Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Neurobiol Learn Mem ; 141: 44-52, 2017 May.
Article in English | MEDLINE | ID: mdl-28341151

ABSTRACT

Schizophrenia is a debilitating brain disorder characterized by disturbances of emotion, perception and cognition. Cognitive impairments predict functional outcome in schizophrenia and are detectable even in the prodromal stage of the disorder. However, our understanding of the underlying neurobiology is limited and procognitive treatments remain elusive. We recently demonstrated that mice heterozygous for an inactivated allele of the schizophrenia-associated Brd1 gene (Brd1+/- mice) display behaviors reminiscent of schizophrenia, including impaired social cognition and long-term memory. Here, we further characterize performance of these mice by following the preclinical guidelines recommended by the 'Measurement and Treatment Research to Improve Cognition in Schizophrenia (MATRICS)' and 'Cognitive Neuroscience Treatment Research to Improve Cognition in Schizophrenia (CNTRICS)' initiatives to maximize translational value. Brd1+/- mice exhibit relational encoding deficits, compromised working and long term memory, as well as impaired executive cognitive functioning with cognitive behaviors relying on medial prefrontal cortex being particularly affected. Akin to patients with schizophrenia, the cognitive deficits displayed by Brd1+/- mice are not global, but selective. Our results underline the value of Brd1+/- mice as a promising tool for studying the neurobiology of cognitive deficits in schizophrenia.


Subject(s)
Cognition Disorders/genetics , Cognition/physiology , Executive Function/physiology , Histone Acetyltransferases/genetics , Schizophrenia/genetics , Alleles , Animals , Behavior, Animal/physiology , Cognition Disorders/physiopathology , Disease Models, Animal , Heterozygote , Male , Memory, Long-Term/physiology , Memory, Short-Term/physiology , Mice , Mice, Knockout , Prefrontal Cortex/physiopathology , Schizophrenia/physiopathology
2.
Eur J Pharmacol ; 761: 79-85, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-25941078

ABSTRACT

Here, we present a preliminary pharmacological characterisation of Lu AF33241, a novel, brain penetrant phosphodiesterase inhibitor of (PDE) 2A and 10A tool compound, in in vitro/in vivo assays indicative of PDE2A and/or PDE10A inhibition, and in vivo models/assays relevant to cognitive processing and antipsychotic-like activity. An assay was also included to investigate potential effects on motor activity. The in vitro selectivity of Lu AF33241 was determined against a panel of PDE enzymes. Lu AF33241 potently inhibited both full-length recombinant hPDE2A (Ki=4.2nM) and hPDE10A (Ki=42nM). The compound moderately inhibited both hPDE1C (Ki=1200nM), hPDE7B (Ki=890nM), and hPDE11A (Ki=1800nM). Lu AF33241 displayed a Ki above 5000nM against all other tested members of the PDE family. Albeit within a narrow dose range, Lu AF33241 attenuated sub-chronic phencyclidine-induced deficits in novel object recognition (3 and 10mg/kg), displayed antipsychotic-like activity in the conditioned avoidance response paradigm (10mg/kg), and did not induce catalepsy within a dose-range of 2-6mg/kg. Further catalepsy studies are needed to investigate a predictive safety window. Lu AF33241 represents a novel PDE2A/PDE10A inhibitor tool compound that may serve to further the understanding of the roles played by these enzymes in various CNS disorders.


Subject(s)
Antipsychotic Agents/pharmacology , Behavior, Animal/drug effects , Blood-Brain Barrier/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 2/antagonists & inhibitors , Phosphodiesterase Inhibitors/pharmacology , Phosphoric Diester Hydrolases/metabolism , Quinoxalines/pharmacology , Triazoles/pharmacology , Animals , Antipsychotic Agents/metabolism , Antipsychotic Agents/toxicity , Avoidance Learning/drug effects , Capillary Permeability , Catalepsy/chemically induced , Cognition/drug effects , Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism , Exploratory Behavior/drug effects , Humans , Male , Motor Activity/drug effects , Phosphodiesterase Inhibitors/metabolism , Phosphodiesterase Inhibitors/toxicity , Quinoxalines/metabolism , Rats, Sprague-Dawley , Rats, Wistar , Recognition, Psychology/drug effects , Triazoles/metabolism
3.
Psychopharmacology (Berl) ; 231(16): 3151-67, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24577516

ABSTRACT

Here, we present the pharmacological characterisation of Lu AF64280, a novel, selective, brain penetrant phosphodiesterase (PDE) 2A inhibitor, in in vitro/in vivo assays indicative of PDE2A inhibition, and in vivo models/assays relevant to cognitive processing or antipsychotic-like activity. The in vitro selectivity of Lu AF64280 was determined against a panel of PDE enzymes and 3',5'-cyclic guanosine monophosphate (cGMP) levels in the hippocampus were determined using in vivo microdialysis. Lu AF64280 potently inhibited hPDE2A (Ki = 20 nM), 50-fold above moderate inhibition of both hPDE9A (Ki = 1,000 nM) and hPDE10A (Ki = 1,800 nM), and displayed a >250-fold selectivity over all other full-length human recombinant PDE family members (Ki above 5,000 nM). Lu AF64280 (20 mg/kg) significantly increased cGMP levels in the hippocampus (p < 0.01 versus vehicle-treated mice), attenuated sub-chronic phencyclidine-induced deficits in novel object exploration in rats (10 mg/kg, p < 0.001 versus vehicle-treated), blocked early postnatal phencyclidine-induced deficits in the intradimensional/extradimensional shift task in rats (1 and 10 mg/kg, p < 0.001 versus vehicle-treated) and attenuated spontaneous P20-N40 auditory gating deficits in DBA/2 mice (20 mg/kg, p < 0.05 versus vehicle-treated). In contrast, Lu AF64280 failed to attenuate phencyclidine-induced hyperactivity in mice, and was devoid of antipsychotic-like activity in the conditioned avoidance response paradigm in rats, at any dose tested. Lu AF64280 represents a novel tool compound for selective PDE2A inhibition that substantiates a critical role of this enzyme in cognitive processes under normal and pathological conditions.


Subject(s)
Antipsychotic Agents/pharmacology , Cognition Disorders/etiology , Cognition Disorders/psychology , Cyclic Nucleotide Phosphodiesterases, Type 2/antagonists & inhibitors , Heterocyclic Compounds, 4 or More Rings/pharmacology , Phosphodiesterase Inhibitors/pharmacology , Schizophrenia/complications , Schizophrenia/drug therapy , Schizophrenic Psychology , Animals , Antipsychotic Agents/pharmacokinetics , Avoidance Learning/drug effects , Behavior, Animal/drug effects , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism , Excitatory Amino Acid Antagonists/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacokinetics , Hippocampus/metabolism , Humans , Hyperkinesis/chemically induced , Hyperkinesis/prevention & control , Male , Mice , Mice, Inbred DBA , Phencyclidine/antagonists & inhibitors , Phencyclidine/pharmacology , Phosphodiesterase Inhibitors/pharmacokinetics , Rats , Recognition, Psychology/drug effects , Recombinant Proteins , Sensory Gating/drug effects
4.
Bioorg Med Chem Lett ; 24(1): 288-93, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24291041

ABSTRACT

In this Letter, we describe a chemical lead optimization campaign starting from a novel, weak α7 nicotinic acetylcholine receptor positive allosteric modulator (PAM) hit from a HTS screen. Exploration of the structure-activity relationships for α7 PAM potency, intrinsic hepatic clearance, the structure-property relationships for lipophilicity, and thermodynamic solubility, led to the identification of Lu AF58801: a potent, orally available, brain penetrant PAM of the α7 nicotinic acetylcholine receptor, showing efficacy in a novel object recognition task in rats treated subchronically with phencyclidine (PCP).


Subject(s)
Brain/drug effects , Cognition Disorders/drug therapy , Cyclopropanes/pharmacology , Drug Discovery , Phenylethyl Alcohol/analogs & derivatives , alpha7 Nicotinic Acetylcholine Receptor/metabolism , Administration, Oral , Allosteric Regulation/drug effects , Animals , Brain/metabolism , Cognition Disorders/chemically induced , Cyclopropanes/administration & dosage , Cyclopropanes/chemistry , Dose-Response Relationship, Drug , Humans , Molecular Structure , Phencyclidine/administration & dosage , Phenylethyl Alcohol/administration & dosage , Phenylethyl Alcohol/chemistry , Phenylethyl Alcohol/pharmacology , Rats , Rats, Inbred Strains , Structure-Activity Relationship
5.
Neuropharmacology ; 73: 183-91, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23748055

ABSTRACT

As affective and cognitive disturbances frequently co-occur in psychiatric disorders, research into opportunities to simultaneously target both entities is warranted. These disorders are typically treated with monoamine reuptake inhibitors (MRIs), whereas ongoing research suggests that symptoms also improve by nicotinic acetylcholine receptor (nAChR) activation. Preclinical studies have corroborated this and also demonstrated a synergistic antidepressant-like action when nAChR agonists and MRIs are combined. Here, we present the in vitro and in vivo profile of NS9775, a combined full α7 nAChR agonist and triple MRI. NS9775 potently inhibited [(3)H]α-bungarotoxin binding in vitro (Ki: 1.8 nM), and ex vivo (ED50: 3.6 mg/kg), showing negligible activity at α4ß2-(Ki: 1720 nM) or α1-containing nAChRs (Ki: 12,200 nM). In α7-expressing oocytes, NS9775 displayed an EC50 value of 280 nM, with a maximal response of 77% relative to a saturating acetylcholine concentration. Furthermore, NS9775 inhibited cortical [(3)H]5-HT, [(3)H]NA and [(3)H]DA uptake equipotently (14-43 nM), and inhibited striatal [(3)H]WIN35,428 binding (ED50: 9.1 mg/kg). Behaviourally in mice, NS9775 (0.3-3.0 mg/kg) reversed scopolamine-induced deficits in a modified Y-maze and MK-801-induced learning deficits in 5-trial inhibitory avoidance. Swim distance in the forced swim test was increased by 30 mg/kg NS9775, and 10 and 30 mg/kg NS9775 reduced digging behaviour in the marble burying paradigm and increased the number of punished crossings in the four plate test. This pro-cognitive, antidepressant-like and anxiolytic-like effect of NS9775 suggests that combining α7 nAChR agonism and triple monoamine reuptake inhibition could be a step in the evolution of pharmacological treatments of affective and/or cognitive disturbances.


Subject(s)
Affective Symptoms/drug therapy , Cognition Disorders/drug therapy , Naphthalenes/pharmacology , Neurotransmitter Uptake Inhibitors/pharmacology , Nicotinic Agonists/pharmacology , Animals , Avoidance Learning/drug effects , Behavior, Animal/drug effects , Brain/diagnostic imaging , Brain/metabolism , Bungarotoxins , Cognition Disorders/chemically induced , Drug Evaluation, Preclinical , Female , Male , Maze Learning/drug effects , Mice , Naphthalenes/therapeutic use , Punishment , Radioligand Assay , Radionuclide Imaging , Tritium
6.
Biochem Pharmacol ; 85(9): 1363-9, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23415905

ABSTRACT

Autoinduction in drug metabolism is a known phenomenon observed when a drug induces the enzymes responsible for its own metabolism. The potency, rate and extent of autoinduction following a given treatment paradigm may have therapeutic implications in clinic as well as for in vivo pharmacological assessments in animals. RO4938581, an imidazo-triazolo-benzodiazepine, is a novel GABAAα5 negative modulator recently pursued for the treatment of cognitive dysfunctions. As circulating plasma levels of RO4938581 were shown to decrease rapidly after repeated dosing in rats, with CYP1A2 being involved in the metabolism of the compound, we examined the potential role of RO4938581-mediated autoinduction of CYP1A2. Incubation of rat hepatocytes with RO4938581 revealed potent CYP1A2 induction with significant increase in enzymatic activity at concentrations of 0.1nM and RO4938581 was shown to be 700-fold more potent than ß-napththoflavone. Ex vivo studies revealed a 7-fold increase in metabolic CYP1A2 activity in liver microsomes prepared from rats administered with 0.1mg/kg of RO4938581 24h before. This induction profile was reflected in vivo in pharmacokinetic studies in rats where an 8-fold reduction in plasma exposure was observed after a second dose. The reduction in plasma exposures due to CYP1A2 autoinduction were confirmed functionally in contextual fear conditioning paradigm in rats, where a positive pharmacological effect observed after acute drug administration disappeared completely after sub-chronic dosing. Together, these findings suggest that RO4938581 possesses potent CYP1A2 autoinductive properties in rats and may serve as a tool for mechanistic metabolism or drug-drug interaction studies encircling this enzyme in rats.


Subject(s)
Benzodiazepines/pharmacology , Cytochromes/biosynthesis , GABA Modulators/pharmacology , Imidazoles/pharmacology , Receptors, GABA-A/metabolism , Animals , Benzodiazepines/pharmacokinetics , Cells, Cultured , Conditioning, Classical/drug effects , Cytochrome P-450 CYP1A2 , Fear/drug effects , GABA Modulators/pharmacokinetics , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Imidazoles/pharmacokinetics , Male , Rats
7.
Front Pharmacol ; 3: 11, 2012.
Article in English | MEDLINE | ID: mdl-22347859

ABSTRACT

Dopamine (DA) containing midbrain neurons play critical roles in several psychiatric and neurological diseases, including schizophrenia and attention deficit hyperactivity disorder, and the substantia nigra pars compacta neurons selectively degenerate in Parkinson's disease. Pharmacological modulation of DA receptors and transporters are well established approaches for treatment of DA-related disorders. Direct modulation of the DA system by influencing the discharge pattern of these autonomously firing neurons has yet to be exploited as a potential therapeutic strategy. Small conductance Ca(2+)-activated K(+) channels (SK channels), in particular the SK3 subtype, are important in the physiology of DA neurons, and agents modifying SK channel activity could potentially affect DA signaling and DA-related behaviors. Here we show that cyclohexyl-[2-(3,5-dimethyl-pyrazol-1-yl)-6-methyl-pyrimidin-4-yl]-amine (CyPPA), a subtype-selective positive modulator of SK channels (SK3 > SK2 > > > SK1, IK), decreased spontaneous firing rate, increased the duration of the apamin-sensitive afterhyperpolarization, and caused an activity-dependent inhibition of current-evoked action potentials in DA neurons from both mouse and rat midbrain slices. Using an immunocytochemically and pharmacologically validated DA release assay employing cultured DA neurons from rats, we show that CyPPA repressed DA release in a concentration-dependent manner with a maximal effect equal to the D2 receptor agonist quinpirole. In vivo studies revealed that systemic administration of CyPPA attenuated methylphenidate-induced hyperactivity and stereotypic behaviors in mice. Taken together, the data accentuate the important role played by SK3 channels in the physiology of DA neurons, and indicate that their facilitation by CyPPA profoundly influences physiological as well as pharmacologically induced hyperdopaminergic behavior.

8.
Pharmacol Biochem Behav ; 100(3): 624-9, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22108649

ABSTRACT

Emerging evidence points to an involvement of nicotinic acetylcholine receptors (nAChRs) in major depression. Nicotine improves symptoms of depression in humans and shows antidepressant-like effects in rodents. Monoamine release is facilitated by nAChR stimulation, and nicotine-evoked serotonin (5-HT) release has been shown to depend on α7 nAChR activation. The α7 nAChR agonist PNU-282987 shows no antidepressant-like activity when tested alone in the mouse forced swim (mFST) or tail suspension tests (mTST). However, in combination with a sub-active dose of the selective 5-HT reuptake inhibitor citalopram, inducing ~50% 5-HT reuptake inhibition, PNU-282987 has shown marked antidepressant-like effects in the mFST. SSR180711 is a recently described α7 nAChR agonist that has shown antidepressant-like activity in the rat forced swim test. To address the possibility that 5-HT reuptake inhibition contributes to the antidepressant-like profile of SSR180711, we compared the behavioural and biochemical profiles of PNU-282987 and SSR180711. In the mFST and mTST, SSR180711 (3-30 mg/kg, s.c.) showed dose-dependent antidepressant-like activity, while PNU-282987 (3-30 mg/kg, s.c.) showed no significant effect. The ED(50) to displace [³H]α-bungarotoxin binding was 1.7 and 5.5 mg/kg for SSR180711 and PNU-282987, respectively, suggesting that both compounds produce near-maximal α7 nAChR occupancy at the highest dose. While PNU-282987 did not affect ex vivo [³H]5-HT uptake, SSR180711 inhibited [³H]5-HT uptake with an ED50 of 30 mg/kg. This degree of inhibition is similar to that observed with a citalopram dose of ~2.4 mg/kg, a dose that is normally not active in the mFST or mTST. This suggests that the antidepressant-like activity of SSR180711 may involve partial 5-HT reuptake inhibition. SSR180711 therefore represents a compound displaying the synergistic effect of α7 nAChR agonism combined with partial 5-HT reuptake inhibition previously described. The addition of α7 nAChR agonism to classical monoamine-based mechanisms may represent a novel option for the improved treatment of major depression.


Subject(s)
Antidepressive Agents/therapeutic use , Benzamides/therapeutic use , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Bridged Bicyclo Compounds/therapeutic use , Nicotinic Agonists/therapeutic use , Receptors, Nicotinic/chemistry , Selective Serotonin Reuptake Inhibitors/therapeutic use , Serotonin Plasma Membrane Transport Proteins/chemistry , Animals , Antidepressive Agents/administration & dosage , Antidepressive Agents/metabolism , Antidepressive Agents/pharmacokinetics , Behavior, Animal/drug effects , Benzamides/administration & dosage , Benzamides/metabolism , Benzamides/pharmacokinetics , Bridged Bicyclo Compounds/administration & dosage , Bridged Bicyclo Compounds/metabolism , Bridged Bicyclo Compounds/pharmacokinetics , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Bridged Bicyclo Compounds, Heterocyclic/metabolism , Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Citalopram/therapeutic use , Depression/drug therapy , Depression/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Synergism , Drug Therapy, Combination , Female , Mice , Mice, Inbred Strains , Molecular Targeted Therapy , Nicotinic Agonists/administration & dosage , Nicotinic Agonists/metabolism , Nicotinic Agonists/pharmacokinetics , Receptors, Nicotinic/administration & dosage , Receptors, Nicotinic/metabolism , Serotonin/metabolism , Tissue Distribution , alpha7 Nicotinic Acetylcholine Receptor
9.
Psychopharmacology (Berl) ; 221(3): 451-68, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22124672

ABSTRACT

RATIONALE: A growing body of evidence suggests that negative modulation of γ-aminobutyric acid (GABA) GABA(A) α5 receptors may be a promising strategy for the treatment of certain facets of cognitive impairment; however, selective modulators of GABA(A) α5 receptors have not yet been tested in "schizophrenia-relevant" cognitive assay/model systems in animals. OBJECTIVES: The objectives of this study were to investigate the potential of RO4938581, a negative modulator of GABA(A) α5 receptors, and to attenuate cognitive impairments induced following sub-chronic (sub-PCP) and early postnatal PCP (neo-PCP) administration in the novel object recognition (NOR) and intra-extradimensional shift (ID/ED) paradigms in rats. Complementary in vitro, ex vivo and in vivo studies were performed to confirm negative modulatory activity of RO4938581 and to investigate animal model validity, concept validity and potential side effect issues, respectively. RESULTS: In vitro studies confirmed the reported negative modulatory activity of RO4938581, whilst immunohistochemical analyses revealed significantly reduced parvalbumin-positive cells in the prefrontal cortex of sub-PCP- and neo-PCP-treated rats. RO4938581 (1 mg/kg) ameliorated both sub-PCP- and neo-PCP-induced cognitive deficits in NOR and ID/ED performance, respectively. In contrast, QH-II-066 (1 and 3 mg/kg), a GABA(A) α5 receptor positive modulator, impaired cognitive performance in the NOR task when administered to vehicle-treated animals. Additional studies revealed that both RO4938581 (1 mg/kg) and QH-II-066 (1 and 3 mg/kg) attenuated amphetamine-induced hyperactivity in rats. CONCLUSIONS: Taken together, these novel findings suggest that negative modulation of GABA(A) α5 receptors may represent an attractive treatment option for the cognitive impairments, and potentially positive symptoms, associated with schizophrenia.


Subject(s)
Benzodiazepines/pharmacology , Cognition Disorders/drug therapy , Imidazoles/pharmacology , Phencyclidine/toxicity , Receptors, GABA-A/drug effects , Amphetamine/pharmacology , Animals , CHO Cells , Central Nervous System Stimulants/pharmacology , Cognition Disorders/chemically induced , Cricetinae , Cricetulus , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Hyperkinesis/chemically induced , Male , Oocytes , Parvalbumins/metabolism , Phencyclidine/administration & dosage , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Rats , Rats, Wistar , Schizophrenia/drug therapy , Schizophrenia/physiopathology , Xenopus laevis
10.
J Psychopharmacol ; 25(10): 1347-56, 2011 Oct.
Article in English | MEDLINE | ID: mdl-20360159

ABSTRACT

Nicotine increases serotonergic and noradrenergic neuronal activity and facilitates serotonin and noradrenaline release. Accordingly, nicotine enhances antidepressant-like actions of reuptake inhibitors selective for serotonin or noradrenaline in the mouse forced swim test and the mouse tail suspension test. Both high-affinity α4ß2 and low-affinity α7 nicotinic acetylcholine receptor subtypes are implicated in nicotine-mediated release of serotonin and noradrenaline. The present study therefore investigated whether selective agonism of α4ß2 or α7 nicotinic acetylcholine receptors would affect the mouse forced swim test activity of two antidepressants with distinct mechanisms of action, namely the selective serotonin reuptake inhibitor citalopram and the noradrenaline reuptake inhibitor reboxetine. Subthreshold and threshold doses of citalopram (3 and 10 mg/kg) or reboxetine (10 and 20 mg/kg) were tested alone and in combination with the novel α4ß2-selective partial nicotinic acetylcholine receptor agonist, NS3956 (0.3 and 1.0 mg/kg) or the α7-selective nicotinic acetylcholine receptor agonist, PNU-282987 (10 and 30 mg/kg). Alone, NS3956 and PNU-282987 were devoid of activity in the mouse forced swim test, but both 1.0 mg/kg NS3956 and 30 mg/kg PNU-282987 enhanced the effect of citalopram and also reboxetine. The data suggest that the activity of citalopram and reboxetine in the mouse forced swim test can be enhanced by agonists at either α4ß2 or α7 nicotinic acetylcholine receptors, suggesting that both nicotinic acetylcholine receptor subtypes may be involved in the nicotine-enhanced action of antidepressants.


Subject(s)
Antidepressive Agents/pharmacology , Azepines/pharmacology , Citalopram/pharmacology , Morpholines/pharmacology , Nicotinic Agonists/pharmacology , Pyridines/pharmacology , Receptors, Nicotinic/physiology , Animals , Benzamides/pharmacology , Bridged Bicyclo Compounds/pharmacology , Female , Male , Mice , Motor Activity/drug effects , Rats , Rats, Wistar , Reboxetine , Swimming , Xenopus laevis , alpha7 Nicotinic Acetylcholine Receptor
11.
Front Psychiatry ; 1: 146, 2010.
Article in English | MEDLINE | ID: mdl-21423454

ABSTRACT

Phencyclidine (PCP) induces a behavioral syndrome in rodents that bears remarkable similarities to some of the core symptoms observed in schizophrenic patients, among those cognitive deficits. The successful alleviation of cognitive impairments associated with schizophrenia (CIAS) has become a major focus of research efforts as they remain largely untreated. The aim of the present study was to investigate the effects of selected antipsychotic and cognition enhancing drugs, namely haloperidol, risperidone, donepezil, and modafinil in an animal model widely used in preclinical schizophrenia research. To this end, the novel object recognition (NOR) task was applied to rats abstinent following sub-chronic treatment with PCP. Rats were administered either PCP (5 mg/kg, i.p.) or vehicle twice a day for 7 days, followed by a 7-day washout period, before testing in NOR. Upon testing, vehicle-treated rats successfully discriminated between novel and familiar objects, an effect abolished in rats that had previously been exposed to PCP treatment. Acute treatment with modafinil (64 mg/kg, p.o.) ameliorated the PCP-induced deficit in novel object exploration, whereas haloperidol (0.1 mg/kg, s.c.), risperidone (0.2 mg/kg, i.p.), and donepezil (3 mg/kg, p.o.) were without significant effect. Given the negligible efficacy of haloperidol and risperidone, and the contradictory data with donepezil to treat CIAS in the clinic, together with the promising preliminary pro-cognitive effects of modafinil in certain subsets of schizophrenic patients, the sub-chronic PCP-NOR abstinence paradigm may represent an attractive option for the identification of potential novel treatments for CIAS.

12.
Behav Pharmacol ; 20(3): 286-95, 2009 May.
Article in English | MEDLINE | ID: mdl-19404193

ABSTRACT

Clinical and preclinical evidence suggest a role of nicotinic acetylcholine receptors in major depression. In humans, both nicotine and the nonselective nicotinic acetylcholine receptor antagonist mecamylamine ameliorate depressive symptoms. Similarly, both drugs produce antidepressant-like effects in rodents. In rats, the most consistent finding is antidepressant-like effects of nicotine, but not mecamylamine. Conversely, in mice, several studies show antidepressant-like effects of mecamylamine, whereas nicotine has shown modest or no effects. These contradictory results might be because of genetic differences. Here, we compared the effects of nicotine and mecamylamine in females and males of NMRI, C57BL/6J and BALB/c mice using the mouse forced swim (mFST) and tail suspension tests (mTST). In the mFST, mecamylamine, but not nicotine, increased swim distance in NMRI mice. In contrast, nicotine, but not mecamylamine, increased swim distance in C57BL/6J mice. Both drugs increased swim distance in BALB/c mice. Effects in the mFST were independent of sex. In the mTST, mecamylamine decreased immobility in NMRI mice only, independent of sex. Nicotine was devoid of effects in the mTST, except in female C57BL/6J mice, where it increased immobility. We hypothesize that nicotine and mecamylamine produce antidepressant-like effects through partially different mechanisms.


Subject(s)
Antidepressive Agents/pharmacology , Hindlimb Suspension , Mecamylamine/pharmacology , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Nicotinic Antagonists/pharmacology , Animals , Antidepressive Agents/therapeutic use , Behavior, Animal/drug effects , Disease Models, Animal , Female , Male , Mice , Mice, Inbred Strains , Motor Activity/drug effects , Sex Factors , Species Specificity , Stress, Psychological/drug therapy , Stress, Psychological/psychology
13.
Psychopharmacology (Berl) ; 205(3): 517-28, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19452140

ABSTRACT

INTRODUCTION: Smoking rates among depressed individuals is higher than among healthy subjects, and nicotine alleviates depressive symptoms. Nicotine increases serotonergic and noradrenergic neuronal activity and facilitates serotonin and noradrenaline release. In mice, acute nicotine administration enhances the activity of antidepressants in the mouse forced swim (mFST) and tail suspension tests. Here, we investigated if this action of nicotine is also reflected in a chronic treatment regimen. MATERIALS AND METHODS: After chronic treatment with nicotine in the drinking water, mice were challenged with nicotine, duloxetine, citalopram, and reboxetine in the mFST. Additionally, 8-OH-DPAT- and clonidine-induced hypothermia was tested in vehicle- and nicotine-pretreated mice, as a measure of 5-HT(1A) and alpha(2)-adrenoceptor function, respectively. Finally, the effects on the brain expression levels of high- and low-affinity nicotinic acetylcholine receptors (nAChRs) and the transporters for serotonin (SERT) and noradrenaline (NET) were assessed using [(3)H]epibatidine, [(3)H]alpha-bungarotoxin, [(3)H]citalopram, and [(3)H]nisoxetine binding, respectively. RESULTS: In the mFST, nicotine-pretreated mice did not show altered response to the nicotine challenge, but increased responses to all three antidepressants tested were observed when compared to mice that had been administered drinking water without nicotine. There was no change in hypothermic responses to 8-OH-DPAT or clonidine. [(3)H]epibatidine binding was significantly increased in all brain regions investigated; whereas, [(3)H]alpha-bungarotoxin, [(3)H]citalopram, and [(3)H]nisoxetine binding were not altered, indicating that chronic oral nicotine increases the expression and/or affinity of high-affinity nAChRs, but not low-affinity nAChRs, SERT, or NET. DISCUSSION: It is suggested that the increased sensitivity to antidepressants after chronic nicotine exposure involves increased high-affinity nAChR-mediated neurotransmission.


Subject(s)
Antidepressive Agents/pharmacology , Brain/drug effects , Bridged Bicyclo Compounds, Heterocyclic/metabolism , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Pyridines/metabolism , Stress, Psychological/metabolism , Animals , Behavior, Animal/drug effects , Binding Sites , Brain/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Female , In Vitro Techniques , Mice , Mice, Inbred Strains , Motor Activity/drug effects , Nicotine/administration & dosage , Nicotinic Agonists/administration & dosage , Norepinephrine/metabolism , Radioligand Assay , Receptors, Nicotinic/biosynthesis , Receptors, Nicotinic/metabolism , Serotonin/metabolism , Stress, Psychological/psychology
14.
Neuropharmacology ; 56(6-7): 1001-9, 2009.
Article in English | MEDLINE | ID: mdl-19233218

ABSTRACT

The core features of schizophrenia include deficits in cognitive processes, such as attention and working memory, subserved by the prefrontal cortex (PFC). These deficits are believed to involve deficient neurotransmission through NMDA receptors, particularly on parvalbumin-containing interneurons, and administration of the NMDA-antagonist phencyclidine (PCP) in rodents is a well validated model of such cognitive deficits. Here we show that repeated PCP treatment (10 mg/kg/day for 10 days) decreased the expression of parvalbumin and synaptophysin mRNA in the mouse PFC, which corresponds to changes seen in patients with schizophrenia. In addition, PCP increased the basal mRNA expression in the PFC of the activity-regulated cytoskeleton-associated protein (Arc), a molecule involved in synaptic plasticity. These molecular changes produced by PCP were accompanied by a behavioral impairment as determined in a modified Y-maze test. Polymorphisms in the alpha(7) nicotinic acetylcholine receptor (nAChR) gene have been linked to schizophrenia. Here we demonstrate that acute administration of the selective alpha(7) nAChR partial agonist SSR180711 dose-dependently reversed the behavioral impairment induced by PCP. Importantly, repeated co-administration of SSR180711 (3 mg/kg) with PCP prevented both the changes in parvalbumin, synaptophysin, and Arc mRNA expression in the PFC, and the behavioral impairment induced by PCP. These results are the first to demonstrate prevention of the deleterious effects induced by repeated PCP treatment. The behavioral and molecular effects of alpha(7) nAChR agonism in this model, particularly the prevention of a decline in parvalbumin mRNA expression, suggest an involvement of the alpha(7) nAChR not only in the symptomatic relief, but also the pathophysiology, of schizophrenia.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Maze Learning/drug effects , Nicotinic Agonists/pharmacology , Phencyclidine/toxicity , Prefrontal Cortex/drug effects , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, Nicotinic/physiology , Animals , Cognition Disorders/chemically induced , Cognition Disorders/metabolism , Cognition Disorders/psychology , Cytoskeletal Proteins/biosynthesis , Cytoskeletal Proteins/genetics , Drug Partial Agonism , Gene Expression/drug effects , Male , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Parvalbumins/biosynthesis , Parvalbumins/genetics , Prefrontal Cortex/metabolism , RNA, Messenger/biosynthesis , Receptors, GABA-A/biosynthesis , Receptors, GABA-A/genetics , Receptors, Metabotropic Glutamate/biosynthesis , Receptors, Metabotropic Glutamate/genetics , Receptors, N-Methyl-D-Aspartate/biosynthesis , Receptors, N-Methyl-D-Aspartate/genetics , Synaptophysin/biosynthesis , Synaptophysin/genetics , alpha7 Nicotinic Acetylcholine Receptor
15.
Behav Brain Res ; 198(2): 481-5, 2009 Mar 17.
Article in English | MEDLINE | ID: mdl-19162078

ABSTRACT

In an effort to investigate the potential antimanic-like activity of K(v)7 channel openers, we decided to test: (1) the subtype non-selective K(v)7 opener retigabine, (2) the K(v)7.4-K(v)7.5 (and K(v)7.5/3 heteromers) preferring channel opener BMS-204352 (Maxipost), and (3) the novel K(v)7.2/3 preferring channel opener ICA-27243, in the amphetamine (AMPH)+chlordiazepoxide (CDP)-induced hyperactivity paradigm in mice, a test often used to assess potential antimanic-like activity of novel compounds. Lithium and lamotrigine were included as positive controls. Pretreatment with lithium attenuated AMPH/CDP-induced hyperactivity, without affecting the activity of AMPH- or CDP-alone, and thus confirmed some predictive validity for the test paradigm. Pretreatment with lamotrigine significantly attenuated AMPH/CDP-induced effects, but also reduced motility when tested in the presence of CDP-alone. Pretreatment with retigabine or ICA-27243 attenuated AMPH/CDP-induced hyperactivity without affecting basal locomotor activity. In contrast, pretreatment with BMS-204352 failed to decrease AMPH/CDP-induced hyperactivity at lower doses (3 and 10 mg/kg). At higher doses BMS-204352 attenuated hyperactivity induced by the AMPH/CDP mix, but only at doses decreasing basal locomotor activity (30 and 60 mg/kg). None of the K(v)7 openers tested significantly affected AMPH-induced hyperactivity. In contrast, retigabine and ICA-27243 were shown to induce significant reductions in motility when administered in combination with CDP-alone. In conclusion, the results with lithium confirm some predictive validity for the test paradigm. However, our data highlight an important confounder for interpreting a role for K(v)7 channels in the alleviation of manic-like symptoms when employing the AMPH/CDP hyperactivity model in mice. It is imperative that relevant control studies (AMPH- and CDP-alone) be incorporated and reported routinely to enable thorough interpretation of data generated by means of this behavioural test.


Subject(s)
Anticonvulsants/therapeutic use , Antimanic Agents/therapeutic use , Bipolar Disorder/drug therapy , Central Nervous System Stimulants/toxicity , Hyperkinesis/drug therapy , KCNQ Potassium Channels/agonists , Amphetamine/toxicity , Animals , Anticonvulsants/administration & dosage , Antimanic Agents/administration & dosage , Benzamides/therapeutic use , Bipolar Disorder/chemically induced , Bipolar Disorder/psychology , Carbamates/therapeutic use , Central Nervous System Stimulants/administration & dosage , Chlordiazepoxide/toxicity , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Hyperkinesis/psychology , Indoles/therapeutic use , Injections, Intraperitoneal , KCNQ Potassium Channels/classification , Lamotrigine , Lithium Compounds/therapeutic use , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Phenylenediamines/therapeutic use , Pyridines/therapeutic use , Triazines/therapeutic use
16.
Eur J Pharmacol ; 602(1): 58-65, 2009 Jan 05.
Article in English | MEDLINE | ID: mdl-18848931

ABSTRACT

The alpha7 (alpha7) nicotinic acetylcholine receptor may represent a drug target for the treatment of disorders associated with working memory/attentional dysfunction. We investigated the effects of three distinct alpha7 nicotinic acetylcholine receptor agonists: 2-methyl-5-(6-phenyl-pyridazin-3-yl)-octahydro-pyrrolo[3,4-c]pyrrole (A-582941; 0.01-0.1 mg/kg), 4-bromophenyl 1,4-diazabicyclo(3.2.2) nonane-4-carboxylate (SSR180711; 0.3-3 mg/kg) and N-[(3R)-1-azabicyclo[2.2.2]oct-3-yl]-4-chlorobenzamide (PNU-282987; 1-10 mg/kg), on scopolamine-induced deficits in a modified Y-maze procedure. Mice were forced to choose one of two visually distinct arms, and were confined there for a 5 min exploration period before being allowed to explore both arms for a 2 min test session, immediately thereafter. The time spent in each arm, entries and total distance travelled were recorded using an automated system. Characterisation experiments showed that scopolamine-treated (1 mg/kg) mice spent less time exploring the unfamiliar arm, when compared with vehicle-treated animals. Combination experiments showed that all three alpha7 agonists ameliorated scopolamine-induced changes in unfamiliar arm exploration. In conclusion, the present data support the idea that alpha7 nicotinic acetylcholine receptors may represent an interesting target for the treatment of conditions associated with attentional/working memory dysfunction.


Subject(s)
Behavior, Animal/drug effects , Maze Learning/drug effects , Nicotinic Agonists/pharmacology , Receptors, Nicotinic/metabolism , Scopolamine/pharmacology , Animals , Dose-Response Relationship, Drug , Male , Memory Disorders/chemically induced , Memory Disorders/drug therapy , Mice , Mice, Inbred C57BL , Nicotinic Agonists/therapeutic use , alpha7 Nicotinic Acetylcholine Receptor
17.
Behav Brain Res ; 197(1): 150-6, 2009 Jan 30.
Article in English | MEDLINE | ID: mdl-18786574

ABSTRACT

Current literature suggests that nicotinic acetylcholine receptors (nAChRs) are involved in major depression. In rodents, antidepressant-like effects of both nicotine and the non-selective nAChR antagonist mecamylamine have been reported. Nicotine increases serotonergic and noradrenergic neuronal activity and facilitates serotonin and noradrenaline release. Thus, we hypothesise that nicotine may enhance the behavioural effects of serotonin (e.g., citalopram) and/or noradrenaline (e.g., reboxetine) reuptake inhibitors. Here, we tested if nicotine enhanced the activity of citalopram or reboxetine in the mouse forced swim test (mFST) and the mouse tail suspension test (mTST). The potential for mecamylamine to augment antidepressant drug action was also investigated. Sub-threshold and threshold doses of citalopram (3 and 10mg/kg) or reboxetine (3, 10 and 20mg/kg) were tested alone and in combination with nicotine (0.3 and 1.0mg/kg) and mecamylamine (1 and 3mg/kg). Locomotor activity experiments were performed to rule out non-specific stimulant effects. Nicotine (1.0mg/kg) enhanced the effect of 10mg/kg citalopram and 20mg/kg reboxetine in the mFST. Similarly, nicotine (1.0mg/kg) enhanced the effect of 3 and 10mg/kg citalopram and 3 and 10mg/kg reboxetine in the mTST. No concomitant locomotor stimulation was observed at the tested dose combinations. Mecamylamine was effective on its own in some tests, but did not augment the effects of citalopram or reboxetine at the doses tested. The data show that nicotine enhances the effects of both serotonin and noradrenaline reuptake inhibitors, possibly reflecting nicotine's facilitating effects on the release of these two neurotransmitters, and indicating that nicotine may enhance antidepressant action.


Subject(s)
Antidepressive Agents/pharmacology , Citalopram/pharmacology , Depressive Disorder/drug therapy , Morpholines/pharmacology , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Adrenergic Uptake Inhibitors/pharmacology , Analysis of Variance , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Interactions , Drug Therapy, Combination , Female , Mecamylamine/pharmacology , Mice , Nicotinic Antagonists/pharmacology , Norepinephrine/metabolism , Reboxetine , Receptors, Nicotinic/drug effects , Receptors, Nicotinic/metabolism , Serotonin/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology
18.
Behav Brain Res ; 198(1): 136-41, 2009 Mar 02.
Article in English | MEDLINE | ID: mdl-19038290

ABSTRACT

The chronic mild stress (CMS) protocol is widely used to evoke depressive-like behaviours in laboratory rats. The aim of the present study was to examine the effects of chronic stress on cognitive performance. About 70% of rats exposed to 7 weeks of chronic mild stress showed a gradual reduction in consumption of a sucrose solution, indicating an anhedonic-like state. The remaining rats did not reduce their sucrose intake, but appeared resilient to the stress-induced effects on sucrose intake. Cognitive profiling of the CMS rats revealed that chronic stress had a negative effect on performance in the spontaneous alternation test, possibly reflecting a deficit in working memory. This effect was independent of whether the stressed rats were anhedonic-like or stress-resilient as measured by their sucrose intake. CMS did not influence performance in passive avoidance and auditory cued fear conditioning, however, in rats displaying an anhedonic-like profile, CMS increased freezing behaviour in contextual fear conditioning.


Subject(s)
Cognition/physiology , Depression/physiopathology , Feeding Behavior/psychology , Memory/physiology , Stress, Physiological , Acoustic Stimulation , Analysis of Variance , Animals , Avoidance Learning/physiology , Body Weight/physiology , Conditioning, Classical/physiology , Cues , Disease Models, Animal , Exploratory Behavior/physiology , Fear/physiology , Fear/psychology , Feeding Behavior/physiology , Freezing Reaction, Cataleptic/physiology , Male , Movement/physiology , Rats , Rats, Wistar , Sucrose/administration & dosage
19.
Peptides ; 26(8): 1394-400, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16042979

ABSTRACT

Neuropeptide Y (NPY) is thought to be implicated in depressive disorders. The mouse forced swim test (FST) is an animal model widely used as a predictor of the efficacy of antidepressant drugs. The present study was undertaken to explore the possible contribution of endogenous serotonin (5-HT) systems in the behavioral effects elicited by NPY in this model. The selective serotonin re-uptake inhibitor (SSRI), fluoxetine, was also tested for comparison. 5-HT was depleted prior to testing by the administration of the tryptophan hydroxylase inhibitor p-chlorophenylalanine (PCPA; 300 mg/kg, i.p., each day for 3 days; control mice received saline-vehicle over the same period). On the fourth day, mice received NPY (3 nmol, I.C.V.), fluoxetine (16 mg/kg, i.p.) or saline injections before testing in the FST. Both NPY and fluoxetine significantly reduced immobility time in saline-treated control animals. Pre-treatment with PCPA significantly blocked the effects of fluoxetine in the FST, confirming the role of endogenous 5-HT. Similarly, pre-treatment with PCPA also significantly attenuated the anti-immobility effects of NPY, thus suggesting a role for 5-HT in the effects of NPY in the FST. Quantitative receptor autoradiography revealed increases in specific [125I][Leu31, Pro34]PYY sites that were sensitive to BIBP3226 (Y1-like sites) in various brain regions. Specific [125I]GR231118 and [125I]PYY(3-36) binding levels were not changed following PCPA treatment, suggesting that depletion of endogenous 5-HT resulted in an apparent increase in the level of Y1 sites in their high-affinity state. Taken together, these results suggest a role for 5-HT-related systems in the antidepressant-like properties of NPY.


Subject(s)
Antidepressive Agents/administration & dosage , Disease Models, Animal , Fenclonine/administration & dosage , Motor Activity/drug effects , Neuropeptide Y/antagonists & inhibitors , Serotonin/physiology , Animals , Fluoxetine/administration & dosage , Fluoxetine/antagonists & inhibitors , Male , Mice , Motor Activity/physiology , Neuropeptide Y/administration & dosage , Neuropeptide Y/physiology , Selective Serotonin Reuptake Inhibitors/administration & dosage , Swimming/physiology
20.
Behav Brain Res ; 141(2): 251-5, 2003 May 15.
Article in English | MEDLINE | ID: mdl-12742262

ABSTRACT

The behavioural phenotype of mice lacking neuropeptide Y (NPY) Y(2)-type receptors was assessed in two well documented animal models of anxiety: namely, the elevated plus maze and the open field. NPY Y(2)-/- mice made more entries into, and spent significantly more time on, the open arms of the elevated plus maze when compared to their wild-type Y(2)+/+ controls (P<0.001). This effect was not due to non-specific changes in locomotor activity as the number of closed arm entries did not differ between groups. In addition, NPY Y(2)-/- mice displayed increased preference for the central area of the open field when compared to Y(2)+/+ animals (P<0.01), whereas total entries did not differ between groups. This study suggests that NPY Y(2) receptors may play an inhibitory role and supports the hypothesis that Y(2) receptors are involved in the regulation of anxiety-like behaviours by NPY.


Subject(s)
Anxiety/psychology , Behavior, Animal/physiology , Receptors, Neuropeptide Y/physiology , Animals , Male , Maze Learning/physiology , Mice , Mice, Knockout , Motor Activity/genetics , Motor Activity/physiology , Phenotype , Receptors, Neuropeptide Y/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...