Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
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
3.
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
4.
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
5.
Neuropsychopharmacology ; 32(1): 1-16, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17019409

ABSTRACT

In this paper, we report on the pharmacological and functional profile of SSR180711 (1,4-Diazabicyclo[3.2.2]nonane-4-carboxylic acid, 4-bromophenyl ester), a new selective alpha7 acetylcholine nicotinic receptor (n-AChRs) partial agonist. SSR180711 displays high affinity for rat and human alpha7 n-AChRs (K(i) of 22+/-4 and 14+/-1 nM, respectively). Ex vivo (3)[H]alpha-bungarotoxin binding experiments demonstrate that SSR180711 rapidly penetrates into the brain (ID(50)=8 mg/kg p.o.). In functional studies performed with human alpha7 n-AChRs expressed in Xenopus oocytes or GH4C1 cells, the compound shows partial agonist effects (intrinsic activity=51 and 36%, EC(50)=4.4 and 0.9 microM, respectively). In rat cultured hippocampal neurons, SSR180711 induced large GABA-mediated inhibitory postsynaptic currents and small alpha-bungarotoxin sensitive currents through the activation of presynaptic and somato-dendritic alpha7 n-AChRs, respectively. In mouse hippocampal slices, the compound increased the amplitude of both glutamatergic (EPSCs) and GABAergic (IPSCs) postsynaptic currents evoked in CA1 pyramidal cells. In rat and mouse hippocampal slices, a concentration of 0.3 muM of SSR180711 increased long-term potentiation (LTP) in the CA1 field. Null mutation of the alpha7 n-AChR gene totally abolished SSR180711-induced modulation of EPSCs, IPSCs and LTP in mice. Intravenous administration of SSR180711 strongly increased the firing rate of single ventral pallidum neurons, extracellularly recorded in anesthetized rats. In microdialysis experiments, administration of the compound (3-10 mg/kg i.p.) dose-dependently increased extracellular acetylcholine (ACh) levels in the hippocampus and prefrontal cortex of freely moving rats. Together, these results demonstrate that SSR180711 is a selective and partial agonist at human, rat and mouse alpha7 n-AChRs, increasing glutamatergic neurotransmission, ACh release and LTP in the hippocampus.


Subject(s)
Nicotinic Agonists/pharmacology , Nicotinic Agonists/pharmacokinetics , Receptors, Nicotinic/physiology , Animals , Animals, Newborn , Binding Sites/drug effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cells, Cultured , Dose-Response Relationship, Drug , Drug Interactions , Gene Expression/drug effects , Gene Expression/physiology , Hippocampus/cytology , Humans , In Vitro Techniques , Membrane Potentials/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/drug effects , Neurons/physiology , Nicotinic Agonists/chemistry , Nicotinic Antagonists/pharmacology , Oocytes/physiology , Patch-Clamp Techniques/methods , Protein Subunits/drug effects , Protein Subunits/physiology , Rats , Rats, Sprague-Dawley , Receptors, Nicotinic/deficiency , Synaptic Transmission/drug effects , Synaptic Transmission/physiology , alpha7 Nicotinic Acetylcholine Receptor , gamma-Aminobutyric Acid/pharmacology
6.
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
7.
J Pharmacol Exp Ther ; 302(2): 731-41, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12130738

ABSTRACT

SL65.0155 [5-(8-amino-7-chloro-2,3-dihydro-1,4-benzodioxin-5-yl)-3-[1-(2-phenyl ethyl)-4-piperidinyl]-1,3,4-oxadiazol-2(3H)-one monohydrochloride] is a novel benzodioxanoxadiazolone compound with high affinity for human 5-hydroxytryptamine (5-HT)(4) receptors (K(i) of 0.6 nM) and good selectivity (greater than 100-fold for all other receptors tested). In cells expressing the 5-HT(4(b)) and 5-HT(4(e)) splice variants, SL65.0155 acted as a partial agonist, stimulating cAMP production with a maximal effect of 40 to 50% of serotonin. However, in the rat esophagus preparation, SL65.0155 acted as a 5-HT(4) antagonist with a pK(b) of 8.81. In addition, SL65.0155 potently improved performance in several tests of learning and memory. In the object recognition task, it improved retention at 24 h when administered i.p. or p.o. (0.001-0.1 mg/kg). This effect was antagonized by the 5-HT(4) antagonist SDZ 205,557, itself without effect, demonstrating that the promnesic effects of SL65.0155 are mediated by 5-HT(4) agonism. SL65.0155 also reversed the cognitive deficits of aged rats in the linear maze task and the scopolamine-induced deficit of mice in the water maze task. Furthermore, the combined administration of an inactive dose of SL65.0155 with the cholinesterase inhibitor rivastigmine resulted in a significant promnesic effect, suggesting a synergistic interaction. SL65.0155 was devoid of unwanted cardiovascular, gastrointestinal, or central nervous system effects with doses up to more than 100-fold higher than those active in the cognitive tests. These results characterize SL65.0155 as a novel promnesic agent acting via 5-HT(4) receptors, with an excellent preclinical profile. Its broad range of activity in cognitive tests and synergism with cholinesterase inhibitors suggest that SL65.0155 represents a promising new agent for the treatment of dementia.


Subject(s)
Cognition/physiology , Cyclic AMP/metabolism , Dioxanes/pharmacology , Maze Learning/physiology , Oxadiazoles/pharmacology , Receptors, Serotonin/physiology , Serotonin Receptor Agonists/pharmacology , Alternative Splicing , Animals , Blood Pressure/drug effects , CHO Cells , COS Cells , Chlorocebus aethiops , Cognition/drug effects , Cricetinae , Esophagus/drug effects , Esophagus/physiology , Gastrointestinal Motility/drug effects , Guinea Pigs , Heart Rate/drug effects , Ileum/drug effects , Ileum/physiology , Maze Learning/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/physiology , Radioligand Assay , Rats , Receptors, Serotonin/drug effects , Receptors, Serotonin/genetics , Receptors, Serotonin, 5-HT4 , Recombinant Proteins/drug effects , Recombinant Proteins/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...