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










Database
Publication year range
1.
J Pharmacol Exp Ther ; 347(2): 438-57, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23965381

ABSTRACT

Impaired transmission through glutamatergic circuits has been postulated to play a role in the underlying pathophysiology of schizophrenia. Furthermore, inhibition of the N-methyl-d-aspartate (NMDA) subtype of ionotropic glutamate receptors (NMDAR) induces a syndrome that recapitulates many of the symptoms observed in patients with schizophrenia. Selective activation of metabotropic glutamate receptor subtype 5 (mGlu5) may provide a novel therapeutic approach for treatment of symptoms associated with schizophrenia through facilitation of transmission through central glutamatergic circuits. Here, we describe the characterization of two novel N-aryl piperazine mGlu5 positive allosteric modulators (PAMs): 2-(4-(2-(benzyloxy)acetyl)piperazin-1-yl)benzonitrile (VU0364289) and 1-(4-(2,4-difluorophenyl)piperazin-1-yl)-2-((4-fluorobenzyl)oxy)ethanone (DPFE). VU0364289 and DPFE induced robust leftward shifts in the glutamate concentration-response curves for Ca(2+) mobilization and extracellular signal-regulated kinases 1 and 2 phosphorylation. Both PAMs displayed micromolar affinity for the common mGlu5 allosteric binding site and high selectivity for mGlu5. VU0364289 and DPFE possessed suitable pharmacokinetic properties for dosing in vivo and produced robust dose-related effects in reversing amphetamine-induced hyperlocomotion, a preclinical model predictive of antipsychotic-like activity. In addition, DPFE enhanced acquisition of contextual fear conditioning in rats and reversed behavioral deficits in a mouse model of NMDAR hypofunction. In contrast, DPFE had no effect on reversing apomorphine-induced disruptions of prepulse inhibition of the acoustic startle reflex. These mGlu5 PAMs also increased monoamine levels in the prefrontal cortex, enhanced performance in a hippocampal-mediated memory task, and elicited changes in electroencephalogram dynamics commensurate with procognitive effects. Collectively, these data support and extend the role for the development of novel mGlu5 PAMs for the treatment of psychosis and cognitive deficits observed in individuals with schizophrenia.


Subject(s)
Antipsychotic Agents/pharmacology , Hyperkinesis/drug therapy , Memory, Short-Term/drug effects , Nootropic Agents/pharmacology , Piperazines/pharmacology , Receptor, Metabotropic Glutamate 5/agonists , Receptors, N-Methyl-D-Aspartate/metabolism , Allosteric Regulation , Animals , Antipsychotic Agents/chemistry , Antipsychotic Agents/pharmacokinetics , Antipsychotic Agents/therapeutic use , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , HEK293 Cells , Humans , Hyperkinesis/metabolism , Hyperkinesis/psychology , Male , Maze Learning/drug effects , Mice , Mice, Knockout , Motor Activity/drug effects , Nootropic Agents/chemistry , Nootropic Agents/pharmacokinetics , Nootropic Agents/therapeutic use , Piperazines/chemistry , Piperazines/pharmacokinetics , Piperazines/therapeutic use , Rats , Rats, Sprague-Dawley , Receptor, Metabotropic Glutamate 5/genetics , Receptors, N-Methyl-D-Aspartate/genetics , Schizophrenia/drug therapy , Schizophrenia/metabolism , Transfection
2.
Handb Exp Pharmacol ; (213): 297-365, 2012.
Article in English | MEDLINE | ID: mdl-23027420

ABSTRACT

Metabotropic glutamate receptors (mGluRs) represent exciting targets for the development of novel therapeutic agents for schizophrenia. Recent studies indicate that selective activation of specific mGluR subtypes may provide potential benefits for not only the positive symptoms, but also the negative symptoms and cognitive impairments observed in individuals with schizophrenia. Although optimization of traditional orthosteric agonists may still offer a feasible approach for the activation of mGluRs, important progress has been made in the discovery of novel subtype-selective allosteric ligands, including positive allosteric modulators (PAMs) of mGluR2 and mGluR5. These allosteric mGluR ligands have improved properties for clinical development and have served as key preclinical tools for a more in-depth understanding of the potential roles of these different mGluR subtypes for the treatment of schizophrenia.


Subject(s)
Receptors, Metabotropic Glutamate/physiology , Schizophrenia/drug therapy , Allosteric Regulation , Animals , Cognition/drug effects , Humans , Ligands , Receptors, Metabotropic Glutamate/analysis , Receptors, Metabotropic Glutamate/chemistry , Receptors, Metabotropic Glutamate/drug effects , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...