Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 794
Filter
1.
Int J Mol Sci ; 24(3)2023 Jan 19.
Article in English | MEDLINE | ID: mdl-36768302

ABSTRACT

Following the glutamatergic theory of schizophrenia and based on our previous study regarding the antipsychotic-like activity of mGlu7 NAMs, we synthesized a new compound library containing 103 members, which were examined for NAM mGlu7 activity in the T-REx 293 cell line expressing a recombinant human mGlu7 receptor. Out of the twenty-two scaffolds examined, active compounds were found only within the quinazolinone chemotype. 2-(2-Chlorophenyl)-6-(2,3-dimethoxyphenyl)-3-methylquinazolin-4(3H)-one (A9-7, ALX-171, mGlu7 IC50 = 6.14 µM) was selective over other group III mGlu receptors (mGlu4 and mGlu8), exhibited satisfactory drug-like properties in preliminary DMPK profiling, and was further tested in animal models of antipsychotic-like activity, assessing the positive, negative, and cognitive symptoms. ALX-171 reversed DOI-induced head twitches and MK-801-induced disruptions of social interactions or cognition in the novel object recognition test and spatial delayed alternation test. On the other hand, the efficacy of the compound was not observed in the MK-801-induced hyperactivity test or prepulse inhibition. In summary, the observed antipsychotic activity profile of ALX-171 justifies the further development of the group of quinazolin-4-one derivatives in the search for a new drug candidate for schizophrenia treatment.


Subject(s)
Antipsychotic Agents , Quinazolinones , Receptors, Metabotropic Glutamate , Schizophrenia , Animals , Humans , Antipsychotic Agents/pharmacology , Antipsychotic Agents/therapeutic use , Dizocilpine Maleate , Quinazolinones/pharmacology , Quinazolinones/therapeutic use , Receptors, Metabotropic Glutamate/drug effects , Receptors, Metabotropic Glutamate/metabolism , Schizophrenia/drug therapy , Schizophrenia/metabolism , Drug Design
2.
J Neurosci ; 41(11): 2344-2359, 2021 03 17.
Article in English | MEDLINE | ID: mdl-33500274

ABSTRACT

Metabotropic glutamate receptor 7 (mGlu7) is an inhibitory heterotrimeric G-protein-coupled receptor that modulates neurotransmitter release and synaptic plasticity at presynaptic terminals in the mammalian central nervous system. Recent studies have shown that rare mutations in glutamate receptors and synaptic scaffold proteins are associated with neurodevelopmental disorders (NDDs). However, the role of presynaptic mGlu7 in the pathogenesis of NDDs remains largely unknown. Recent whole-exome sequencing (WES) studies in families with NDDs have revealed that several missense mutations (c.1865G>A:p.R622Q; c.461T>C:p.I154T; c.1972C>T:p.R658W and c.2024C>A:p.T675K) or a nonsense mutation (c.1757G>A:p.W586X) in the GRM7 gene may be linked to NDDs. In the present study, we investigated the mechanistic links between GRM7 point mutations and NDD pathology. We find that the pathogenic GRM7 I154T and R658W/T675K mutations lead to the degradation of the mGlu7 protein. In particular, the GRM7 R658W/T675K mutation results in a lack of surface mGlu7 expression in heterologous cells and cultured neurons isolated from male and female rat embryos. We demonstrate that the expression of mGlu7 variants or exposure to mGlu7 antagonists impairs axon outgrowth through the mitogen-activated protein kinase (MAPK)-cAMP-protein kinase A (PKA) signaling pathway during early neuronal development, which subsequently leads to a decrease in the number of presynaptic terminals in mature neurons. Treatment with an mGlu7 agonist restores the pathologic phenotypes caused by mGlu7 I154T but not by mGlu7 R658W/T675K because of its lack of neuronal surface expression. These findings provide evidence that stable neuronal surface expression of mGlu7 is essential for neural development and that mGlu7 is a promising therapeutic target for NDDs.SIGNIFICANCE STATEMENT Neurodevelopmental disorders (NDDs) affect brain development and function by multiple etiologies. Metabotropic glutamate receptor 7 (mGlu7) is a receptor that controls excitatory neurotransmission and synaptic plasticity. Since accumulating evidence indicates that the GRM7 gene locus is associated with NDD risk, we analyzed the functional effects of human GRM7 variants identified in patients with NDDs. We demonstrate that stable neuronal surface expression of mGlu7 is essential for axon outgrowth and presynaptic terminal development in neurons. We found that mitogen-activated protein kinase (MAPK)-cAMP-protein kinase A (PKA) signaling and subsequent cytoskeletal dynamics are defective because of the degradation of mGlu7 variants. Finally, we show that the defects caused by mGlu7 I154T can be reversed by agonists, providing the rationale for proposing mGlu7 as a potential therapeutic target for NDDs.


Subject(s)
Axons/pathology , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/pathology , Point Mutation/genetics , Presynaptic Terminals , Receptors, Metabotropic Glutamate/genetics , Animals , Axons/drug effects , Cell Count , Cell Survival , Cyclic AMP-Dependent Protein Kinases/biosynthesis , Cyclic AMP-Dependent Protein Kinases/genetics , Female , Gene Expression Regulation , Male , Neurons/metabolism , Neurons/pathology , Pregnancy , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/biosynthesis , Receptors, Metabotropic Glutamate/drug effects , Signal Transduction/genetics , Synapses/pathology , Exome Sequencing
3.
Proc Natl Acad Sci U S A ; 116(13): 6441-6450, 2019 03 26.
Article in English | MEDLINE | ID: mdl-30867285

ABSTRACT

Currently approved antidepressant drugs often take months to take full effect, and ∼30% of depressed patients remain treatment resistant. In contrast, ketamine, when administered as a single subanesthetic dose, exerts rapid and sustained antidepressant actions. Preclinical studies indicate that the ketamine metabolite (2R,6R)-hydroxynorketamine [(2R,6R)-HNK] is a rapid-acting antidepressant drug candidate with limited dissociation properties and abuse potential. We assessed the role of group II metabotropic glutamate receptor subtypes 2 (mGlu2) and 3 (mGlu3) in the antidepressant-relevant actions of (2R,6R)-HNK using behavioral, genetic, and pharmacological approaches as well as cortical quantitative EEG (qEEG) measurements in mice. Both ketamine and (2R,6R)-HNK prevented mGlu2/3 receptor agonist (LY379268)-induced body temperature increases in mice lacking the Grm3, but not Grm2, gene. This action was not replicated by NMDA receptor antagonists or a chemical variant of ketamine that limits metabolism to (2R,6R)-HNK. The antidepressant-relevant behavioral effects and 30- to 80-Hz qEEG oscillation (gamma-range) increases resultant from (2R,6R)-HNK administration were prevented by pretreatment with an mGlu2/3 receptor agonist and absent in mice lacking the Grm2, but not Grm3-/-, gene. Combined subeffective doses of the mGlu2/3 receptor antagonist LY341495 and (2R,6R)-HNK exerted synergistic increases on gamma oscillations and antidepressant-relevant behavioral actions. These findings highlight that (2R,6R)-HNK exerts antidepressant-relevant actions via a mechanism converging with mGlu2 receptor signaling and suggest enhanced cortical gamma oscillations as a marker of target engagement relevant to antidepressant efficacy. Moreover, these results support the use of (2R,6R)-HNK and inhibitors of mGlu2 receptor function in clinical trials for treatment-resistant depression either alone or in combination.


Subject(s)
Antidepressive Agents/pharmacology , Depression/drug therapy , Ketamine/pharmacology , Receptors, Metabotropic Glutamate/drug effects , Amino Acids/antagonists & inhibitors , Animals , Behavior, Animal/drug effects , Bridged Bicyclo Compounds, Heterocyclic/antagonists & inhibitors , Disease Models, Animal , Drug Resistance , Female , Fever , Ketamine/administration & dosage , Ketamine/chemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Metabotropic Glutamate/genetics , Receptors, Metabotropic Glutamate/metabolism , Receptors, N-Methyl-D-Aspartate/drug effects
4.
J Neurophysiol ; 125(6): 2339-2355, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33978492

ABSTRACT

The activity of central pattern-generating networks (CPGs) may change under the control exerted by various neurotransmitters and modulators to adapt its behavioral outputs to different environmental demands. Although the mechanisms underlying this control have been well established in invertebrates, most of their synaptic and cellular bases are not yet well understood in vertebrates. Gymnotus omarorum, a pulse-type gymnotiform electric fish, provides a well-suited vertebrate model to investigate these mechanisms. G. omarorum emits rhythmic and stereotyped electric organ discharges (EODs), which function in both perception and communication, under the command of an electromotor CPG. This nucleus is composed of electrotonically coupled intrinsic pacemaker cells, which pace the rhythm, and bulbospinal projecting relay cells that contribute to organize the pattern of the muscle-derived effector activation that produce the EOD. Descending influences target CPG neurons to produce adaptive behavioral electromotor responses to different environmental challenges. We used electrophysiological and pharmacological techniques in brainstem slices of G. omarorum to investigate the underpinnings of the fast transmitter control of its electromotor CPG. We demonstrate that pacemaker, but not relay cells, are endowed with ionotropic and metabotropic glutamate receptor subtypes. We also show that glutamatergic control of the CPG likely involves two types of synapses contacting pacemaker cells, one type containing both α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) and N-methyl-d-aspartate (NMDA) receptors and the other one only-NMDA receptor. Fast neurotransmitter control of vertebrate CPGs seems to exploit the kinetics of the involved postsynaptic receptors to command different behavioral outputs. The prospect of common neural designs to control CPG activity in vertebrates is discussed.NEW & NOTEWORTHY Underpinnings of neuromodulation of central pattern-generating networks (CPG) have been well characterized in many species. The effects of fast neurotransmitter systems remain, however, poorly understood. This research uses in vitro electrophysiological and pharmacological techniques to show that the neurotransmitter control of a vertebrate CPG in gymnotiform fish involves the convergence of only-NMDA and AMPA-NMDA glutamatergic synapses onto neurons that pace the rhythm. These inputs may organize different behavioral outputs according to their distinct functional properties.


Subject(s)
Biological Clocks/physiology , Central Pattern Generators/metabolism , Electrophysiological Phenomena/physiology , Excitatory Amino Acid Agonists/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Gymnotiformes/physiology , Receptors, Ionotropic Glutamate/metabolism , Receptors, Metabotropic Glutamate/metabolism , Animals , Biological Clocks/drug effects , Central Pattern Generators/drug effects , Electric Stimulation , Electrophysiological Phenomena/drug effects , Gymnotiformes/metabolism , Receptors, Ionotropic Glutamate/drug effects , Receptors, Metabotropic Glutamate/drug effects
5.
Neurobiol Learn Mem ; 181: 107445, 2021 05.
Article in English | MEDLINE | ID: mdl-33895349

ABSTRACT

In rodent models of smoking during pregnancy, early postnatal nicotine exposure results in impaired hippocampus-dependent memory, but the underlying mechanism remains elusive. Given that hippocampal cholinergic systems modulate memory and rapid development of hippocampal cholinergic systems occurs during nicotine exposure, here we investigated its impacts on cholinergic function. Both nicotinic and muscarinic activation produce transient or long-lasting depression of excitatory synaptic transmission in the hippocampal CA1 region. We found that postnatal nicotine exposure impairs both the induction and nicotinic modulation of NMDAR-dependent long-term depression (LTD). Activation of muscarinic receptors decreases excitatory synaptic transmission and CA1 network activity in both wild-type and α2 knockout mice. These muscarinic effects are still observed in nicotine-exposed mice. M1 muscarinic receptor activity is required for mGluR-dependent LTD. Early postnatal nicotine exposure has no effect on mGluR-dependent LTD induction, suggesting that it has no effect on the function of m1 muscarinic receptors involved in this form of LTD. Our results demonstrate that early postnatal nicotine exposure has more pronounced effects on nicotinic function than muscarinic function in the hippocampal CA1 region. Thus, impaired hippocampus-dependent memory may arise from the developmental disruption of nicotinic cholinergic systems in the hippocampal CA1 region.


Subject(s)
CA1 Region, Hippocampal/drug effects , Long-Term Synaptic Depression/drug effects , Nicotine/pharmacology , Nicotinic Agonists/pharmacology , Receptor, Muscarinic M1/drug effects , Receptors, Nicotinic/drug effects , Animals , Animals, Newborn , CA1 Region, Hippocampal/growth & development , CA1 Region, Hippocampal/metabolism , Cigarette Smoking , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Female , Lactation , Long-Term Synaptic Depression/physiology , Male , Maternal Exposure , Memory/drug effects , Memory/physiology , Mice , Mice, Knockout , Receptor, Muscarinic M1/metabolism , Receptors, Metabotropic Glutamate/drug effects , Receptors, Metabotropic Glutamate/metabolism , Receptors, Muscarinic/drug effects , Receptors, Muscarinic/metabolism , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Nicotinic/metabolism
6.
Am J Physiol Regul Integr Comp Physiol ; 318(3): R545-R564, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31967862

ABSTRACT

Astrocytic excitatory amino acid transporters (EAATs) are critical to restraining synaptic and neuronal activity in the nucleus tractus solitarii (nTS). Relief of nTS EAAT restraint generates two opposing effects, an increase in neuronal excitability that reduces blood pressure and breathing and an attenuation in afferent [tractus solitarius (TS)]-driven excitatory postsynaptic current (EPSC) amplitude. Although the former is due, in part, to activation of ionotropic glutamate receptors, there remains a substantial contribution from another unidentified glutamate receptor. In addition, the mechanism(s) by which EAAT inhibition reduced TS-EPSC amplitude is unknown. Metabotropic glutamate receptors (mGluRs) differentially modulate nTS excitability. Activation of group I mGluRs on nTS neuron somas leads to depolarization, whereas group II/III mGluRs on sensory afferents decrease TS-EPSC amplitude. Thus we hypothesize that EAATs control postsynaptic excitability and TS-EPSC amplitude via restraint of mGluR activation. To test this hypothesis, we used in vivo recording, brain slice electrophysiology, and imaging of glutamate release and TS-afferent Ca2+. Results show that EAAT blockade in the nTS with (3S)-3-[[3-[[4-(trifluoromethyl)benzoyl]amino]phenyl]methoxy]-l-aspartic acid (TFB-TBOA) induced group I mGluR-mediated depressor, bradycardic, and apneic responses that were accompanied by neuronal depolarization, elevated discharge, and increased spontaneous synaptic activity. Conversely, upon TS stimulation TFB-TBOA elevated extracellular glutamate to decrease presynaptic Ca2+ and TS-EPSC amplitude via activation of group II/III mGluRs. Together, these data suggest an important role of EAATs in restraining mGluR activation and overall cardiorespiratory function.


Subject(s)
Amino Acid Transport System X-AG/drug effects , Aspartic Acid/analogs & derivatives , Astrocytes/metabolism , Neurons/metabolism , Receptors, Metabotropic Glutamate/metabolism , Amino Acid Transport System X-AG/metabolism , Animals , Aspartic Acid/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Glutamic Acid/metabolism , Neurons/drug effects , Receptors, Metabotropic Glutamate/drug effects , Solitary Nucleus/metabolism , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
7.
Cereb Cortex ; 29(8): 3241-3252, 2019 07 22.
Article in English | MEDLINE | ID: mdl-30137253

ABSTRACT

The fragile X mental retardation protein (FMRP) is an RNA-binding protein involved in translational regulation of mRNAs that play key roles in synaptic morphology and plasticity. The functional absence of FMRP causes the fragile X syndrome (FXS), the most common form of inherited intellectual disability and the most common monogenic cause of autism. No effective treatment is available for FXS. We recently identified the Phosphodiesterase 2A (Pde2a) mRNA as a prominent target of FMRP. PDE2A enzymatic activity is increased in the brain of Fmr1-KO mice, a recognized model of FXS, leading to decreased levels of cAMP and cGMP. Here, we pharmacologically inhibited PDE2A in Fmr1-KO mice and observed a rescue both of the maturity of dendritic spines and of the exaggerated hippocampal mGluR-dependent long-term depression. Remarkably, PDE2A blockade rescued the social and communicative deficits of both mouse and rat Fmr1-KO animals. Importantly, chronic inhibition of PDE2A in newborn Fmr1-KO mice followed by a washout interval, resulted in the rescue of the altered social behavior observed in adolescent mice. Altogether, these results reveal the key role of PDE2A in the physiopathology of FXS and suggest that its pharmacological inhibition represents a novel therapeutic approach for FXS.


Subject(s)
Animal Communication , Cyclic Nucleotide Phosphodiesterases, Type 2/metabolism , Dendritic Spines/drug effects , Fragile X Syndrome/enzymology , Hippocampus/drug effects , Imidazoles/pharmacology , Long-Term Synaptic Depression/drug effects , Neurons/drug effects , Phosphodiesterase Inhibitors/pharmacology , Social Behavior , Triazines/pharmacology , Animals , Animals, Newborn , Cyclic AMP/metabolism , Cyclic GMP/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 2/antagonists & inhibitors , Dendritic Spines/pathology , Embryo, Mammalian , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Fragile X Mental Retardation Protein/genetics , Fragile X Syndrome/genetics , Fragile X Syndrome/pathology , Fragile X Syndrome/physiopathology , Gene Knockout Techniques , Hippocampus/metabolism , Mice , Mice, Knockout , Neurons/metabolism , Neurons/pathology , Primary Cell Culture , Rats , Receptors, Metabotropic Glutamate/drug effects , Receptors, Metabotropic Glutamate/metabolism
8.
J Neurosci ; 38(19): 4462-4470, 2018 05 09.
Article in English | MEDLINE | ID: mdl-29593052

ABSTRACT

NMDA receptors (NMDARs) mediate both long-term potentiation and long-term depression (LTD) and understanding how a single receptor can initiate both phenomena remains a major question in neuroscience. A prominent hypothesis implicates the NMDAR subunit composition, specifically GluN2A and GluN2B, in dictating the rules of synaptic plasticity. However, studies testing this hypothesis have yielded inconsistent and often contradictory results, especially for LTD. These inconsistent results may be due to challenges in the interpretation of subunit-selective pharmacology and in dissecting out the contributions of differential channel properties versus the interacting proteins unique to GluN2A or GluN2B. In this study, we address the pharmacological and biochemical challenges by using a single-neuron genetic approach to delete NMDAR subunits in conditional knock-out mice. In addition, the recently discovered non-ionotropic nature of NMDAR-dependent LTD allowed the rigorous assessment of unique subunit contributions to NMDAR-dependent LTD while eliminating the variable of differential charge transfer. Here we find that neither the GluN2A nor the GluN2B subunit is strictly necessary for either non-ionotropic or ionotropic LTD.SIGNIFICANCE STATEMENT NMDA receptors are key regulators of bidirectional synaptic plasticity. Understanding the mechanisms regulating bidirectional plasticity will guide development of therapeutic strategies to treat the dysfunctional synaptic plasticity in multiple neuropsychiatric disorders. Because of the unique properties of the NMDA receptor GluN2 subunits, they have been postulated to differentially affect synaptic plasticity. However, there has been significant controversy regarding the roles of the GluN2 subunits in synaptic long term depression (LTD). Using single-neuron knock-out of the GluN2 subunits, we show that LTD requires neither GluN2A nor GluN2B.


Subject(s)
Long-Term Synaptic Depression/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Female , Male , Mice , Mice, Knockout , Negative Results , Neuronal Plasticity/physiology , Neurons/metabolism , Receptors, Metabotropic Glutamate/drug effects , Receptors, Metabotropic Glutamate/physiology , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism
9.
J Neurosci ; 37(17): 4618-4634, 2017 04 26.
Article in English | MEDLINE | ID: mdl-28363980

ABSTRACT

G-protein ßγ subunits (Gßγ) interact with presynaptic proteins and regulate neurotransmitter release downstream of Ca2+ influx. To accomplish their roles in sensory signaling, photoreceptor synapses use specialized presynaptic proteins that support neurotransmission at active zone structures known as ribbons. While several G-protein coupled receptors (GPCRs) influence synaptic transmission at ribbon synapses of cones and other retinal neurons, it is unknown whether Gßγ contributes to these effects. We tested whether activation of one particular GPCR, a metabotropic glutamate receptor (mGluR), can reduce cone synaptic transmission via Gßγ in tiger salamander retinas. In recordings from horizontal cells, we found that an mGluR agonist (L-AP4) reduced cone-driven light responses and mEPSC frequency. In paired recordings of cones and horizontal cells, L-AP4 slightly reduced cone ICa (∼10%) and caused a larger reduction in cone-driven EPSCs (∼30%). Proximity ligation assay revealed direct interactions between SNAP-25 and Gßγ subunits in retinal synaptic layers. Pretreatment with the SNAP-25 cleaving protease BoNT/A inhibited L-AP4 effects on synaptic transmission, as did introduction of a peptide derived from the SNAP-25 C terminus. Introducing Gßγ subunits directly into cones reduced EPSC amplitude. This effect was inhibited by BoNT/A, supporting a role for Gßγ/SNAP-25 interactions. However, the mGluR-dependent reduction in ICa was not mimicked by Gßγ, indicating that this effect was independent of Gßγ. The finding that synaptic transmission at cone ribbon synapses is regulated by Gßγ/SNAP-25 interactions indicates that these mechanisms are shared by conventional and ribbon-type synapses. Gßγ liberated from other photoreceptor GPCRs is also likely to regulate synaptic transmission.SIGNIFICANCE STATEMENT Dynamic regulation of synaptic transmission by presynaptic G-protein coupled receptors shapes information flow through neural circuits. At the first synapse in the visual system, presynaptic metabotropic glutamate receptors (mGluRs) regulate cone photoreceptor synaptic transmission, although the mechanisms and functional impact of this are unclear. We show that mGluRs regulate light response encoding across the cone synapse, accomplished in part by triggering G-protein ßγ subunits (Gßγ) interactions with SNAP-25, a core component of the synaptic vesicle fusion machinery. In addition to revealing a role in visual processing, this provides the first demonstration that Gßγ/SNAP-25 interactions regulate synaptic function at a ribbon-type synapse, contributing to an emerging picture of the ubiquity of Gßγ/SNARE interactions in regulating synaptic transmission throughout the nervous system.


Subject(s)
Ambystoma/physiology , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/metabolism , Receptors, Metabotropic Glutamate/metabolism , Retinal Cone Photoreceptor Cells/physiology , SNARE Proteins/metabolism , Synapses/physiology , Synaptic Transmission/physiology , Animals , Excitatory Postsynaptic Potentials/physiology , Female , Male , Receptors, Metabotropic Glutamate/drug effects , Retinal Horizontal Cells/metabolism , Retinal Horizontal Cells/physiology
10.
Mol Pharmacol ; 93(5): 526-540, 2018 05.
Article in English | MEDLINE | ID: mdl-29545267

ABSTRACT

Allosteric modulation of metabotropic glutamate receptor 2 (mGlu2) has demonstrated efficacy in preclinical rodent models of several brain disorders, leading to industry and academic drug discovery efforts. Although the pharmacology and binding sites of some mGlu2 allosteric modulators have been characterized previously, questions remain about the nature of the allosteric mechanism of cooperativity with glutamate and whether structurally diverse allosteric modulators bind in an identical manner to specific allosteric sites. To further investigate the in vitro pharmacology of mGlu2 allosteric modulators, we developed and characterized a novel mGlu2 positive allosteric modulator (PAM) radioligand in parallel with functional studies of a structurally diverse set of mGlu2 PAMs and negative allosteric modulators (NAMs). Using an operational model of allosterism to analyze the functional data, we found that PAMs affect both the affinity and efficacy of glutamate at mGlu2, whereas NAMs predominantly affect the efficacy of glutamate in our assay system. More importantly, we found that binding of a novel mGlu2 PAM radioligand was inhibited by multiple structurally diverse PAMs and NAMs, indicating that they may bind to the mGlu2 allosteric site labeled with the novel mGlu2 PAM radioligand; however, further studies suggested that these allosteric modulators do not all interact with the radioligand in an identical manner. Together, these findings provide new insights into the binding sites and modes of efficacy of different structurally and functionally distinct mGlu2 allosteric modulators and suggest that different ligands either interact with distinct sites or adapt different binding poses to shared allosteric site(s).


Subject(s)
Receptors, Metabotropic Glutamate/drug effects , Allosteric Regulation , Allosteric Site , Animals , Cell Line , Glutamic Acid/metabolism , HEK293 Cells , Humans , Ligands , Mutagenesis , Protein Binding , Radioligand Assay , Rats , Receptors, Metabotropic Glutamate/genetics , Receptors, Metabotropic Glutamate/metabolism
11.
Bioorg Med Chem Lett ; 28(19): 3194-3196, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30146095

ABSTRACT

This work describes the rational amelioration of Cytochrome P450 4/5 (CYP3A4/5) induction through the Pregnane-X Receptor (PXR) pathway in a series of compounds that modulate the metabotropic glutamate Receptor 2 (mGluR2) via an allosteric mechanism. The compounds were initially shown to induce CYP3A4/5 via the gold-standard induction assay measured in primary human hepatocytes. This was followed up by testing the compounds in a PXR assay which correlated well with the assay in primary cells. Further, one of the compounds was crystallized with PXR (pdb code 6DUP). Analysis of this co-crystal structure, together with previously published PXR co-crystal structures, lead to modification ideas. The compounds synthesized based on these ideas were shown not to be CYP3A4/5 inducers. The mGluR2 activity of the resulting compounds was maintained.


Subject(s)
Cytochrome P-450 CYP3A/biosynthesis , Pregnane X Receptor/physiology , Receptors, Metabotropic Glutamate/drug effects , Allosteric Regulation , Animals , Crystallography, X-Ray , Enzyme Induction/physiology , Humans , Pregnane X Receptor/chemistry , Rats
12.
Bioorg Med Chem Lett ; 28(15): 2641-2646, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29958762

ABSTRACT

Previous reports from our laboratory disclosed the structure and activity of a novel 1H-pyrazolo[4,3-b]pyridine-3-amine scaffold (VU8506) which showed excellent potency, selectivity and in vivo efficacy in preclinical rodent models of Parkinson's disease. Unfortunately, this compound suffered from significant CYP1A2 induction as measured through upstream AhR activation (125-fold) and thus was precluded from further advancement in chronic studies. Herein, we report a new scaffold developed recently which was systematically studied in order to mitigate the CYP1A2 liabilities presented in the earlier scaffolds. We have identified a novel structure that maintains the potency and selectivity of other mGlu4 PAMs, leading to 9i (hmGlu4 EC50 = 43 nM; AhR activation = 2.3-fold).


Subject(s)
Cytochrome P-450 CYP1A2 Inducers/pharmacology , Cytochrome P-450 CYP1A2/biosynthesis , Drug Discovery , Pyrazoles/chemistry , Pyrazoles/pharmacology , Pyridines/chemistry , Pyridines/pharmacology , Receptors, Metabotropic Glutamate/drug effects , Allosteric Regulation/drug effects , Animals , Antiparkinson Agents/pharmacology , Enzyme Induction/drug effects , Humans , Rats , Receptors, Metabotropic Glutamate/metabolism , Structure-Activity Relationship
13.
Bioorg Med Chem ; 26(17): 4797-4803, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30143366

ABSTRACT

Isoxazolo[3,4-d] pyridazinones ([3,4-d]s) are selective positive modulators of the metabotropic glutamate receptors (mGluRs) subtypes 2 and 4, with no functional cross reactivity at mGluR1a, mGLuR5 or mGluR8. Modest binding for two of the [3,4-d]s is observed at the allosteric fenobam mGluR5 site, but not sufficient to translate into a functional effect. The structure activity relationship (SAR) for mGluR2 and mGluR4 are distinct: the compounds which select for mGluR2 all contain fluorine on the N-6 aryl group. Furthermore, the [3,4-d]s in this study showed no significant binding at inhibitory GABAA, nor excitatory NMDA receptors, and previously we had disclosed that they lack significant activity at the System Xc-Antiporter. A homology model based on Conn's mGluR1 crystal structure was examined, and suggested explanations for a preference for allosteric over orthosteric binding, subtype selectivity, and suggested avenues for optimization of efficacy as a reasonable working hypothesis.


Subject(s)
Isoxazoles/chemistry , Pyridazines/chemistry , Pyridazines/pharmacology , Receptors, Metabotropic Glutamate/drug effects , Allosteric Regulation , Animals , Receptors, Metabotropic Glutamate/chemistry , Structure-Activity Relationship
14.
J Neurosci ; 36(18): 4910-6, 2016 05 04.
Article in English | MEDLINE | ID: mdl-27147646

ABSTRACT

UNLABELLED: Type 1 metabotropic glutamate receptor (mGluR1)-dependent signaling at parallel fiber to Purkinje neuron synapses is critical for cerebellar function. In a mouse model of human spino-cerebellar ataxia type 1 (early SCA1, 12 weeks) we find prolonged parallel fiber mGluR1-dependent synaptic currents and calcium signaling. Acute treatment with a low dose of the potent and specific activity-dependent mGluR1-negative allosteric modulator JNJ16259685 shortened the prolonged mGluR1 currents and rescued the moderate ataxia. Our results provide exciting new momentum for developing mGluR1-based pharmacology to treat ataxia. SIGNIFICANCE STATEMENT: Ataxia is a progressive and devastating degenerative movement disorder commonly associated with loss of cerebellar function and with no known cure. In the early stages of a mouse model of human spinocerebellar ataxia type 1, SCA1, where mice exhibit only moderate motor impairment, we detect excess "gain of function" of metabotropic glutamate receptor signaling at an important cerebellar synapse. Because careful control of this type of signaling is critical for cerebellar function in mice and humans, we sought to remove the excess signaling with a powerful, readily available pharmacological modulator. Remarkably, this pharmacological treatment acutely restored normal motor function in the ataxic mice. Our results pave the way for exploring a new avenue for early treatment of human ataxias.


Subject(s)
Receptors, Metabotropic Glutamate/genetics , Signal Transduction , Spinocerebellar Ataxias/genetics , Spinocerebellar Ataxias/physiopathology , Synapses , Animals , Behavior, Animal/drug effects , Calcium Signaling/physiology , Female , Male , Mice , Movement/drug effects , Purkinje Cells/metabolism , Quinolines/pharmacology , Receptors, Metabotropic Glutamate/drug effects , Receptors, Metabotropic Glutamate/metabolism , Signal Transduction/drug effects , Spinocerebellar Ataxias/psychology , Synapses/drug effects
15.
Addict Biol ; 22(2): 275-290, 2017 Mar.
Article in English | MEDLINE | ID: mdl-26549202

ABSTRACT

N-Methyl-d-aspartate receptors (NMDARs) are major targets of both acute and chronic alcohol, as well as regulators of plasticity in a number of brain regions. Aberrant plasticity may contribute to the treatment resistance and high relapse rates observed in alcoholics. Recent work suggests that chronic alcohol treatment preferentially modulates both the expression and subcellular localization of NMDARs containing the GluN2B subunit. Signaling through synaptic and extrasynaptic GluN2B-NMDARs has already been implicated in the pathophysiology of various other neurological disorders. NMDARs interact with a large number of proteins at the glutamate synapse, and a better understanding of how alcohol modulates this proteome is needed. We employed a discovery-based proteomic approach in subcellular fractions of hippocampal tissue from chronic intermittent alcohol (CIE)-exposed C57Bl/6J mice to gain insight into alcohol-induced changes in GluN2B signaling complexes. Protein enrichment analyses revealed changes in the association of post-synaptic proteins, including scaffolding, glutamate receptor and PDZ-domain binding proteins with GluN2B. In particular, GluN2B interaction with metabotropic glutamate (mGlu)1/5 receptor-dependent long-term depression (LTD)-associated proteins such as Arc and Homer 1 was increased, while GluA2 was decreased. Accordingly, we found a lack of mGlu1/5 -induced LTD while α1 -adrenergic receptor-induced LTD remained intact in hippocampal CA1 following CIE. These data suggest that CIE specifically disrupts mGlu1/5 -LTD, representing a possible connection between NMDAR and mGlu receptor signaling. These studies not only demonstrate a new way in which alcohol can modulate plasticity in the hippocampus but also emphasize the utility of this discovery-based proteomic approach to generate new hypotheses regarding alcohol-related mechanisms.


Subject(s)
Central Nervous System Depressants/pharmacology , Ethanol/pharmacology , Hippocampus/drug effects , Long-Term Synaptic Depression/drug effects , Receptors, Metabotropic Glutamate/drug effects , Receptors, N-Methyl-D-Aspartate/drug effects , Animals , Central Nervous System Depressants/administration & dosage , Cytoskeletal Proteins/drug effects , Cytoskeletal Proteins/metabolism , Ethanol/administration & dosage , Hippocampus/metabolism , Homer Scaffolding Proteins/drug effects , Homer Scaffolding Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/drug effects , Nerve Tissue Proteins/metabolism , Proteome/drug effects , Proteome/metabolism , Receptors, Metabotropic Glutamate/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction
16.
Bioorg Med Chem Lett ; 26(9): 2289-92, 2016 May 01.
Article in English | MEDLINE | ID: mdl-27013388

ABSTRACT

This letter describes the re-exploration of the mGlu1 PAM Ro 07-11401 scaffold through a multi-dimensional, iterative parallel synthesis approach. Unlike recent series of mGlu1 PAMs with robust SAR, the SAR around the Ro 07-11401 structure was incredibly steep (only ∼6 of 200 analogs displayed mGlu1 PAM activity), and reminiscent of the CPPHA mGlu5 PAM scaffold. Despite the steep SAR, two new thiazole derivatives were discovered with improved in vitro DMPK profiles and ∼3- to 4-fold improvement in CNS exposure (Kps 1.01-1.19); albeit, with a ∼3-fold diminution in mGlu1 PAM potency, yet comparable efficacy (∼5-fold leftward shift of the glutamate concentration-response curve at 10µM). Thus, this effort has provided additional CNS penetrant mGlu1 PAM tools in a different chemotype than the VU0486321 scaffold. These compounds will permit a better understanding of the pharmacology and therapeutic potential of selective mGlu1 activation, while highlighting the steep SAR challenges that can often be encountered in GPCR allosteric modulator discovery.


Subject(s)
Central Nervous System/drug effects , Excitatory Amino Acid Antagonists/pharmacology , Receptors, Metabotropic Glutamate/drug effects , Animals , Central Nervous System/metabolism , Drug Discovery , Excitatory Amino Acid Antagonists/metabolism , Humans , Rats , Structure-Activity Relationship
17.
J Neurosci ; 33(2): 495-506a, 2013 Jan 09.
Article in English | MEDLINE | ID: mdl-23303930

ABSTRACT

Anomalies in prefrontal cortex (PFC) function are posited to underpin difficulties in learning to suppress drug-seeking behavior during abstinence. Because group 1 metabotropic glutamate receptors (mGluRs) regulate drug-related learning, we assayed the consequences of extended access to intravenous cocaine (6 h/d; 0.25 mg/infusion for 10 d) on the PFC expression of group 1 mGluRs and the relevance of observed changes for cocaine seeking. After protracted withdrawal, cocaine-experienced animals exhibited a time-dependent intensification of cue-induced cocaine-seeking behavior and an impaired extinction of this behavior. These behavioral phenomena were associated with a time-dependent reduction in mGluR1/5 expression within ventromedial PFC (vmPFC) of cocaine-experienced animals exposed to extinction testing but not in untested ones. Interestingly, pharmacological manipulations of vmPFC mGluR1/5 produced no immediate effects on cue-induced cocaine-seeking behavior but produced residual effects on a subsequent test for cocaine seeking. At 3 d withdrawal, cocaine-experienced rats infused intra-vmPFC with mGluR1/5 antagonists, either before or after an initial test for cocaine seeking, persisted in their cocaine seeking akin to cocaine-experienced rats in protracted withdrawal. Conversely, cocaine-experienced rats infused with an mGluR1/5 agonist before the initial test for cocaine-seeking at 30 d withdrawal exhibited a facilitation of extinction learning. These data indicate that cue-elicited deficits in vmPFC group 1 mGluR function mediate resistance to extinction during protracted withdrawal from a history of extensive cocaine self-administration and pose pharmacological stimulation of these receptors as a potential approach to facilitate learned suppression of drug-seeking behavior that may aid drug abstinence.


Subject(s)
Cocaine-Related Disorders/physiopathology , Extinction, Psychological/physiology , Prefrontal Cortex/physiology , Receptors, Metabotropic Glutamate/physiology , Substance Withdrawal Syndrome/physiopathology , Animals , Blotting, Western , Cocaine-Related Disorders/psychology , Conditioning, Operant , Cues , Excitatory Amino Acid Antagonists/pharmacology , Extinction, Psychological/drug effects , Injections , Male , Methoxyhydroxyphenylglycol/analogs & derivatives , Methoxyhydroxyphenylglycol/pharmacology , Prefrontal Cortex/drug effects , Pyridines/administration & dosage , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/biosynthesis , Receptors, Metabotropic Glutamate/drug effects , Recurrence , Self Administration , Substance Withdrawal Syndrome/psychology
18.
Mol Pharmacol ; 86(6): 774-85, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25326002

ABSTRACT

Although G protein-coupled receptors are primarily known for converting extracellular signals into intracellular responses, some receptors, such as the group 1 metabotropic glutamate receptor, mGlu5, are also localized on intracellular membranes where they can mediate both overlapping and unique signaling effects. Thus, besides "ligand bias," whereby a receptor's signaling modality can shift from G protein dependence to independence, canonical mGlu5 receptor signaling can also be influenced by "location bias" (i.e., the particular membrane and/or cell type from which it signals). Because mGlu5 receptors play important roles in both normal development and in disorders such as Fragile X syndrome, autism, epilepsy, addiction, anxiety, schizophrenia, pain, dyskinesias, and melanoma, a large number of drugs are being developed to allosterically target this receptor. Therefore, it is critical to understand how such drugs might be affecting mGlu5 receptor function on different membranes and in different brain regions. Further elucidation of the site(s) of action of these drugs may determine which signal pathways mediate therapeutic efficacy.


Subject(s)
Receptor, Metabotropic Glutamate 5/physiology , Receptors, Metabotropic Glutamate/physiology , Signal Transduction/physiology , Animals , Arrestins/physiology , Calcium/metabolism , Humans , Phosphorylation , Receptor, Metabotropic Glutamate 5/analysis , Receptor, Metabotropic Glutamate 5/chemistry , Receptor, Metabotropic Glutamate 5/drug effects , Receptors, Metabotropic Glutamate/analysis , Receptors, Metabotropic Glutamate/chemistry , Receptors, Metabotropic Glutamate/drug effects , beta-Arrestins
19.
Mov Disord ; 29(6): 715-9, 2014 May.
Article in English | MEDLINE | ID: mdl-24591264

ABSTRACT

Levodopa-induced dyskinesias (LIDs) represent one major motor disability of Parkinson's disease (PD) therapy. Thus, research effort is still devoted to finding agents that may improve parkinsonism and concomitantly reduce or avoid dyskinesia. Rodent and nonhuman primate models provide useful tools to study the molecular and neuronal bases of LIDs. Among the various strategies investigated recently, the use of drugs targeting metabotropic glutamate receptors has received large attention. In particular, use of antagonists of the subtype 5 of metabotropic glutamate receptors revealed promising preclinical and clinical results.


Subject(s)
Antiparkinson Agents/adverse effects , Dyskinesia, Drug-Induced/etiology , Dyskinesia, Drug-Induced/therapy , Levodopa/adverse effects , Receptors, Metabotropic Glutamate/metabolism , Animals , Disease Models, Animal , Humans , Parkinson Disease/drug therapy , Receptors, Metabotropic Glutamate/drug effects
20.
Neurochem Res ; 39(10): 1876-94, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25146900

ABSTRACT

Glutamate, the primary excitatory neurotransmitter in the central nervous system (CNS), exerts neuromodulatory actions via the activation of metabotropic glutamate (mGlu) receptors. There are eight known mGlu receptor subtypes (mGlu1-8), which are widely expressed throughout the brain, and are divided into three groups (I-III), based on signalling pathways and pharmacological profiles. Group III mGlu receptors (mGlu4/6/7/8) are primarily, although not exclusively, localised on presynaptic terminals, where they act as both auto- and hetero-receptors, inhibiting the release of neurotransmitter. Until recently, our understanding of the role of individual group III mGlu receptor subtypes was hindered by a lack of subtype-selective pharmacological tools. Recent advances in the development of both orthosteric and allosteric group III-targeting compounds, however, have prompted detailed investigations into the possible functional role of these receptors within the CNS, and revealed their involvement in a number of pathological conditions, such as epilepsy, anxiety and Parkinson's disease. The heterogeneous expression of group III mGlu receptor subtypes throughout the brain, as well as their distinct distribution at glutamatergic and GABAergic synapses, makes them ideal targets for therapeutic intervention. This review summarises the advances in subtype-selective pharmacology, and discusses the individual roles of group III mGlu receptors in physiology, and their potential involvement in disease.


Subject(s)
Receptors, Metabotropic Glutamate/drug effects , Allosteric Site , Hippocampus/physiology , Humans , Ligands , Nervous System Diseases/physiopathology , Neuronal Plasticity/physiology , Protein Conformation , Receptors, Metabotropic Glutamate/chemistry , Receptors, Metabotropic Glutamate/physiology , Synaptic Transmission/physiology
SELECTION OF CITATIONS
SEARCH DETAIL