Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 200
Filter
1.
bioRxiv ; 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39229143

ABSTRACT

Objective: GRIN1 -related neurodevelopmental disorder ( GRIN1 -NDD) is characterized by clinically significant variation in the GRIN1 gene, which encodes the obligatory GluN1 subunit of N-methyl-D-aspartate receptors (NMDARs). The identified p.Tyr647Ser (Y647S) variant - carried by a 33-year-old female with seizures and intellectual disability - is located in the M3 helix in the GluN1 transmembrane domain. This study builds upon initial in vitro investigations of the functional impacts of the GRIN1 Y647S variant and examines its in vivo consequences in a mouse model. Methods: To investigate in vitro functional impacts of NMDARs containing GluN1-Y647S variant subunits, GluN1-Y647S was co-expressed with wildtype GluN2A or GluN2B subunits in Xenopus laevis oocytes and HEK cells. Grin1 Y647S/+ mice were created by CRISPR-Cas9 endonuclease-mediated transgenesis and the molecular, electrophysiological, and behavioural consequences of the variant were examined. Results: In vitro , NMDARs containing GluN1-Y647S show altered sensitivity to endogenous agonists and negative allosteric modulators, and reduced cell surface trafficking. Grin1 Y647S/+ mice displayed a reduction in whole brain GluN1 levels and deficiency in NMDAR-mediated synaptic transmission in the hippocampus. Behaviourally, Grin1 Y647S/+ mice exhibited spontaneous seizures, altered vocalizations, muscle strength, sociability, and problem-solving. Interpretation: The Y647S variant confers a complex in vivo phenotype, which reflects largely diminished properties of NMDAR function. As a result, Grin1 Y647S/+ mice display atypical behaviour in domains relevant to the clinical characteristics of GRIN1 -NDD and the individual carrying the variant. Ultimately, the characterization of Grin1 Y647S/+ mice accomplished in the present work expands our understanding of the mechanisms underlying GRIN1 -NDD and provides a foundation for the development of novel therapeutics.

2.
Nature ; 632(8023): 209-217, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39085540

ABSTRACT

Glutamate transmission and activation of ionotropic glutamate receptors are the fundamental means by which neurons control their excitability and neuroplasticity1. The N-methyl-D-aspartate receptor (NMDAR) is unique among all ligand-gated channels, requiring two ligands-glutamate and glycine-for activation. These receptors function as heterotetrameric ion channels, with the channel opening dependent on the simultaneous binding of glycine and glutamate to the extracellular ligand-binding domains (LBDs) of the GluN1 and GluN2 subunits, respectively2,3. The exact molecular mechanism for channel gating by the two ligands has been unclear, particularly without structures representing the open channel and apo states. Here we show that the channel gate opening requires tension in the linker connecting the LBD and transmembrane domain (TMD) and rotation of the extracellular domain relative to the TMD. Using electron cryomicroscopy, we captured the structure of the GluN1-GluN2B (GluN1-2B) NMDAR in its open state bound to a positive allosteric modulator. This process rotates and bends the pore-forming helices in GluN1 and GluN2B, altering the symmetry of the TMD channel from pseudofourfold to twofold. Structures of GluN1-2B NMDAR in apo and single-liganded states showed that binding of either glycine or glutamate alone leads to distinct GluN1-2B dimer arrangements but insufficient tension in the LBD-TMD linker for channel opening. This mechanistic framework identifies a key determinant for channel gating and a potential pharmacological strategy for modulating NMDAR activity.


Subject(s)
Glutamic Acid , Glycine , Ion Channel Gating , Receptors, N-Methyl-D-Aspartate , Animals , Rats , Allosteric Regulation , Cryoelectron Microscopy , Glutamic Acid/metabolism , Glycine/metabolism , Ligands , Models, Molecular , Oocytes/metabolism , Protein Domains , Protein Multimerization , Protein Subunits/metabolism , Protein Subunits/chemistry , Receptors, N-Methyl-D-Aspartate/chemistry , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, N-Methyl-D-Aspartate/ultrastructure , Rotation , Xenopus laevis
3.
Glia ; 72(9): 1707-1724, 2024 09.
Article in English | MEDLINE | ID: mdl-38864289

ABSTRACT

Astrocytes play an essential role in regulating synaptic transmission. This study describes a novel form of modulation of excitatory synaptic transmission in the mouse hippocampus by astrocytic G-protein-coupled receptors (GPCRs). We have previously described astrocytic glutamate release via protease-activated receptor-1 (PAR1) activation, although the regulatory mechanisms for this are complex. Through electrophysiological analysis and modeling, we discovered that PAR1 activation consistently increases the concentration and duration of glutamate in the synaptic cleft. This effect was not due to changes in the presynaptic glutamate release or alteration in glutamate transporter expression. However, blocking group II metabotropic glutamate receptors (mGluR2/3) abolished PAR1-mediated regulation of synaptic glutamate concentration, suggesting a role for this GPCR in mediating the effects of PAR1 activation on glutamate release. Furthermore, activation of mGluR2/3 causes glutamate release through the TREK-1 channel in hippocampal astrocytes. These data show that astrocytic GPCRs engage in a novel regulatory mechanism to shape the time course of synaptically-released glutamate in excitatory synapses of the hippocampus.


Subject(s)
Astrocytes , CA1 Region, Hippocampal , Glutamic Acid , Mice, Inbred C57BL , Receptor, PAR-1 , Receptors, Metabotropic Glutamate , Synapses , Animals , Receptors, Metabotropic Glutamate/metabolism , Astrocytes/metabolism , Glutamic Acid/metabolism , Synapses/metabolism , CA1 Region, Hippocampal/metabolism , Receptor, PAR-1/metabolism , Mice , Excitatory Postsynaptic Potentials/physiology , Excitatory Postsynaptic Potentials/drug effects , Male , Synaptic Transmission/physiology , Synaptic Transmission/drug effects , Potassium Channels, Tandem Pore Domain/metabolism
4.
medRxiv ; 2024 May 07.
Article in English | MEDLINE | ID: mdl-38766179

ABSTRACT

Genetic variants in genes GRIN1 , GRIN2A , GRIN2B , and GRIN2D , which encode subunits of the N-methyl-D-aspartate receptor (NMDAR), have been associated with severe and heterogeneous neurologic diseases. Missense variants in these genes can result in gain or loss of the NMDAR function, requiring opposite therapeutic treatments. Computational methods that predict pathogenicity and molecular functional effects are therefore crucial for accurate diagnosis and therapeutic applications. We assembled missense variants: 201 from patients, 631 from general population, and 159 characterized by electrophysiological readouts showing whether they can enhance or reduce the receptor function. This includes new functional data from 47 variants reported here, for the first time. We found that pathogenic/benign variants and variants that increase/decrease the channel function were distributed unevenly on the protein structure, with spatial proximity to ligands bound to the agonist and antagonist binding sites being key predictive features. Leveraging distances from ligands, we developed two independent machine learning-based predictors for NMDAR missense variants: a pathogenicity predictor which outperforms currently available predictors (AUC=0.945, MCC=0.726), and the first binary predictor of molecular function (increase or decrease) (AUC=0.809, MCC=0.523). Using these, we reclassified variants of uncertain significance in the ClinVar database and refined a previous genome-informed epidemiological model to estimate the birth incidence of molecular mechanism-defined GRIN disorders. Our findings demonstrate that distance from ligands is an important feature in NMDARs that can enhance variant pathogenicity prediction and enable functional prediction. Further studies with larger numbers of phenotypically and functionally characterized variants will enhance the potential clinical utility of this method.

5.
Methods Mol Biol ; 2799: 55-77, 2024.
Article in English | MEDLINE | ID: mdl-38727903

ABSTRACT

NMDA-type ionotropic glutamate receptors are critically involved in many brain functions and are implicated in a variety of brain disorders. Seven NMDA receptor subunits exist (GluN1, GluN2A-D, and GluN3A-B) that assemble into tetrameric receptor subtypes with distinct functional properties and physiological roles. The majority NMDA receptors are composed of two GluN1 and two GluN2 subunits, which can assemble into four diheteromeric receptors subtypes composed of GluN1 and one type of GluN2 subunit (e.g., GluN1/2A), and presumably also six triheteromeric receptor subtypes composed of GluN1 and two different GluN2 subunits (e.g., GluN1/2A/2B). Furthermore, the GluN1 subunit exists as eight splice variants (e.g., GluN1-1a and GluN1-1b isoforms), and two different GluN1 isoforms can co-assemble to also form triheteromeric NMDA receptors (e.g., GluN1-1a/1b/2A). Here, we describe a method to faithfully express triheteromeric NMDA receptors in heterologous expression systems by controlling the identity of two of the four subunits. This method overcomes the problem that co-expression of three different NMDA receptor subunits generates two distinct diheteromeric receptor subtypes as well as one triheteromeric receptor subtype, thereby confounding studies that require a homogenous population of triheteromeric NMDA receptors. The method has been applied to selectively express recombinant triheteromeric GluN1/2A/2B, GluN1/2A/2C, GluN1/2B/2D, GluN1-1a/GluN1-1b/2A, GluN1-1a/GluN1-1b/2B receptors with negligible co-expression of the respective diheteromeric receptor subtypes. This method therefore enables quantitative evaluation of functional and pharmacological properties of triheteromeric NMDA receptors, some of which are abundant NMDA receptor subtypes in the adult brain.


Subject(s)
Protein Isoforms , Protein Subunits , Receptors, N-Methyl-D-Aspartate , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, N-Methyl-D-Aspartate/genetics , Humans , Protein Subunits/metabolism , Protein Subunits/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , HEK293 Cells , Animals , Cell Membrane/metabolism , Gene Expression
6.
Cell Mol Life Sci ; 81(1): 153, 2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38538865

ABSTRACT

N-methyl-D-aspartate receptors (NMDARs) are members of the glutamate receptor family and participate in excitatory postsynaptic transmission throughout the central nervous system. Genetic variants in GRIN genes encoding NMDAR subunits are associated with a spectrum of neurological disorders. The M3 transmembrane helices of the NMDAR couple directly to the agonist-binding domains and form a helical bundle crossing in the closed receptors that occludes the pore. The M3 functions as a transduction element whose conformational change couples ligand binding to opening of an ion conducting pore. In this study, we report the functional consequences of 48 de novo missense variants in GRIN1, GRIN2A, and GRIN2B that alter residues in the M3 transmembrane helix. These de novo variants were identified in children with neurological and neuropsychiatric disorders including epilepsy, developmental delay, intellectual disability, hypotonia and attention deficit hyperactivity disorder. All 48 variants in M3 for which comprehensive testing was completed produce a gain-of-function (28/48) compared to loss-of-function (9/48); 11 variants had an indeterminant phenotype. This supports the idea that a key structural feature of the M3 gate exists to stabilize the closed state so that agonist binding can drive channel opening. Given that most M3 variants enhance channel gating, we assessed the potency of FDA-approved NMDAR channel blockers on these variant receptors. These data provide new insight into the structure-function relationship of the NMDAR gate, and suggest that variants within the M3 transmembrane helix produce a gain-of-function.


Subject(s)
Epilepsy , Receptors, N-Methyl-D-Aspartate , Child , Humans , Epilepsy/genetics , Mutation, Missense , Phenotype , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction
7.
J Neurophysiol ; 131(4): 768-777, 2024 04 01.
Article in English | MEDLINE | ID: mdl-38380828

ABSTRACT

NMDA-type glutamate receptors (NMDARs) play a crucial role in synaptogenesis, circuit development, and synaptic plasticity, serving as fundamental components in cellular models of learning and memory. Their dysregulation has been implicated in several neurological disorders and synaptopathies. NMDARs are heterotetrameric complexes composed of two GluN1 and two GluN2 subunits. The composition of GluN2 subunits determines the main biophysical properties of the channel, such as calcium permeability and gating kinetics, and influences the ability of the receptor to interact with postsynaptic proteins involved in normal synaptic physiology and plasticity, including scaffolding proteins and signaling molecules. During early development, NMDARs in the forebrain contain solely the GluN2B subunit, a necessary subunit for proper synaptogenesis and synaptic plasticity. As the animal matures, the expression of the GluN2A subunit increases, leading to a partial replacement of GluN2B-containing synaptic NMDARs with GluN2A-containing receptors. The switch in the synaptic GluN2A-to-GluN2B ratio has a significant impact on the kinetics of excitatory postsynaptic currents and diminishes the synaptic plasticity capacity. In this study, we present findings indicating that GluN2A expression occurs earlier in a mouse model of fragile X syndrome (FXS). This altered timing of GluN2A expression affects various important parameters of NMDAR-mediated excitatory postsynaptic currents, including maximal current amplitude, decay time, and response to consecutive stimuli delivered in close temporal proximity. These observations suggest that the early expression of GluN2A during a critical period when synapses and circuits are developing could be an underlying factor contributing to the formation of pathological circuits in the FXS mouse model.NEW & NOTEWORTHY NMDA receptors (NMDARs) play important roles in synaptic transmission and are involved in multiple neurological disorders. During development, GluN2A in the forebrain becomes incorporated into previously GluN2B-dominated NMDARs, leading to the "GluN2A/GluN2B ratio switch." This is a crucial step for normal brain development. Here we present findings indicating that GluN2A expression occurs earlier in the fragile X mouse and this could be an underlying factor contributing to the pathology found in the fragile X model.


Subject(s)
Fragile X Syndrome , Receptors, N-Methyl-D-Aspartate , Mice , Animals , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/physiology , Synaptic Transmission , Neuronal Plasticity
8.
Hum Mol Genet ; 33(4): 355-373, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-37944084

ABSTRACT

GRID1 and GRID2 encode the enigmatic GluD1 and GluD2 proteins, which form tetrameric receptors that play important roles in synapse organization and development of the central nervous system. Variation in these genes has been implicated in neurodevelopmental phenotypes. We evaluated GRID1 and GRID2 human variants from the literature, ClinVar, and clinical laboratories and found that many of these variants reside in intolerant domains, including the amino terminal domain of both GRID1 and GRID2. Other conserved regions, such as the M3 transmembrane domain, show different intolerance between GRID1 and GRID2. We introduced these variants into GluD1 and GluD2 cDNA and performed electrophysiological and biochemical assays to investigate the mechanisms of dysfunction of GRID1/2 variants. One variant in the GRID1 distal amino terminal domain resides at a position predicted to interact with Cbln2/Cbln4, and the variant disrupts complex formation between GluD1 and Cbln2, which could perturb its role in synapse organization. We also discovered that, like the lurcher mutation (GluD2-A654T), other rare variants in the GRID2 M3 domain create constitutively active receptors that share similar pathogenic phenotypes. We also found that the SCHEMA schizophrenia M3 variant GluD1-A650T produced constitutively active receptors. We tested a variety of compounds for their ability to inhibit constitutive currents of GluD receptor variants and found that pentamidine potently inhibited GluD2-T649A constitutive channels (IC50 50 nM). These results identify regions of intolerance to variation in the GRID genes, illustrate the functional consequences of GRID1 and GRID2 variants, and suggest how these receptors function normally and in disease.


Subject(s)
Central Nervous System , Receptors, Glutamate , Humans , Central Nervous System/metabolism , Mutation , Protein Domains , Receptors, Glutamate/metabolism
9.
Neuropsychopharmacology ; 49(1): 51-66, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37369776

ABSTRACT

N-methyl-D-aspartate (NMDA) receptors mediate a slow component of excitatory synaptic transmission, are widely distributed throughout the central nervous system, and regulate synaptic plasticity. NMDA receptor modulators have long been considered as potential treatments for psychiatric disorders including depression and schizophrenia, neurodevelopmental disorders such as Rett Syndrome, and neurodegenerative conditions such as Alzheimer's disease. New interest in NMDA receptors as therapeutic targets has been spurred by the findings that certain inhibitors of NMDA receptors produce surprisingly rapid and robust antidepressant activity by a novel mechanism, the induction of changes in the brain that well outlast the presence of drug in the body. These findings are driving research into an entirely new paradigm for using NMDA receptor antagonists in a host of related conditions. At the same time positive allosteric modulators of NMDA receptors are being pursued for enhancing synaptic function in diseases that feature NMDA receptor hypofunction. While there is great promise, developing the therapeutic potential of NMDA receptor modulators must also navigate the potential significant risks posed by the use of such agents. We review here the emerging pharmacology of agents that target different NMDA receptor subtypes, offering new avenues for capturing the therapeutic potential of targeting this important receptor class.


Subject(s)
Psychiatry , Schizophrenia , Humans , Receptors, N-Methyl-D-Aspartate/metabolism , Central Nervous System , Brain/metabolism
10.
bioRxiv ; 2023 Dec 06.
Article in English | MEDLINE | ID: mdl-38106229

ABSTRACT

Background: N-methyl-D-aspartate receptor (NMDAR) are amino acid receptors that are well studied in brain physiology; however, their role in kidney is poorly understood. Nonetheless, NMDAR inhibitors can increase serum K+ and reduce GFR, which suggests they have an important physiological role in the kidney. We hypothesized that NMDARs in the distal nephron induce afferent-arteriole vasodilation through the vasodilator mechanism connecting-tubule-glomerular feedback (CNTGF) that involves ENaC activation. Methods and results: Using a tubule-specific transcriptome database combined with molecular biology and microscopy techniques, we showed kidney expression of NMDAR subunits along the nephron and specifically in ENaC-positive cells. This receptor is expressed in both male and female mice, with higher abundance in females (p=0.02). Microperfusing NMDAR agonists into the connecting tubule induced afferent-arteriole vasodilation (EC50 10.7 vs. 24.5 mM; p<0.001) that was blunted or eliminated with the use of NMDAR blocker MK-801 or with the ENaC inhibitor Benzamil, indicating a dependence on CNTGF of the NMDAR-induced vasodilation. In vivo, we confirmed this CNTGF-associated vasodilation using kidney micropuncture (Stop-flow pressure 37.9±2.6 vs. 28.6±1.9 mmHg, NMDAR agonist vs vehicle; p<0.01). We explored NMDAR and ENaC channel interaction by using mpkCCD cells and split-open connecting tubules. We observed increased amiloride-sensitive current following NMDAR activation that was prevented by MK-801 (1.14 vs. 0.4 µAmp; p=0.03). In split-open tubules, NMDAR activation increased ENaC activity (Npo Vehicle vs. NMDA; p=0.04). Conclusion: NMDARs are expressed along the nephron, including ENaC-positive cells, with higher expression in females. Epithelial NMDAR mediates renal vasodilation through the connecting-tubule-glomerular feedback, by increasing ENaC activity.

11.
Cell Mol Life Sci ; 80(11): 345, 2023 Nov 03.
Article in English | MEDLINE | ID: mdl-37921875

ABSTRACT

AMPA receptors are members of the glutamate receptor family and mediate a fast component of excitatory synaptic transmission at virtually all central synapses. Thus, their functional characteristics are a critical determinant of brain function. We evaluate intolerance of each GRIA gene to genetic variation using 3DMTR and report here the functional consequences of 52 missense variants in GRIA1-4 identified in patients with various neurological disorders. These variants produce changes in agonist EC50, response time course, desensitization, and/or receptor surface expression. We predict that these functional and localization changes will have important consequences for circuit function, and therefore likely contribute to the patients' clinical phenotype. We evaluated the sensitivity of variant receptors to AMPAR-selective modulators including FDA-approved drugs to explore potential targeted therapeutic options.


Subject(s)
Nervous System Diseases , Humans , Nervous System Diseases/genetics , Synaptic Transmission/physiology , Receptors, AMPA/genetics , Receptors, AMPA/metabolism , Synapses/metabolism
12.
Commun Biol ; 6(1): 952, 2023 09 19.
Article in English | MEDLINE | ID: mdl-37723282

ABSTRACT

N-methyl-D-aspartate receptors (NMDARs) are ligand-gated ionotropic glutamate receptors that mediate a calcium-permeable component to fast excitatory neurotransmission. NMDARs are heterotetrameric assemblies of two obligate GluN1 subunits (GRIN1) and two GluN2 subunits (GRIN2A-GRIN2D). Sequencing data shows that 43% (297/679) of all currently known NMDAR disease-associated genetic variants are within the GRIN2A gene, which encodes the GluN2A subunit. Here, we show that unlike missense GRIN2A variants, individuals affected with disease-associated null GRIN2A variants demonstrate a transient period of seizure susceptibility that begins during infancy and diminishes near adolescence. We show increased circuit excitability and CA1 pyramidal cell output in juvenile mice of both Grin2a+/- and Grin2a-/- mice. These alterations in somatic spiking are not due to global upregulation of most Grin genes (including Grin2b). Deeper evaluation of the developing CA1 circuit led us to uncover age- and Grin2a gene dosing-dependent transient delays in the electrophysiological maturation programs of parvalbumin (PV) interneurons. We report that Grin2a+/+ mice reach PV cell electrophysiological maturation between the neonatal and juvenile neurodevelopmental timepoints, with Grin2a+/- mice not reaching PV cell electrophysiological maturation until preadolescence, and Grin2a-/- mice not reaching PV cell electrophysiological maturation until adulthood. Overall, these data may represent a molecular mechanism describing the transient nature of seizure susceptibility in disease-associated null GRIN2A patients.


Subject(s)
Calcium , Parvalbumins , Receptors, N-Methyl-D-Aspartate , Animals , Mice , Hippocampus , Interneurons , Parvalbumins/genetics , Seizures , Receptors, N-Methyl-D-Aspartate/genetics
13.
Epilepsia ; 64(11): 2968-2981, 2023 11.
Article in English | MEDLINE | ID: mdl-37577761

ABSTRACT

OBJECTIVE: To investigate the clinical features and potential pathogenesis mechanism of de novo CLPTM1 variants associated with epilepsy. METHODS: Identify de novo genetic variants associated with epilepsy by reanalyzing trio-based whole-exome sequencing data. We analyzed the clinical characteristics of patients with these variants and performed functional in vitro studies in cells expressing mutant complementary DNA for these variants using whole-cell voltage-clamp current recordings and outside-out patch-clamp recordings from transiently transfected human embryonic kidney (HEK) cells. RESULTS: Two de novo missense variants related to epilepsy were identified in the CLPTM1 gene. Functional studies indicated that CLPTM1-p.R454H and CLPTM1-p.R568Q variants reduced the γ-aminobutyric acid A receptor (GABAA R) current response amplitude recorded under voltage clamp compared to the wild-type receptors. These variants also reduced the charge transfer and altered the time course of desensitization and deactivation following rapid removal of GABA. The surface expression of the GABAA R γ2 subunit from the CLPTM1-p.R568Q group was significantly reduced compared to CLPTM1-WT. SIGNIFICANCE: This is the first report of functionally relevant variants within the CLPTM1 gene. Patch-clamp recordings showed that these de novo CLPTM1 variants reduce GABAA R currents and charge transfer, which should promote excitation and hypersynchronous activity. This study may provide insights into the molecular mechanisms of the CLPTM1 variants underlying the patients' phenotypes, as well as for exploring potential therapeutic targets for epilepsy.


Subject(s)
Epilepsy , Receptors, GABA-A , Humans , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Epilepsy/genetics , Mutation, Missense/genetics , Phenotype , gamma-Aminobutyric Acid , Membrane Proteins/genetics , Membrane Proteins/metabolism
14.
ACS Chem Neurosci ; 14(17): 3059-3076, 2023 09 06.
Article in English | MEDLINE | ID: mdl-37566734

ABSTRACT

Subunit-selective inhibition of N-methyl-d-aspartate receptors (NMDARs) is a promising therapeutic strategy for several neurological disorders, including epilepsy, Alzheimer's and Parkinson's disease, depression, and acute brain injury. We previously described the dihydroquinoline-pyrazoline (DQP) analogue 2a (DQP-26) as a potent NMDAR negative allosteric modulator with selectivity for GluN2C/D over GluN2A/B. However, moderate (<100-fold) subunit selectivity, inadequate cell-membrane permeability, and poor brain penetration complicated the use of 2a as an in vivo probe. In an effort to improve selectivity and the pharmacokinetic profile of the series, we performed additional structure-activity relationship studies of the succinate side chain and investigated the use of prodrugs to mask the pendant carboxylic acid. These efforts led to discovery of the analogue (S)-(-)-2i, also referred to as (S)-(-)-DQP-997-74, which exhibits >100- and >300-fold selectivity for GluN2C- and GluN2D-containing NMDARs (IC50 0.069 and 0.035 µM, respectively) compared to GluN2A- and GluN2B-containing receptors (IC50 5.2 and 16 µM, respectively) and has no effects on AMPA, kainate, or GluN1/GluN3 receptors. Compound (S)-(-)-2i is 5-fold more potent than (S)-2a. In addition, compound 2i shows a time-dependent enhancement of inhibitory actions at GluN2C- and GluN2D-containing NMDARs in the presence of the agonist glutamate, which could attenuate hypersynchronous activity driven by high-frequency excitatory synaptic transmission. Consistent with this finding, compound 2i significantly reduced the number of epileptic events in a murine model of tuberous sclerosis complex (TSC)-induced epilepsy that is associated with upregulation of the GluN2C subunit. Thus, 2i represents a robust tool for the GluN2C/D target validation. Esterification of the succinate carboxylate improved brain penetration, suggesting a strategy for therapeutic development of this series for NMDAR-associated neurological conditions.


Subject(s)
Receptors, N-Methyl-D-Aspartate , Synaptic Transmission , Mice , Animals , Receptors, N-Methyl-D-Aspartate/metabolism , Structure-Activity Relationship , Synaptic Transmission/physiology , Glutamic Acid/pharmacology , Brain/metabolism
15.
J Neurochem ; 2023 Aug 30.
Article in English | MEDLINE | ID: mdl-37649269

ABSTRACT

N-methyl-D-aspartate receptors (NMDAR), ionotropic glutamate receptors, mediate a slow component of excitatory synaptic transmission in the central nervous system and play a key role in normal brain function and development. Genetic variations in GRIN genes encoding NMDAR subunits that alter the receptor's functional characteristics are associated with a wide range of neurological and neuropsychiatric conditions. Pathological GRIN variants located in the M2 re-entrant loop lining the channel pore cause significant functional changes, the most consequential alteration being a reduction in voltage-dependent Mg2+ inhibition. Voltage-dependent Mg2+ block is a unique feature of NMDAR biology whereby channel activation requires both ligand binding and postsynaptic membrane depolarization. Thus, loss of NMDAR Mg2+ block will have a profound impact on synaptic function and plasticity. Here, we choose 11 missense variants within the GRIN1, GRIN2A, and GRIN2B genes that alter residues located in the M2 loop and significantly reduce Mg2+ inhibition. Each variant was evaluated for tolerance to genetic variation using the 3-dimensional structure and assessed for functional rescue pharmacology via electrophysiological recordings. Three FDA-approved NMDAR drugs-memantine, dextromethorphan, and ketamine-were chosen based on their ability to bind near the M2 re-entrant loop, potentially rectifying dysregulated NMDAR function by supplementing the reduced voltage-dependent Mg2+ block. These results provide insight of structural determinants of FDA-approved NMDAR drugs at their binding sites in the channel pore and may further define conditions necessary for the use of such agents as potential rescue pharmacology.

17.
Hum Mol Genet ; 32(19): 2857-2871, 2023 09 16.
Article in English | MEDLINE | ID: mdl-37369021

ABSTRACT

Advances in sequencing technology have generated a large amount of genetic data from patients with neurological conditions. These data have provided diagnosis of many rare diseases, including a number of pathogenic de novo missense variants in GRIN genes encoding N-methyl-d-aspartate receptors (NMDARs). To understand the ramifications for neurons and brain circuits affected by rare patient variants, functional analysis of the variant receptor is necessary in model systems. For NMDARs, this functional analysis needs to assess multiple properties in order to understand how variants could impact receptor function in neurons. One can then use these data to determine whether the overall actions will increase or decrease NMDAR-mediated charge transfer. Here, we describe an analytical and comprehensive framework by which to categorize GRIN variants as either gain-of-function (GoF) or loss-of-function (LoF) and apply this approach to GRIN2B variants identified in patients and the general population. This framework draws on results from six different assays that assess the impact of the variant on NMDAR sensitivity to agonists and endogenous modulators, trafficking to the plasma membrane, response time course and channel open probability. We propose to integrate data from multiple in vitro assays to arrive at a variant classification, and suggest threshold levels that guide confidence. The data supporting GoF and LoF determination are essential to assessing pathogenicity and patient stratification for clinical trials as personalized pharmacological and genetic agents that can enhance or reduce receptor function are advanced. This approach to functional variant classification can generalize to other disorders associated with missense variants.


Subject(s)
Nervous System Diseases , Receptors, N-Methyl-D-Aspartate , Humans , Receptors, N-Methyl-D-Aspartate/genetics , Receptors, N-Methyl-D-Aspartate/metabolism , Mutation, Missense/genetics , Nervous System Diseases/metabolism , Neurons/metabolism , Models, Biological
18.
Glia ; 71(8): 1921-1946, 2023 08.
Article in English | MEDLINE | ID: mdl-37029775

ABSTRACT

Astrocyte maturation is crucial to proper brain development and function. This maturation process includes the ramification of astrocytic morphology and the establishment of astrocytic domains. While this process has been well-studied, the mechanisms by which astrocyte maturation is initiated are not well understood. GPR37L1 is an astrocyte-specific G protein-coupled receptor (GPCR) that is predominantly expressed in mature astrocytes and has been linked to the modulation of seizure susceptibility in both humans and mice. To investigate the role of GPR37L1 in astrocyte biology, RNA-seq analyses were performed on astrocytes immunopanned from P7 Gpr37L1-/- knockout (L1KO) mouse cortex and compared to those from wild-type (WT) mouse cortex. These RNA-seq studies revealed that pathways involved in central nervous system development were altered and that L1KO cortical astrocytes express lower amounts of mature astrocytic genes compared to WT astrocytes. Immunohistochemical studies of astrocytes from L1KO mouse brain revealed that these astrocytes exhibit overall shorter total process length, and are also less complex and spaced further apart from each other in the mouse cortex. This work sheds light on how GPR37L1 regulates cellular processes involved in the control of astrocyte biology and maturation.


Subject(s)
Astrocytes , Receptors, G-Protein-Coupled , Humans , Mice , Animals , Astrocytes/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Seizures/metabolism
19.
Cell Mol Life Sci ; 80(4): 110, 2023 Mar 31.
Article in English | MEDLINE | ID: mdl-37000222

ABSTRACT

The short pre-M1 helix within the S1-M1 linker (also referred to as the pre-M1 linker) between the agonist-binding domain (ABD, S1) and the M1 transmembrane helix of the NMDA receptor (NMDAR) is devoid of missense variants within the healthy population but is a locus for de novo pathogenic variants associated with neurological disorders. Several de novo variants within this helix have been identified in patients presenting early in life with intellectual disability, developmental delay, and/or epilepsy. In this study, we evaluated functional properties for twenty variants within the pre-M1 linker in GRIN1, GRIN2A, and GRIN2B genes, including six novel missense variants. The effects of pre-M1 variants on agonist potency, sensitivity to endogenous allosteric modulators, response time course, channel open probability, and surface expression were assessed. Our data indicated that virtually all of the variants evaluated altered channel function, and multiple variants had profound functional consequences, which may contribute to the neurological conditions in the patients harboring the variants in this region. These data strongly suggest that the residues within the pre-M1 helix play a key role in channel gating and are highly intolerant to genetic variation.


Subject(s)
Epilepsy , Intellectual Disability , Receptors, N-Methyl-D-Aspartate , Humans , Epilepsy/genetics , Mutation, Missense/genetics , Receptors, N-Methyl-D-Aspartate/metabolism
20.
ACS Chem Neurosci ; 14(5): 917-935, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36779874

ABSTRACT

Many cases of accidental death associated with drug overdose are due to chronic opioid use, tolerance, and addiction. Analgesic tolerance is characterized by a decreased response to the analgesic effects of opioids, requiring increasingly higher doses to maintain the desired level of pain relief. Overactivation of GluN2B-containing N-methyl-d-Aspartate receptors is thought to play a key role in mechanisms underlying cellular adaptation that takes place in the development of analgesic tolerance. Herein, we describe a novel GluN2B-selective negative allosteric modulator, EU93-108, that shows high potency and brain penetrance. We describe the structural basis for binding at atomic resolution. This compound possesses intrinsic analgesic properties in the rodent tail immersion test. EU93-108 has an acute and significant anodyne effect, whereby morphine when combined with EU93-108 produces a higher tail flick latency compared to that of morphine alone. These data suggest that engagement of GluN2B as a target has utility in the treatment of pain, and EU93-108 could serve as an appropriate tool compound to interrogate this hypothesis. Future structure-activity relationship work around this scaffold could give rise to compounds that can be co-administered with opioids to diminish the onset of tolerance due to chronic opioid use, thereby modifying their utility.


Subject(s)
Analgesia , Morphine , Animals , Analgesics, Opioid/pharmacology , Analgesics, Opioid/therapeutic use , Receptors, N-Methyl-D-Aspartate/metabolism , Rodentia/metabolism , Analgesics/pharmacology , Analgesics/therapeutic use , Pain/drug therapy , Pain/metabolism , Dose-Response Relationship, Drug
SELECTION OF CITATIONS
SEARCH DETAIL