1
|
Forouzanfar F, Ahmadzadeh AM, Pourbagher-Shahri AM, Gorji A. Significance of NMDA receptor-targeting compounds in neuropsychological disorders: An In-depth Review. Eur J Pharmacol 2025:177690. [PMID: 40315950 DOI: 10.1016/j.ejphar.2025.177690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 04/16/2025] [Accepted: 04/29/2025] [Indexed: 05/04/2025]
Abstract
N-methyl-D-aspartate receptors (NMDARs), a subclass of glutamate-gated ion channels, play an integral role in the maintenance of synaptic plasticity and excitation-inhibition balance within the central nervous system (CNS). Any irregularities in NMDAR functions, whether hypo-activation or over-activation, can destabilize neural networks and impair CNS function. Several decades of experimental and clinical investigations have demonstrated that NMDAR dysfunction is implicated in the pathophysiology of various neurological disorders. Despite designing a long list of compounds that differentially modulate NMDARs, success in developing drugs that can selectively and effectively regulate various NMDAR subtypes while showing encouraging efficacy in clinical settings remains limited. A better understanding of the basic mechanism of NMDAR function, particularly its selective regulation in pathological conditions, could aid in designing effective drugs for the treatment of neurological conditions. Here, we reviewed the experimental and clinical investigations that studied the effects of available NMDAR modulators in various neurological disorders and weighed up the pros and cons of the use of these substances on the improvement of functional outcomes of these disorders. Despite numerous efforts to develop NMDAR modulatory drugs that did not produce the desired outcomes, NMDARs remain a significant target for advancing novel drugs to treat neurological disorders. This article reviews the complexity of NMDAR signaling dysfunction in different neurological diseases, the efforts taken to examine designed compounds targeting specific subtypes of NMDARs, including challenges accompanied by using these substances, and the potential enhancements in drug discovery for NMDAR modulatory compounds by innovative technologies.
Collapse
Affiliation(s)
- Fatemeh Forouzanfar
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Mahmoud Ahmadzadeh
- Transplant Research Center, Clinical Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Radiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Mohammad Pourbagher-Shahri
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Gorji
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran; Department of Neurosurgery, Münster University, Münster, Germany; Epilepsy Research Center, Münster University, Münster, Germany.
| |
Collapse
|
2
|
Kang H, Epstein M, Banke TG, Perszyk R, Simorowski N, Paladugu S, Liotta DC, Traynelis SF, Furukawa H. Structural basis for channel gating and blockade in tri-heteromeric GluN1-2B-2D NMDA receptor. Neuron 2025; 113:991-1005.e5. [PMID: 39954679 PMCID: PMC11968220 DOI: 10.1016/j.neuron.2025.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 12/09/2024] [Accepted: 01/21/2025] [Indexed: 02/17/2025]
Abstract
Discrete activation of N-methyl-D-aspartate receptor (NMDAR) subtypes by glutamate and the co-agonist glycine is fundamental to neuroplasticity. A distinct variant, the tri-heteromeric receptor, comprising glycine-binding GluN1 and two types of glutamate-binding GluN2 subunits, exhibits unique pharmacological characteristics, notably enhanced sensitivity to the anti-depressant channel blocker S-(+)-ketamine. Despite its significance, the structural mechanisms underlying ligand gating and channel blockade of tri-heteromeric NMDARs remain poorly understood. Here, we identify and characterize tri-heteromeric GluN1-2B-2D NMDAR in the adult brain, resolving its structures in the activated, inhibited, and S-(+)-ketamine-blocked states. These structures reveal the ligand-dependent conformational dynamics that modulate the tension between the extracellular domain and transmembrane channels, governing channel gating and blockade. Additionally, we demonstrate that the inhibitor (S)-DQP-997-74 selectively decouples linker tension in GluN2D, offering insights into subtype-selective targeting for cognitive modulation.
Collapse
Affiliation(s)
- Hyunook Kang
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Max Epstein
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Tue G Banke
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Riley Perszyk
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Noriko Simorowski
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Srinu Paladugu
- Department of Chemistry, Emory University, Atlanta, GA, USA
| | | | - Stephen F Traynelis
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA; Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, GA, USA
| | - Hiro Furukawa
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
3
|
Van Maldegem M, Vohryzek J, Atasoy S, Alnagger N, Cardone P, Bonhomme V, Vanhaudenhuyse A, Demertzi A, Jaquet O, Bahri MA, Nunez P, Kringelbach ML, Stamatakis EA, Luppi AI. Connectome harmonic decomposition tracks the presence of disconnected consciousness during ketamine-induced unresponsiveness. Br J Anaesth 2025; 134:1088-1104. [PMID: 39933965 PMCID: PMC11947573 DOI: 10.1016/j.bja.2024.12.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/22/2024] [Accepted: 12/07/2024] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Ketamine, in doses suitable to induce anaesthesia in humans, gives rise to a unique state of unresponsiveness accompanied by vivid experiences and sensations, making it possible to disentangle the correlated but distinct concepts of conscious awareness and behavioural responsiveness. This distinction is often overlooked in the study of consciousness. METHODS The mathematical framework of connectome harmonic decomposition (CHD) was used to view functional magnetic resonance imaging (fMRI) signals during ketamine-induced unresponsiveness as distributed patterns across spatial scales. The connectome harmonic signature of this particular state was mapped onto signatures of other states of consciousness for comparison. RESULTS An increased prevalence of fine-grained connectome harmonics was found in fMRI signals obtained during ketamine-induced unresponsiveness, indicating higher granularity. After statistical assessment, the ketamine sedation harmonic signature showed alignment with signatures of LSD-induced (fixed effect =0.0113 [0.0099, 0.0127], P<0.001) or ketamine-induced (fixed effect =0.0087 [0.0071, 0.0103], P<0.001) psychedelic states, and misalignment with signatures seen in unconscious individuals owing to propofol sedation (fixed effect =-0.0213 [-0.0245, -0.0181], P<0.001) or brain injury (fixed effect =-0.0205 [-0.0234, -0.0178], P<0.001). CONCLUSIONS The CHD framework, which only requires resting-state fMRI data and can be applied retrospectively, has the ability to track alterations in conscious awareness in the absence of behavioural responsiveness on a group level. This is possible because of ketamine's unique property of decoupling these two facets, and is important for consciousness and anaesthesia research.
Collapse
Affiliation(s)
- Milan Van Maldegem
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK; Department of Physiology, Development & Neuroscience, University of Cambridge, Cambridge, UK; Division of Anaesthesia, University of Cambridge, Cambridge, UK.
| | - Jakub Vohryzek
- Centre for Eudaimonia and Human Flourishing, Department of Psychiatry, University of Oxford, Oxford, UK; Centre for Brain and Cognition, Computational Neuroscience Group, Department of Information and Communication Technologies, Universitat Pompeu Fabra, Barcelona, Spain
| | - Selen Atasoy
- Centre for Eudaimonia and Human Flourishing, Department of Psychiatry, University of Oxford, Oxford, UK; Centre for Music in the Brain, Aarhus University, Aarhus, Denmark
| | - Naji Alnagger
- Coma Science Group, GIGA-Consciousness, University of Liege, Liege, Belgium; Centre du Cerveau, University Hospital of Liege, Liege, Belgium
| | - Paolo Cardone
- Coma Science Group, GIGA-Consciousness, University of Liege, Liege, Belgium; Centre du Cerveau, University Hospital of Liege, Liege, Belgium
| | - Vincent Bonhomme
- Anaesthesia and Perioperative Neuroscience, GIGA-Consciousness, University of Liege, Liege, Belgium; Department of Anesthesia and Intensive Care Medicine, University Hospital of Liege, Liege, Belgium
| | - Audrey Vanhaudenhuyse
- Conscious Care Lab, GIGA-Consciousness, University of Liege, Liege, Belgium; Algology Interdisciplinary Centre, University Hospital of Liege, Liege, Belgium
| | - Athena Demertzi
- Physiology of Cognition Lab, GIGA-CRC Human Imaging Unit, University of Liege, Liege, Belgium; Psychology and Neuroscience of Cognition Research Unit, University of Liege, Liege, Belgium
| | - Oceane Jaquet
- Department of Anesthesia and Intensive Care Medicine, University Hospital of Liege, Liege, Belgium
| | | | - Pablo Nunez
- Coma Science Group, GIGA-Consciousness, University of Liege, Liege, Belgium; Centre du Cerveau, University Hospital of Liege, Liege, Belgium
| | - Morten L Kringelbach
- Centre for Eudaimonia and Human Flourishing, Department of Psychiatry, University of Oxford, Oxford, UK; Centre for Music in the Brain, Aarhus University, Aarhus, Denmark
| | - Emmanuel A Stamatakis
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK; Division of Anaesthesia, University of Cambridge, Cambridge, UK
| | - Andrea I Luppi
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK; Division of Anaesthesia, University of Cambridge, Cambridge, UK; Centre for Eudaimonia and Human Flourishing, Department of Psychiatry, University of Oxford, Oxford, UK; Division of Information Engineering, University of Cambridge, Cambridge, UK; St John's College, University of Cambridge, Cambridge, UK
| |
Collapse
|
4
|
Zhao F, Atxabal U, Mariottini S, Yi F, Lotti JS, Layeux MS, Currier C, Maderia MP, Cornelison LE, Anderson CM, Schultz EP, Zhang Z, Jiang L, Gao Z, Liu N, Woodahl EL, Bunch L, Hansen KB, Clausen RP. Design of ( R)-3-(5-Thienyl)carboxamido-2-aminopropanoic Acid Derivatives as Novel NMDA Receptor Glycine Site Agonists: Variation in Molecular Geometry to Improve Potency and Augment GluN2 Subunit-Specific Activity. J Med Chem 2025; 68:3572-3590. [PMID: 39847708 PMCID: PMC11832032 DOI: 10.1021/acs.jmedchem.4c02715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/16/2024] [Accepted: 01/08/2025] [Indexed: 01/25/2025]
Abstract
NMDA receptor ligands have therapeutic potential in neurological and psychiatric disorders. We designed (R)-3-(5-thienyl)carboxamido-2-aminopropanoic acid derivatives with nanomolar agonist potencies at NMDA receptor subtypes (GluN12/A-D). These compounds are superagonists at GluN1/2C compared to glycine and partial to full agonists at GluN1/2A and GluN1/2D but display functional antagonism at GluN1/2B due to low agonist efficacy. Notably, 8d display 864% agonist efficacy at GluN1/2C relative to glycine, and 8j has high potency at GluN1/2A (0.018 μM), GluN1/2C (0.0029 μM), and GluN1/2D (0.016 μM). We evaluated the binding mode in the glycine site using molecular modeling and mutagenesis. In vitro absorption, distribution, metabolism, and excretion (ADME) assays predict high metabolic stability but poor blood-brain barrier permeability. However, an ester prodrug for the carboxylate group of 7j display moderately high blood-brain barrier permeability. The thiophenecarboxamide agonists expand the synthetic pharmacology of NMDA receptors and provide structural insights that facilitate the design of GluN1 agonists with GluN2 subunit-specific activity.
Collapse
Affiliation(s)
- Fabao Zhao
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK-2100, Denmark
- Department
of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry
of Education), School of Pharmaceutical Sciences, Shandong University, 250012 Jinan, Shandong P. R. China
| | - Unai Atxabal
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK-2100, Denmark
| | - Sofia Mariottini
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK-2100, Denmark
| | - Feng Yi
- Center
for Structural and Functional Neuroscience, Center for Biomolecular
Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, Montana 59812, United States
| | - James S. Lotti
- Center
for Structural and Functional Neuroscience, Center for Biomolecular
Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, Montana 59812, United States
| | - Michael S. Layeux
- Center
for Structural and Functional Neuroscience, Center for Biomolecular
Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, Montana 59812, United States
| | - Chandler Currier
- Center
for Structural and Functional Neuroscience, Center for Biomolecular
Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, Montana 59812, United States
| | - Matthew P. Maderia
- Center
for Structural and Functional Neuroscience, Center for Biomolecular
Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, Montana 59812, United States
| | - Lauren E. Cornelison
- Center
for Structural and Functional Neuroscience, Center for Biomolecular
Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, Montana 59812, United States
| | - Carly M. Anderson
- Center
for Structural and Functional Neuroscience, Center for Biomolecular
Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, Montana 59812, United States
| | - Eric P. Schultz
- Center
for Structural and Functional Neuroscience, Center for Biomolecular
Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, Montana 59812, United States
| | - Zhucheng Zhang
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK-2100, Denmark
| | - Liyang Jiang
- Department
of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry
of Education), School of Pharmaceutical Sciences, Shandong University, 250012 Jinan, Shandong P. R. China
| | - Zhen Gao
- Department
of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry
of Education), School of Pharmaceutical Sciences, Shandong University, 250012 Jinan, Shandong P. R. China
| | - Na Liu
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK-2100, Denmark
- Department
of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry
of Education), School of Pharmaceutical Sciences, Shandong University, 250012 Jinan, Shandong P. R. China
| | - Erica L. Woodahl
- L.S.
Skaggs Institute for Health Innovation, Department of Biomedical and
Pharmaceutical Sciences, University of Montana, Missoula, Montana 59812, United States
| | - Lennart Bunch
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK-2100, Denmark
| | - Kasper B. Hansen
- Center
for Structural and Functional Neuroscience, Center for Biomolecular
Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, Montana 59812, United States
| | - Rasmus P. Clausen
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, DK-2100, Denmark
| |
Collapse
|
5
|
Montanucci L, Brünger T, Bhattarai N, Boßelmann CM, Kim S, Allen JP, Zhang J, Klöckner C, Krey I, Fariselli P, May P, Lemke JR, Myers SJ, Yuan H, Traynelis SF, Lal D. Ligand distances as key predictors of pathogenicity and function in NMDA receptors. Hum Mol Genet 2025; 34:128-139. [PMID: 39535073 PMCID: PMC11780861 DOI: 10.1093/hmg/ddae156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 10/10/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Genetic variants in the 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 and neurodevelopmental disorders, including early onset epilepsy, developmental and epileptic encephalopathy, intellectual disability, and autism spectrum disorders. 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 of missense variants are therefore crucial for therapeutic applications. We assembled 223 missense variants from patients, 631 control variants from the general population, and 160 missense variants characterized by electrophysiological readouts that show whether they can enhance or reduce the function of the receptor. This includes new functional data from 33 variants reported here, for the first time. By mapping these variants onto the NMDAR protein structures, 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 a key predictive feature for both variant pathogenicity and molecular functional consequences. Leveraging distances from ligands, we developed two machine-learning based predictors for NMDA variants: a pathogenicity predictor which outperforms currently available predictors and the first molecular function (increase/decrease) predictor. Our findings can have direct application to patient care by improving diagnostic yield for genetic neurodevelopmental disorders and by guiding personalized treatment informed by the knowledge of the molecular disease mechanism.
Collapse
Affiliation(s)
- Ludovica Montanucci
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, 1133 John Freeman Blvd, Houston, TX 77030, United States
| | - Tobias Brünger
- Cologne Center for Genomics, University of Cologne, University Hospital Cologne, Weyertal 115b, Cologne 50937, Germany
| | - Nisha Bhattarai
- Epilepsy Center, Neurological Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44106, United States
| | - Christian M Boßelmann
- Epilepsy Center, Neurological Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44106, United States
| | - Sukhan Kim
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 100 Woodruff Circle, Atlanta, GA 30322, United States
- Center for Functional Evaluation of Rare Variants (CFERV), Emory University School of Medicine, 100 Woodruff Circle, Atlanta, GA 30322, United States
| | - James P Allen
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 100 Woodruff Circle, Atlanta, GA 30322, United States
| | - Jing Zhang
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 100 Woodruff Circle, Atlanta, GA 30322, United States
| | - Chiara Klöckner
- Institute of Human Genetics, University of Leipzig Hospitals and Clinics, Philipp-Rosenthal-street 55, Leipzig 04103, Germany
| | - Ilona Krey
- Institute of Human Genetics, University of Leipzig Hospitals and Clinics, Philipp-Rosenthal-street 55, Leipzig 04103, Germany
| | - Piero Fariselli
- Department of Medical Sciences, University of Torino, Via Santena 19,Torino, 10123, Italy
| | - Patrick May
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 7 Av. des Hauts-Fourneaux, Esch-sur-Alzette, 4362, Luxembourg
| | - Johannes R Lemke
- Institute of Human Genetics, University of Leipzig Hospitals and Clinics, Philipp-Rosenthal-street 55, Leipzig 04103, Germany
| | - Scott J Myers
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 100 Woodruff Circle, Atlanta, GA 30322, United States
- Center for Functional Evaluation of Rare Variants (CFERV), Emory University School of Medicine, 100 Woodruff Circle, Atlanta, GA 30322, United States
| | - Hongjie Yuan
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 100 Woodruff Circle, Atlanta, GA 30322, United States
- Center for Functional Evaluation of Rare Variants (CFERV), Emory University School of Medicine, 100 Woodruff Circle, Atlanta, GA 30322, United States
| | - Stephen F Traynelis
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 100 Woodruff Circle, Atlanta, GA 30322, United States
- Center for Functional Evaluation of Rare Variants (CFERV), Emory University School of Medicine, 100 Woodruff Circle, Atlanta, GA 30322, United States
| | - Dennis Lal
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, 1133 John Freeman Blvd, Houston, TX 77030, United States
- Epilepsy Center, Neurological Institute, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH 44106, United States
- Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology (M.I.T.) and Harvard, 415 Main St, Cambridge, MA 02142, United States
- Stanley Center for Psychiatric Research, Broad Institute of Harvard and M.I.T, 415 Main St., Cambridge, MA 02142, United States
| |
Collapse
|
6
|
He M, Wollmuth LP. Regulation of NMDAR activation efficiency by environmental factors and subunit composition. J Gen Physiol 2025; 157:e202413637. [PMID: 39576244 PMCID: PMC11586625 DOI: 10.1085/jgp.202413637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/28/2024] [Accepted: 11/07/2024] [Indexed: 11/24/2024] Open
Abstract
NMDA receptors (NMDAR) convert the major excitatory neurotransmitter glutamate into a synaptic signal. A key question is how efficiently the ion channel opens in response to the rapid exposure to presynaptic glutamate release. Here, we applied glutamate to single channel outside-out patches and measured the successes of channel openings and the latency to first opening to assay the activation efficiency of NMDARs under different physiological conditions and with different human subunit compositions. For GluN1/GluN2A receptors, we find that various factors, including intracellular ATP and GTP, can enhance the efficiency of activation presumably via the intracellular C-terminal domain. Notably, an energy-based internal solution or increasing the time between applications to increase recovery time improved efficiency. However, even under these optimized conditions and with a 1-s glutamate application, there remained around 10-15% inefficiency. Channel activation became more inefficient with brief synaptic-like pulses of glutamate at 2 ms. Of the different NMDAR subunit compositions, GluN2B-containing NMDARs showed the lowest success rate and longest latency to first openings, highlighting that they display the most distinct activation mechanism. In contrast, putative triheteromeric GluN1/GluN2A/GluN2B receptors showed high activation efficiency. Despite the low open probability, NMDARs containing either GluN2C or GluN2D subunits displayed high activation efficiency, nearly comparable with that for GluN2A-containing receptors. These results highlight that activation efficiency in NMDARs can be regulated by environmental surroundings and varies across different subunits.
Collapse
Affiliation(s)
- Miaomiao He
- Graduate Program in Biochemistry and Structural Biology, Stony Brook University, Stony Brook, NY, USA
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA
- Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY, USA
| | - Lonnie P. Wollmuth
- Department of Neurobiology and Behavior, Stony Brook University, Stony Brook, NY, USA
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY, USA
- Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
7
|
Tumdam R, Hussein Y, Garin-Shkolnik T, Stern S. NMDA Receptors in Neurodevelopmental Disorders: Pathophysiology and Disease Models. Int J Mol Sci 2024; 25:12366. [PMID: 39596430 PMCID: PMC11594297 DOI: 10.3390/ijms252212366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/08/2024] [Accepted: 11/15/2024] [Indexed: 11/28/2024] Open
Abstract
N-methyl-D-aspartate receptors (NMDARs) are critical components of the mammalian central nervous system, involved in synaptic transmission, plasticity, and neurodevelopment. This review focuses on the structural and functional characteristics of NMDARs, with a particular emphasis on the GRIN2 subunits (GluN2A-D). The diversity of GRIN2 subunits, driven by alternative splicing and genetic variants, significantly impacts receptor function, synaptic localization, and disease manifestation. The temporal and spatial expression of these subunits is essential for typical neural development, with each subunit supporting distinct phases of synaptic formation and plasticity. Disruptions in their developmental regulation are linked to neurodevelopmental disorders, underscoring the importance of understanding these dynamics in NDD pathophysiology. We explore the physiological properties and developmental regulation of these subunits, highlighting their roles in the pathophysiology of various NDDs, including ASD, epilepsy, and schizophrenia. By reviewing current knowledge and experimental models, including mouse models and human-induced pluripotent stem cells (hiPSCs), this article aims to elucidate different approaches through which the intricacies of NMDAR dysfunction in NDDs are currently being explored. The comprehensive understanding of NMDAR subunit composition and their mutations provides a foundation for developing targeted therapeutic strategies to address these complex disorders.
Collapse
Affiliation(s)
- Roshan Tumdam
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3103301, Israel
| | - Yara Hussein
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3103301, Israel
| | | | - Shani Stern
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3103301, Israel
| |
Collapse
|
8
|
Bender PA, Chakraborty S, Durham RJ, Berka V, Carrillo E, Jayaraman V. Bi-directional allosteric pathway in NMDA receptor activation and modulation. Nat Commun 2024; 15:8841. [PMID: 39396999 PMCID: PMC11471786 DOI: 10.1038/s41467-024-53181-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 10/04/2024] [Indexed: 10/15/2024] Open
Abstract
N-methyl-D-aspartate (NMDA) receptors are ionotropic glutamate receptors involved in learning and memory. NMDA receptors primarily comprise two GluN1 and two GluN2 subunits. The GluN2 subunit dictates biophysical receptor properties, including the extent of receptor activation and desensitization. GluN2A- and GluN2D-containing receptors represent two functional extremes. To uncover the conformational basis of their functional divergence, we utilize single-molecule fluorescence resonance energy transfer to probe the extracellular domains of these receptor subtypes under resting and ligand-bound conditions. We find that the conformational profile of the GluN2 amino-terminal domain correlates with the disparate functions of GluN2A- and GluN2D-containing receptors. Changes at the pre-transmembrane segments inversely correlate with those observed at the amino-terminal domain, confirming direct allosteric communication between these domains. Additionally, binding of a positive allosteric modulator at the transmembrane domain shifts the conformational profile of the amino-terminal domain towards the active state, revealing a bidirectional allosteric pathway between extracellular and transmembrane domains.
Collapse
Affiliation(s)
- Paula A Bender
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, USA
- Department of Biochemistry and Molecular Biology, Center for Membrane Biology, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Subhajit Chakraborty
- Department of Biochemistry and Molecular Biology, Center for Membrane Biology, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Ryan J Durham
- Department of Biochemistry and Molecular Biology, Center for Membrane Biology, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Vladimir Berka
- Department of Biochemistry and Molecular Biology, Center for Membrane Biology, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Elisa Carrillo
- Department of Biochemistry and Molecular Biology, Center for Membrane Biology, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Vasanthi Jayaraman
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX, USA.
- Department of Biochemistry and Molecular Biology, Center for Membrane Biology, University of Texas Health Science Center at Houston, Houston, TX, USA.
| |
Collapse
|
9
|
Wu Y, Zhu Z, Yang J, Wang J, Ji T, Zhu H, Peng W, Chen M, Zhao H. Insights into the terahertz response of L-glutamic acid and its receptor. Analyst 2024; 149:4605-4614. [PMID: 39037577 DOI: 10.1039/d4an00697f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
L-Glutamic acid (L-Glu) is a basic unit of proteins and also serves as an important neurotransmitter in the central nervous system. Its structural properties are critical for biological functions and selective receptor recognition. Although this molecule has been extensively studied, the low frequency vibrational behavior that is closely related to conformational changes and the intermolecular interactions between L-Glu and its receptors are still unclear. In this study, we acquired the fingerprint spectrum of L-Glu by using air plasma terahertz (THz) time-domain spectroscopy in the 0.5-18 THz range. The low frequency vibrational characteristics of L-Glu were investigated through density functional theory (DFT) calculations. The THz responses of the ligand binding domain of the NMDAR-L-Glu complex were studied by the ONIOM method, with a focus on discussing the normal modes and interactions of ligand L-Glu and water molecules. The results illustrate that THz spectroscopy exhibits a sensitive response to the influence of L-Glu on the structure of the NMDAR. The water molecules in proteins have various strong vibration modes in the THz band, showing specificity, diversity and complexity of vibrational behavior. There is potential for influencing and regulating the structural stability of the NMDAR-L-Glu complex through water molecules.
Collapse
Affiliation(s)
- Yu Wu
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China.
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhongjie Zhu
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.
| | - Jinrong Yang
- East China Normal University, Shanghai 200241, China
| | - Jie Wang
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.
| | - Te Ji
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.
| | - Huachun Zhu
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.
| | - Weiwei Peng
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.
| | - Min Chen
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China.
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.
| | - Hongwei Zhao
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai 201800, China.
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China.
| |
Collapse
|
10
|
Bleier J, Furtado de Mendonca PR, Habrian CH, Stanley C, Vyklicky V, Isacoff EY. Subtype-specific conformational landscape of NMDA receptor gating. Cell Rep 2024; 43:114634. [PMID: 39154344 PMCID: PMC11446236 DOI: 10.1016/j.celrep.2024.114634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/22/2024] [Accepted: 07/30/2024] [Indexed: 08/20/2024] Open
Abstract
N-methyl-D-aspartate receptors are ionotropic glutamate receptors that mediate synaptic transmission and plasticity. Variable GluN2 subunits in diheterotetrameric receptors with identical GluN1 subunits set very different functional properties. To understand this diversity, we use single-molecule fluorescence resonance energy transfer (smFRET) to measure the conformations of the ligand binding domain and modulatory amino-terminal domain of the common GluN1 subunit in receptors with different GluN2 subunits. Our results demonstrate a strong influence of the GluN2 subunits on GluN1 rearrangements, both in non-agonized and partially agonized activation intermediates, which have been elusive to structural analysis, and in the fully liganded state. Chimeric analysis reveals structural determinants that contribute to these subtype differences. Our study provides a framework for understanding the conformational landscape that supports highly divergent levels of activity, desensitization, and agonist potency in receptors with different GluN2s and could open avenues for the development of subtype-specific modulators.
Collapse
Affiliation(s)
- Julia Bleier
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | | | - Chris H Habrian
- Biophysics Graduate Group, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Cherise Stanley
- Department of Molecular & Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Vojtech Vyklicky
- Department of Molecular & Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Ehud Y Isacoff
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Molecular & Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Weill Neurohub, University of California, Berkeley, Berkeley, CA 94720, USA; Molecular Biology & Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA.
| |
Collapse
|
11
|
Montanucci L, Brünger T, Bhattarai N, Boßelmann CM, Kim S, Allen JP, Zhang J, Klöckner C, Fariselli P, May P, Lemke JR, Myers SJ, Yuan H, Traynelis SF, Lal D. Distances from ligands as main predictive features for pathogenicity and functional effect of variants in NMDA receptors. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.05.06.24306939. [PMID: 38766179 PMCID: PMC11100844 DOI: 10.1101/2024.05.06.24306939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
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.
Collapse
|
12
|
Bender PA, Chakraborty S, Durham RJ, Berka V, Carrillo E, Jayaraman V. Bi-directional allosteric pathway in NMDA receptor activation and modulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.16.589813. [PMID: 38659769 PMCID: PMC11042370 DOI: 10.1101/2024.04.16.589813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
N-methyl-D-aspartate (NMDA) receptors are ionotropic glutamate receptors involved in learning and memory. NMDA receptors primarily comprise two GluN1 and two GluN2 subunits. The GluN2 subunit dictates biophysical receptor properties, including the extent of receptor activation and desensitization. GluN2A- and GluN2D-containing receptors represent two functional extremes. To uncover the conformational basis of their functional divergence, we utilized single-molecule fluorescence resonance energy transfer to probe the extracellular domains of these receptor subtypes under resting and ligand-bound conditions. We find that the conformational profile of the GluN2 amino-terminal domain correlates with the disparate functions of GluN2A- and GluN2D-containing receptors. Changes at the pre-transmembrane segments inversely correlate with those observed at the amino-terminal domain, confirming direct allosteric communication between these domains. Additionally, binding of a positive allosteric modulator at the transmembrane domain shifts the conformational profile of the amino-terminal domain towards the active state, revealing a bidirectional allosteric pathway between extracellular and transmembrane domains.
Collapse
|
13
|
Xu Y, Song R, Perszyk RE, Chen W, Kim S, Park KL, Allen JP, Nocilla KA, Zhang J, XiangWei W, Tankovic A, McDaniels ED, Sheikh R, Mizu RK, Karamchandani MM, Hu C, Kusumoto H, Pecha J, Cappuccio G, Gaitanis J, Sullivan J, Shashi V, Petrovski S, Jauss RT, Lee HK, Bozarth X, Lynch DR, Helbig I, Pierson TM, Boerkoel CF, Myers SJ, Lemke JR, Benke TA, Yuan H, Traynelis SF. De novo GRIN variants in M3 helix associated with neurological disorders control channel gating of NMDA receptor. Cell Mol Life Sci 2024; 81:153. [PMID: 38538865 PMCID: PMC10973091 DOI: 10.1007/s00018-023-05069-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 11/23/2023] [Accepted: 11/27/2023] [Indexed: 05/18/2024]
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.
Collapse
Affiliation(s)
- Yuchen Xu
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Neurology, The First Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Rui Song
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Neurology, Qilu Hospital of Shandong University, Jinan, Shandong, 250012, China
| | - Riley E Perszyk
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Wenjuan Chen
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Psychiatry, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Sukhan Kim
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Center for Functional Evaluation of Rare Variants (CFERV), Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Kristen L Park
- Departments of Pediatrics and Neurology, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, USA
| | - James P Allen
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Kelsey A Nocilla
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Jing Zhang
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Wenshu XiangWei
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Pediatrics and Pediatric Epilepsy Center, Peking University First Hospital, Beijing, China
| | - Anel Tankovic
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Ellington D McDaniels
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Rehan Sheikh
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Ruth K Mizu
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Manish M Karamchandani
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Chun Hu
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Hirofumi Kusumoto
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Joseph Pecha
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Gerarda Cappuccio
- Department of Translational Medicine, Section of Pediatrics, Federico II University, Via Pansini 5, 80131, Naples, Italy
- Department of Pediatrics-Neurology, Baylor College of Medicine, Houston, TX, USA
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA
| | - John Gaitanis
- Hasbro Children's Hospital, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Jennifer Sullivan
- Department of Pediatrics, Division of Medical Genetics, Duke University, Durham, NC, USA
| | - Vandana Shashi
- Department of Pediatrics, Division of Medical Genetics, Duke University, Durham, NC, USA
| | - Slave Petrovski
- Ce Department of Medicine, University of Melbourne, Austin Health, Melbourne, VIC, Australia
- Centre for Genomics Research, Discovery Sciences, AstraZeneca, BioPharmaceuticals R&D, Cambridge, UK
| | - Robin-Tobias Jauss
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany
| | - Hyun Kyung Lee
- Provincial Medical Genetics Program, Department of Medical Genetics, University of British Columbia, Children's and Women's Health Centre of BC, Vancouver, B.C, V6H 3N1, Canada
| | - Xiuhua Bozarth
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA, USA
- Division of Pediatric Neurology, Department of Neurology, Seattle Children's Hospital, University of Washington, Seattle, WA, USA
| | - David R Lynch
- Departments of Pediatrics and Neurology, Perelman School of Medicine, Children's Hospital of Philadelphia, University of Pennsylvania, Philadelphia, PA, USA
| | - Ingo Helbig
- Division of Child Neurology, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Tyler Mark Pierson
- Department of Pediatrics and Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Center for the Undiagnosed Patient, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Cornelius F Boerkoel
- Provincial Medical Genetics Program, Department of Medical Genetics, University of British Columbia, Children's and Women's Health Centre of BC, Vancouver, B.C, V6H 3N1, Canada
| | - Scott J Myers
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Center for Functional Evaluation of Rare Variants (CFERV), Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Johannes R Lemke
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany
- Center for Rare Diseases, University of Leipzig Medical Center, Leipzig, Germany
| | - Timothy A Benke
- Departments of Pediatrics and Neurology, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, USA
| | - Hongjie Yuan
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Center for Functional Evaluation of Rare Variants (CFERV), Emory University School of Medicine, Atlanta, GA, 30322, USA.
| | - Stephen F Traynelis
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Center for Functional Evaluation of Rare Variants (CFERV), Emory University School of Medicine, Atlanta, GA, 30322, USA.
- Emory Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| |
Collapse
|
14
|
Bleier J, de Mendonca PRF, Habrian C, Stanley C, Vyklicky V, Isacoff EY. Conformational basis of subtype-specific allosteric control of NMDA receptor gating. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.10.579740. [PMID: 38370786 PMCID: PMC10871359 DOI: 10.1101/2024.02.10.579740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
N-methyl-D-aspartate receptors are ionotropic glutamate receptors that are integral to synaptic transmission and plasticity. Variable GluN2 subunits in diheterotetrameric receptors with identical GluN1 subunits set very different functional properties, which support their individual physiological roles in the nervous system. To understand the conformational basis of this diversity, we assessed the conformation of the common GluN1 subunit in receptors with different GluN2 subunits using single-molecule fluorescence resonance energy transfer (smFRET). We established smFRET sensors in the ligand binding domain and modulatory amino-terminal domain to study an apo-like state and partially liganded activation intermediates, which have been elusive to structural analysis. Our results demonstrate a strong, subtype-specific influence of apo and glutamate-bound GluN2 subunits on GluN1 rearrangements, suggesting a conformational basis for the highly divergent levels of receptor activity, desensitization and agonist potency. Chimeric analysis reveals structural determinants that contribute to the subtype differences. Our study provides a framework for understanding GluN2-dependent functional properties and could open new avenues for subtype-specific modulation.
Collapse
Affiliation(s)
- Julia Bleier
- Helen Wills Neuroscience Institute, University of California, Berkeley, California, 94720 USA
| | | | - Chris Habrian
- Department of Molecular & Cell Biology, University of California, Berkeley, California, 94720 USA
- Current address: Department of Molecular and Cellular Physiology, Stanford University School of Medicine, 279 Campus Drive, Stanford, CA 94305, USA
| | - Cherise Stanley
- Department of Molecular & Cell Biology, University of California, Berkeley, California, 94720 USA
| | - Vojtech Vyklicky
- Department of Molecular & Cell Biology, University of California, Berkeley, California, 94720 USA
- Current address: DIANA Biotechnologies, a.s. Průmyslová 596, 252 50 Vestec, Czech Republic
| | - Ehud Y. Isacoff
- Helen Wills Neuroscience Institute, University of California, Berkeley, California, 94720 USA
- Department of Molecular & Cell Biology, University of California, Berkeley, California, 94720 USA
- Weill Neurohub, University of California, Berkeley, California, 94720 USA
- Molecular Biology & Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, California, 94720, USA
| |
Collapse
|
15
|
Huang X, Chen W, Zhu S. Expression and Purification of Mammalian NMDA Receptor Protein for Functional Characterization. Methods Mol Biol 2024; 2799:13-27. [PMID: 38727900 DOI: 10.1007/978-1-0716-3830-9_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2024]
Abstract
N-methyl-D-aspartate (NMDA) receptors are critical for brain function and serve as drug targets for the treatment of neurological and psychiatric disorders. They typically form the tetrameric assembly of GluN1-GluN2 (2A to 2D) subtypes, with their diverse three-dimensional conformations linked with the physiologically relevant function in vivo. Purified proteins of tetrameric assembled NMDA receptors have broad applications in the structural elucidation, hybridoma technology for antibody production, and high-throughput drug screening. However, obtaining sufficient quantity and monodisperse NMDA receptor protein is still technically challenging. Here, we summarize a paradigm for the expression and purification of diverse NMDA receptor subtypes, with detailed descriptions on screening constructs by fluorescence size-exclusion chromatography (FSEC), generation of recombinant baculovirus, expression in the eukaryotic expression system, protein purification by affinity chromatography and size-exclusion chromatography (SEC), biochemical and functional validation assays.
Collapse
Affiliation(s)
- Xuejing Huang
- Department of Neurology, The First Affiliated Hospital, Institute of Neuroscience, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian, China
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Wanjin Chen
- Department of Neurology, The First Affiliated Hospital, Institute of Neuroscience, Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian, China
| | - Shujia Zhu
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
16
|
Myers SJ, Yuan H, Perszyk RE, Zhang J, Kim S, Nocilla KA, Allen JP, Bain JM, Lemke JR, Lal D, Benke TA, Traynelis SF. Classification of missense variants in the N-methyl-d-aspartate receptor GRIN gene family as gain- or loss-of-function. Hum Mol Genet 2023; 32:2857-2871. [PMID: 37369021 PMCID: PMC10508039 DOI: 10.1093/hmg/ddad104] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/30/2023] [Accepted: 06/01/2023] [Indexed: 06/29/2023] Open
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.
Collapse
Affiliation(s)
- Scott J Myers
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
- The Center for Functional Evaluation of Rare Variants (CFERV), Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Hongjie Yuan
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
- The Center for Functional Evaluation of Rare Variants (CFERV), Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Riley E Perszyk
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jing Zhang
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Sukhan Kim
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Kelsey A Nocilla
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - James P Allen
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
- The Center for Functional Evaluation of Rare Variants (CFERV), Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jennifer M Bain
- Department of Neurology, Division of Child Neurology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Johannes R Lemke
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig 04103, Germany
- Center for Rare Diseases, University of Leipzig Medical Center, Leipzig 04103, Germany
| | - Dennis Lal
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
- Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Cologne Center for Genomics (CCG), Medical Faculty of the University of Cologne, Köln 50923, Germany
| | - Timothy A Benke
- Department of Pediatrics, Pharmacology and Neurology, University of Colorado School of Medicine, and Children’s Hospital Colorado, Aurora, CO 80045, USA
| | - Stephen F Traynelis
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
- The Center for Functional Evaluation of Rare Variants (CFERV), Emory University School of Medicine, Atlanta, GA 30322, USA
- Emory Neurodegenerative Disease Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
17
|
Miles SA, Nillama JA, Hunter L. Tinker, Tailor, Soldier, Spy: The Diverse Roles That Fluorine Can Play within Amino Acid Side Chains. Molecules 2023; 28:6192. [PMID: 37687021 PMCID: PMC10489206 DOI: 10.3390/molecules28176192] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/17/2023] [Accepted: 08/17/2023] [Indexed: 09/10/2023] Open
Abstract
Side chain-fluorinated amino acids are useful tools in medicinal chemistry and protein science. In this review, we outline some general strategies for incorporating fluorine atom(s) into amino acid side chains and for elaborating such building blocks into more complex fluorinated peptides and proteins. We then describe the diverse benefits that fluorine can offer when located within amino acid side chains, including enabling 19F NMR and 18F PET imaging applications, enhancing pharmacokinetic properties, controlling molecular conformation, and optimizing target-binding.
Collapse
Affiliation(s)
| | | | - Luke Hunter
- School of Chemistry, The University of New South Wales (UNSW), Sydney 2052, Australia
| |
Collapse
|
18
|
Piniella D, Zafra F. Functional crosstalk of the glycine transporter GlyT1 and NMDA receptors. Neuropharmacology 2023; 232:109514. [PMID: 37003571 DOI: 10.1016/j.neuropharm.2023.109514] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/10/2023] [Accepted: 03/20/2023] [Indexed: 04/03/2023]
Abstract
NMDA-type glutamate receptors (NMDARs) constitute one of the main glutamate (Glu) targets in the central nervous system and are involved in synaptic plasticity, which is the molecular substrate of learning and memory. Hypofunction of NMDARs has been associated with schizophrenia, while overstimulation causes neuronal death in neurodegenerative diseases or in stroke. The function of NMDARs requires coincidental binding of Glu along with other cellular signals such as neuronal depolarization, and the presence of other endogenous ligands that modulate their activity by allosterism. Among these allosteric modulators are zinc, protons and Gly, which is an obligatory co-agonist. These characteristics differentiate NMDARs from other receptors, and their structural bases have begun to be established in recent years. In this review we focus on the crosstalk between Glu and glycine (Gly), whose concentration in the NMDAR microenvironment is maintained by various Gly transporters that remove or release it into the medium in a regulated manner. The GlyT1 transporter is particularly involved in this task, and has become a target of great interest for the treatment of schizophrenia since its inhibition leads to an increase in synaptic Gly levels that enhances the activity of NMDARs. However, the only drug that has completed phase III clinical trials did not yield the expected results. Notwithstanding, there are additional drugs that continue to be investigated, and it is hoped that knowledge gained from the recently published 3D structure of GlyT1 may allow the rational design of more effective new drugs.
Collapse
Affiliation(s)
- Dolores Piniella
- Centro de Biología Molecular Severo Ochoa, Facultad de Ciencias, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain; IdiPAZ, Institute of Health Carlos III (ISCIII), Spain
| | - Francisco Zafra
- Centro de Biología Molecular Severo Ochoa, Facultad de Ciencias, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain; IdiPAZ, Institute of Health Carlos III (ISCIII), Spain.
| |
Collapse
|
19
|
Chou TH, Kang H, Simorowski N, Traynelis SF, Furukawa H. Structural insights into assembly and function of GluN1-2C, GluN1-2A-2C, and GluN1-2D NMDARs. Mol Cell 2022; 82:4548-4563.e4. [PMID: 36309015 PMCID: PMC9722627 DOI: 10.1016/j.molcel.2022.10.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/02/2022] [Accepted: 10/05/2022] [Indexed: 11/06/2022]
Abstract
Neurotransmission mediated by diverse subtypes of N-methyl-D-aspartate receptors (NMDARs) is fundamental for basic brain functions and development as well as neuropsychiatric diseases and disorders. NMDARs are glycine- and glutamate-gated ion channels that exist as heterotetramers composed of obligatory GluN1 and GluN2(A-D) and/or GluN3(A-B). The GluN2C and GluN2D subunits form ion channels with distinct properties and spatio-temporal expression patterns. Here, we provide the structures of the agonist-bound human GluN1-2C NMDAR in the presence and absence of the GluN2C-selective positive allosteric potentiator (PAM), PYD-106, the agonist-bound GluN1-2A-2C tri-heteromeric NMDAR, and agonist-bound GluN1-2D NMDARs by single-particle electron cryomicroscopy. Our analysis shows unique inter-subunit and domain arrangements of the GluN2C NMDARs, which contribute to functional regulation and formation of the PAM binding pocket and is distinct from GluN2D NMDARs. Our findings here provide the fundamental blueprint to study GluN2C- and GluN2D-containing NMDARs, which are uniquely involved in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Tsung-Han Chou
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Hyunook Kang
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Noriko Simorowski
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Stephen F Traynelis
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Hiro Furukawa
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
20
|
Chang A, Liu JM, Nguyen K, Kumar PR. Expression optimization, purification, and biophysical characterization of a GluN2D-containing NMDA receptor. Protein Expr Purif 2022; 198:106129. [PMID: 35752385 DOI: 10.1016/j.pep.2022.106129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 11/15/2022]
Abstract
N-methyl-d-aspartate (NMDA) receptors are hetero-tetrameric ion channels typically consisting of two GluN1 and two GluN2 subunits. A GluN2D subunit containing NMDA receptor dysfunction has been implicated in several neurological diseases, including schizophrenia; however, the lack of a purified GluN2D containing NMDA receptor has been a hurdle for structural and biophysical studies. Here, we present expression and purification strategies to generate human GluN2D containing NMDA receptor, confirm its hetero-tetrameric form using fluorescence size exclusion chromatography (FSEC) and evaluated its suitability for structural studies. The purification methodology outlined here will help in the development of GluN2D specific channel modulators and enable structure activity relationship (SAR) studies.
Collapse
Affiliation(s)
- Aram Chang
- Physical Biochemistry and Molecular Design, Biotherapeutics and Medicinal Sciences, Biogen, Cambridge, MA, 02142, USA
| | - Justin M Liu
- Physical Biochemistry and Molecular Design, Biotherapeutics and Medicinal Sciences, Biogen, Cambridge, MA, 02142, USA
| | - Katrina Nguyen
- Physical Biochemistry and Molecular Design, Biotherapeutics and Medicinal Sciences, Biogen, Cambridge, MA, 02142, USA
| | - P Rajesh Kumar
- Physical Biochemistry and Molecular Design, Biotherapeutics and Medicinal Sciences, Biogen, Cambridge, MA, 02142, USA.
| |
Collapse
|
21
|
Tajima N, Simorowski N, Yovanno RA, Regan MC, Michalski K, Gómez R, Lau AY, Furukawa H. Development and characterization of functional antibodies targeting NMDA receptors. Nat Commun 2022; 13:923. [PMID: 35177668 PMCID: PMC8854693 DOI: 10.1038/s41467-022-28559-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 01/27/2022] [Indexed: 12/13/2022] Open
Abstract
N-methyl-D-aspartate receptors (NMDARs) are critically involved in basic brain functions and neurodegeneration as well as tumor invasiveness. Targeting specific subtypes of NMDARs with distinct activities has been considered an effective therapeutic strategy for neurological disorders and diseases. However, complete elimination of off-target effects of small chemical compounds has been challenging and thus, there is a need to explore alternative strategies for targeting NMDAR subtypes. Here we report identification of a functional antibody that specifically targets the GluN1-GluN2B NMDAR subtype and allosterically down-regulates ion channel activity as assessed by electrophysiology. Through biochemical analysis, x-ray crystallography, single-particle electron cryomicroscopy, and molecular dynamics simulations, we show that this inhibitory antibody recognizes the amino terminal domain of the GluN2B subunit and increases the population of the non-active conformational state. The current study demonstrates that antibodies may serve as specific reagents to regulate NMDAR functions for basic research and therapeutic objectives. Selective targeting individual subtypes of N-methyl-D-aspartate receptors (NMDARs) is a desirable therapeutic strategy for neurological disorders. Here, the authors report identification of a functional antibody that specifically targets and allosterically down-regulates ion channel activity of the GluN1—GluN2B NMDAR subtype.
Collapse
Affiliation(s)
- Nami Tajima
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, 11724, USA
| | - Noriko Simorowski
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, 11724, USA
| | - Remy A Yovanno
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, 725 N. Wolfe Street, WBSB 706, Baltimore, MD, 21205, USA
| | - Michael C Regan
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, 11724, USA
| | - Kevin Michalski
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, 11724, USA
| | - Ricardo Gómez
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, 11724, USA
| | - Albert Y Lau
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, 725 N. Wolfe Street, WBSB 706, Baltimore, MD, 21205, USA.
| | - Hiro Furukawa
- W.M. Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, 11724, USA.
| |
Collapse
|
22
|
Rapid Regulation of Glutamate Transport: Where Do We Go from Here? Neurochem Res 2022; 47:61-84. [PMID: 33893911 PMCID: PMC8542062 DOI: 10.1007/s11064-021-03329-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/08/2021] [Accepted: 04/13/2021] [Indexed: 01/03/2023]
Abstract
Glutamate is the predominant excitatory neurotransmitter in the mammalian central nervous system (CNS). A family of five Na+-dependent transporters maintain low levels of extracellular glutamate and shape excitatory signaling. Shortly after the research group of the person being honored in this special issue (Dr. Baruch Kanner) cloned one of these transporters, his group and several others showed that their activity can be acutely (within minutes to hours) regulated. Since this time, several different signals and post-translational modifications have been implicated in the regulation of these transporters. In this review, we will provide a brief introduction to the distribution and function of this family of glutamate transporters. This will be followed by a discussion of the signals that rapidly control the activity and/or localization of these transporters, including protein kinase C, ubiquitination, glutamate transporter substrates, nitrosylation, and palmitoylation. We also include the results of our attempts to define the role of palmitoylation in the regulation of GLT-1 in crude synaptosomes. In some cases, the mechanisms have been fairly well-defined, but in others, the mechanisms are not understood. In several cases, contradictory phenomena have been observed by more than one group; we describe these studies with the goal of identifying the opportunities for advancing the field. Abnormal glutamatergic signaling has been implicated in a wide variety of psychiatric and neurologic disorders. Although recent studies have begun to link regulation of glutamate transporters to the pathogenesis of these disorders, it will be difficult to determine how regulation influences signaling or pathophysiology of glutamate without a better understanding of the mechanisms involved.
Collapse
|
23
|
Hansen KB, Wollmuth LP, Bowie D, Furukawa H, Menniti FS, Sobolevsky AI, Swanson GT, Swanger SA, Greger IH, Nakagawa T, McBain CJ, Jayaraman V, Low CM, Dell'Acqua ML, Diamond JS, Camp CR, Perszyk RE, Yuan H, Traynelis SF. Structure, Function, and Pharmacology of Glutamate Receptor Ion Channels. Pharmacol Rev 2021; 73:298-487. [PMID: 34753794 PMCID: PMC8626789 DOI: 10.1124/pharmrev.120.000131] [Citation(s) in RCA: 361] [Impact Index Per Article: 90.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Many physiologic effects of l-glutamate, the major excitatory neurotransmitter in the mammalian central nervous system, are mediated via signaling by ionotropic glutamate receptors (iGluRs). These ligand-gated ion channels are critical to brain function and are centrally implicated in numerous psychiatric and neurologic disorders. There are different classes of iGluRs with a variety of receptor subtypes in each class that play distinct roles in neuronal functions. The diversity in iGluR subtypes, with their unique functional properties and physiologic roles, has motivated a large number of studies. Our understanding of receptor subtypes has advanced considerably since the first iGluR subunit gene was cloned in 1989, and the research focus has expanded to encompass facets of biology that have been recently discovered and to exploit experimental paradigms made possible by technological advances. Here, we review insights from more than 3 decades of iGluR studies with an emphasis on the progress that has occurred in the past decade. We cover structure, function, pharmacology, roles in neurophysiology, and therapeutic implications for all classes of receptors assembled from the subunits encoded by the 18 ionotropic glutamate receptor genes. SIGNIFICANCE STATEMENT: Glutamate receptors play important roles in virtually all aspects of brain function and are either involved in mediating some clinical features of neurological disease or represent a therapeutic target for treatment. Therefore, understanding the structure, function, and pharmacology of this class of receptors will advance our understanding of many aspects of brain function at molecular, cellular, and system levels and provide new opportunities to treat patients.
Collapse
Affiliation(s)
- Kasper B Hansen
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Lonnie P Wollmuth
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Derek Bowie
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Hiro Furukawa
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Frank S Menniti
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Alexander I Sobolevsky
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Geoffrey T Swanson
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Sharon A Swanger
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Ingo H Greger
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Terunaga Nakagawa
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Chris J McBain
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Vasanthi Jayaraman
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Chian-Ming Low
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Mark L Dell'Acqua
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Jeffrey S Diamond
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Chad R Camp
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Riley E Perszyk
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Hongjie Yuan
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Stephen F Traynelis
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| |
Collapse
|
24
|
Menga A, Favia M, Spera I, Vegliante MC, Gissi R, De Grassi A, Laera L, Campanella A, Gerbino A, Carrà G, Canton M, Loizzi V, Pierri CL, Cormio G, Mazzone M, Castegna A. N-acetylaspartate release by glutaminolytic ovarian cancer cells sustains protumoral macrophages. EMBO Rep 2021; 22:e51981. [PMID: 34260142 PMCID: PMC8419692 DOI: 10.15252/embr.202051981] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 06/10/2021] [Accepted: 06/21/2021] [Indexed: 02/01/2023] Open
Abstract
Glutaminolysis is known to correlate with ovarian cancer aggressiveness and invasion. However, how this affects the tumor microenvironment is elusive. Here, we show that ovarian cancer cells become addicted to extracellular glutamine when silenced for glutamine synthetase (GS), similar to naturally occurring GS-low, glutaminolysis-high ovarian cancer cells. Glutamine addiction elicits a crosstalk mechanism whereby cancer cells release N-acetylaspartate (NAA) which, through the inhibition of the NMDA receptor, and synergistically with IL-10, enforces GS expression in macrophages. In turn, GS-high macrophages acquire M2-like, tumorigenic features. Supporting this in␣vitro model, in silico data and the analysis of ascitic fluid isolated from ovarian cancer patients prove that an M2-like macrophage phenotype, IL-10 release, and NAA levels positively correlate with disease stage. Our study uncovers the unprecedented role of glutamine metabolism in modulating macrophage polarization in highly invasive ovarian cancer and highlights the anti-inflammatory, protumoral function of NAA.
Collapse
Affiliation(s)
- Alessio Menga
- Department of Molecular Biotechnologies and Health SciencesUniversity of TurinTurinItaly
- Department of Biosciences, Biotechnologies and BiopharmaceuticsUniversity of BariBariItaly
- Molecular Biotechnology CenterTurinItaly
| | - Maria Favia
- Department of Biosciences, Biotechnologies and BiopharmaceuticsUniversity of BariBariItaly
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly
| | - Iolanda Spera
- Department of Biosciences, Biotechnologies and BiopharmaceuticsUniversity of BariBariItaly
| | - Maria C Vegliante
- Haematology and Cell Therapy UnitIRCCS‐Istituto Tumori ‘Giovanni Paolo II'BariItaly
| | - Rosanna Gissi
- Department of Biosciences, Biotechnologies and BiopharmaceuticsUniversity of BariBariItaly
| | - Anna De Grassi
- Department of Biosciences, Biotechnologies and BiopharmaceuticsUniversity of BariBariItaly
| | - Luna Laera
- Department of Biosciences, Biotechnologies and BiopharmaceuticsUniversity of BariBariItaly
| | - Annalisa Campanella
- Department of Biosciences, Biotechnologies and BiopharmaceuticsUniversity of BariBariItaly
| | - Andrea Gerbino
- Department of Biosciences, Biotechnologies and BiopharmaceuticsUniversity of BariBariItaly
| | - Giovanna Carrà
- Molecular Biotechnology CenterTurinItaly
- Department of Clinical and Biological SciencesUniversity of TurinOrbassanoItaly
| | - Marcella Canton
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza ‐ IRPPadovaItaly
| | - Vera Loizzi
- Policlinico University of Bari “Aldo Moro”BariItaly
| | - Ciro L Pierri
- Department of Biosciences, Biotechnologies and BiopharmaceuticsUniversity of BariBariItaly
| | - Gennaro Cormio
- Policlinico University of Bari “Aldo Moro”BariItaly
- Gynecologic Oncology UnitIRCCSIstituto Tumori Giovanni Paolo IIBariItaly
| | - Massimiliano Mazzone
- Department of Molecular Biotechnologies and Health SciencesUniversity of TurinTurinItaly
- Molecular Biotechnology CenterTurinItaly
- Laboratory of Tumor Inflammation and AngiogenesisCenter for Cancer BiologyDepartment of OncologyKU LeuvenLeuvenBelgium
| | - Alessandra Castegna
- Department of Biosciences, Biotechnologies and BiopharmaceuticsUniversity of BariBariItaly
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza ‐ IRPPadovaItaly
| |
Collapse
|
25
|
McDaniel MJ, Ogden KK, Kell SA, Burger PB, Liotta DC, Traynelis SF. NMDA receptor channel gating control by the pre-M1 helix. J Gen Physiol 2021; 152:151592. [PMID: 32221541 PMCID: PMC7141592 DOI: 10.1085/jgp.201912362] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 08/09/2019] [Accepted: 02/14/2020] [Indexed: 01/10/2023] Open
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.
Collapse
Affiliation(s)
- Miranda J McDaniel
- Department of Pharmacology, Rollins Research Center, Emory University, Atlanta, GA
| | - Kevin K Ogden
- Department of Pharmacology, Rollins Research Center, Emory University, Atlanta, GA
| | - Steven A Kell
- Department of Pharmacology, Rollins Research Center, Emory University, Atlanta, GA.,Department of Chemistry, Emory University, Atlanta, GA
| | | | | | - Stephen F Traynelis
- Department of Pharmacology, Rollins Research Center, Emory University, Atlanta, GA
| |
Collapse
|
26
|
Furukawa H, Simorowski N, Michalski K. Effective production of oligomeric membrane proteins by EarlyBac-insect cell system. Methods Enzymol 2021; 653:3-19. [PMID: 34099177 DOI: 10.1016/bs.mie.2020.12.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Despite major advances in methodologies for membrane protein production over the last two decades, there remain challenging protein complexes that are technically difficult to yield by conventional recombinant expression methods. A large number of these proteins are multimeric membrane proteins from eukaryotic species, which are required to pass through stringent quality control mechanisms of host cells for proper folding and complex assembly. Here, we describe the development procedure to improve the production efficiency of multi-oligomeric membrane protein complexes in insect cells and recombinant baculovirus, which involves screening of promoters, enhancers, and untranslated regions for expression levels, using calcium homeostasis modulator (CALHM) and N-methyl-d-aspartate receptor (NMDAR) proteins as examples. We demonstrate that our insect cell expression strategy is effective in expression of both multi-homomeric CALHM proteins and multi-heteromeric NMDARs.
Collapse
Affiliation(s)
- Hiro Furukawa
- WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, United States.
| | - Noriko Simorowski
- WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, United States
| | - Kevin Michalski
- WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, United States
| |
Collapse
|
27
|
Strong KL, Epplin MP, Ogden KK, Burger PB, Kaiser TM, Wilding TJ, Kusumoto H, Camp CR, Shaulsky G, Bhattacharya S, Perszyk RE, Menaldino DS, McDaniel MJ, Zhang J, Le P, Banke TG, Hansen KB, Huettner JE, Liotta DC, Traynelis SF. Distinct GluN1 and GluN2 Structural Determinants for Subunit-Selective Positive Allosteric Modulation of N-Methyl-d-aspartate Receptors. ACS Chem Neurosci 2021; 12:79-98. [PMID: 33326224 DOI: 10.1021/acschemneuro.0c00561] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
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.
Collapse
Affiliation(s)
- Katie L. Strong
- Department of Pharmacology, Emory University, 1510 Clifton Road, Atlanta, Georgia 30322, United States
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Matthew P. Epplin
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Kevin K. Ogden
- Department of Pharmacology, Emory University, 1510 Clifton Road, Atlanta, Georgia 30322, United States
| | - Pieter B. Burger
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Thomas M. Kaiser
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Timothy J. Wilding
- Department of Cell Biology and Physiology, Washington University, St Louis, Missouri 63110, United States
| | - Hiro Kusumoto
- Department of Pharmacology, Emory University, 1510 Clifton Road, Atlanta, Georgia 30322, United States
| | - Chad R. Camp
- Department of Pharmacology, Emory University, 1510 Clifton Road, Atlanta, Georgia 30322, United States
| | - Gil Shaulsky
- Department of Pharmacology, Emory University, 1510 Clifton Road, Atlanta, Georgia 30322, United States
| | - Subhrajit Bhattacharya
- Department of Drug Discovery and Development, Auburn University, Auburn, Alabama 36849, United States
| | - Riley E. Perszyk
- Department of Pharmacology, Emory University, 1510 Clifton Road, Atlanta, Georgia 30322, United States
| | - David S. Menaldino
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Miranda J. McDaniel
- Department of Pharmacology, Emory University, 1510 Clifton Road, Atlanta, Georgia 30322, United States
| | - Jing Zhang
- Department of Pharmacology, Emory University, 1510 Clifton Road, Atlanta, Georgia 30322, United States
| | - Phuong Le
- Department of Pharmacology, Emory University, 1510 Clifton Road, Atlanta, Georgia 30322, United States
| | - Tue G. Banke
- Department of Pharmacology, Emory University, 1510 Clifton Road, Atlanta, Georgia 30322, United States
| | - Kasper B. Hansen
- Department of Pharmacology, Emory University, 1510 Clifton Road, Atlanta, Georgia 30322, United States
- Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Division for Biological Sciences, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
| | - James E. Huettner
- Department of Cell Biology and Physiology, Washington University, St Louis, Missouri 63110, United States
| | - Dennis C. Liotta
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Stephen F. Traynelis
- Department of Pharmacology, Emory University, 1510 Clifton Road, Atlanta, Georgia 30322, United States
| |
Collapse
|
28
|
Maolanon A, Papangelis A, Kawiecki D, Mou TC, Syrenne JT, Yi F, Hansen KB, Clausen RP. Stereoselective synthesis of novel 2'-(S)-CCG-IV analogues as potent NMDA receptor agonists. Eur J Med Chem 2020; 212:113099. [PMID: 33383257 DOI: 10.1016/j.ejmech.2020.113099] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 12/03/2020] [Accepted: 12/07/2020] [Indexed: 11/24/2022]
Abstract
We developed a versatile stereoselective route for the synthesis of new 2'-(S)-CCG-IV analogues. The route allows for late stage diversification and thereby provides access to a great variety of conformationally restricted cyclopropyl glutamate analogues. A selection of the 2'-(S)-CCG-IV analogues were evaluated using two-electrode voltage-clamp electrophysiology at recombinant GluN1/GluN2A-D receptors, demonstrating that agonists can be developed with GluN2 subunit-dependent potency and agonist efficacy. We also describe a crystal structure of the GluN2A agonist binding domain in complex with 2'-butyl-(S)-CCG-IV that determines the position of 2'-substituents in (S)-CCG-IV agonists in the glutamate binding site and provides further insight to the structural determinants of their agonist efficacy. The stereoselective synthesis described here enables versatile and straight-forward modifications to diverse analogues of interest for the development of potent subtype-specific NMDA receptor agonists and other applications.
Collapse
Affiliation(s)
- Alex Maolanon
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK, 2100, Copenhagen, Denmark
| | - Athanasios Papangelis
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK, 2100, Copenhagen, Denmark
| | - David Kawiecki
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK, 2100, Copenhagen, Denmark
| | - Tung-Chung Mou
- Division of Biological Sciences, University of Montana, Missoula, MT, 59812, USA; Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, MT, 59812, USA
| | - Jed T Syrenne
- Division of Biological Sciences, University of Montana, Missoula, MT, 59812, USA; Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, MT, 59812, USA; Center for Structural and Functional Neuroscience, University of Montana, Missoula, MT, 59812, USA
| | - Feng Yi
- Division of Biological Sciences, University of Montana, Missoula, MT, 59812, USA; Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, MT, 59812, USA; Center for Structural and Functional Neuroscience, University of Montana, Missoula, MT, 59812, USA
| | - Kasper B Hansen
- Division of Biological Sciences, University of Montana, Missoula, MT, 59812, USA; Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, MT, 59812, USA; Center for Structural and Functional Neuroscience, University of Montana, Missoula, MT, 59812, USA
| | - Rasmus P Clausen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK, 2100, Copenhagen, Denmark.
| |
Collapse
|
29
|
Kalev-Zylinska ML, Morel-Kopp MC, Ward CM, Hearn JI, Hamilton JR, Bogdanova AY. Ionotropic glutamate receptors in platelets: opposing effects and a unifying hypothesis. Platelets 2020; 32:998-1008. [PMID: 33284715 DOI: 10.1080/09537104.2020.1852542] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Ionotropic glutamate receptors include α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPAR), kainate receptors (KAR), and N-methyl-D-aspartate receptors (NMDAR). All function as cation channels; AMPAR and KAR are more permeable to sodium and NMDAR to calcium ions. Compared to the brain, receptor assemblies in platelets are unusual, suggesting distinctive functionalities.There is convincing evidence that AMPAR and KAR amplify platelet function and thrombus formation in vitro and in vivo. Transgenic mice lacking GluA1 and GluK2 (AMPAR and KAR subunits, respectively) have longer bleeding times and prolonged time to thrombosis in an arterial model. In humans, rs465566 KAR gene polymorphism associates with altered in vitro platelet responses suggesting enhanced aspirin effect. The NMDAR contribution to platelet function is less well defined. NMDA at low concentrations (≤10 μM) inhibits platelet aggregation and high concentrations (≥100 μM) have no effect. However, open NMDAR channel blockers interfere with platelet activation and aggregation induced by other agonists in vitro; anti-GluN1 antibodies interfere with thrombus formation under high shear rates ex vivo; and rats vaccinated with GluN1 develop iron deficiency anemia suggestive of mild chronic bleeding. In this review, we summarize data on glutamate receptors in platelets and propose a unifying model that reconciles some of the opposing effects observed.
Collapse
Affiliation(s)
- Maggie L Kalev-Zylinska
- Blood and Cancer Biology Laboratory, Department of Molecular Medicine & Pathology, University of Auckland, Auckland, New Zealand.,Department of Pathology and Laboratory Medicine, LabPlus Haematology, Auckland City Hospital, Auckland, New Zealand
| | - Marie-Christine Morel-Kopp
- Department of Haematology and Transfusion Medicine, Royal North Shore Hospital, Sydney, Australia.,Northern Blood Research Centre, Kolling Institute, University of Sydney, Sydney, Australia
| | - Christopher M Ward
- Department of Haematology and Transfusion Medicine, Royal North Shore Hospital, Sydney, Australia.,Northern Blood Research Centre, Kolling Institute, University of Sydney, Sydney, Australia
| | - James I Hearn
- Blood and Cancer Biology Laboratory, Department of Molecular Medicine & Pathology, University of Auckland, Auckland, New Zealand
| | - Justin R Hamilton
- Australian Centre for Blood Diseases, Monash University, Melbourne, Australia
| | - Anna Y Bogdanova
- Red Blood Cell Research Group, Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zürich, Switzerland.,Zurich Center for Integrative Human Physiology, University of Zurich, Zürich, Switzerland
| |
Collapse
|
30
|
A comprehensive description of GluN2B-selective N-methyl-D-aspartate (NMDA) receptor antagonists. Eur J Med Chem 2020; 200:112447. [DOI: 10.1016/j.ejmech.2020.112447] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 05/06/2020] [Accepted: 05/08/2020] [Indexed: 12/17/2022]
|
31
|
Chou TH, Tajima N, Romero-Hernandez A, Furukawa H. Structural Basis of Functional Transitions in Mammalian NMDA Receptors. Cell 2020; 182:357-371.e13. [PMID: 32610085 PMCID: PMC8278726 DOI: 10.1016/j.cell.2020.05.052] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 04/22/2020] [Accepted: 05/27/2020] [Indexed: 12/18/2022]
Abstract
Excitatory neurotransmission meditated by glutamate receptors including N-methyl-D-aspartate receptors (NMDARs) is pivotal to brain development and function. NMDARs are heterotetramers composed of GluN1 and GluN2 subunits, which bind glycine and glutamate, respectively, to activate their ion channels. Despite importance in brain physiology, the precise mechanisms by which activation and inhibition occur via subunit-specific binding of agonists and antagonists remain largely unknown. Here, we show the detailed patterns of conformational changes and inter-subunit and -domain reorientation leading to agonist-gating and subunit-dependent competitive inhibition by providing multiple structures in distinct ligand states at 4 Å or better. The structures reveal that activation and competitive inhibition by both GluN1 and GluN2 antagonists occur by controlling the tension of the linker between the ligand-binding domain and the transmembrane ion channel of the GluN2 subunit. Our results provide detailed mechanistic insights into NMDAR pharmacology, activation, and inhibition, which are fundamental to the brain physiology.
Collapse
Affiliation(s)
- Tsung-Han Chou
- WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Nami Tajima
- WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Annabel Romero-Hernandez
- WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; Watson School of Biological Sciences, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Hiro Furukawa
- WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; Watson School of Biological Sciences, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
32
|
Wang JX, Irvine MW, Burnell ES, Sapkota K, Thatcher RJ, Li M, Simorowski N, Volianskis A, Collingridge GL, Monaghan DT, Jane DE, Furukawa H. Structural basis of subtype-selective competitive antagonism for GluN2C/2D-containing NMDA receptors. Nat Commun 2020; 11:423. [PMID: 31969570 PMCID: PMC6976569 DOI: 10.1038/s41467-020-14321-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 12/18/2019] [Indexed: 12/15/2022] Open
Abstract
N-Methyl-D-aspartate receptors (NMDARs) play critical roles in the central nervous system. Their heterotetrameric composition generates subtypes with distinct functional properties and spatio-temporal distribution in the brain, raising the possibility for subtype-specific targeting by pharmacological means for treatment of neurological diseases. While specific compounds for GluN2A and GluN2B-containing NMDARs are well established, those that target GluN2C and GluN2D are currently underdeveloped with low potency and uncharacterized binding modes. Here, using electrophysiology and X-ray crystallography, we show that UBP791 ((2S*,3R*)-1-(7-(2-carboxyethyl)phenanthrene-2-carbonyl)piperazine-2,3-dicarboxylic acid) inhibits GluN2C/2D with 40-fold selectivity over GluN2A-containing receptors, and that a methionine and a lysine residue in the ligand binding pocket (GluN2D-Met763/Lys766, GluN2C-Met736/Lys739) are the critical molecular elements for the subtype-specific binding. These findings led to development of UBP1700 ((2S*,3R*)-1-(7-(2-carboxyvinyl)phenanthrene-2-carbonyl)piperazine-2,3-dicarboxylic acid) which shows over 50-fold GluN2C/2D-selectivity over GluN2A with potencies in the low nanomolar range. Our study shows that the L-glutamate binding site can be targeted for GluN2C/2D-specific inhibition.
Collapse
Affiliation(s)
- Jue Xiang Wang
- WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
- Watson School of Biological Sciences, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| | - Mark W Irvine
- Glutamate Research Group, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK
| | - Erica S Burnell
- Glutamate Research Group, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK
- School of Chemistry, National University of Ireland Galway, Galway, H91TK33, Ireland
| | - Kiran Sapkota
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5800, USA
| | - Robert J Thatcher
- Glutamate Research Group, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK
| | - Minjun Li
- WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| | - Noriko Simorowski
- WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| | - Arturas Volianskis
- Centre for Neuroscience and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Graham L Collingridge
- Glutamate Research Group, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK
- Tanz Centre for Research in Neurodegenerative Diseases, Department of Physiology, University of Toronto, Krembil Discovery Tower, 60 Leonard Avenue, Toronto, ON, M5T 0S8, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Avenue, Toronto, ON, M5G 1X5, Canada
| | - Daniel T Monaghan
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5800, USA
| | - David E Jane
- Glutamate Research Group, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK.
| | - Hiro Furukawa
- WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA.
- Watson School of Biological Sciences, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA.
| |
Collapse
|
33
|
GRIN2D/GluN2D NMDA receptor: Unique features and its contribution to pediatric developmental and epileptic encephalopathy. Eur J Paediatr Neurol 2020; 24:89-99. [PMID: 31918992 PMCID: PMC7035963 DOI: 10.1016/j.ejpn.2019.12.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 12/06/2019] [Indexed: 12/14/2022]
Abstract
N-methyl-d-aspartate receptors (NMDARs), a subset of ligand-gated ionotropic glutamate receptors, are critical for learning, memory, and neuronal development. However, when NMDAR subunits are mutated, a host of neuropathological conditions can occur, including epilepsy. Recently, genetic variation within the GRIN2D gene, which encodes the GluN2D subunit of the NMDAR, has been associated with a set of early-onset neurological diseases, notably developmental and epileptic encephalopathy (DEE). Importantly, patients with GRIN2D variants are largely refractory to conventional anti-epileptic drug (AED) treatment, highlighting the need to further understand the distinctive characteristics of GluN2D in neurological and pathological functions. In this review, we first summarize GluN2D's unique spatial and temporal expression patterns, electrophysiological profiles, and contributions to both pre- and postsynaptic signaling. Next, we review thirteen unique case studies from DEE patients harboring ten different causal GRIN2D variants. These patients are highly heterogenous, manifesting multiple seizure types, electroencephalographic recordings, and neurological and developmental outcomes. Lastly, this review concludes by highlighting the difficulty in treating patients with DEE-associated GRIN2D variants, and stresses the need for selective therapeutic agents delivered within a precise time window.
Collapse
|
34
|
Polli FS, Kohlmeier KA. Alterations in NMDAR-mediated signaling within the laterodorsal tegmental nucleus are associated with prenatal nicotine exposure. Neuropharmacology 2019; 158:107744. [DOI: 10.1016/j.neuropharm.2019.107744] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 07/23/2019] [Accepted: 08/18/2019] [Indexed: 12/18/2022]
|
35
|
Isoform-specific Inhibition of N-methyl-D-aspartate Receptors by Bile Salts. Sci Rep 2019; 9:10068. [PMID: 31296930 PMCID: PMC6624251 DOI: 10.1038/s41598-019-46496-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 06/18/2019] [Indexed: 12/27/2022] Open
Abstract
The N-methyl-D-aspartate subfamily of ionotropic glutamate receptors (NMDARs) is well known for its important roles in the central nervous system (CNS), e.g. learning and memory formation. Besides the CNS, NMDARs are also expressed in numerous peripheral tissues including the pancreas, kidney, stomach, and blood cells, where an understanding of their physiological and pathophysiological roles is only evolving. Whereas subunit composition increases functional diversity of NMDARs, a great number of endogenous cues tune receptor signaling. Here, we characterized the effects of the steroid bile salts cholate and chenodeoxycholate (CDC) on recombinantly expressed NMDARs of defined molecular composition. CDC inhibited NMDARs in an isoform-dependent manner, preferring GluN2D and GluN3B over GluN2A and GluN2B receptors. Determined IC50 values were in the range of bile salt serum concentrations in severe cholestatic disease states, pointing at a putative pathophysiological significance of the identified receptor modulation. Both pharmacological and molecular simulation analyses indicate that CDC acts allosterically on GluN2D, whereas it competes with agonist binding on GluN3B receptors. Such differential modes of inhibition may allow isoform-specific targeted interference with the NMDAR/bile salt interaction. In summary, our study provides further molecular insight into the modulation of NMDARs by endogenous steroids and points at a putative pathophysiological role of the receptors in cholestatic disease.
Collapse
|
36
|
Dissecting diverse functions of NMDA receptors by structural biology. Curr Opin Struct Biol 2019; 54:34-42. [PMID: 30703613 DOI: 10.1016/j.sbi.2018.12.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 12/09/2018] [Accepted: 12/20/2018] [Indexed: 12/18/2022]
Abstract
N-Methyl-d-aspartate receptors (NMDARs) are glutamate-gated ion channels, which are critically involved in brain development, learning and memory, cognition, as well as a number of neurological diseases and disorders. Structural biology of NMDARs has been challenging due to technical difficulties associated with assembling a number of different membrane protein subunits. Here, we review historical X-ray crystallographic studies on isolated extracellular domains, which are still the most effective mean to delineate compound binding modes, as well as the most recent studies using electron cryo-microscopy (cryo-EM). A number of NMDAR structures accumulated over the past 15 years provide insights into the hetero-tetrameric assembly pattern, pharmacological specificities elicited by subtypes and alternative splicing, and potential patterns of conformational dynamics; however, many more important unanswered questions remain.
Collapse
|
37
|
Regan MC, Zhu Z, Yuan H, Myers SJ, Menaldino DS, Tahirovic YA, Liotta DC, Traynelis SF, Furukawa H. Structural elements of a pH-sensitive inhibitor binding site in NMDA receptors. Nat Commun 2019; 10:321. [PMID: 30659174 PMCID: PMC6338780 DOI: 10.1038/s41467-019-08291-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 12/18/2018] [Indexed: 12/31/2022] Open
Abstract
Context-dependent inhibition of N-methyl-D-aspartate (NMDA) receptors has important therapeutic implications for the treatment of neurological diseases that are associated with altered neuronal firing and signaling. This is especially true in stroke, where the proton concentration in the afflicted area can increase by an order of magnitude. A class of allosteric inhibitors, the 93-series, shows greater potency against GluN1-GluN2B NMDA receptors in such low pH environments, allowing targeted therapy only within the ischemic region. Here we map the 93-series compound binding site in the GluN1-GluN2B NMDA receptor amino terminal domain and show that the interaction of the N-alkyl group with a hydrophobic cage of the binding site is critical for pH-dependent inhibition. Mutation of residues in the hydrophobic cage alters pH-dependent potency, and remarkably, can convert inhibitors into potentiators. Our study provides a foundation for the development of highly specific neuroprotective compounds for the treatment of neurological diseases. Context-dependent inhibition of NMDA receptors has important therapeutic implications for treatment of neurological diseases. Here, the authors use structural biology and biophysics to describe the basis for pH-dependent inhibition for a class of allosteric NMDAR inhibitors, called the 93-series.
Collapse
Affiliation(s)
- Michael C Regan
- WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| | - Zongjian Zhu
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, 30322, USA.,Department of Neonatology, First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shaanxi, China
| | - Hongjie Yuan
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Scott J Myers
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Dave S Menaldino
- Department of Chemistry, Emory University, Atlanta, GA, 30322, USA
| | | | - Dennis C Liotta
- Department of Chemistry, Emory University, Atlanta, GA, 30322, USA
| | - Stephen F Traynelis
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Hiro Furukawa
- WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA.
| |
Collapse
|
38
|
Wells G, Yuan H, McDaniel MJ, Kusumoto H, Snyder JP, Liotta DC, Traynelis SF. The GluN2B-Glu413Gly NMDA receptor variant arising from a de novo GRIN2B mutation promotes ligand-unbinding and domain opening. Proteins 2018; 86:1265-1276. [PMID: 30168177 PMCID: PMC6774441 DOI: 10.1002/prot.25595] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 07/17/2018] [Accepted: 08/18/2018] [Indexed: 12/19/2022]
Abstract
N-methyl-D-aspartate (NMDA) receptors are transmembrane glutamate-binding ion channels that mediate neurotransmission in mammals. NMDA receptor subunits are tetrameric complexes of GluN1 and GluN2A-D subunits, encoded by the GRIN gene family. Of these subunits, GluN2B is suggested to be required for normal development of the central nervous system. A mutation identified in a patient with developmental delay, E413G, resides in the GluN2B ligand-binding domain and substantially reduces glutamate potency by an unknown mechanism. GluN2B Gly413, though near the agonist, is not in van der Waals contact with glutamate. Visual analysis of the GluN2B structure with the E413G mutation modeled suggests that replacement of Glu with Gly at this position increases solvent access to the ligand-binding domain. This was confirmed by molecular modeling, which showed that the ligand is more mobile in GluN2B-E413G than WT GluN2B. Evaluation of agonist occupancy using random accelerated molecular dynamics (RAMD) simulations predicts that the glutamate exits the binding-site more rapidly for GluN2B-E413G than WT receptors. This analysis was extended to other binding-site mutations, which produced qualitative agreement between experimentally determined EC50 values, deactivation time constants, and ligand motion within the binding-site. Furthermore, long sub-microsecond molecular dynamics simulations of the bi-lobed ligand-binding domain revealed that it adopted a cleft-open ligand-free state more often for GluN2B-E413G than wild-type GluN2B. This is consistent with the idea that L-glutamate binding is altered such that the ligand-binding domain occupies the open-cleft conformation associated with the closed channel.
Collapse
Affiliation(s)
- Gordon Wells
- African Health Research Institute, Steyn Lab, K-RITH Tower, Nelson R. Mandela Medical School, Durban, South Africa
- Department of Chemistry, Emory University, Atlanta, Georgia
| | - Hongjie Yuan
- Department of Pharmacology, Emory University, Atlanta, Georgia
| | | | | | - James P Snyder
- Department of Chemistry, Emory University, Atlanta, Georgia
| | | | | |
Collapse
|
39
|
Morris PG, Mishina M, Jones S. Altered Synaptic and Extrasynaptic NMDA Receptor Properties in Substantia Nigra Dopaminergic Neurons From Mice Lacking the GluN2D Subunit. Front Cell Neurosci 2018; 12:354. [PMID: 30364232 PMCID: PMC6193106 DOI: 10.3389/fncel.2018.00354] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Accepted: 09/20/2018] [Indexed: 11/13/2022] Open
Abstract
N-methyl-D-aspartate receptors (NMDARs) are ubiquitously expressed in the mammalian brain and are essential for neuronal development, survival and plasticity. GluN2 subunit composition has a profound effect on the properties of NMDARs. In substantia nigra dopaminergic (SNc-DA) neurons, pharmacological experiments suggest that the relatively rare GluN2D subunits form functional synaptic and extrasynaptic NMDARs. Given the importance of establishing this point, mice lacking the GluN2D subunit (Grin2D-null) were used in this study to further explore the contribution of the GluN2D subunit to NMDAR responses. Significantly less DQP-1105-sensitive NMDAR-EPSC and significantly more ifenprodil-sensitive NMDAR-EPSC was observed in SNc-DA neurons from Grin2D-null mice, indicating that in these animals a small population of synaptic GluN2D subunits is replaced with GluN2B. Significantly larger currents were seen in response to higher concentrations (1–10 mM) of NMDA in SNc-DA neurons from Grin2D-null mice, as well as significantly more desensitization: these data are consistent with the presence of GluN2D-containing whole-cell NMDARs in SNc-DA neurons, with low conductance and little desensitization. Brief applications of NMDA evoked responses that were significantly less sensitive to DQP-1105 in slices from Grin2D-null mice. Tonic NMDAR activity in response to ambient extracellular glutamate, determined by the sensitivity of tonic current to D-AP5 (50 μM), was significantly less in SNc-DA neurons from Grin2D-null mice. In the presence of the glutamate transporter blocker TBOA (30 μM), the D-AP5-sensitive current was also significantly less in Grin2D-null mice. Taken together, these data support the evidence for GluN2D subunit expression in functional NMDARs at both synaptic and extrasynaptic locations in SNc-DA neurons.
Collapse
Affiliation(s)
- Paul G Morris
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| | - Masayoshi Mishina
- Brain Science Laboratory, The Research Organization of Science and Technology, Ritsumeikan University, Shiga, Japan
| | - Susan Jones
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
40
|
Gibb AJ, Ogden KK, McDaniel MJ, Vance KM, Kell SA, Butch C, Burger P, Liotta DC, Traynelis SF. A structurally derived model of subunit-dependent NMDA receptor function. J Physiol 2018; 596:4057-4089. [PMID: 29917241 PMCID: PMC6117563 DOI: 10.1113/jp276093] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 06/11/2018] [Indexed: 12/14/2022] Open
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. 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.
Collapse
Affiliation(s)
- Alasdair J Gibb
- Department of Neuroscience, Physiology and Pharmacology, University College London, Gower Street, London, WC1E 6BT, UK
| | - Kevin K Ogden
- Department of Pharmacology, Emory University School of Medicine, Rollins Research Center, 1510 Clifton Road, Atlanta, GA, 30322, USA
| | - Miranda J McDaniel
- Department of Pharmacology, Emory University School of Medicine, Rollins Research Center, 1510 Clifton Road, Atlanta, GA, 30322, USA
| | - Katie M Vance
- Department of Pharmacology, Emory University School of Medicine, Rollins Research Center, 1510 Clifton Road, Atlanta, GA, 30322, USA
| | - Steven A Kell
- Department of Chemistry, Emory University School, 1515 Dickey Drive, Atlanta, GA, 30322, USA
| | - Chris Butch
- Department of Chemistry, Emory University School, 1515 Dickey Drive, Atlanta, GA, 30322, USA
| | - Pieter Burger
- Department of Chemistry, Emory University School, 1515 Dickey Drive, Atlanta, GA, 30322, USA
| | - Dennis C Liotta
- Department of Chemistry, Emory University School, 1515 Dickey Drive, Atlanta, GA, 30322, USA
| | - Stephen F Traynelis
- Department of Pharmacology, Emory University School of Medicine, Rollins Research Center, 1510 Clifton Road, Atlanta, GA, 30322, USA
| |
Collapse
|
41
|
Hansen KB, Yi F, Perszyk RE, Furukawa H, Wollmuth LP, Gibb AJ, Traynelis SF. Structure, function, and allosteric modulation of NMDA receptors. J Gen Physiol 2018; 150:1081-1105. [PMID: 30037851 PMCID: PMC6080888 DOI: 10.1085/jgp.201812032] [Citation(s) in RCA: 366] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 07/03/2018] [Indexed: 12/22/2022] Open
Abstract
Hansen et al. review recent structural data that have provided insight into the function and allosteric modulation of NMDA receptors. NMDA-type glutamate receptors are ligand-gated ion channels that mediate a Ca2+-permeable component of excitatory neurotransmission in the central nervous system (CNS). They are expressed throughout the CNS and play key physiological roles in synaptic function, such as synaptic plasticity, learning, and memory. NMDA receptors are also implicated in the pathophysiology of several CNS disorders and more recently have been identified as a locus for disease-associated genomic variation. NMDA receptors exist as a diverse array of subtypes formed by variation in assembly of seven subunits (GluN1, GluN2A-D, and GluN3A-B) into tetrameric receptor complexes. These NMDA receptor subtypes show unique structural features that account for their distinct functional and pharmacological properties allowing precise tuning of their physiological roles. Here, we review the relationship between NMDA receptor structure and function with an emphasis on emerging atomic resolution structures, which begin to explain unique features of this receptor.
Collapse
Affiliation(s)
- Kasper B Hansen
- Department of Biomedical and Pharmaceutical Sciences and Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, MT
| | - Feng Yi
- Department of Biomedical and Pharmaceutical Sciences and Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, MT
| | - Riley E Perszyk
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA
| | - Hiro Furukawa
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY
| | - Lonnie P Wollmuth
- Departments of Neurobiology & Behavior and Biochemistry & Cell Biology, Stony Brook University, Stony Brook, NY
| | - Alasdair J Gibb
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - Stephen F Traynelis
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|
42
|
Maolanon AR, Risgaard R, Wang SY, Snoep Y, Papangelis A, Yi F, Holley D, Barslund AF, Svenstrup N, Hansen KB, Clausen RP. Subtype-Specific Agonists for NMDA Receptor Glycine Binding Sites. ACS Chem Neurosci 2017; 8:1681-1687. [PMID: 28514141 DOI: 10.1021/acschemneuro.7b00117] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
A series of analogues based on serine as lead structure were designed, and their agonist activities were evaluated at recombinant NMDA receptor subtypes (GluN1/2A-D) using two-electrode voltage-clamp (TEVC) electrophysiology. Pronounced variation in subunit-selectivity, potency, and agonist efficacy was observed in a manner that was dependent on the GluN2 subunit in the NMDA receptor. In particular, compounds 15a and 16a are potent GluN2C-specific superagonists at the GluN1 subunit with agonist efficacies of 398% and 308% compared to glycine. This study demonstrates that subunit-selectivity among glycine site NMDA receptor agonists can be achieved and suggests that glycine-site agonists can be developed as pharmacological tool compounds to study GluN2C-specific effects in NMDA receptor-mediated neurotransmission.
Collapse
Affiliation(s)
- Alex R. Maolanon
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Rune Risgaard
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Shuang-Yan Wang
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Yoran Snoep
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Athanasios Papangelis
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Feng Yi
- Department
of Biomedical and Pharmaceutical Sciences and Center for Biomolecular
Structure and Dynamics, University of Montana, Missoula, Montana 59812, United States
| | - David Holley
- Department
of Biomedical and Pharmaceutical Sciences and Center for Biomolecular
Structure and Dynamics, University of Montana, Missoula, Montana 59812, United States
| | - Anne F. Barslund
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
- Neuroscience Drug Discovery, H. Lundbeck
A/S, Ottiliavej 9, 2500 Valby, Denmark
| | - Niels Svenstrup
- Neuroscience Drug Discovery, H. Lundbeck
A/S, Ottiliavej 9, 2500 Valby, Denmark
| | - Kasper B. Hansen
- Department
of Biomedical and Pharmaceutical Sciences and Center for Biomolecular
Structure and Dynamics, University of Montana, Missoula, Montana 59812, United States
| | - Rasmus P. Clausen
- Department
of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| |
Collapse
|
43
|
Sun W, Hansen KB, Jahr CE. Allosteric Interactions between NMDA Receptor Subunits Shape the Developmental Shift in Channel Properties. Neuron 2017; 94:58-64.e3. [PMID: 28384476 DOI: 10.1016/j.neuron.2017.03.018] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 03/03/2017] [Accepted: 03/10/2017] [Indexed: 11/15/2022]
Abstract
During development of the central nervous system, there is a shift in the subunit composition of NMDA receptors (NMDARs) resulting in a dramatic acceleration of NMDAR-mediated synaptic currents. This shift coincides with upregulation of the GluN2A subunit and triheteromeric GluN1/2A/2B receptors with fast deactivation kinetics, whereas expression of diheteromeric GluN1/2B receptors with slower deactivation kinetics is decreased. Here, we show that allosteric interactions occur between the glutamate-binding GluN2 subunits in triheteromeric GluN1/2A/2B NMDARs. This allosterism is dominated by the GluN2A subunit and results in functional properties not predicted by those of diheteromeric GluN1/2A and GluN1/2B NMDARs. These findings suggest that GluN1/2A/2B NMDARs may maintain some signaling properties of the GluN2B subunit while having the kinetic properties of GluN1/2A NMDARs and highlight the complexity in NMDAR signaling created by diversity in subunit composition.
Collapse
Affiliation(s)
- Weinan Sun
- Vollum Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239, USA
| | - Kasper B Hansen
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Dr., Missoula, MT 59812, USA
| | - Craig E Jahr
- Vollum Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR 97239, USA.
| |
Collapse
|
44
|
Dai J, Zhou HX. Semiclosed Conformations of the Ligand-Binding Domains of NMDA Receptors during Stationary Gating. Biophys J 2017; 111:1418-1428. [PMID: 27705765 DOI: 10.1016/j.bpj.2016.08.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 08/02/2016] [Accepted: 08/12/2016] [Indexed: 12/26/2022] Open
Abstract
NMDA receptors are tetrameric ligand-gated ion channels. In the continuous presence of saturating agonists, NMDA receptors undergo stationary gating, in which the channel stochastically switches between an open state that permits ion conductance and a closed state that prevents permeation. The ligand-binding domains (LBDs) of the four subunits are expected to have closed clefts in the channel-open state. On the other hand, there is little knowledge about the conformational status of the LBDs in the channel-closed state during stationary gating. To probe the latter conformational status, Kussius and Popescu engineered interlobe disulfide cross-links in NMDA receptors and found that the cross-linking produced stationary gating kinetics that differed only subtly from that produced by agonist binding. These authors assumed that the cross-linking immobilized the LBDs in cleft-closed conformations, and consequently concluded that throughout stationary gating, agonist-bound LBDs also stayed predominantly in cleft-closed conformations and made only infrequent excursions to cleft-open conformations. Here, by calculating the conformational free energies of cross-linked and agonist-bound LBDs, we assess whether cross-linking actually traps the LBDs in cleft-closed conformations and delineate semiclosed conformations of agonist-bound LBDs that may potentially be thermodynamically and kinetically important during stationary gating. Our free-energy results show that the cross-linked LBDs are not locked in the fully closed form; rather, they sample semiclosed conformations almost as readily as the agonist-bound LBDs. Several lines of reasoning suggest that LBDs are semiclosed in the channel-closed state during stationary gating. Our free-energy simulations suggest possible structural details of such semiclosed LBD conformations, including intra- and intermolecular interactions that serve as alternatives to those in the cleft-closed conformations.
Collapse
Affiliation(s)
- Jian Dai
- Department of Physics, Florida State University, Tallahassee, Florida; Institute of Molecular Biophysics, Florida State University, Tallahassee, Florida
| | - Huan-Xiang Zhou
- Department of Physics, Florida State University, Tallahassee, Florida; Institute of Molecular Biophysics, Florida State University, Tallahassee, Florida.
| |
Collapse
|
45
|
Patel M, Rangan A. Role of the locus coeruleus in the emergence of power law wake bouts in a model of the brainstem sleep-wake system through early infancy. J Theor Biol 2017; 426:82-95. [PMID: 28552556 DOI: 10.1016/j.jtbi.2017.05.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 04/10/2017] [Accepted: 05/22/2017] [Indexed: 01/02/2023]
Abstract
Infant rats randomly cycle between the sleeping and waking states, which are tightly correlated with the activity of mutually inhibitory brainstem sleep and wake populations. Bouts of sleep and wakefulness are random; from P2-P10, sleep and wake bout lengths are exponentially distributed with increasing means, while during P10-P21, the sleep bout distribution remains exponential while the distribution of wake bouts gradually transforms to power law. The locus coeruleus (LC), via an undeciphered interaction with sleep and wake populations, has been shown experimentally to be responsible for the exponential to power law transition. Concurrently during P10-P21, the LC undergoes striking physiological changes - the LC exhibits strong global 0.3 Hz oscillations up to P10, but the oscillation frequency gradually rises and synchrony diminishes from P10-P21, with oscillations and synchrony vanishing at P21 and beyond. In this work, we construct a biologically plausible Wilson Cowan-style model consisting of the LC along with sleep and wake populations. We show that external noise and strong reciprocal inhibition can lead to switching between sleep and wake populations and exponentially distributed sleep and wake bout durations as during P2-P10, with the parameters of inhibition between the sleep and wake populations controlling mean bout lengths. Furthermore, we show that the changing physiology of the LC from P10-P21, coupled with reciprocal excitation between the LC and wake population, can explain the shift from exponential to power law of the wake bout distribution. To our knowledge, this is the first study that proposes a plausible biological mechanism, which incorporates the known changing physiology of the LC, for tying the developing sleep-wake circuit and its interaction with the LC to the transformation of sleep and wake bout dynamics from P2-P21.
Collapse
Affiliation(s)
- Mainak Patel
- Department of Mathematics, College of William and Mary, Williamsburg, VA, USA.
| | - Aaditya Rangan
- Courant Institute of Mathematical Sciences, New York University, NYC, USA.
| |
Collapse
|
46
|
Chen W, Tankovic A, Burger PB, Kusumoto H, Traynelis SF, Yuan H. Functional Evaluation of a De Novo GRIN2A Mutation Identified in a Patient with Profound Global Developmental Delay and Refractory Epilepsy. Mol Pharmacol 2017; 91:317-330. [PMID: 28126851 PMCID: PMC5363715 DOI: 10.1124/mol.116.106781] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Accepted: 01/23/2017] [Indexed: 12/22/2022] Open
Abstract
The N-methyl-d-aspartate receptor (NMDAR), a ligand-gated ionotropic glutamate receptor, plays important roles in normal brain development and a wide range of neurologic disorders, including epilepsy. Here, we evaluate for the first time the functional properties of a de novo GRIN2A missense mutation (p.M817V) in the pre-M4 linker in a child with profound global developmental delay and refractory epilepsy. Electrophysiologic recordings revealed that the mutant GluN2A(M817V)-containing receptors showed enhanced agonist potency, reduced sensitivity to endogenous negative inhibitors (Mg2+, proton, and zinc), prolonged synaptic-like response time course, increased single-channel mean open time, and increased channel open probability. These results suggest that the gain-of-function M817V mutation causes overactivation of NMDAR and drives neuronal hyperexcitability, which may contribute to the patient's observed epileptic phenotype. Molecular modeling of the closed channel conformation reveals that this mutation weakens the interaction between GluN2 transmembrane helix M4 and two GluN1 transmembrane helices, and increases atomic fluctuation or movement of the pre-M1 region of GluN1 subunit, suggesting a mechanism by which channel function is enhanced. The functional changes of this mutation on agonist potency occur when the mutation is introduced into all other GluN2 subunits, suggesting a conserved role of this residue in control of NMDAR function through interactions of membrane spanning GluN2 and GluN1 helices. A number of NMDAR-targeted drugs including U.S. Food and Drug Association-approved NMDAR channel blockers were evaluated for their ability to inhibit receptors containing GluN2A(M817V) as a first step to exploring the potential for rescue pharmacology and personalized medicine.
Collapse
Affiliation(s)
- Wenjuan Chen
- Department of Pharmacology, Rollins Research Center, School of Medicine, Emory University, Atlanta, Georgia (W.C., A.T., H.K., S.F.T., H.Y.); Department of Neurology, Xiangya Hospital, Central South University, Changsha, People's Republic of China (W.C.); Department of Chemistry, Emory University, Atlanta, Georgia (P.B.B.); Center for Functional Evaluation of Rare Variants (CFERV), Rollins Research Center, School of Medicine, Emory University, Atlanta, Georgia (S.F.T., H.Y.)
| | - Anel Tankovic
- Department of Pharmacology, Rollins Research Center, School of Medicine, Emory University, Atlanta, Georgia (W.C., A.T., H.K., S.F.T., H.Y.); Department of Neurology, Xiangya Hospital, Central South University, Changsha, People's Republic of China (W.C.); Department of Chemistry, Emory University, Atlanta, Georgia (P.B.B.); Center for Functional Evaluation of Rare Variants (CFERV), Rollins Research Center, School of Medicine, Emory University, Atlanta, Georgia (S.F.T., H.Y.)
| | - Pieter B Burger
- Department of Pharmacology, Rollins Research Center, School of Medicine, Emory University, Atlanta, Georgia (W.C., A.T., H.K., S.F.T., H.Y.); Department of Neurology, Xiangya Hospital, Central South University, Changsha, People's Republic of China (W.C.); Department of Chemistry, Emory University, Atlanta, Georgia (P.B.B.); Center for Functional Evaluation of Rare Variants (CFERV), Rollins Research Center, School of Medicine, Emory University, Atlanta, Georgia (S.F.T., H.Y.)
| | - Hirofumi Kusumoto
- Department of Pharmacology, Rollins Research Center, School of Medicine, Emory University, Atlanta, Georgia (W.C., A.T., H.K., S.F.T., H.Y.); Department of Neurology, Xiangya Hospital, Central South University, Changsha, People's Republic of China (W.C.); Department of Chemistry, Emory University, Atlanta, Georgia (P.B.B.); Center for Functional Evaluation of Rare Variants (CFERV), Rollins Research Center, School of Medicine, Emory University, Atlanta, Georgia (S.F.T., H.Y.)
| | - Stephen F Traynelis
- Department of Pharmacology, Rollins Research Center, School of Medicine, Emory University, Atlanta, Georgia (W.C., A.T., H.K., S.F.T., H.Y.); Department of Neurology, Xiangya Hospital, Central South University, Changsha, People's Republic of China (W.C.); Department of Chemistry, Emory University, Atlanta, Georgia (P.B.B.); Center for Functional Evaluation of Rare Variants (CFERV), Rollins Research Center, School of Medicine, Emory University, Atlanta, Georgia (S.F.T., H.Y.)
| | - Hongjie Yuan
- Department of Pharmacology, Rollins Research Center, School of Medicine, Emory University, Atlanta, Georgia (W.C., A.T., H.K., S.F.T., H.Y.); Department of Neurology, Xiangya Hospital, Central South University, Changsha, People's Republic of China (W.C.); Department of Chemistry, Emory University, Atlanta, Georgia (P.B.B.); Center for Functional Evaluation of Rare Variants (CFERV), Rollins Research Center, School of Medicine, Emory University, Atlanta, Georgia (S.F.T., H.Y.)
| |
Collapse
|
47
|
Gao K, Tankovic A, Zhang Y, Kusumoto H, Zhang J, Chen W, XiangWei W, Shaulsky GH, Hu C, Traynelis SF, Yuan H, Jiang Y. A de novo loss-of-function GRIN2A mutation associated with childhood focal epilepsy and acquired epileptic aphasia. PLoS One 2017; 12:e0170818. [PMID: 28182669 PMCID: PMC5300259 DOI: 10.1371/journal.pone.0170818] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 01/11/2017] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE N-methyl-D-aspartate receptors (NMDAR) subunit GRIN2A/GluN2A mutations have been identified in patients with various neurological diseases, such as epilepsy and intellectual disability / developmental delay (ID/DD). In this study, we investigated the phenotype and underlying molecular mechanism of a GRIN2A missense mutation identified by next generation sequencing on idiopathic focal epilepsy using in vitro electrophysiology. METHODS Genomic DNA of patients with epilepsy and ID/DD were sequenced by targeted next-generation sequencing within 300 genes related to epilepsy and ID/DD. The effects of one missense GRIN2A mutation on NMDAR function were evaluated by two-electrode voltage clamp current recordings and whole cell voltage clamp current recordings. RESULTS We identified one de novo missense GRIN2A mutation (Asp731Asn, GluN2A(D731N)) in a child with unexplained epilepsy and DD. The D731N mutation is located in a portion of the agonist-binding domain (ABD) in the GluN2A subunit, which is the binding pocket for agonist glutamate. This residue in the ABD is conserved among vertebrate species and all other NMDAR subunits, suggesting an important role in receptor function. The proband shows developmental delay as well as EEG-confirmed seizure activity. Functional analyses reveal that the GluN2A(D731N) mutation decreases glutamate potency by over 3,000-fold, reduces amplitude of current response, shortens synaptic-like response time course, and decreases channel open probability, while enhancing sensitivity to negative allosteric modulators, including extracellular proton and zinc inhibition. The combined effects reduce NMDAR function. SIGNIFICANCE We identified a de novo missense mutation in the GRIN2A gene in a patient with childhood focal epilepsy and acquired epileptic aphasia. The mutant decreases NMDAR activation suggesting NMDAR hypofunction may contribute to the epilepsy pathogenesis.
Collapse
Affiliation(s)
- Kai Gao
- Department of Pediatrics and Pediatric Epilepsy Center, Peking University First Hospital, Beijing, China
| | - Anel Tankovic
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Yujia Zhang
- Department of Pediatrics and Pediatric Epilepsy Center, Peking University First Hospital, Beijing, China
| | - Hirofumi Kusumoto
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Jin Zhang
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Wenjuan Chen
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, United States of America
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Wenshu XiangWei
- Department of Pediatrics and Pediatric Epilepsy Center, Peking University First Hospital, Beijing, China
| | - Gil H. Shaulsky
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Chun Hu
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Stephen F. Traynelis
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, United States of America
- Center for Functional Evaluation of Rare Variant (CFERV), Emory University School of Medicine, Atlanta, GA, United States of America
| | - Hongjie Yuan
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, United States of America
- Center for Functional Evaluation of Rare Variant (CFERV), Emory University School of Medicine, Atlanta, GA, United States of America
| | - Yuwu Jiang
- Department of Pediatrics and Pediatric Epilepsy Center, Peking University First Hospital, Beijing, China
- Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing, China
| |
Collapse
|
48
|
Molecular Mechanism of Disease-Associated Mutations in the Pre-M1 Helix of NMDA Receptors and Potential Rescue Pharmacology. PLoS Genet 2017; 13:e1006536. [PMID: 28095420 PMCID: PMC5240934 DOI: 10.1371/journal.pgen.1006536] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 12/13/2016] [Indexed: 12/31/2022] Open
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.
Collapse
|
49
|
Prediction of N-Methyl-D-Aspartate Receptor GluN1-Ligand Binding Affinity by a Novel SVM-Pose/SVM-Score Combinatorial Ensemble Docking Scheme. Sci Rep 2017; 7:40053. [PMID: 28059133 PMCID: PMC5216401 DOI: 10.1038/srep40053] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 11/30/2016] [Indexed: 01/24/2023] Open
Abstract
The glycine-binding site of the N-methyl-D-aspartate receptor (NMDAR) subunit GluN1 is a potential pharmacological target for neurodegenerative disorders. A novel combinatorial ensemble docking scheme using ligand and protein conformation ensembles and customized support vector machine (SVM)-based models to select the docked pose and to predict the docking score was generated for predicting the NMDAR GluN1-ligand binding affinity. The predicted root mean square deviation (RMSD) values in pose by SVM-Pose models were found to be in good agreement with the observed values (n = 30, r2 = 0.928–0.988, = 0.894–0.954, RMSE = 0.002–0.412, s = 0.001–0.214), and the predicted pKi values by SVM-Score were found to be in good agreement with the observed values for the training samples (n = 24, r2 = 0.967, = 0.899, RMSE = 0.295, s = 0.170) and test samples (n = 13, q2 = 0.894, RMSE = 0.437, s = 0.202). When subjected to various statistical validations, the developed SVM-Pose and SVM-Score models consistently met the most stringent criteria. A mock test asserted the predictivity of this novel docking scheme. Collectively, this accurate novel combinatorial ensemble docking scheme can be used to predict the NMDAR GluN1-ligand binding affinity for facilitating drug discovery.
Collapse
|
50
|
Hansen KB, Yi F, Perszyk RE, Menniti FS, Traynelis SF. NMDA Receptors in the Central Nervous System. Methods Mol Biol 2017; 1677:1-80. [PMID: 28986865 DOI: 10.1007/978-1-4939-7321-7_1] [Citation(s) in RCA: 119] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
NMDA-type glutamate receptors are ligand-gated ion channels that mediate a major component of excitatory neurotransmission in the central nervous system (CNS). They are widely distributed at all stages of development and are critically involved in normal brain functions, including neuronal development and synaptic plasticity. NMDA receptors are also implicated in the pathophysiology of numerous neurological and psychiatric disorders, such as ischemic stroke, traumatic brain injury, Alzheimer's disease, epilepsy, mood disorders, and schizophrenia. For these reasons, NMDA receptors have been intensively studied in the past several decades to elucidate their physiological roles and to advance them as therapeutic targets. Seven NMDA receptor subunits exist that assemble into a diverse array of tetrameric receptor complexes, which are differently regulated, have distinct regional and developmental expression, and possess a wide range of functional and pharmacological properties. The diversity in subunit composition creates NMDA receptor subtypes with distinct physiological roles across neuronal cell types and brain regions, and enables precise tuning of synaptic transmission. Here, we will review the relationship between NMDA receptor structure and function, the diversity and significance of NMDA receptor subtypes in the CNS, as well as principles and rules by which NMDA receptors operate in the CNS under normal and pathological conditions.
Collapse
Affiliation(s)
- Kasper B Hansen
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT, USA. .,Center for Biomolecular Structure and Dynamics, University of Montana, Missoula, MT, USA.
| | - Feng Yi
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, Missoula, MT, USA
| | - Riley E Perszyk
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, USA
| | - Frank S Menniti
- MindImmune Therapeutics, Inc., George & Anne Ryan Institute for Neuroscience, Kingston, RI, USA
| | - Stephen F Traynelis
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|