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.
J Pharmacol Exp Ther ; 361(1): 68-86, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28138040

ABSTRACT

The ability of the N-methyl-d-aspartate receptor antagonist ketamine to alleviate symptoms in patients suffering from treatment-resistant depression (TRD) is well documented. In this paper, we directly compare in vivo biologic responses in rodents elicited by a recently discovered metabotropic glutamate (mGlu) 2/3 receptor antagonist 2-amino-3-[(3,4-difluorophenyl)sulfanylmethyl]-4-hydroxy-bicyclo[3.1.0]hexane-2,6-dicarboxylic acid (LY3020371) with those produced by ketamine. Both LY3020371 and ketamine increased the number of spontaneously active dopamine cells in the ventral tegmental area of anesthetized rats, increased O2 in the anterior cingulate cortex, promoted wakefulness, enhanced the efflux of biogenic amines in the prefrontal cortex, and produced antidepressant-related behavioral effects in rodent models. The ability of LY3020371 to produce antidepressant-like effects in the forced-swim assay in rats was associated with cerebrospinal fluid (CSF) drug levels that matched concentrations required for functional antagonist activity in native rat brain tissue preparations. Metabolomic pathway analyses from analytes recovered from rat CSF and hippocampus demonstrated that both LY3020371 and ketamine activated common pathways involving GRIA2 and ADORA1. A diester analog of LY3020371 [bis(((isopropoxycarbonyl)oxy)-methyl) (1S,2R,3S,4S,5R,6R)-2-amino-3-(((3,4-difluorophenyl)thio)methyl)-4-hydroxy-bicyclo[3.1.0]hexane-2,6-dicarboxylate (LY3027788)] was an effective oral prodrug; when given orally, it recapitulated effects of intravenous doses of LY3020371 in the forced-swim and wake-promotion assays, and augmented the antidepressant-like effects of fluoxetine or citalopram without altering plasma or brain levels of these compounds. The broad overlap of biologic responses produced by LY3020371 and ketamine supports the hypothesis that mGlu2/3 receptor blockade might be a novel therapeutic approach for the treatment of TRD patients. LY3020371 and LY3027788 represent molecules that are ready for clinical tests of this hypothesis.


Subject(s)
Antidepressive Agents/therapeutic use , Excitatory Amino Acid Antagonists/therapeutic use , Ketamine/therapeutic use , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Antidepressive Agents/pharmacology , Depression/drug therapy , Depression/metabolism , Depression/psychology , Excitatory Amino Acid Antagonists/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Ketamine/pharmacology , Male , Rats , Rats, Sprague-Dawley , Rats, Wistar , Receptors, Metabotropic Glutamate/metabolism , Receptors, Metabotropic Glutamate/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Treatment Outcome
2.
J Pharmacol Exp Ther ; 358(1): 71-82, 2016 07.
Article in English | MEDLINE | ID: mdl-27189960

ABSTRACT

Ketamine is a rapidly acting antidepressant in patients with treatment-resistant depression (TRD). Although the mechanisms underlying these effects are not fully established, inquiry to date has focused on the triggering of synaptogenesis transduction pathways via glutamatergic mechanisms. Preclinical data suggest that blockade of metabotropic glutamate (mGlu2/3) receptors shares many overlapping features and mechanisms with ketamine and may also provide rapid efficacy for TRD patients. Central dopamine circuitry is recognized as an end target for mood regulation and hedonic valuation and yet has been largely neglected in mechanistic studies of antidepressant-relevant effects of ketamine. Herein, we evaluated the changes in dopaminergic neurotransmission after acute administration of ketamine and the mGlu2/3 receptor antagonist LY341495 [(2S)-2-amino-2-[(1S,2S)-2-carboxycycloprop-1-yl]-3-(xanth-9-yl) propanoic acid ] in preclinical models using electrophysiologic, neurochemical, and behavioral endpoints. When given acutely, both ketamine and LY341495, but not the selective serotonin reuptake inhibitor (SSRI) citalopram, increased the number of spontaneously active dopamine neurons in the ventral tegmental area (VTA), increased extracellular levels of dopamine in the nucleus accumbens and prefrontal cortex, and enhanced the locomotor stimulatory effects of the dopamine D2/3 receptor agonist quinpirole. Further, both ketamine and LY341495 reduced immobility time in the tail-suspension assay in CD1 mice, which are relatively resistant to SSRI antidepressants. Both the VTA neuronal activation and the antidepressant phenotype induced by ketamine and LY341495 were attenuated by the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor antagonist 1,2,3,4-tetrahydro-6-nitro-2,3-dioxo- (9CI)-benzo[f]quinoxaline-7-sulfonamide, indicating AMPA-dependent effects. These findings provide another overlapping mechanism of action of ketamine and mGlu2/3 receptor antagonism that differentiates them from conventional antidepressants and thus support the potential rapidly acting antidepressant actions of mGlu2/3 receptor antagonism in patients.


Subject(s)
Amino Acids/pharmacology , Antidepressive Agents/pharmacology , Dopamine/metabolism , Ketamine/pharmacology , Receptors, Metabotropic Glutamate/antagonists & inhibitors , Xanthenes/pharmacology , Action Potentials/drug effects , Animals , Dose-Response Relationship, Drug , Excitatory Amino Acid Antagonists/pharmacology , Male , Mice, Inbred BALB C , Microdialysis , Motor Activity/drug effects , Neurons/drug effects , Neurons/metabolism , Nucleus Accumbens/drug effects , Nucleus Accumbens/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Quinoxalines/pharmacology , Rats, Wistar , Receptors, AMPA/antagonists & inhibitors , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism
3.
J Pharmacol Exp Ther ; 351(2): 448-56, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25187432

ABSTRACT

Scopolamine produces rapid and significant symptom improvement in patients with depression, and most notably in patients who do not respond to current antidepressant treatments. Scopolamine is a nonselective muscarinic acetylcholine receptor antagonist, and it is not known which one or more of the five receptor subtypes in the muscarinic family are mediating these therapeutic effects. We used the mouse forced-swim test, an antidepressant detecting assay, in wild-type and transgenic mice in which each muscarinic receptor subtype had been genetically deleted to define the relevant receptor subtypes. Only the M1 and M2 knockout (KO) mice had a blunted response to scopolamine in the forced-swim assay. In contrast, the effects of the tricyclic antidepressant imipramine were not significantly altered by gene deletion of any of the five muscarinic receptors. The muscarinic antagonists biperiden, pirenzepine, and VU0255035 (N-[3-oxo-3-[4-(4-pyridinyl)-1-piper azinyl]propyl]-2,1,3-benzothiadiazole-4-sulfonamide) with selectivity for M1 over M2 receptors also demonstrated activity in the forced-swim test, which was attenuated in M1 but not M2 receptor KO mice. An antagonist with selectivity of M2 over M1 receptors (SCH226206 [(2-amino-3-methyl-phenyl)-[4-[4-[[4-(3 chlorophenyl)sulfonylphenyl]methyl]-1-piperidyl]-1-piperidyl]methanone]) was also active in the forced-swim assay, and the effects were deleted in M2 (-/-) mice. Brain exposure and locomotor activity in the KO mice demonstrated that these behavioral effects of scopolamine are pharmacodynamic in nature. These data establish muscarinic M1 and M2 receptors as sufficient to generate behavioral effects consistent with an antidepressant phenotype and therefore as potential targets in the antidepressant effects of scopolamine.


Subject(s)
Antidepressive Agents/pharmacology , Receptor, Muscarinic M1/metabolism , Receptor, Muscarinic M2/metabolism , Scopolamine/pharmacology , Animals , Brain/drug effects , Brain/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout/metabolism , Motor Activity/drug effects , Muscarinic Antagonists/pharmacology , Rats , Rats, Sprague-Dawley , Swimming/physiology
4.
J Pharmacol Exp Ther ; 344(2): 501-10, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23197772

ABSTRACT

Dopamine D(3) receptors have eluded definitive linkage to neurologic and psychiatric disorders since their cloning over 20 years ago. We report a new method that does not employ a radiolabel for simultaneously defining in vivo receptor occupancy of D(3) and D(2) receptors in rat brain after systemic dosing using the tracer epidepride (N-[[(2S)-1-ethylpyrrolidin-2-yl]methyl]-5-iodo-2,3-dimethoxybenzamide). Decreases in epidepride binding in lobule 9 of cerebellum (rich in D(3) receptors) were compared with nonspecific binding in the lateral cerebellum. The in vivo occupancy of the dopamine D(3) receptors was dose dependently increased by SB-277011A (trans-N-[4-[2-(6-cyano-1,2,3,4-tetrahydroisoquinolin-2-yl)ethyl]cyclohexyl]-4-quinolinecarboxamide) and U99194 (2,3-dihydro-5,6-dimethoxy- N,N-dipropyl-1H-inden-2-amine). Both antagonists increased extracellular levels of acetylcholine (ACh) in the medial prefrontal cortex of rats and modified brain-tissue levels of ACh and choline. Consistent with these findings, the D(3) receptor antagonists enhanced the acquisition of learning of rats either alone or in the presence of the norepinephrine uptake blocker reboxetine as with the attention-deficit-hyperactivity disorder (ADHD) drug methylphenidate. Like reboxetine, the D(3) receptor antagonists also prevented deficits induced by scopolamine in object recognition memory of rats. Mice in which the dopamine transporter (DAT) has been deleted exhibit hyperactivity that is normalized by compounds that are effective in the treatment of ADHD. Both D(3) receptor antagonists decreased the hyperactivity of DAT(-/-) mice without affecting the activity of wild type controls. The present findings indicate that dopamine D(3) receptor antagonists engender cognition-enhancing and hyperactivity-dampening effects. Thus, D(3) receptor blockade could be considered as a novel treatment approach for cognitive deficits and hyperactivity syndromes, including those observed in ADHD.


Subject(s)
Attention Deficit Disorder with Hyperactivity/drug therapy , Behavior, Animal/drug effects , Brain Chemistry/drug effects , Brain/drug effects , Dopamine Antagonists/pharmacology , Receptors, Dopamine D3/antagonists & inhibitors , Animals , Attention Deficit Disorder with Hyperactivity/metabolism , Brain/metabolism , Dopamine Antagonists/chemistry , Dopamine Antagonists/pharmacokinetics , Dopamine Antagonists/therapeutic use , Dopamine D2 Receptor Antagonists , Indans/chemistry , Indans/pharmacokinetics , Indans/pharmacology , Indans/therapeutic use , Male , Microdialysis , Molecular Structure , Nitriles/chemistry , Nitriles/pharmacokinetics , Nitriles/pharmacology , Nitriles/therapeutic use , Pattern Recognition, Visual/drug effects , Protein Binding , Rats , Rats, Sprague-Dawley , Rats, Wistar , Tetrahydroisoquinolines/chemistry , Tetrahydroisoquinolines/pharmacokinetics , Tetrahydroisoquinolines/pharmacology , Tetrahydroisoquinolines/therapeutic use
5.
Mol Psychiatry ; 11(2): 187-95, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16231039

ABSTRACT

Atomoxetine has been approved by the FDA as the first new drug in 30 years for the treatment of attention deficit/hyperactivity disorder (ADHD). As a selective norepinephrine uptake inhibitor and a nonstimulant, atomoxetine has a different mechanism of action from the stimulant drugs used up to now for the treatment of ADHD. Since brain acetylcholine (ACh) has been associated with memory, attention and motivation, processes dysregulated in ADHD, we investigated the effects of atomoxetine on cholinergic neurotransmission. We showed here that, in rats, atomoxetine (0.3-3 mg/kg, i.p.),--increases in vivo extracellular levels of ACh in cortical but not subcortical brain regions. The marked increase of cortical ACh induced by atomoxetine was dependent upon norepinephrine alpha-1 and/or dopamine D1 receptor activation. We observed similar increases in cortical and hippocampal ACh release with methylphenidate (1 and 3 mg/kg, i.p.)--currently the most commonly prescribed medication for the treatment of ADHD--and with the norepinephrine uptake inhibitor reboxetine (3-30 mg/kg, i.p.). Since drugs that increase cholinergic neurotransmission are used in the treatment of cognitive dysfunction and dementias, we also investigated the effects of atomoxetine on memory tasks. We showed that, consistent with its cortical procholinergic and catecholamine-enhancing profile, atomoxetine (1-3 mg/kg, p.o.) significantly ameliorated performance in the object recognition test and the radial arm-maze test.


Subject(s)
Adrenergic Uptake Inhibitors/pharmacology , Cholinergic Fibers/drug effects , Maze Learning/drug effects , Pattern Recognition, Visual/drug effects , Propylamines/pharmacology , Acetylcholine/metabolism , Animals , Atomoxetine Hydrochloride , Central Nervous System Stimulants/pharmacology , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Cholinergic Fibers/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Male , Methylphenidate/pharmacology , Microdialysis , Morpholines/pharmacology , Norepinephrine/metabolism , Rats , Rats, Sprague-Dawley , Rats, Wistar , Reboxetine , Receptors, Dopamine/drug effects , Receptors, Dopamine/metabolism
6.
J Pharmacol Exp Ther ; 294(2): 688-700, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10900249

ABSTRACT

LY426965 [(2S)-(+)-1-cyclohexyl-4-[4-(2-methoxyphenyl)-1-piperazinyl]2-methyl- 2-phenyl-1-butanone monohydrochloride] is a novel compound with high affinity for the cloned human 5-hydroxytryptamine (HT)(1A) receptor (K(i) = 4.66 nM) and 20-fold or greater selectivity over other serotonin and nonserotonin receptor subtypes. Both in vitro and in vivo studies indicate that LY426965 is a full antagonist and has no partial agonist properties. LY426965 did not stimulate [(35)S]guanosine-5'-O-(3-thio) triphosphate (GTPgammaS) binding to homogenates of cells expressing the cloned human 5-HT(1A) receptor in vitro but did inhibit 300 nM 5-HT-stimulated [(35)S]GTPgammaS binding with a K(i) value of 3.07 nM. After both p.o. and s.c. administration, LY426965 blocked the lower lip retraction, flat body posture, hypothermia, and increase in rat serum corticosterone induced by the 5-HT(1A) agonist 8-OH-DPAT (8-hydroxy-2-dipropylaminotetralin). In pigeons, LY426965 dose-dependently blocked the stimulus cue induced by 8-OH-DPAT but had no 8-OH-DPAT-like discriminative properties. LY426965 completely reversed the effects of nicotine withdrawal on the auditory startle reflex in rats. In microdialysis experiments, LY426965 administered together with fluoxetine significantly increased extracellular levels of serotonin above those achievable with fluoxetine alone. In electrophysiological studies, the administration of LY426965 produced a slight elevation of the firing rate of 5-HT neurons in the dorsal raphe nucleus of anesthetized rats and both blocked and reversed the effects of fluoxetine on 5-HT neuronal activity. These preclinical results indicate that LY426965 is a selective, full 5-HT(1A) antagonist that may have clinical use as pharmacotherapy for smoking cessation and depression and related disorders.


Subject(s)
Fluoxetine/pharmacology , Nicotine/adverse effects , Piperidines/pharmacology , Receptors, Serotonin/drug effects , Selective Serotonin Reuptake Inhibitors/pharmacology , Serotonin Antagonists/pharmacology , Substance Withdrawal Syndrome/drug therapy , 8-Hydroxy-2-(di-n-propylamino)tetralin/antagonists & inhibitors , 8-Hydroxy-2-(di-n-propylamino)tetralin/pharmacology , Acoustic Stimulation , Animals , Body Temperature/drug effects , Columbidae , Corticosterone/blood , Depression/drug therapy , Discrimination Learning/drug effects , Drug Interactions , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Lip/drug effects , Male , Microdialysis , Neurons/drug effects , Neurons/physiology , Posture , Rats , Rats, Long-Evans , Rats, Sprague-Dawley , Receptors, Serotonin/metabolism , Receptors, Serotonin, 5-HT1 , Reflex, Startle/drug effects , Serotonin/pharmacology , Serotonin Antagonists/metabolism , Serotonin Receptor Agonists/pharmacology , Smoking Cessation , Substance Withdrawal Syndrome/etiology , Sulfur Radioisotopes
SELECTION OF CITATIONS
SEARCH DETAIL
...