Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Sci Rep ; 9(1): 18045, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31792284

ABSTRACT

Glycogen synthase kinase 3 (GSK3) has been identified as a promising target for the treatment of Alzheimer's disease (AD), where abnormal activation of this enzyme has been associated with hyperphosphorylation of tau proteins. This study describes the effects of the selective GSK3 inhibitor, SAR502250, in models of neuroprotection and neuropsychiatric symptoms (NPS) associated with AD. In P301L human tau transgenic mice, SAR502250 attenuated tau hyperphosphorylation in the cortex and spinal cord. SAR502250 prevented the increase in neuronal cell death in rat embryonic hippocampal neurons following application of the neurotoxic peptide, Aß25-35. In behavioral studies, SAR502250 improved the cognitive deficit in aged transgenic APP(SW)/Tau(VLW) mice or in adult mice after infusion of Aß25-35. It attenuated aggression in the mouse defense test battery and improved depressive-like state of mice in the chronic mild stress procedure after 4 weeks of treatment. Moreover, SAR502250 decreased hyperactivity produced by psychostimulants. In contrast, the drug failed to modify anxiety-related behaviors or sensorimotor gating deficit. This profile confirms the neuroprotective effects of GSK3 inhibitors and suggests an additional potential in the treatment of some NPS associated with AD.


Subject(s)
Aggression/drug effects , Alzheimer Disease/drug therapy , Cognitive Dysfunction/drug therapy , Glycogen Synthase Kinase 3/antagonists & inhibitors , Neuroprotective Agents/pharmacology , Alzheimer Disease/complications , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Apoptosis/drug effects , Behavior, Animal/drug effects , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Cognitive Dysfunction/etiology , Cognitive Dysfunction/pathology , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Humans , Male , Mice , Mice, Transgenic , Neurons/drug effects , Neurons/pathology , Neuroprotective Agents/therapeutic use , Phosphorylation/drug effects , Primary Cell Culture , Rats , Spinal Cord/drug effects , Spinal Cord/pathology , tau Proteins/genetics , tau Proteins/metabolism
2.
Sci Rep ; 8(1): 2416, 2018 02 05.
Article in English | MEDLINE | ID: mdl-29403000

ABSTRACT

Enhancing endogenous cannabinoid (eCB) signaling has been considered as a potential strategy for the treatment of stress-related conditions. Fatty acid amide hydrolase (FAAH) represents the primary degradation enzyme of the eCB anandamide (AEA), oleoylethanolamide (OEA) and palmitoylethanolamide (PEA). This study describes a potent reversible FAAH inhibitor, SSR411298. The drug acts as a selective inhibitor of FAAH, which potently increases hippocampal levels of AEA, OEA and PEA in mice. Despite elevating eCB levels, SSR411298 did not mimic the interoceptive state or produce the behavioral side-effects (memory deficit and motor impairment) evoked by direct-acting cannabinoids. When SSR411298 was tested in models of anxiety, it only exerted clear anxiolytic-like effects under highly aversive conditions following exposure to a traumatic event, such as in the mouse defense test battery and social defeat procedure. Results from experiments in models of depression showed that SSR411298 produced robust antidepressant-like activity in the rat forced-swimming test and in the mouse chronic mild stress model, restoring notably the development of inadequate coping responses to chronic stress. This preclinical profile positions SSR411298 as a promising drug candidate to treat diseases such as post-traumatic stress disorder, which involves the development of maladaptive behaviors.


Subject(s)
Amidohydrolases/antagonists & inhibitors , Anti-Anxiety Agents/pharmacology , Anxiety Disorders/drug therapy , Cannabinoid Receptor Agonists/metabolism , Carbamates/pharmacology , Dioxanes/pharmacology , Enzyme Inhibitors/pharmacology , Receptors, Cannabinoid/genetics , Stress, Psychological/drug therapy , Acute Disease , Amides , Amidohydrolases/genetics , Amidohydrolases/metabolism , Animals , Anti-Anxiety Agents/chemical synthesis , Anxiety Disorders/physiopathology , Arachidonic Acids/metabolism , Carbamates/chemical synthesis , Chronic Disease , Dioxanes/chemical synthesis , Endocannabinoids/metabolism , Enzyme Inhibitors/chemical synthesis , Ethanolamines/metabolism , Female , Gene Expression , Male , Mice , Oleic Acids/metabolism , Palmitic Acids/metabolism , Polyunsaturated Alkamides/metabolism , Rats, Sprague-Dawley , Receptors, Cannabinoid/metabolism , Stress, Psychological/physiopathology
3.
Sci Rep ; 6: 35320, 2016 10 13.
Article in English | MEDLINE | ID: mdl-27734956

ABSTRACT

Normalization of altered glutamate neurotransmission through activation of the mGluR2 has emerged as a new approach to treat schizophrenia. These studies describe a potent brain penetrant mGluR2 positive allosteric modulator (PAM), SAR218645. The compound behaves as a selective PAM of mGluR2 in recombinant and native receptor expression systems, increasing the affinity of glutamate at mGluR2 as inferred by competition and GTPγ35S binding assays. SAR218645 augmented the mGluR2-mediated response to glutamate in a rat recombinant mGluR2 forced-coupled Ca2+ mobilization assay. SAR218645 potentiated mGluR2 agonist-induced contralateral turning. When SAR218645 was tested in models of the positive symptoms of schizophrenia, it reduced head twitch behavior induced by DOI, but it failed to inhibit conditioned avoidance and hyperactivity using pharmacological and transgenic models. Results from experiments in models of the cognitive symptoms associated with schizophrenia showed that SAR218645 improved MK-801-induced episodic memory deficits in rats and attenuated working memory impairment in NMDA Nr1neo-/- mice. The drug reversed disrupted latent inhibition and auditory-evoked potential in mice and rats, respectively, two endophenotypes of schizophrenia. This profile positions SAR218645 as a promising candidate for the treatment of cognitive symptoms of patients with schizophrenia, in particular those with abnormal attention and sensory gating abilities.


Subject(s)
Attention/drug effects , Cognition Disorders/drug therapy , Cognition/drug effects , Indans/pharmacology , Memory/drug effects , Oxazoles/pharmacology , Pyrimidines/pharmacology , Receptors, AMPA/chemistry , Schizophrenia/drug therapy , Allosteric Site , Amphetamines/pharmacology , Animals , Calcium/metabolism , Cerebral Cortex/metabolism , Cyclic AMP/metabolism , Dizocilpine Maleate/chemistry , Dizocilpine Maleate/pharmacology , Electroconvulsive Therapy , HEK293 Cells , Humans , Indans/therapeutic use , Male , Maze Learning , Memory, Short-Term/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxazoles/therapeutic use , Phenotype , Pyrimidines/therapeutic use , Rats , Rats, Sprague-Dawley
4.
Sci Rep ; 5: 7642, 2015 Jan 06.
Article in English | MEDLINE | ID: mdl-25560837

ABSTRACT

Monoacylglycerol lipase (MAGL) represents a primary degradation enzyme of the endogenous cannabinoid (eCB), 2-arachidonoyglycerol (2-AG). This study reports a potent covalent MAGL inhibitor, SAR127303. The compound behaves as a selective and competitive inhibitor of mouse and human MAGL, which potently elevates hippocampal levels of 2-AG in mice. In vivo, SAR127303 produces antinociceptive effects in assays of inflammatory and visceral pain. In addition, the drug alters learning performance in several assays related to episodic, working and spatial memory. Moreover, long term potentiation (LTP) of CA1 synaptic transmission and acetylcholine release in the hippocampus, two hallmarks of memory function, are both decreased by SAR127303. Although inactive in acute seizure tests, repeated administration of SAR127303 delays the acquisition and decreases kindled seizures in mice, indicating that the drug slows down epileptogenesis, a finding deserving further investigation to evaluate the potential of MAGL inhibitors as antiepileptics. However, the observation that 2-AG hydrolysis blockade alters learning and memory performance, suggests that such drugs may have limited value as therapeutic agents.


Subject(s)
Analgesics/pharmacology , Arachidonic Acids/metabolism , Carbamates/pharmacology , Endocannabinoids/metabolism , Glycerides/metabolism , Learning/drug effects , Memory, Short-Term/drug effects , Monoacylglycerol Lipases/metabolism , Sulfonamides/pharmacology , Acetylcholine/metabolism , Administration, Oral , Analgesics/chemistry , Analgesics/therapeutic use , Animals , Arachidonic Acids/chemistry , Binding Sites , Brain/metabolism , Cannabinoid Receptor Antagonists/pharmacology , Carbamates/chemistry , Carbamates/therapeutic use , Chromatography, High Pressure Liquid , Crystallography, X-Ray , Disease Models, Animal , Electric Stimulation , Endocannabinoids/chemistry , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Glycerides/chemistry , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Hydrolysis , In Vitro Techniques , Long-Term Potentiation/drug effects , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mice, SCID , Monoacylglycerol Lipases/antagonists & inhibitors , Pain/drug therapy , Pain/pathology , Piperidines/pharmacology , Protein Structure, Tertiary , Pyrazoles/pharmacology , Rimonabant , Seizures/drug therapy , Seizures/pathology , Sulfonamides/chemistry , Sulfonamides/therapeutic use
5.
Article in English | MEDLINE | ID: mdl-24782829

ABSTRACT

Disruption of circadian clock enhances the risk of metabolic syndrome, obesity, and type 2 diabetes. Circadian clocks rely on a highly regulated network of transcriptional and translational loops that drive clock-controlled gene expression. Among these transcribed clock genes are cryptochrome (CRY) family members, which comprise Cry1 and Cry2. While the metabolic effects of deletion of several core components of the clock gene machinery have been well characterized, those of selective inactivation of Cry1 or Cry2 genes have not been described. In this study, we demonstrate that ablation of Cry1, but not Cry2, prevents high-fat diet (HFD)-induced obesity in mice. Despite similar caloric intake, Cry1 (-/-) mice on HFD gained markedly less weight (-18%) at the end of the 16-week experiment and displayed reduced fat accumulation compared to wild-type (WT) littermates (-61%), suggesting increased energy expenditure. Analysis of serum lipid and glucose profiles showed no difference between Cry1 (-/-) and WT mice. Both Cry1 (-/-) and Cry2 (-/-) mice are indistinguishable from WT controls in body weight, fat and protein contents, and food consumption when they are allowed unlimited access to a standard rodent diet. We conclude that although CRY signaling may not be essential for the maintenance of energy homeostasis under steady-state nutritional conditions, Cry1 may play a role in readjusting energy balance under changing nutritional circumstances. These studies reinforce the important role of circadian clock genes in energy homeostasis and suggest that Cry1 is a plausible target for anti-obesity therapy.

6.
Bioorg Med Chem Lett ; 23(23): 6269-73, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24139584

ABSTRACT

Lead optimization guided by histamine H3 receptor (H3R) affinity and calculated physico-chemical properties enabled simultaneous improvement in potency and PK properties leading to the identification of a potent, selective, devoid of hERG issues, orally bioavailable, and CNS penetrable H3R antagonist/inverse agonist 3h. The compound was active in forced-swimming tests suggesting its potential therapeutic utility as an anti-depressive agent. This Letter further includes its cardiovascular and neuropsychological/behavioral safety assessments.


Subject(s)
Amides/chemistry , Antidepressive Agents/chemistry , Histamine Antagonists/chemistry , Pyrrolidines/chemistry , Amides/pharmacokinetics , Amides/pharmacology , Animals , Antidepressive Agents/pharmacokinetics , Antidepressive Agents/pharmacology , CHO Cells , Cricetulus , Histamine Antagonists/pharmacokinetics , Histamine Antagonists/pharmacology , Humans , Kinetics , Male , Mice , Pyrrolidines/pharmacokinetics , Pyrrolidines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Histamine H3/chemistry , Receptors, Histamine H3/metabolism
7.
Bioorg Med Chem Lett ; 23(22): 6141-5, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24076172

ABSTRACT

Previous studies have shown that compound 1 displayed high affinity towards histamine H3 receptor (H3R), (human (h-H3R), K(i)=8.6 nM, rhesus monkey (rh-H3R), K(i)=1.2 nM, and rat (r-H3R), K(i)=16.5 nM), but exhibited high affinity for hERG channel. Herein, we report the discovery of a novel, potent, and highly selective H3R antagonist/inverse agonist 5a(SS) (SAR110068) with acceptable hERG channel selectivity and desirable pharmacological and pharmacokinetic properties through lead optimization sequence. The significant awakening effects of 5a(SS) on sleep-wake cycles studied by using EEG recording in rats during their light phase support its potential therapeutic utility in human sleep-wake disorders.


Subject(s)
Histamine H3 Antagonists/pharmacology , Pyrans/pharmacology , Pyrrolidines/pharmacology , Sleep Disorders, Circadian Rhythm/drug therapy , Administration, Oral , Animals , Dogs , Drug Stability , Guinea Pigs , Haplorhini , Histamine H3 Antagonists/chemistry , Histamine H3 Antagonists/pharmacokinetics , Humans , Male , Mice , Pyrans/chemistry , Pyrans/pharmacokinetics , Pyrrolidines/chemistry , Pyrrolidines/pharmacokinetics , Rabbits , Rats , Rats, Sprague-Dawley , Receptors, Histamine H3/chemistry , Receptors, Histamine H3/metabolism , Sheep , Sleep Disorders, Circadian Rhythm/metabolism , Substrate Specificity
8.
Pharmacol Biochem Behav ; 102(2): 203-14, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22542742

ABSTRACT

SAR110894 is a novel histamine H3-R ligand, displaying high and selective affinity for human, rat or mouse H3-Rs. SAR110894 is a potent H3-R antagonist at native receptors, reversing R-α-methylhistamine-induced inhibition of electrical field stimulation contraction in the guinea-pig ileum. Additionally, SAR110894 inhibited constitutive GTPγS binding at human H3-Rs demonstrating inverse agonist properties. In behavioral models addressing certain aspects of cognitive impairment associated with schizophrenia (CIAS) and attention deficit/hyperactivity disorder (ADHD), SAR110894 improved memory performances in several variants of the object recognition task in mice (0.3-3 mg/kg, p.o.) or rats (0.3-1 mg/kg, p.o.). Moreover, SAR110894 (1 mg/kg, p.o.) reversed a deficit in working memory in the Y-maze test, following an acute low dose of phencyclidine (PCP) (0.5 mg/kg, i.p.) in mice sensitized by repeated treatment with a high dose of PCP (10 mg/kg, i.p.). In the latent inhibition (LI) model, SAR110894 potentiated LI in saline-treated rats (1 and 3 mg/kg, i.p.) and reversed abnormally persistent LI induced by neonatal nitric oxide synthase (NOS) inhibition in rodents (0.3-3 mg/kg, i.p.). In a social novelty discrimination task in rats, SAR110894 attenuated selective attention deficit induced by neonatal PCP treatment (3 and 10 mg/kg, p.o.) or a parametric modification of the procedure (3 and 10 mg/kg, p.o.). SAR110894 showed efficacy in several animal models related to the cognitive deficits in Alzheimer's disease (AD). It prevented the occurrence of episodic memory deficit induced by scopolamine in rats (0.01-10 mg/kg, p.o.) or by the central infusion of the toxic amyloid fragment ß25₋35 in the object recognition test in mice (1 and 3 mg/kg, p.o.). Altogether, these findings suggest that SAR110894 may be of therapeutic interest for the treatment of the cognitive symptoms of AD, schizophrenia and certain aspects of ADHD.


Subject(s)
Cognition/drug effects , Histamine H3 Antagonists/pharmacology , Animals , Female , Histamine H3 Antagonists/therapeutic use , Maze Learning , Mice , Rats , Rats, Sprague-Dawley , Rats, Wistar , Schizophrenia/drug therapy
9.
Eur J Med Chem ; 46(9): 4035-41, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21689868

ABSTRACT

Synthesis and biological evaluation of new derivatives of Morphine-6-Glucuronide (M6G) are described. M6G is an active metabolite of morphine which displays more analgesia than morphine with a superior side effect profile but with a less efficiently BBB penetration. These phenomena could be explained by the presence of the glucuronide moiety, which confers a higher hydrophilic character compare to morphine. In this context, we have prepared three analogues of M6G possessing a tetrazole, an oxadiazole, and a triazolopyrimidine moiety instead of the carboxylic acid function on position 5 of the sugar. These three analogues showed higher analgesic properties than morphine and M6G even by oral administration.


Subject(s)
Analgesics, Opioid/chemical synthesis , Analgesics, Opioid/pharmacology , Morphine Derivatives/chemical synthesis , Morphine Derivatives/pharmacology , Analgesics, Opioid/pharmacokinetics , Animals , Blood-Brain Barrier , Magnetic Resonance Spectroscopy , Male , Mice , Morphine Derivatives/pharmacokinetics , Spectrometry, Mass, Electrospray Ionization
10.
Psychopharmacology (Berl) ; 215(1): 149-63, 2011 May.
Article in English | MEDLINE | ID: mdl-21181124

ABSTRACT

RATIONALE: The psychotomimetic effects of cannabis are believed to be mediated via cannabinoid CB1 receptors. Furthermore, studies have implicated CB1 receptors in the pathophysiology of schizophrenia. OBJECTIVE: These studies investigated the effects of the CB1 receptor antagonist, AVE1625, in acute pharmacological and neurodevelopmental models of schizophrenia. AVE1625 was administered to rodents alone or as a co-treatment with clinically used antipsychotic drugs (APDs). METHODS: The antipsychotic potential of AVE1625 was tested using psychotomimetic-induced hyperactivity and latent inhibition (LI) deficit models. The procognitive profile was assessed using hole board, novel object recognition, auditory evoked potential, and LI techniques. In addition, the side-effect profile was established by measuring catalepsy, antipsychotic-induced weight gain, plasma levels of prolactin, and anxiogenic potential. RESULTS: AVE1625 (1, 3, and 10 mg/kg ip), reversed abnormally persistent LI induced by MK-801 or neonatal nitric oxide synthase inhibition in rodents, and improved both working and episodic memory. AVE1625 was not active in positive symptom models but importantly, it did not diminish the efficacy of APDs. It also decreased catalepsy and weight gain induced by APDs, suggesting that it may decrease APD-induced extrapyramidal side effects (EPS) and compliance. Unlike other CB1 antagonists, AVE1625 did not produce anxiogenic-like effects. CONCLUSIONS: These preclinical data suggest that AVE1625 may be useful to treat the cognitive deficits in schizophrenia and as a co-treatment with currently available antipsychotics. In addition, an improved side-effect profile was seen, with potential to ameliorate the EPS and weight gain issues with currently available treatments.


Subject(s)
Antipsychotic Agents/adverse effects , Antipsychotic Agents/therapeutic use , Cognition/drug effects , Hydrocarbons, Halogenated/therapeutic use , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Schizophrenia/drug therapy , Sulfonamides/therapeutic use , Acoustic Stimulation , Amphetamine/pharmacology , Animals , Antipsychotic Agents/administration & dosage , Anxiety/chemically induced , Anxiety/prevention & control , Behavior, Animal/drug effects , Catalepsy/chemically induced , Catalepsy/prevention & control , Conditioning, Classical/drug effects , Disease Models, Animal , Drug Evaluation, Preclinical , Drug Therapy, Combination , Evoked Potentials, Auditory/drug effects , Hydrocarbons, Halogenated/administration & dosage , Hydrocarbons, Halogenated/adverse effects , Male , Mice , Mice, Inbred Strains , Rats , Rats, Sprague-Dawley , Schizophrenia/metabolism , Schizophrenia/physiopathology , Sulfonamides/administration & dosage , Sulfonamides/adverse effects , Weight Gain/drug effects
11.
Pharmacol Biochem Behav ; 98(3): 405-11, 2011 May.
Article in English | MEDLINE | ID: mdl-21115031

ABSTRACT

The current work extends our previous findings in stress-related disorders, but also addresses the impact of a neurokinin-2 (NK2) antagonist on cognition. Besides efficacy in mood disorders, an NK2 antagonist may have the potential to lack the disinhibitory components and adverse side effects associated with existing clinical treatments. Saredutant (3-30 mg/kg, per os, p.o.) was tested for anxiolytic-like potential in three mouse models: holeboard, stress-induced hyperthermia (SIH) and four-plate. In the holeboard model saredutant (30 mg/kg) showed a trend to increase head dipping without affecting general activity. In the SIH model, saredutant demonstrated a significant reduction in stress-induced temperature at 30 mg/kg, while the number of punished crossings in the four-plate was increased at all doses tested (3-30 mg/kg). While chlordiazepoxide (CDP) demonstrated anxiolytic-like effects in these models, the adverse side effects of benzodiazepines, such as sedation, disinhibition and cognitive deficits are well-documented. Saredutant produced no detrimental effect in three models of cognition: Morris Water Maze (MWM) in rats, spontaneous alternation in a Y-maze in mice and novel objection recognition in mice. In contrast, the benzodiazepine, diazepam (DZM), produced cognitive impairments. NK2 receptor antagonists like saredutant may therefore yield beneficial effects for mood disorders without the adverse effects of current treatments.


Subject(s)
Antidepressive Agents/pharmacology , Benzamides/pharmacology , Cognition/drug effects , Piperidines/pharmacology , Receptors, Neurokinin-2/antagonists & inhibitors , Stress, Physiological/drug effects , Animals , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Wistar
12.
Psychopharmacology (Berl) ; 202(1-3): 385-96, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18709358

ABSTRACT

RATIONALE: SSR103800 and SSR504734 are novel glycine transport 1 (GlyT1) inhibitors with therapeutic potential for the treatment of schizophrenia. OBJECTIVE: The present studies investigated the effects of GlyT1 inhibitors in acute pharmacological and neurodevelopmental models of schizophrenia using latent inhibition in the rat; these latent inhibition (LI) models are believed to be predictive for treatments of positive, negative, and cognitive aspects of schizophrenia. MATERIALS AND METHODS: LI, the poorer conditioning to a previously irrelevant stimulus, was measured in a conditioned emotional response procedure in male rats. The effects of SSR103800 or SSR504734 (both at 1, 3, and 10 mg/kg, i.p.) were determined on amphetamine-induced disrupted LI, MK-801-induced abnormally persistent LI, and neurodevelopmentally induced abnormally persistent LI in adult animals that had been neonatally treated with a nitric oxide synthase inhibitor. RESULTS: SSR103800 (1 and 3 mg/kg) and SSR504734 (1 and 10 mg/kg) potentiated LI under conditions where LI was not present in nontreated controls and SSR103800 (1 mg/kg) reversed amphetamine-induced disrupted LI while not affecting LI on its own. Additionally, SSR103800 (1 and 3 mg/kg) and SSR504734 (3 and 10 mg/kg) reversed abnormally persistent LI induced by MK-801. In the neurodevelopmental model, SSR504734 (3 and 10 mg/kg) reverted the LI back to control (normal) levels. CONCLUSIONS: These preclinical data, from acute and neurodevelopmental models, suggest that GlyT1 inhibition may exhibit activity in the positive, negative, and cognitive symptom domains of schizophrenia.


Subject(s)
Aging/psychology , Antipsychotic Agents/pharmacology , Cognition/drug effects , Glycine Plasma Membrane Transport Proteins/antagonists & inhibitors , Reflex, Startle/drug effects , Schizophrenia/drug therapy , Schizophrenic Psychology , Amphetamine/antagonists & inhibitors , Amphetamine/toxicity , Animals , Animals, Newborn , Benzamides/pharmacology , Central Nervous System Stimulants/toxicity , Conditioning, Operant/drug effects , Dizocilpine Maleate/antagonists & inhibitors , Dizocilpine Maleate/toxicity , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/toxicity , Haloperidol/pharmacology , Male , Nitroarginine/pharmacology , Piperidines/pharmacology , Rats , Rats, Wistar
13.
Pharmacol Biochem Behav ; 91(1): 47-58, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18621075

ABSTRACT

On native human, rat and mouse glycine transporter-1(GlyT1), SSR130800 behaves as a selective inhibitor with IC50 values of 1.9, 5.3 and 6.8 nM, respectively. It reversibly blocked glycine uptake in mouse brain cortical homogenates, increased extracellular levels of glycine in the rat prefrontal cortex, and potentiated NMDA-mediated excitatory postsynaptic currents in rat hippocampal slices. SSR103800 (30 mg/kg, p.o.) decreased MK-801- and PCP-induced locomotor hyperactivity in rodents. SSR103800 (1 and 10 mg/kg, p.o.) attenuated social recognition deficit in adult rats induced by neonatal injections of PCP (10 mg/kg, s.c., on post-natal day 7, 9 and 11). SSR103800 (3 mg/kg, p.o.) counteracted the deficit in short-term visual episodic-like memory induced by a low challenge dose of PCP (1 mg/kg, i.p.), in PCP-sensitized rats (10 mg/kg, i.p.). SSR103800 (30 mg/kg, i.p.) increased the prepulse inhibition of the startle reflex in DBA/1J mice. SSR103800 decreased defensive- and despair-related behaviors in the tonic immobility test in gerbils (10 and 30 mg/kg, p.o.) and in the forced-swimming procedure in rats (1 and 3 mg/kg, p.o.), respectively. These findings suggest that SSR103800 may have a therapeutic potential in the management of the core symptoms of schizophrenia and comorbid depression states.


Subject(s)
Antipsychotic Agents/pharmacology , Glycine Plasma Membrane Transport Proteins/antagonists & inhibitors , Schizophrenia/drug therapy , Animals , Antidepressive Agents/pharmacology , Discrimination, Psychological/drug effects , Dizocilpine Maleate/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Extracellular Space/drug effects , Extracellular Space/metabolism , Gerbillinae , Glycine/metabolism , Male , Mice , Motor Activity/drug effects , N-Methylaspartate/physiology , Phencyclidine/pharmacology , Recognition, Psychology/drug effects , Reflex, Startle/drug effects , Stereoisomerism , Swimming/psychology
14.
Neuropsychopharmacology ; 33(3): 574-87, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17460614

ABSTRACT

The characterization of the first selective orally active and brain-penetrant beta3-adrenoceptor agonist, SR58611A (amibegron), has opened new possibilities for exploring the involvement of this receptor in stress-related disorders. By using a battery of tests measuring a wide range of anxiety-related behaviors in rodents, including the mouse defense test battery, the elevated plus-maze, social interaction, stress-induced hyperthermia, four-plate, and punished drinking tests, we demonstrated for the first time that the stimulation of the beta3 receptor by SR58611A resulted in robust anxiolytic-like effects, with minimal active doses ranging from 0.3 to 10 mg/kg p.o., depending on the procedure. These effects paralleled those obtained with the prototypical benzodiazepine anxiolytic diazepam or chlordiazepoxide. Moreover, when SR58611A was tested in acute or chronic models of depression in rodents, such as the forced-swimming and the chronic mild stress tests, it produced antidepressant-like effects, which were comparable in terms of the magnitude of the effects to those of the antidepressant fluoxetine or imipramine. Supporting these behavioral data, SR58611A modified spontaneous sleep parameters in a manner comparable to that observed with fluoxetine. Importantly, SR58611A was devoid of side effects related to cognition (as shown in the Morris water maze and object recognition tasks), motor activity (in the rotarod), alcohol interaction, or physical dependence. Antagonism studies using pharmacological tools targeting a variety of neurotransmitters involved in anxiety and depression and the use of mice lacking the beta3 adrenoceptor suggested that these effects of SR58611A are mediated by beta3 adrenoceptors. Taken as a whole, these findings indicate that the pharmacological stimulation of beta3 adrenoceptors may represent an innovative approach for the treatment of anxiety and depressive disorders.


Subject(s)
Adrenergic beta-3 Receptor Agonists , Adrenergic beta-Agonists/therapeutic use , Anxiety Disorders/drug therapy , Depressive Disorder/drug therapy , Tetrahydronaphthalenes/therapeutic use , Adrenergic beta-Agonists/administration & dosage , Aggression/drug effects , Animals , Anti-Anxiety Agents/pharmacology , Antidepressive Agents, Second-Generation/pharmacology , Antidepressive Agents, Tricyclic/pharmacology , Anxiety Disorders/psychology , Behavior, Animal/drug effects , Cognition/drug effects , Depressive Disorder/psychology , Diazepam/pharmacology , Ethanol/pharmacology , Exploratory Behavior/drug effects , Fluoxetine/pharmacology , Gerbillinae , Imipramine/pharmacology , Interpersonal Relations , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Rats, Wistar , Receptors, Adrenergic, beta-3/genetics , Sleep/drug effects , Substance-Related Disorders/psychology , Swimming/psychology , Tetrahydronaphthalenes/administration & dosage
15.
Neuropsychopharmacology ; 32(1): 17-34, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16936709

ABSTRACT

SSR180711 (4-bromophenyl 1,4diazabicyclo(3.2.2) nonane-4-carboxylate, monohydrochloride) is a selective alpha7 nicotinic receptor (n-AChR) partial agonist. Based on the purported implication of this receptor in cognitive deficits associated with schizophrenia, the present study assessed efficacy of SSR180711 (i.p. and p.o.) in different types of learning and memory involved in this pathology. SSR180711 enhanced episodic memory in the object recognition task in rats and mice (MED: 0.3 mg/kg), an effect mediated by the alpha7 n-AChR, as it was no longer seen in mice lacking this receptor. Efficacy was retained after repeated treatment (eight administrations over 5 days, 1 mg/kg), indicating lack of tachyphylaxia. SSR180711 also reversed (MED: 0.3 mg/kg) MK-801-induced deficits in retention of episodic memory in rats (object recognition). The drug reversed (MED: 0.3 mg/kg) selective attention impaired by neonatal phencyclidine (PCP) treatment and restored MK-801- or PCP-induced memory deficits in the Morris or linear maze (MED: 1-3 mg/kg). In neurochemical and electrophysiological correlates of antipsychotic drug action, SSR180711 increased extracellular levels of dopamine in the prefrontal cortex (MED: 1 mg/kg) and enhanced (3 mg/kg) spontaneous firing of retrosplenial cortex neurons in rats. Selectivity of SSR180711 was confirmed as these effects were abolished by methyllycaconitine (3 mg/kg, i.p. and 1 mg/kg, i.v., respectively), a selective alpha7 n-AChR antagonist. Additional antidepressant-like properties of SSR180711 were demonstrated in the forced-swimming test in rats (MED: 1 mg/kg), the maternal separation-induced ultrasonic vocalization paradigm in rat pups (MED: 3 mg/kg) and the chronic mild stress procedure in mice (10 mg/kg o.d. for 3 weeks). Taken together, these findings characterize SSR180711 as a promising new agent for the treatment of cognitive symptoms of schizophrenia. The antidepressant-like properties of SSR180711 are of added interest, considering the high prevalence of depressive symptoms in schizophrenic patients.


Subject(s)
Cognition Disorders/drug therapy , Nicotinic Agonists/therapeutic use , Receptors, Nicotinic/physiology , Analysis of Variance , Animals , Animals, Newborn , Behavior, Animal/drug effects , Cognition Disorders/etiology , Disease Models, Animal , Dizocilpine Maleate/pharmacology , Dopamine/metabolism , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Interactions , Excitatory Amino Acid Antagonists/pharmacology , Exploratory Behavior/drug effects , Female , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Phencyclidine/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Nicotinic/deficiency , Recognition, Psychology/drug effects , Schizophrenia/complications , alpha7 Nicotinic Acetylcholine Receptor
SELECTION OF CITATIONS
SEARCH DETAIL
...