB accumulation in the dorsal striatum. In summary, our data provide evidence that GS39783 attenuates the acute behavioral effects of cocaine exposure in rodents and in addition prevents the induction of selective long-term adaptive changes in dopaminergic signaling pathways. Further investigation of GABA(B) receptor-positive modulation as a novel therapeutic strategy for the treatment of cocaine dependence and possibly other drugs of abuse is therefore warranted.


Subject(s)
Brain/drug effects , Cocaine-Related Disorders/metabolism , Cocaine/antagonists & inhibitors , Dopamine/metabolism , GABA Agonists/pharmacology , Receptors, GABA-B/drug effects , Animals , Baclofen/pharmacology , Behavior, Addictive/drug therapy , Behavior, Addictive/metabolism , Behavior, Addictive/physiopathology , Brain/metabolism , Brain/physiopathology , Cocaine/pharmacology , Cocaine-Related Disorders/drug therapy , Cocaine-Related Disorders/physiopathology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/physiopathology , Cyclic AMP Response Element-Binding Protein/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclopentanes/pharmacology , Cyclopentanes/therapeutic use , Disease Models, Animal , Dopamine Uptake Inhibitors/adverse effects , Dopamine Uptake Inhibitors/antagonists & inhibitors , Dopamine and cAMP-Regulated Phosphoprotein 32/drug effects , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Dose-Response Relationship, Drug , GABA Agonists/therapeutic use , Hyperkinesis/chemically induced , Hyperkinesis/drug therapy , Hyperkinesis/metabolism , Male , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-fos/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Receptors, GABA-B/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
16.
Neuropsychopharmacology ; 31(6): 1112-22, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16237391

ABSTRACT

Regulation of neurotransmission via group-III metabotropic glutamate receptors (mGluR4, -6, -7, and -8) has recently been implicated in the pathophysiology of affective disorders, such as major depression and anxiety. For instance, mice with a targeted deletion of the gene for mGluR7 (mGluR7-/-) showed antidepressant and anxiolytic-like effects in a variety of stress-related paradigms, including the forced swim stress and the stress-induced hyperthermia tests. Deletion of mGluR7 reduces also amygdala- and hippocampus-dependent conditioned fear and aversion responses. Since the hypothalamic-pituitary-adrenal (HPA) axis regulates the stress response we investigate whether parameters of the HPA axis at the levels of selected mRNA transcripts and endocrine hormones are altered in mGluR7-deficient mice. Over all, mGluR7-/- mice showed only moderately lower serum levels of corticosterone and ACTH compared with mGluR7+/+ mice. More strikingly however, we found strong evidence for upregulated glucocorticoid receptor (GR)-dependent feedback suppression of the HPA axis in mice with mGluR7 deficiency: (i) mRNA transcripts of GR were significantly upregulated in the hippocampus of mGluR7-/- animals, (ii) similar increases were seen with 5-HT1A receptor transcripts, which are thought to be directly controlled by the transcription factor GR and finally (iii) mGluR7-/- mice showed elevated sensitivity to dexamethasone-induced suppression of serum corticosterone when compared with mGluR7+/+ animals. These results indicate that mGluR7 deficiency causes dysregulation of HPA axis parameters, which may account, at least in part, for the phenotype of mGluR7-/- mice in animal models for anxiety and depression. In addition, we present evidence that protein levels of brain-derived neurotrophic factor are also elevated in the hippocampus of mGluR7-/- mice, which we discuss in the context of the antidepressant-like phenotype found in those animals. We conclude that genetic ablation of mGluR7 in mice interferes at multiple sites in the neuronal circuitry and molecular pathways implicated in affective disorders.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Hippocampus/metabolism , Hypothalamo-Hypophyseal System/metabolism , Pituitary-Adrenal System/metabolism , Receptors, Metabotropic Glutamate/deficiency , Stress, Physiological/metabolism , Adrenocorticotropic Hormone/blood , Animals , Body Weight/drug effects , Body Weight/genetics , Brain-Derived Neurotrophic Factor/genetics , Dexamethasone/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Glucocorticoids/pharmacology , Hippocampus/drug effects , Hypothalamo-Hypophyseal System/drug effects , Immunoassay/methods , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pituitary-Adrenal System/drug effects , RNA, Messenger/metabolism , Radioimmunoassay/methods , Receptor, Serotonin, 5-HT1A/genetics , Receptor, Serotonin, 5-HT1A/metabolism , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods
17.
Proc Natl Acad Sci U S A ; 102(51): 18712-7, 2005 Dec 20.
Article in English | MEDLINE | ID: mdl-16339898

ABSTRACT

Metabotropic glutamate receptor (mGluR) subtypes (mGluR1 to mGluR8) act as important pre- and postsynaptic regulators of neurotransmission in the CNS. These receptors consist of two domains, an extracellular region containing the orthosteric agonist site and a transmembrane heptahelical domain involved in G protein activation and recognition of several recently synthesized pharmacological modulators. The presynaptic receptor mGluR7 shows the highest evolutionary conservation within the family, but no selective pharmacological tool was known. Here we characterize an mGluR7-selective agonist, N,N'-dibenzhydrylethane-1,2-diamine dihydrochloride (AMN082), which directly activates receptor signaling via an allosteric site in the transmembrane domain. At transfected mammalian cells expressing mGluR7, AMN082 potently inhibits cAMP accumulation and stimulates GTPgammaS binding (EC50-values, 64-290 nM) with agonist efficacies comparable with those of L-2-amino-4-phosphonobutyrate (L-AP4) and superior to those of L-glutamate. AMN082 (< or = 10 microM) failed to show appreciable activating or inhibitory effects at other mGluR subtypes and selected ionotropic GluRs. Chimeric receptor studies position the binding site of AMN082 in the transmembrane region of mGluR7, and we demonstrate that this allosteric agonist has little, if any, effect on the potency of orthosteric ligands. Here we provide evidence for full agonist activity mediated by the heptahelical domain of family 3 G protein-coupled receptors (which have mGluR-like structure) that may lead to drug development opportunities. Further, AMN082 is orally active, penetrates the blood-brain barrier, and elevates the plasma stress hormones corticosterone and corticotropin in an mGluR7-dependent fashion. Therefore, AMN082 is a valuable tool for unraveling the role of mGluR7 in stress-related CNS disorders.


Subject(s)
Benzhydryl Compounds/pharmacology , Receptors, Metabotropic Glutamate/agonists , Receptors, Metabotropic Glutamate/metabolism , Signal Transduction/drug effects , Stress, Physiological/physiopathology , Allosteric Regulation , Allosteric Site/drug effects , Animals , CHO Cells , Cricetinae , Hormones/metabolism , Receptors, Metabotropic Glutamate/chemistry
18.
Neurosci Biobehav Rev ; 29(4-5): 571-625, 2005.
Article in English | MEDLINE | ID: mdl-15890404

ABSTRACT

Since its introduction almost 20 years ago, the tail suspension test has become one of the most widely used models for assessing antidepressant-like activity in mice. The test is based on the fact that animals subjected to the short-term, inescapable stress of being suspended by their tail, will develop an immobile posture. Various antidepressant medications reverse the immobility and promote the occurrence of escape-related behaviour. This review focuses on the utility this test as part of a research program aimed at understanding the mechanism of action of antidepressants. We discuss the inherent difficulties in modeling depression in rodents. We describe how the tail suspension differs from the closely related forced swim test. Further, we address some key issues associated with using the TST as a model of antidepressant action. We discuss issues regarding whether it satisfies criteria to be a valid model for assessing depression-related behavioural traits. We elaborate on the tests' ease of use, strain differences observed in the test and gender effects in the test. We focus on the utility of the test for genetic analysis. Furthermore, we discuss the concept of whether immobility maybe a behavioural trait relevant to depression. All of the available pharmacological data using the test in genetically modified mice is collated. Special attention is given to selective breeding programs such as the Rouen 'depressed' mice which have been bred for high and low immobility in the tail suspension test. We provide an extensive pooling of the pharmacological studies published to date using the test. Finally, we provide novel pharmacological validation of an automated system (Bioseb) for assessing immobility. Taken together, we conclude that the tail suspension test is a useful test for assessing the behavioural effects of antidepressant compounds and other pharmacological and genetic manipulations relevant to depression.


Subject(s)
Antidepressive Agents/therapeutic use , Depressive Disorder/drug therapy , Disease Models, Animal , Hindlimb Suspension/methods , Analysis of Variance , Animals , Behavior, Animal/drug effects , Depressive Disorder/genetics , Dose-Response Relationship, Drug , Humans , Immobilization , Male , Mice , Mice, Inbred Strains , Predictive Value of Tests , Reproducibility of Results
19.
Neuroreport ; 16(3): 307-10, 2005 Feb 28.
Article in English | MEDLINE | ID: mdl-15706241

ABSTRACT

Metabotropic GABAB receptors predominantly function as heterodimers of GABAB(1) and GABAB(2) subunits, but GABAB(1) can also form functional receptors in the absence of GABAB(2). Mice lacking the GABAB(1) subunit have altered behavioural responses in tests for anxiety and depression. In these studies, we investigated anxiety and depression in GABAB(2)-deficient mice. We compared the effects directly with that of genetic deletion of the GABAB(1) receptor subunit. Both GABAB(1) and GABAB(2)-deficient mice were found to be more anxious than wild type in the light-dark box paradigm. In contrast, these mice exhibited an antidepressant-like behaviour in the forced swim test. Taken together, these data suggest that heterodimeric GABAB(1,2) receptors are required for the normal regulation of emotional behaviour.


Subject(s)
Anxiety/genetics , Depression/genetics , Receptors, GABA-B/deficiency , Receptors, GABA-B/genetics , Animals , Behavior, Animal/physiology , Exploratory Behavior/physiology , Immobilization/physiology , Mice , Mice, Knockout , Motor Activity/genetics , Protein Subunits/deficiency , Protein Subunits/genetics , Reaction Time/physiology , Swimming
20.
Eur J Pharmacol ; 497(1): 119-20, 2004 Aug 16.
Article in English | MEDLINE | ID: mdl-15321743

ABSTRACT

Recently, we demonstrated that mice lacking the GABA(B(1)) subunit were more anxious than wild-type animals in several behavioural paradigms, most notably in the light-dark test. In an attempt to assess the effects of classical benzodiazepine anxiolytics on anxiety-like behaviour observed in these mice, animals were administered either chlordiazepoxide (10 mg/kg, p.o.) or diazepam (7.5 mg/kg, p.o.) prior to testing in the light-dark box. Surprisingly, in contrast with the wild-type mice, neither benzodiazepines decreased anxiety-like behaviour in GABA(B(1))(-/-) mice. These data suggest that targeted deletion of GABA(B(1)) subunit alters GABA(A) receptor function in vivo.


Subject(s)
Anti-Anxiety Agents/pharmacology , Chlordiazepoxide/pharmacology , Diazepam/pharmacology , Receptors, GABA-B/physiology , Animals , Anti-Anxiety Agents/therapeutic use , Anxiety/drug therapy , Anxiety/genetics , Anxiety/psychology , Chlordiazepoxide/therapeutic use , Diazepam/therapeutic use , Female , Male , Mice , Mice, Knockout , Motor Activity/drug effects , Receptors, GABA-A/physiology , Receptors, GABA-B/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...