Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
ACS Chem Neurosci ; 12(1): 79-98, 2021 01 06.
Article in English | MEDLINE | ID: mdl-33326224

ABSTRACT

N-Methyl-d-aspartate receptors (NMDARs) are ionotropic ligand-gated glutamate receptors that mediate fast excitatory synaptic transmission in the central nervous system (CNS). Several neurological disorders may involve NMDAR hypofunction, which has driven therapeutic interest in positive allosteric modulators (PAMs) of NMDAR function. Here we describe modest changes to the tetrahydroisoquinoline scaffold of GluN2C/GluN2D-selective PAMs that expands activity to include GluN2A- and GluN2B-containing recombinant and synaptic NMDARs. These new analogues are distinct from GluN2C/GluN2D-selective compounds like (+)-(3-chlorophenyl)(6,7-dimethoxy-1-((4-methoxyphenoxy)methyl)-3,4-dihydroisoquinolin-2(1H)-yl)methanone (CIQ) by virtue of their subunit selectivity, molecular determinants of action, and allosteric regulation of agonist potency. The (S)-enantiomers of two analogues (EU1180-55, EU1180-154) showed activity at NMDARs containing all subunits (GluN2A, GluN2B, GluN2C, GluN2D), whereas the (R)-enantiomers were primarily active at GluN2C- and GluN2D-containing NMDARs. Determination of the actions of enantiomers on triheteromeric receptors confirms their unique pharmacology, with greater activity of (S) enantiomers at GluN2A/GluN2D and GluN2B/GluN2D subunit combinations than (R) enantiomers. Evaluation of the (S)-EU1180-55 and EU1180-154 response of chimeric kainate/NMDA receptors revealed structural determinants of action within the pore-forming region and associated linkers. Scanning mutagenesis identified structural determinants within the GluN1 pre-M1 and M1 regions that alter the activity of (S)-EU1180-55 but not (R)-EU1180-55. By contrast, mutations in pre-M1 and M1 regions of GluN2D perturb the actions of only the (R)-EU1180-55 but not the (S) enantiomer. Molecular modeling supports the idea that the (S) and (R) enantiomers interact distinctly with GluN1 and GluN2 pre-M1 regions, suggesting that two distinct sites exist for these NMDAR PAMs, each of which has different functional effects.


Subject(s)
Receptors, N-Methyl-D-Aspartate , Synaptic Transmission , Allosteric Regulation , Models, Molecular , Receptors, N-Methyl-D-Aspartate/metabolism
2.
J Gen Physiol ; 152(4)2020 04 06.
Article in English | MEDLINE | ID: mdl-32221541

ABSTRACT

The NMDA receptor (NMDAR) is an ionotropic glutamate receptor formed from the tetrameric assembly of GluN1 and GluN2 subunits. Within the flexible linker between the agonist binding domain (ABD) and the M1 helix of the pore-forming transmembrane helical bundle lies a two-turn, extracellular pre-M1 helix positioned parallel to the plasma membrane and in van der Waals contact with the M3 helix thought to constitute the channel gate. The pre-M1 helix is tethered to the bilobed ABD, where agonist-induced conformational changes initiate activation. Additionally, it is a locus for de novo mutations associated with neurological disorders, is near other disease-associated de novo sites within the transmembrane domain, and is a structural determinant of subunit-selective modulators. To investigate the role of the pre-M1 helix in channel gating, we performed scanning mutagenesis across the GluN2A pre-M1 helix and recorded whole-cell macroscopic and single channel currents from HEK293 cell-attached patches. We identified two residues at which mutations perturb channel open probability, the mean open time, and the glutamate deactivation time course. We identified a subunit-specific network of aromatic amino acids located in and around the GluN2A pre-M1 helix to be important for gating. Based on these results, we are able to hypothesize about the role of the pre-M1 helix in other NMDAR subunits based on sequence and structure homology. Our results emphasize the role of the pre-M1 helix in channel gating, implicate the surrounding amino acid environment in this mechanism, and suggest unique subunit-specific contributions of pre-M1 helices to GluN1 and GluN2 gating.


Subject(s)
Ion Channel Gating/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Amino Acid Sequence , Amino Acids/metabolism , Animals , Binding Sites/physiology , Cell Line , HEK293 Cells , Humans , Protein Domains/physiology , Protein Subunits/metabolism , Xenopus
3.
J Physiol ; 596(17): 4057-4089, 2018 09.
Article in English | MEDLINE | ID: mdl-29917241

ABSTRACT

KEY POINTS: The kinetics of NMDA receptor (NMDAR) signalling are a critical aspect of the physiology of excitatory synaptic transmission in the brain. Here we develop a mechanistic description of NMDAR function based on the receptor tetrameric structure and the principle that each agonist-bound subunit must undergo some rate-limiting conformational change after agonist binding, prior to channel opening. By fitting this mechanism to single channel data using a new MATLAB-based software implementation of maximum likelihood fitting with correction for limited time resolution, rate constants were derived for this mechanism that reflect distinct structural changes and predict the properties of macroscopic and synaptic NMDAR currents. The principles applied here to develop a mechanistic description of the heterotetrameric NMDAR, and the software used in this analysis, can be equally applied to other heterotetrameric glutamate receptors, providing a unifying mechanistic framework to understanding the physiology of glutamate receptor signalling in the brain. ABSTRACT: NMDA receptors (NMDARs) are tetrameric complexes comprising two glycine-binding GluN1 and two glutamate-binding GluN2 subunits. Four GluN2 subunits encoded by different genes can produce up to 10 different di- and triheteromeric receptors. In addition, some neurological patients contain a de novo mutation or inherited rare variant in only one subunit. There is currently no mechanistic framework to describe tetrameric receptor function that can be extended to receptors with two different GluN1 or GluN2 subunits. Here we use the structural features of glutamate receptors to develop a mechanism describing both single channel and macroscopic NMDAR currents. We propose that each agonist-bound subunit undergoes some rate-limiting conformational change after agonist binding, prior to channel opening. We hypothesize that this conformational change occurs within a triad of interactions between a short helix preceding the M1 transmembrane helix, the highly conserved M3 motif encoded by the residues SYTANLAAF, and the linker preceding the M4 transmembrane helix of the adjacent subunit. Molecular dynamics simulations suggest that pre-M1 helix motion is uncorrelated between subunits, which we interpret to suggest independent subunit-specific conformational changes may influence these pre-gating steps. According to this interpretation, these conformational changes are the main determinants of the key kinetic properties of NMDA receptor activation following agonist binding, and so these steps sculpt their physiological role. We show that this structurally derived tetrameric model describes both single channel and macroscopic data, giving a new approach to interpreting functional properties of synaptic NMDARs that provides a logical framework to understanding receptors with non-identical subunits.


Subject(s)
Glutamic Acid/metabolism , Ion Channel Gating , Receptors, N-Methyl-D-Aspartate/chemistry , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptic Transmission , HEK293 Cells , Humans , Molecular Dynamics Simulation , Protein Conformation , Protein Multimerization , Protein Subunits
4.
PLoS Genet ; 13(1): e1006536, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28095420

ABSTRACT

N-methyl-D-aspartate receptors (NMDARs), ligand-gated ionotropic glutamate receptors, play key roles in normal brain development and various neurological disorders. Here we use standing variation data from the human population to assess which protein domains within NMDAR GluN1, GluN2A and GluN2B subunits show the strongest signal for being depleted of missense variants. We find that this includes the GluN2 pre-M1 helix and linker between the agonist-binding domain (ABD) and first transmembrane domain (M1). We then evaluate the functional changes of multiple missense mutations in the NMDAR pre-M1 helix found in children with epilepsy and developmental delay. We find mutant GluN1/GluN2A receptors exhibit prolonged glutamate response time course for channels containing 1 or 2 GluN2A-P552R subunits, and a slow rise time only for receptors with 2 mutant subunits, suggesting rearrangement of one GluN2A pre-M1 helix is sufficient for rapid activation. GluN2A-P552R and analogous mutations in other GluN subunits increased the agonist potency and slowed response time course, suggesting a functionally conserved role for this residue. Although there is no detectable change in surface expression or open probability for GluN2A-P552R, the prolonged response time course for receptors that contained GluN2A-P552R increased charge transfer for synaptic-like activation, which should promote excitotoxic damage. Transfection of cultured neurons with GluN2A-P552R prolonged EPSPs, and triggered pronounced dendritic swelling in addition to excitotoxicity, which were both attenuated by memantine. These data implicate the pre-M1 region in gating, provide insight into how different subunits contribute to gating, and suggest that mutations in the pre-M1 helix can compromise neuronal health. Evaluation of FDA-approved NMDAR inhibitors on the mutant NMDAR-mediated current response and neuronal damage provides a potential clinical path to treat individuals harboring similar mutations in NMDARs.


Subject(s)
Ion Channel Gating , Mutation, Missense , Nerve Tissue Proteins/metabolism , Nervous System Diseases/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Cells, Cultured , Excitatory Amino Acid Antagonists/pharmacology , Glutamic Acid/metabolism , HEK293 Cells , Humans , Memantine/pharmacology , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Neurons/metabolism , Neurons/physiology , Protein Domains , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/chemistry , Receptors, N-Methyl-D-Aspartate/genetics , Xenopus
5.
Mol Pharmacol ; 90(6): 689-702, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27625038

ABSTRACT

N-methyl-d-aspartate receptors (NMDARs) are ionotropic glutamatergic receptors that have been implicated in learning, development, and neuropathological conditions. They are typically composed of GluN1 and GluN2A-D subunits. Whereas a great deal is known about the role of GluN2A- and GluN2B-containing NMDARs, much less is known about GluN2D-containing NMDARs. Here we explore the subunit composition of synaptic NMDARs on hippocampal interneurons. GluN2D mRNA was detected by single-cell PCR and in situ hybridization in diverse interneuron subtypes in the CA1 region of the hippocampus. The GluN2D subunit was detectable by immunoblotting and immunohistochemistry in all subfields of the hippocampus in young and adult mice. In whole-cell patch-clamp recordings from acute hippocampal slices, (+)-CIQ, the active enantiomer of the positive allosteric modulator CIQ, significantly enhanced the amplitude of the NMDAR component of miniature excitatory postsynaptic currents (mEPSCs) in CA1 interneurons but not in pyramidal cells. (+)-CIQ had no effect in slices from Grin2d-/- mice, suggesting that GluN2D-containing NMDARs participate in excitatory synaptic transmission onto hippocampal interneurons. The time course of the NMDAR component of the mEPSC was unaffected by (+)-CIQ potentiation and was not accelerated in slices from Grin2d-/- mice compared with wild-type, suggesting that GluN2D does not detectably slow the NMDAR EPSC time course at this age. (+)-CIQ increased the activity of CA1 interneurons as detected by the rate and net charge transfer of spontaneous inhibitory postsynaptic currents (sIPSCs) recorded from CA1 pyramidal cells. These data provide evidence that interneurons contain synaptic NMDARs possessing a GluN2D subunit, which can influence interneuron function and signal processing.


Subject(s)
Hippocampus/cytology , Interneurons/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synaptic Transmission , Allosteric Regulation/drug effects , Animals , CA1 Region, Hippocampal/drug effects , CA1 Region, Hippocampal/metabolism , Excitatory Postsynaptic Potentials/drug effects , Interneurons/drug effects , Ion Channel Gating/drug effects , Isoquinolines/pharmacology , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Real-Time Polymerase Chain Reaction , Receptors, N-Methyl-D-Aspartate/genetics , Stereoisomerism , Synaptic Transmission/drug effects , Time Factors , Xenopus laevis
6.
Neuropharmacology ; 100: 2-16, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26231830

ABSTRACT

Mutations in hundreds of genes contribute to cognitive and behavioral dysfunction associated with developmental brain disorders (DBDs). Due to the sheer number of risk factors available for study combined with the cost of developing new animal models, it remains an open question how genes should be prioritized for in-depth neurobiological investigations. Recent reviews have argued that priority should be given to frequently mutated genes commonly found in sporadic DBD patients. Intrigued by this idea, we explored to what extent "high priority" risk factors have been studied in animals in an effort to assess their potential for generating valuable preclinical models capable of advancing the neurobiological understanding of DBDs. We found that in-depth whole animal studies are lacking for many high priority genes, with relatively few neurobiological studies performed in construct valid animal models aimed at understanding the pathological substrates associated with disease phenotypes. However, some high priority risk factors have been extensively studied in animal models and they have generated novel insights into DBD patho-neurobiology while also advancing early pre-clinical therapeutic treatment strategies. We suggest that prioritizing model development toward genes frequently mutated in non-specific DBD populations will accelerate the understanding of DBD patho-neurobiology and drive novel therapeutic strategies. This article is part of the Special Issue entitled 'Synaptopathy--from Biology to Therapy'.


Subject(s)
Brain Diseases/genetics , Disease Models, Animal , Neurodevelopmental Disorders/genetics , Synapses/genetics , Animals , Humans , Mice , Mice, Knockout , Mutation , Phenotype , Research Design , Risk Factors
8.
Neuron ; 81(5): 1084-1096, 2014 Mar 05.
Article in English | MEDLINE | ID: mdl-24607230

ABSTRACT

NMDA receptors are tetrameric ligand-gated ion channels comprised of GluN1, GluN2, and GluN3 subunits. Two different GluN2 subunits have been identified in most NMDA receptor-expressing cells, and the majority of native receptors are triheteromers containing two GluN1 and two different GluN2. In contrast to diheteromeric NMDA receptors, little is known about the function of triheteromers. We developed a method to provide selective cell-surface expression of recombinant GluN1/GluN2A/GluN2B triheteromers and compared properties of these receptors with those of GluN1/GluN2A and GluN1/GluN2B diheteromers. We show that glutamate deactivation of triheteromers is distinct from those of GluN1/GluN2A and GluN1/GluN2B and reveal modulation of triheteromers by subunit-selective antagonists ifenprodil, CP-101,606, TCN-201, and extracellular Zn(2+). Furthermore, kinetic measurements suggest variation in the ifenprodil binding site of triheteromers compared to GluN1/GluN2B diheteromers. This work provides insight into the distinct properties of GluN1/GluN2A/GluN2B triheteromers, which are presumably the most abundant NMDA receptors in the adult forebrain.


Subject(s)
Piperidines/pharmacology , Receptors, N-Methyl-D-Aspartate/physiology , Animals , Dimerization , Excitatory Amino Acid Antagonists/metabolism , Excitatory Amino Acid Antagonists/pharmacology , Glutamic Acid/physiology , HEK293 Cells , Humans , Oocytes/cytology , Patch-Clamp Techniques , Piperidines/metabolism , Protein Binding/physiology , Rats , Receptors, N-Methyl-D-Aspartate/chemistry , Receptors, N-Methyl-D-Aspartate/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Sulfonamides/pharmacology , Trace Elements/pharmacology , Xenopus laevis , Zinc/pharmacology
9.
Neuropsychopharmacology ; 39(3): 625-37, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24008353

ABSTRACT

NMDA receptors are glutamate receptor ion channels that contribute to synaptic plasticity and are important for many forms of learning and memory. In the amygdala, NMDA receptors are critical for the acquisition, retention, and extinction of classically conditioned fear responses. Although the GluN2B subunit has been implicated in both the acquisition and extinction of conditioned fear, GluN2C-knockout mice show reduced conditioned fear responses. Moreover, D-cycloserine (DCS), which facilitates fear extinction, selectively enhances the activity of GluN2C-containing NMDA receptors. To further define the contribution of GluN2C receptors to fear learning, we infused the GluN2C/GluN2D-selective potentiator CIQ bilaterally into the basolateral amygdala (3, 10, or 30 µg/side) following either fear conditioning or fear extinction training. CIQ both increased the expression of conditioned fear 24 h later and enhanced the extinction of the previously conditioned fear response. These results support a critical role for GluN2C receptors in the amygdala in the consolidation of learned fear responses and suggest that increased activity of GluN2C receptors may underlie the therapeutic actions of DCS.


Subject(s)
Amygdala/metabolism , Conditioning, Psychological/physiology , Extinction, Psychological/physiology , Fear/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Retention, Psychology/physiology , Amygdala/drug effects , Animals , Area Under Curve , Benzimidazoles/pharmacology , Conditioning, Psychological/drug effects , Dose-Response Relationship, Drug , Extinction, Psychological/drug effects , Fear/drug effects , Gene Expression Regulation/drug effects , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Microinjections , Motor Activity/drug effects , Oocytes , Receptors, N-Methyl-D-Aspartate/genetics , Retention, Psychology/drug effects , Xenopus
10.
J Med Chem ; 56(13): 5351-81, 2013 Jul 11.
Article in English | MEDLINE | ID: mdl-23627311

ABSTRACT

We describe here the synthesis and evaluation of a series of tetrahydroisoquinolines that show subunit-selective potentiation of NMDA receptors containing the GluN2C or GluN2D subunits. Bischler-Napieralski conditions were employed in the key step for the conversion of acyclic amides to the corresponding tetrahydroisoquinoline-containing analogs. Compounds were evaluated using both two-electrode voltage clamp recordings from Xenopus laevis oocytes and imaging of mammalian BHK cells loaded with Ca(2+)-sensitive dyes. The most potent analogues had EC50 values of 300 nM and showed over 2-fold potentiation of the response to maximally effective concentrations of glutamate and glycine but had no effect on responses from NMDA receptors containing the GluN2A or GluN2B subunits AMPA, kainate, and GABA or glycine receptors or a variety of other potential targets. These compounds represent a potent class of small molecule subunit-selective potentiators of NMDA receptors.


Subject(s)
Excitatory Amino Acid Agonists/pharmacology , Receptors, N-Methyl-D-Aspartate/agonists , Tetrahydroisoquinolines/pharmacology , Animals , Cell Line , Excitatory Amino Acid Agonists/chemical synthesis , Female , Glutamic Acid/pharmacology , Glycine/pharmacology , Membrane Potentials/drug effects , Models, Chemical , Molecular Structure , Oocytes/drug effects , Oocytes/metabolism , Oocytes/physiology , Protein Subunits/agonists , Protein Subunits/genetics , Protein Subunits/metabolism , Rats , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Structure-Activity Relationship , Tetrahydroisoquinolines/chemical synthesis , Tetrahydroisoquinolines/chemistry , Xenopus laevis
11.
Mol Pharmacol ; 84(1): 114-27, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23625947

ABSTRACT

N-methyl-d-aspartate (NMDA) receptors are ligand-gated ion channels assembled from GluN1 and GluN2 subunits. We used a series of N-hydroxypyrazole-5-glycine (NHP5G) partial agonists at the GluN2 glutamate binding site as tools to study activation of GluN1/GluN2A and GluN1/GluN2D NMDA receptor subtypes. Using two-electrode voltage-clamp electrophysiology, fast-application patch-clamp, and single-channel recordings, we show that propyl- and ethyl-substituted NHP5G agonists have a broad range of agonist efficacies relative to the full agonist glutamate (<1-72%). Crystal structures of the agonist binding domains (ABDs) of GluN2A and GluN2D do not reveal any differences in the overall domain conformation induced by binding of the full agonist glutamate or the partial agonist propyl-NHP5G, which is strikingly different from ABD structures of 2-amino-3-(3-hydroxy-5-methylisoxazol-4-yl)propanoate (AMPA) and kainate receptors bound to full and partial agonists. Subsequent evaluation of relative NHP5G agonist efficacy at GluN2A-GluN2D chimeric subunits implicates the amino-terminal domain (ATD) as a strong determinant of agonist efficacy, suggesting that interdomain interactions between the ABD and the ATD may be a central element in controlling the manner by which agonist binding leads to channel opening. We propose that variation in the overall receptor conformation, which is strongly influenced by the nature of interdomain interactions in resting and active states, mediates differences in agonist efficacy and partial agonism at the GluN2 subunits.


Subject(s)
Excitatory Amino Acid Agonists/pharmacology , Glutamic Acid/chemistry , Glutamic Acid/metabolism , Receptors, N-Methyl-D-Aspartate/chemistry , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Binding Sites , Cells, Cultured , Female , Glycine/pharmacology , HEK293 Cells , Humans , Protein Structure, Tertiary , Protein Subunits/chemistry , Protein Subunits/metabolism , Receptors, Kainic Acid/metabolism , Xenopus laevis/metabolism , alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/pharmacology
12.
Mol Pharmacol ; 83(5): 1045-56, 2013 May.
Article in English | MEDLINE | ID: mdl-23455314

ABSTRACT

N-methyl-D-aspartate (NMDA) receptors are glutamate-gated ion channels whose function is critical for normal excitatory synaptic transmission in the brain and whose dysfunction has been implicated in several neurologic conditions. NMDA receptor function is subject to extensive allosteric regulation both by endogenous compounds and by exogenous small molecules. Elucidating the structural determinants and mechanism of action by which allosteric regulators control gating will enhance our understanding of NMDA receptor activation and facilitate the development of novel therapeutics. Here, we investigated the structural determinants for (3-chlorophenyl)(6,7-dimethoxy-1-((4-methoxyphenoxy)methyl)-3,4-dihydroisoquinolin-2(1H)-yl)methanone (CIQ), a GluN2C/2D-selective positive allosteric modulator. We show that CIQ does not bind to the amino-terminal domain of the NMDA receptor and does not share structural determinants with modulators acting at the agonist-binding domain dimer interface or ion channel pore. Rather, we identified critical determinants of CIQ modulation in the region near the first transmembrane helix of GluN2D, including in a putative pre-M1 cuff helix that may influence channel gating. We also show that mutations within the GluN2D pre-M1 region alter open probability of the NMDA receptor. These results suggest a novel site of action for potentiation of NMDA receptors by small molecules and implicate the pre-M1 region in NMDA receptor gating.


Subject(s)
Ion Channel Gating/physiology , Receptors, N-Methyl-D-Aspartate/metabolism , Allosteric Regulation , Amino Acid Sequence , Animals , Binding Sites , Cell Line , Cell Membrane/metabolism , Diffusion , Glutamic Acid/metabolism , HEK293 Cells , Humans , Ion Channels/metabolism , Molecular Sequence Data , Oocytes/metabolism , Protein Structure, Secondary , Xenopus laevis
13.
J Neurosci ; 32(18): 6197-208, 2012 May 02.
Article in English | MEDLINE | ID: mdl-22553026

ABSTRACT

NMDA receptors are ligand-gated ion channels that mediate excitatory neurotransmission in the brain and are involved in numerous neuropathological conditions. NMDA receptors are activated upon simultaneous binding of coagonists glycine and glutamate to the GluN1 and GluN2 subunits, respectively. Subunit-selective modulation of NMDA receptor function by ligand binding to modulatory sites distinct from the agonist binding sites could allow pharmacological intervention with therapeutically beneficial mechanisms. Here, we show the mechanism of action for 3-chloro-4-fluoro-N-[(4-[(2-(phenylcarbonyl)hydrazino)carbonyl]phenyl)methyl]-benzenesulfonamide (TCN-201), a new GluN1/GluN2A-selective NMDA receptor antagonist whose inhibition can be surmounted by glycine. Electrophysiological recordings from chimeric and mutant rat NMDA receptors suggest that TCN-201 binds to a novel allosteric site located at the dimer interface between the GluN1 and GluN2 agonist binding domains. Furthermore, we demonstrate that occupancy of this site by TCN-201 inhibits NMDA receptor function by reducing glycine potency. TCN-201 is therefore a negative allosteric modulator of glycine binding.


Subject(s)
Glycine/chemistry , Glycine/metabolism , Receptors, N-Methyl-D-Aspartate/chemistry , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Binding Sites , HEK293 Cells , Humans , Protein Binding , Protein Subunits , Rats
14.
Trends Pharmacol Sci ; 32(12): 726-33, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21996280

ABSTRACT

N-Methyl-D-aspartate (NMDA) receptors are tetrameric ion channels containing two of four possible GluN2 subunits. These receptors have been implicated for decades in neurological diseases such as stroke, traumatic brain injury, dementia and schizophrenia. The GluN2 subunits substantially contribute to functional diversity of NMDA receptors and are distinctly expressed during development and among brain regions. Thus, subunit-selective antagonists and modulators that differentially target the GluN2 subunit might provide an opportunity to pharmacologically modify the function of select groups of neurons for therapeutic gain. A flurry of clinical, functional and chemical studies have together reinvigorated efforts to identify subunit-selective modulators of NMDA receptor function, resulting in a handful of new compounds that appear to act at novel sites. Here, we review the properties of new emerging classes of subunit-selective NMDA receptor modulators, which we predict will mark the beginning of a productive period of progress for NMDA receptor pharmacology.


Subject(s)
Membrane Transport Modulators/pharmacology , Receptors, N-Methyl-D-Aspartate/agonists , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Allosteric Regulation/drug effects , Animals , Binding Sites , Humans , Ligands , Membrane Transport Modulators/chemistry , Membrane Transport Modulators/metabolism , Membrane Transport Modulators/therapeutic use , Protein Conformation , Protein Isoforms/agonists , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Subunits/agonists , Protein Subunits/antagonists & inhibitors , Protein Subunits/chemistry , Protein Subunits/metabolism , Receptors, N-Methyl-D-Aspartate/chemistry , Receptors, N-Methyl-D-Aspartate/metabolism
15.
Mol Pharmacol ; 80(5): 782-95, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21807990

ABSTRACT

The compound 4-(5-(4-bromophenyl)-3-(6-methyl-2-oxo-4-phenyl-1,2-dihydroquinolin-3-yl)-4,5-dihydro-1H-pyrazol-1-yl)-4-oxobutanoic acid (DQP-1105) is a representative member of a new class of N-methyl-d-aspartate (NMDA) receptor antagonists. DQP-1105 inhibited GluN2C- and GluN2D-containing receptors with IC(50) values that were at least 50-fold lower than those for recombinant GluN2A-, GluN2B-, GluA1-, or GluK2-containing receptors. Inhibition was voltage-independent and could not be surmounted by increasing concentrations of either coagonist, glutamate or glycine, consistent with a noncompetitive mechanism of action. DQP-1105 inhibited single-channel currents in excised outside-out patches without significantly changing mean open time or single-channel conductance, suggesting that DQP inhibits a pregating step without changing the stability of the open pore conformation and thus channel closing rate. Evaluation of DQP-1105 inhibition of chimeric NMDA receptors identified two key residues in the lower lobe of the GluN2 agonist binding domain that control the selectivity of DQP-1105. These data suggest a mechanism for this new class of inhibitors and demonstrate that ligands can access, in a subunit-selective manner, a new site located in the lower, membrane-proximal portion of the agonist-binding domain.


Subject(s)
Excitatory Amino Acid Antagonists/pharmacology , Glutamic Acid/metabolism , Pyrazoles/pharmacology , Quinolones/pharmacology , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Animals , Cells, Cultured , Cricetinae , DNA, Complementary , Excitatory Amino Acid Antagonists/chemistry , Humans , Patch-Clamp Techniques , Pyrazoles/chemistry , Quinolones/chemistry , Rats , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Structure-Activity Relationship
16.
Nat Commun ; 1: 90, 2010 Oct 05.
Article in English | MEDLINE | ID: mdl-20981015

ABSTRACT

NMDA receptors are tetrameric complexes of NR1 and NR2A-D subunits that mediate excitatory synaptic transmission and have a role in neurological disorders. In this article, we identify a novel subunit-selective potentiator of NMDA receptors containing the NR2C or NR2D subunit, which could allow selective modification of circuit function in regions expressing NR2C/D subunits. The substituted tetrahydroisoquinoline CIQ (3-chlorophenyl)(6,7-dimethoxy-1-((4-methoxyphenoxy)methyl)-3,4-dihydroisoquinolin-2(1H)-yl)methanone) enhances receptor responses two-fold with an EC(50) of 3 µM by increasing channel opening frequency without altering mean open time or EC(50) values for glutamate or glycine. The actions of CIQ depend on a single residue in the M1 region (NR2D Thr592) and on the linker between the N-terminal domain and agonist binding domain. CIQ potentiates native NR2D-containing NMDA receptor currents from subthalamic neurons. Our identification of a subunit-selective NMDA receptor modulator reveals a new class of pharmacological tools with which to probe the role of NR2C- and NR2D-containing NMDA receptors in brain function and disease.


Subject(s)
Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Glutamic Acid/metabolism , Glycine/metabolism , HEK293 Cells , Humans , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley
17.
Pharmacol Rev ; 62(3): 405-96, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20716669

ABSTRACT

The mammalian ionotropic glutamate receptor family encodes 18 gene products that coassemble to form ligand-gated ion channels containing an agonist recognition site, a transmembrane ion permeation pathway, and gating elements that couple agonist-induced conformational changes to the opening or closing of the permeation pore. Glutamate receptors mediate fast excitatory synaptic transmission in the central nervous system and are localized on neuronal and non-neuronal cells. These receptors regulate a broad spectrum of processes in the brain, spinal cord, retina, and peripheral nervous system. Glutamate receptors are postulated to play important roles in numerous neurological diseases and have attracted intense scrutiny. The description of glutamate receptor structure, including its transmembrane elements, reveals a complex assembly of multiple semiautonomous extracellular domains linked to a pore-forming element with striking resemblance to an inverted potassium channel. In this review we discuss International Union of Basic and Clinical Pharmacology glutamate receptor nomenclature, structure, assembly, accessory subunits, interacting proteins, gene expression and translation, post-translational modifications, agonist and antagonist pharmacology, allosteric modulation, mechanisms of gating and permeation, roles in normal physiological function, as well as the potential therapeutic use of pharmacological agents acting at glutamate receptors.


Subject(s)
Ion Channels/physiology , Receptors, Glutamate/physiology , Gene Expression , Humans , Ion Channels/chemistry , Ion Channels/genetics , Protein Processing, Post-Translational , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/physiology , Receptors, Glutamate/chemistry , Receptors, Glutamate/genetics , Synaptic Transmission , Terminology as Topic
18.
J Pharmacol Exp Ther ; 333(3): 650-62, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20197375

ABSTRACT

N-Methyl-D-aspartate (NMDA) receptors are ligand-gated ion channels that mediate a slow, Ca(2+)-permeable component of excitatory synaptic transmission in the central nervous system and play a pivotal role in synaptic plasticity, neuronal development, and several neurological diseases. We describe a fluorescence-based assay that measures NMDA receptor-mediated changes in intracellular calcium in a BHK-21 cell line stably expressing NMDA receptor NR2D with NR1 under the control of a tetracycline-inducible promoter (Tet-On). The assay selectively identifies allosteric modulators by using supramaximal concentrations of glutamate and glycine to minimize detection of competitive antagonists. The assay is validated by successfully identifying known noncompetitive, but not competitive NMDA receptor antagonists among 1800 screened compounds from two small focused libraries, including the commercially available library of pharmacologically active compounds. Hits from the primary screen are validated through a secondary screen that used two-electrode voltage-clamp recordings on recombinant NMDA receptors expressed in Xenopus laevis oocytes. This strategy identified several novel modulators of NMDA receptor function, including the histamine H3 receptor antagonists clobenpropit and iodophenpropit, as well as the vanilloid receptor transient receptor potential cation channel, subfamily V, member 1 (TRPV1) antagonist capsazepine. These compounds are noncompetitive antagonists and the histamine H3 receptor ligand showed submicromolar potency at NR1/NR2B NMDA receptors, which raises the possibility that compounds can be developed that act with high potency on both glutamate and histamine receptor systems simultaneously. Furthermore, it is possible that some actions attributed to histamine H3 receptor inhibition in vivo may also involve NMDA receptor antagonism.


Subject(s)
Histamine H3 Antagonists/pharmacology , Imidazoles/pharmacology , Isothiuronium/analogs & derivatives , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Thiourea/analogs & derivatives , Aniline Compounds , Animals , Cell Line , Cricetinae , Drug Evaluation, Preclinical , Electrophysiology , Excitatory Amino Acid Antagonists/pharmacology , Fluorescent Dyes , Humans , Isothiuronium/pharmacology , Microscopy, Fluorescence , Oocytes/drug effects , Patch-Clamp Techniques , Piperidines/pharmacology , Radioligand Assay , Receptors, N-Methyl-D-Aspartate/biosynthesis , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/genetics , Structure-Activity Relationship , Thiourea/pharmacology , Xanthenes , Xenopus laevis
19.
J Neurosci ; 29(39): 12045-58, 2009 Sep 30.
Article in English | MEDLINE | ID: mdl-19793963

ABSTRACT

NMDA receptors comprised of different NR2 subunits exhibit strikingly unique biophysical and pharmacological properties. Here, we report that the extracellular amino-terminal domain (ATD) of the NR2 subunit controls pharmacological and kinetic properties of recombinant NMDA receptors, such as agonist potency, deactivation time course, open probability (P(OPEN)), and mean open/shut duration. Using ATD deletion mutants of NR2A, NR2B, NR2C, NR2D, and chimeras of NR2A and NR2D with interchanged ATD [NR2A-(2D-ATD) and NR2D-(2A-ATD)], we show that the ATD contributes to the low glutamate potency of NR2A-containing NMDA receptors and the high glutamate potency of NR2D-containing receptors. The ATD influences the deactivation time courses of NMDA receptors, as removal of the ATD from NR2A slows the deactivation rate, while removal of the ATD from NR2B, NR2C and NR2D accelerates the deactivation rate. Open probability also is influenced by the ATD. Removal of the ATD from NR2A or replacement of the NR2A-ATD with that of NR2D decreases P(OPEN) in single-channel recordings from outside-out patches of HEK 293 cells. In contrast, deletion of the ATD from NR2D or replacement of the NR2D ATD with that of NR2A increases P(OPEN) and mean open duration. These data demonstrate the modular nature of NMDA receptors, and show that the ATD of the different NR2 subunits plays an important role in fine-tuning the functional properties of the individual NMDA receptor subtypes.


Subject(s)
Protein Subunits/chemistry , Protein Subunits/physiology , Receptors, N-Methyl-D-Aspartate/chemistry , Receptors, N-Methyl-D-Aspartate/physiology , Amino Acid Sequence/physiology , Animals , Cell Line , Female , Humans , Molecular Sequence Data , Protein Structure, Secondary/physiology , Protein Structure, Tertiary/physiology , Xenopus laevis
20.
J Pharmacol Exp Ther ; 323(2): 606-13, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17687070

ABSTRACT

5-Hydroxytryptamine (5-HT; serotonin) is a potent vasoconstrictor and smooth muscle mitogen. Substances that produce similar responses also stimulate production of superoxide. We sought to determine whether 5-HT stimulates production of superoxide. 5-HT can be metabolized by cytochrome P450 to nitric oxide (NO), which scavenges superoxide. Thus, we hypothesized that inhibiting cytochrome P450 would potentiate 5-HT-induced contraction and reveal 5-HT-stimulated superoxide. In isolated tissue bath experiments using endotheliumintact rat aorta, the cytochrome P450 inhibitor ketoconazole (KTZ; 1-50 microM) caused a maximum 8-fold leftward shift in the 5-HT concentration-response curve that was not observed when aorta were stimulated with phenylephrine or KCl. 5-HT did not stimulate concentration-dependent increases in superoxide levels as measured by a lucigenin-enhanced chemiluminescent superoxide assay. KTZ (10 microM) did not reveal 5-HT-stimulated superoxide. The NO inhibitor N(omega)-nitro-L-arginine (L-NNA) (100 microM) with or without KTZ (10 microM) potentiated 5-HT-induced contraction independently of NADPH oxidase-derived superoxide but also did not reveal 5-HT-stimulated superoxide. Metabolism of 5-HT to NO depends on catalase, but the catalase inhibitor 3-amino-1,2,4-triazole (50 mM) attenuated 5-HT-induced contraction. Removal of endothelium did not alter the effects of KTZ on 5-HT-induced contraction, and, in endothelium-intact aorta, KTZ did not decrease acetylcholine-induced relaxation. Unlike KTZ, the cytochrome P450 inhibitors 1-aminobenzotriazole (0.5 mM) and clotrimazole (10 microM) did not potentiate 5-HT-induced contraction. Moreover, 14,15-epoxyeicosa-5(Z)-enoic acid (10 microM), an epoxyeicosatrienoic acid antagonist, caused a small rightward shift in the 5-HT concentration-response curve. These data suggest KTZ acts by a potentially novel mechanism to potentiate 5-HT-induced contraction.


Subject(s)
Aorta, Thoracic/drug effects , Cytochrome P-450 Enzyme Inhibitors , Enzyme Inhibitors/pharmacology , Ketoconazole/pharmacology , Serotonin/pharmacology , Vasoconstriction/drug effects , Animals , Aorta, Thoracic/physiology , Catalase/physiology , Drug Synergism , Male , Nitric Oxide/metabolism , Nitric Oxide Synthase/physiology , Nitroarginine/pharmacology , Rats , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT2A/physiology , Serotonin/metabolism , Superoxides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...