51
|
Galambos AR, Papp ZT, Boldizsár I, Zádor F, Köles L, Harsing LG, Al-Khrasani M. Glycine Transporter 1 Inhibitors: Predictions on Their Possible Mechanisms in the Development of Opioid Analgesic Tolerance. Biomedicines 2024; 12:421. [PMID: 38398023 PMCID: PMC10886540 DOI: 10.3390/biomedicines12020421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/06/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
The development of opioid tolerance in patients on long-term opioid analgesic treatment is an unsolved matter in clinical practice thus far. Dose escalation is required to restore analgesic efficacy, but at the price of side effects. Intensive research is ongoing to elucidate the underlying mechanisms of opioid analgesic tolerance in the hope of maintaining opioid analgesic efficacy. N-Methyl-D-aspartate receptor (NMDAR) antagonists have shown promising effects regarding opioid analgesic tolerance; however, their use is limited by side effects (memory dysfunction). Nevertheless, the GluN2B receptor remains a future target for the discovery of drugs to restore opioid efficacy. Mechanistically, the long-term activation of µ-opioid receptors (MORs) initiates receptor phosphorylation, which triggers β-arrestin-MAPKs and NOS-GC-PKG pathway activation, which ultimately ends with GluN2B receptor overactivation and glutamate release. The presence of glutamate and glycine as co-agonists is a prerequisite for GluN2B receptor activation. The extrasynaptic localization of the GluN2B receptor means it is influenced by the glycine level, which is regulated by astrocytic glycine transporter 1 (GlyT1). Enhanced astrocytic glycine release by reverse transporter mechanisms as a consequence of high glutamate levels or unconventional MOR activation on astrocytes could further activate the GluN2B receptor. GlyT1 inhibitors might inhibit this condition, thereby reducing opioid tolerance.
Collapse
Affiliation(s)
- Anna Rita Galambos
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvá-rad tér 4, H-1445 Budapest, Hungary; (A.R.G.); (Z.T.P.); (I.B.); (F.Z.)
| | - Zsolt Tamás Papp
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvá-rad tér 4, H-1445 Budapest, Hungary; (A.R.G.); (Z.T.P.); (I.B.); (F.Z.)
| | - Imre Boldizsár
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvá-rad tér 4, H-1445 Budapest, Hungary; (A.R.G.); (Z.T.P.); (I.B.); (F.Z.)
| | - Ferenc Zádor
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvá-rad tér 4, H-1445 Budapest, Hungary; (A.R.G.); (Z.T.P.); (I.B.); (F.Z.)
| | - László Köles
- Department of Oral Biology, Semmelweis University, H-1089 Budapest, Hungary;
| | - Laszlo G. Harsing
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvá-rad tér 4, H-1445 Budapest, Hungary; (A.R.G.); (Z.T.P.); (I.B.); (F.Z.)
| | - Mahmoud Al-Khrasani
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvá-rad tér 4, H-1445 Budapest, Hungary; (A.R.G.); (Z.T.P.); (I.B.); (F.Z.)
| |
Collapse
|
52
|
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
|
53
|
Zhang PP, Benske TM, Ahn LY, Schaffer AE, Paton JC, Paton AW, Mu TW, Wang YJ. Adapting the endoplasmic reticulum proteostasis rescues epilepsy-associated NMDA receptor variants. Acta Pharmacol Sin 2024; 45:282-297. [PMID: 37803141 PMCID: PMC10789767 DOI: 10.1038/s41401-023-01172-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 09/17/2023] [Indexed: 10/08/2023]
Abstract
The GRIN genes encoding N-methyl-D-aspartate receptor (NMDAR) subunits are remarkably intolerant to variation. Many pathogenic NMDAR variants result in their protein misfolding, inefficient assembly, reduced surface expression, and impaired function on neuronal membrane, causing neurological disorders including epilepsy and intellectual disability. Here, we investigated the proteostasis maintenance of NMDARs containing epilepsy-associated variations in the GluN2A subunit, including M705V and A727T. In the transfected HEK293T cells, we showed that the two variants were targeted to the proteasome for degradation and had reduced functional surface expression. We demonstrated that the application of BIX, a known small molecule activator of an HSP70 family chaperone BiP (binding immunoglobulin protein) in the endoplasmic reticulum (ER), dose-dependently enhanced the functional surface expression of the M705V and A727T variants in HEK293T cells. Moreover, BIX (10 μM) increased the surface protein levels of the M705V variant in human iPSC-derived neurons. We revealed that BIX promoted folding, inhibited degradation, and enhanced anterograde trafficking of the M705V variant by modest activation of the IRE1 pathway of the unfolded protein response. Our results suggest that adapting the ER proteostasis network restores the folding, trafficking, and function of pathogenic NMDAR variants, representing a potential treatment for neurological disorders resulting from NMDAR dysfunction.
Collapse
Affiliation(s)
- Pei-Pei Zhang
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Taylor M Benske
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Lucie Y Ahn
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Ashleigh E Schaffer
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - James C Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Adrienne W Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Ting-Wei Mu
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.
| | - Ya-Juan Wang
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA.
| |
Collapse
|
54
|
Kysilov B, Kuchtiak V, Hrcka Krausova B, Balik A, Korinek M, Fili K, Dobrovolski M, Abramova V, Chodounska H, Kudova E, Bozikova P, Cerny J, Smejkalova T, Vyklicky L. Disease-associated nonsense and frame-shift variants resulting in the truncation of the GluN2A or GluN2B C-terminal domain decrease NMDAR surface expression and reduce potentiating effects of neurosteroids. Cell Mol Life Sci 2024; 81:36. [PMID: 38214768 PMCID: PMC10786987 DOI: 10.1007/s00018-023-05062-6] [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: 09/09/2023] [Revised: 11/16/2023] [Accepted: 11/21/2023] [Indexed: 01/13/2024]
Abstract
N-methyl-D-aspartate receptors (NMDARs) play a critical role in normal brain function, and variants in genes encoding NMDAR subunits have been described in individuals with various neuropsychiatric disorders. We have used whole-cell patch-clamp electrophysiology, fluorescence microscopy and in-silico modeling to explore the functional consequences of disease-associated nonsense and frame-shift variants resulting in the truncation of GluN2A or GluN2B C-terminal domain (CTD). This study characterizes variant NMDARs and shows their reduced surface expression and synaptic localization, altered agonist affinity, increased desensitization, and reduced probability of channel opening. We also show that naturally occurring and synthetic steroids pregnenolone sulfate and epipregnanolone butanoic acid, respectively, enhance NMDAR function in a way that is dependent on the length of the truncated CTD and, further, is steroid-specific, GluN2A/B subunit-specific, and GluN1 splice variant-specific. Adding to the previously described effects of disease-associated NMDAR variants on the receptor biogenesis and function, our results improve the understanding of the molecular consequences of NMDAR CTD truncations and provide an opportunity for the development of new therapeutic neurosteroid-based ligands.
Collapse
Affiliation(s)
- Bohdan Kysilov
- Laboratory of Cellular Neurophysiology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14200, Prague 4, Czech Republic
- Stony Brook University, Stony Brook, 100 Nicolls Road, NY, 11794, USA
| | - Viktor Kuchtiak
- Laboratory of Cellular Neurophysiology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14200, Prague 4, Czech Republic
- Faculty of Science, Charles University, Albertov 2038, 12800, Prague 2, Czech Republic
| | - Barbora Hrcka Krausova
- Laboratory of Cellular Neurophysiology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14200, Prague 4, Czech Republic
| | - Ales Balik
- Laboratory of Cellular Neurophysiology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14200, Prague 4, Czech Republic
| | - Miloslav Korinek
- Laboratory of Cellular Neurophysiology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14200, Prague 4, Czech Republic
| | - Klevinda Fili
- Laboratory of Cellular Neurophysiology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14200, Prague 4, Czech Republic
- Third Faculty of Medicine, Charles University, Ruska 87, 10000, Prague 10, Czech Republic
| | - Mark Dobrovolski
- Laboratory of Cellular Neurophysiology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14200, Prague 4, Czech Republic
- Third Faculty of Medicine, Charles University, Ruska 87, 10000, Prague 10, Czech Republic
| | - Vera Abramova
- Laboratory of Cellular Neurophysiology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14200, Prague 4, Czech Republic
- Third Faculty of Medicine, Charles University, Ruska 87, 10000, Prague 10, Czech Republic
| | - Hana Chodounska
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo Nam. 2, 16610, Prague 6, Czech Republic
| | - Eva Kudova
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo Nam. 2, 16610, Prague 6, Czech Republic
| | - Paulina Bozikova
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Prumyslova 595, 25250, Vestec, Czech Republic
| | - Jiri Cerny
- Laboratory of Cellular Neurophysiology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14200, Prague 4, Czech Republic
| | - Tereza Smejkalova
- Laboratory of Cellular Neurophysiology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14200, Prague 4, Czech Republic.
| | - Ladislav Vyklicky
- Laboratory of Cellular Neurophysiology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14200, Prague 4, Czech Republic.
| |
Collapse
|
55
|
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
|
56
|
Mӓnnikkӧ R, Kullmann DM. Structure-function and pharmacologic aspects of ion channels relevant to neurologic channelopathies. HANDBOOK OF CLINICAL NEUROLOGY 2024; 203:1-23. [PMID: 39174242 DOI: 10.1016/b978-0-323-90820-7.00009-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
Ion channels are membrane proteins that allow the passage of ions across the membrane. They characteristically contain a pore where the selectivity of certain ion species is determined and gates that open and close the pore are found. The pore is often connected to additional domains or subunits that regulate its function. Channels are grouped into families based on their selectivity for specific ions and the stimuli that control channel opening and closing, such as voltage or ligands. Ion channels are fundamental to the electrical properties of excitable tissues. Dysfunction of channels can lead to abnormal electrical signaling of neurons and muscle cells, accompanied by clinical manifestations, known as channelopathies. Many naturally occurring toxins target ion channels and affect excitable cells where the channels are expressed. Furthermore, ion channels, as membrane proteins and key regulators of a number of physiologic functions, are an important target for drugs in clinical use. In this chapter, we give a general overview of the classification, genetics and structure-function features of the main ion channel families, and address some pharmacologic aspects relevant to neurologic channelopathies.
Collapse
Affiliation(s)
- Roope Mӓnnikkӧ
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom.
| | - Dimitri M Kullmann
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom.
| |
Collapse
|
57
|
Yi F, Traynelis SF, Hansen KB. Selective Cell-Surface Expression of Triheteromeric NMDA Receptors. Methods Mol Biol 2024; 2799:55-77. [PMID: 38727903 PMCID: PMC11311247 DOI: 10.1007/978-1-0716-3830-9_5] [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] [Indexed: 06/01/2024]
Abstract
NMDA-type ionotropic glutamate receptors are critically involved in many brain functions and are implicated in a variety of brain disorders. Seven NMDA receptor subunits exist (GluN1, GluN2A-D, and GluN3A-B) that assemble into tetrameric receptor subtypes with distinct functional properties and physiological roles. The majority NMDA receptors are composed of two GluN1 and two GluN2 subunits, which can assemble into four diheteromeric receptors subtypes composed of GluN1 and one type of GluN2 subunit (e.g., GluN1/2A), and presumably also six triheteromeric receptor subtypes composed of GluN1 and two different GluN2 subunits (e.g., GluN1/2A/2B). Furthermore, the GluN1 subunit exists as eight splice variants (e.g., GluN1-1a and GluN1-1b isoforms), and two different GluN1 isoforms can co-assemble to also form triheteromeric NMDA receptors (e.g., GluN1-1a/1b/2A). Here, we describe a method to faithfully express triheteromeric NMDA receptors in heterologous expression systems by controlling the identity of two of the four subunits. This method overcomes the problem that co-expression of three different NMDA receptor subunits generates two distinct diheteromeric receptor subtypes as well as one triheteromeric receptor subtype, thereby confounding studies that require a homogenous population of triheteromeric NMDA receptors. The method has been applied to selectively express recombinant triheteromeric GluN1/2A/2B, GluN1/2A/2C, GluN1/2B/2D, GluN1-1a/GluN1-1b/2A, GluN1-1a/GluN1-1b/2B receptors with negligible co-expression of the respective diheteromeric receptor subtypes. This method therefore enables quantitative evaluation of functional and pharmacological properties of triheteromeric NMDA receptors, some of which are abundant NMDA receptor subtypes in the adult brain.
Collapse
Affiliation(s)
- Feng Yi
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, China
| | - Stephen F Traynelis
- Department of Pharmacology, Emory University School of Medicine, Atlanta, GA, USA
| | - Kasper B Hansen
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT, USA.
| |
Collapse
|
58
|
Zheng W, Liu X. Modeling and Simulation of the NMDA Receptor at Coarse-Grained and Atomistic Levels. Methods Mol Biol 2024; 2799:269-280. [PMID: 38727913 DOI: 10.1007/978-1-0716-3830-9_15] [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 glutamate-gated excitatory channels that play essential roles in brain functions. While high-resolution structures were solved for an allosterically inhibited form of functional NMDA receptor, other key functional states (particularly the active open-channel state) have not yet been resolved at atomic resolutions. To decrypt the molecular mechanism of the NMDA receptor activation, structural modeling and simulation are instrumental in providing detailed information about the dynamics and energetics of the receptor in various functional states. In this chapter, we describe coarse-grained modeling of the NMDA receptor using an elastic network model and related modeling/analysis tools (e.g., normal mode analysis, flexibility and hotspot analysis, cryo-EM flexible fitting, and transition pathway modeling) based on available structures. Additionally, we show how to build an atomistic model of the active-state receptor with targeted molecular dynamics (MD) simulation and explore its energetics and dynamics with conventional MD simulation. Taken together, these modeling and simulation can offer rich structural and dynamic information which will guide experimental studies of the activation of this key receptor.
Collapse
Affiliation(s)
- Wenjun Zheng
- Department of Physics, State University of New York at Buffalo, Buffalo, NY, USA.
| | - Xing Liu
- Department of Physics, State University of New York at Buffalo, Buffalo, NY, USA
| |
Collapse
|
59
|
Iacobucci GJ, Popescu GK. Estimating the Ca 2+ Block of NMDA Receptors with Single-Channel Electrophysiology. Methods Mol Biol 2024; 2799:151-175. [PMID: 38727907 PMCID: PMC11942295 DOI: 10.1007/978-1-0716-3830-9_9] [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] [Indexed: 07/03/2024]
Abstract
In vertebrate central neurons, NMDA receptors are glutamate- and glycine-gated ion channels that allow the passage of Na+ and Ca2+ ions into the cell when these neurotransmitters are simultaneously present. The passage of Ca2+ is critical for initiating the cellular processes underlying various forms of synaptic plasticity. These Ca2+ ions can autoregulate the NMDA receptor signal through multiple distinct mechanisms to reduce the total flux of cations. One such mechanism is the ability of Ca2+ ions to exclude the passage of Na+ ions resulting in a reduced unitary current conductance. In contrast to the well-characterized Mg2+ block, this "channel block" mechanism is voltage-independent. In this chapter, we discuss theoretical and experimental considerations for the study of channel block by Ca2+ using single-channel patch-clamp electrophysiology. We focus on two classic methodologies to quantify the dependence of unitary channel conductance on external concentrations of Ca2+ as the basis for quantifying Ca2+ block.
Collapse
Affiliation(s)
- Gary J Iacobucci
- Department of Biochemistry, School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA.
| | - Gabriela K Popescu
- Department of Biochemistry, School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA.
| |
Collapse
|
60
|
Vinnakota C, Schroeder A, Du X, Ikeda K, Ide S, Mishina M, Hudson M, Jones NC, Sundram S, Hill RA. Understanding the role of the NMDA receptor subunit, GluN2D, in mediating NMDA receptor antagonist-induced behavioral disruptions in male and female mice. J Neurosci Res 2024; 102:e25257. [PMID: 37814998 PMCID: PMC10953441 DOI: 10.1002/jnr.25257] [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: 05/08/2023] [Revised: 07/24/2023] [Accepted: 09/23/2023] [Indexed: 10/11/2023]
Abstract
Noncompetitive NMDA receptor (NMDAR) antagonists like phencyclidine (PCP) and ketamine cause psychosis-like symptoms in healthy humans, exacerbate schizophrenia symptoms in people with the disorder, and disrupt a range of schizophrenia-relevant behaviors in rodents, including hyperlocomotion. This is negated in mice lacking the GluN2D subunit of the NMDAR, suggesting the GluN2D subunit mediates the hyperlocomotor effects of these drugs. However, the role of GluN2D in mediating other schizophrenia-relevant NMDAR antagonist-induced behavioral disturbances, and in both sexes, is unclear. This study aimed to investigate the role of the GluN2D subunit in mediating schizophrenia-relevant behaviors induced by a range of NMDA receptor antagonists. Using both male and female GluN2D knockout (KO) mice, we examined the effects of the NMDAR antagonist's PCP, the S-ketamine enantiomer (S-ket), and the ketamine metabolite R-norketamine (R-norket) on locomotor activity, anxiety-related behavior, and recognition and short-term spatial memory. GluN2D-KO mice showed a blunted locomotor response to R-norket, S-ket, and PCP, a phenotype present in both sexes. GluN2D-KO mice of both sexes showed an anxious phenotype and S-ket, R-norket, and PCP showed anxiolytic effects that were dependent on sex and genotype. S-ket disrupted spatial recognition memory in females and novel object recognition memory in both sexes, independent of genotype. This datum identifies a role for the GluN2D subunit in sex-specific effects of NMDAR antagonists and on the differential effects of the R- and S-ket enantiomers.
Collapse
Affiliation(s)
- Chitra Vinnakota
- Department of PsychiatryMonash UniversityClaytonVictoriaAustralia
| | - Anna Schroeder
- Department of PsychiatryMonash UniversityClaytonVictoriaAustralia
| | - Xin Du
- Department of PsychiatryMonash UniversityClaytonVictoriaAustralia
| | - Kazutaka Ikeda
- Addictive Substance ProjectTokyo Metropolitan Institute of Medical ScienceTokyoJapan
| | - Soichiro Ide
- Addictive Substance ProjectTokyo Metropolitan Institute of Medical ScienceTokyoJapan
| | - Masayoshi Mishina
- Brain Science Laboratory, The Research Organization of Science and TechnologyRitsumeikan UniversityKusatsuJapan
| | - Matthew Hudson
- Department of NeuroscienceMonash UniversityClaytonVictoriaAustralia
| | | | - Suresh Sundram
- Department of PsychiatryMonash UniversityClaytonVictoriaAustralia
- Mental Health ProgramMonash HealthClaytonVictoriaAustralia
| | - Rachel Anne Hill
- Department of PsychiatryMonash UniversityClaytonVictoriaAustralia
| |
Collapse
|
61
|
Shah NM, Ghazaryan N, Gonzaga NL, Paclibar CG, Biju AP, Liang C, Mukherjee J. Glutamate's Effects on the N-Methyl-D-Aspartate (NMDA) Receptor Ion Channel in Alzheimer's Disease Brain: Challenges for PET Radiotracer Development for Imaging the NMDA Ion Channel. Molecules 2023; 29:20. [PMID: 38202606 PMCID: PMC10779680 DOI: 10.3390/molecules29010020] [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: 12/03/2023] [Revised: 12/13/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
In an effort to further understand the challenges facing in vivo imaging probe development for the N-methyl-D-aspartate (NMDA) receptor ion channel, we have evaluated the effect of glutamate on the Alzheimer's disease (AD) brain. Human post-mortem AD brain slices of the frontal cortex and anterior cingulate were incubated with [3H]MK-801 and adjacent sections were tested for Aβ and Tau. The binding of [3H]MK-801 was measured in the absence and presence of glutamate and glycine. Increased [3H]MK-801 binding in AD brains was observed at baseline and in the presence of glutamate, indicating a significant increase (>100%) in glutamate-induced NMDA ion channel activity in AD brains compared to cognitively normal brains. The glycine effect was lower, suggesting a decrease of the co-agonist effect of glutamate and glycine in the AD brain. Our preliminary findings suggest that the targeting of the NMDA ion channel as well as the glutamate site may be appropriate in the diagnosis and treatment of AD. However, the low baseline levels of [3H]MK-801 binding in the frontal cortex and anterior cingulate in the absence of glutamate and glycine indicate significant hurdles for in vivo imaging probe development and validation.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jogeshwar Mukherjee
- Preclinical Imaging, Department of Radiological Sciences, University of California-Irvine, Irvine, CA 92697, USA; (N.M.S.); (N.G.); (N.L.G.); (C.G.P.); (A.P.B.); (C.L.)
| |
Collapse
|
62
|
Remya C, Variyar EJ, Omkumar RV, Sadasivan C, Dileep KV. Unveiling the molecular basis of lobeline's allosteric regulation of NMDAR: insights from molecular modeling. Sci Rep 2023; 13:22418. [PMID: 38104236 PMCID: PMC10725453 DOI: 10.1038/s41598-023-49835-2] [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: 09/28/2023] [Accepted: 12/12/2023] [Indexed: 12/19/2023] Open
Abstract
Neurological and psychiatric disorders contribute significantly to the global disease burden, adversely affecting the quality of life for both patients and their families. Impaired glutamatergic signaling is considered to be a major cause for most of the neurological and psychiatric disorders. Glutamate receptors are over activated in excitotoxic conditions, leading to dysregulation of Ca2+ homeostasis, triggering the production of free radicals and oxidative stress, mitochondrial dysfunction and eventually cell death. Excitotoxicity primarily results from the overactivity of NMDARs, a subtype of ionotropic glutamate receptors, due to their pronounced Ca2+ permeability and conductance characteristics. NMDAR antagonists are suggested to have therapeutic use as they can prevent excitotoxicity. Our previous studies demonstrated lobeline, an alkaloid, exerts neuroprotective action in excitotoxic conditions by blocking NMDAR. However, the atomic level interactions of lobeline with NMDAR was not characterized yet. Structural comparison of lobeline with a known NMDAR antagonist ifenprodil, followed by molecular docking and dynamics simulations revealed that lobeline could bind to the ifenprodil binding site i.e., in the heterodimer interface of GluN1-GluN2B subunits and exert ifenprodil like activities. By in silico structure guided modifications on lobeline and subsequent free energy calculations, we propose putative NMDAR antagonists derived from lobeline.
Collapse
Affiliation(s)
- Chandran Remya
- Laboratory for Computational and Structural Biology, Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur, Kerala, 680005, India
| | - E J Variyar
- Department of Biotechnology and Microbiology, Kannur University, Dr. Janaki Ammal Campus, Thalassery, Kerala, 670661, India
| | - R V Omkumar
- Neurobiology Division, Rajiv Gandhi Centre for Biotechnology, Thycaud PO, Trivandrum, Kerala, 695014, India
| | - C Sadasivan
- Department of Biotechnology and Microbiology, Kannur University, Dr. Janaki Ammal Campus, Thalassery, Kerala, 670661, India
- Inter University Centre for Bioscience, Kannur University, Dr. Janaki Ammal Campus, Thalassery, Kerala, 670661, India
| | - K V Dileep
- Laboratory for Computational and Structural Biology, Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur, Kerala, 680005, India.
| |
Collapse
|
63
|
Yu Y, Wu K, Yang X, Long J, Chang C. Terahertz Photons Improve Cognitive Functions in Posttraumatic Stress Disorder. RESEARCH (WASHINGTON, D.C.) 2023; 6:0278. [PMID: 38111677 PMCID: PMC10726292 DOI: 10.34133/research.0278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 11/12/2023] [Indexed: 12/20/2023]
Abstract
Posttraumatic stress disorder (PTSD) is a serious psychosis leading to cognitive impairment. To restore cognitive functions for patients, the main treatments are based on medication or rehabilitation training but with limited effectiveness and strong side effects. Here, we demonstrate a new treatment approach for PTSD by using terahertz (THz) photons stimulating the hippocampal CA3 subregion. We verified that this method can nonthermally restore cognitive function in PTSD rats in vivo. After THz photon irradiation, the PTSD rats' recognitive index improved by about 10% in a novel object recognition test, the PTSD rats' accuracy improved by about 100% in a shuttler box test, the PTSD rats' numbers to identify target box was about 5 times lower in a Barnes maze test, and the rate of staying in new arm increased by approximately 40% in a Y-maze test. Further experimental studies found that THz photon (34.5 THz) irradiation could improve the expression of NR2B (increased by nearly 40%) and phosphorylated NR2B (increased by about 50%). In addition, molecular dynamics simulations showed that THz photons at a frequency of 34.5 THz are mainly absorbed by the pocket of glutamate receptors rather than by glutamate molecules. Moreover, the binding between glutamate receptors and glutamate molecules was increased by THz photons. This study offers a nondrug, nonthermal approach to regulate the binding between the excitatory neurotransmitter (glutamate) and NR2B. By increasing synaptic plasticity, it effectively improves the cognitive function of animals with PTSD, providing a promising treatment strategy for NR2B-related cognitive disorders.
Collapse
Affiliation(s)
- Yun Yu
- School of Life Science and Technology,
Xi’an Jiaotong University, Xi’an 710049, China
- Innovation Laboratory of Terahertz Biophysics,
National Innovation Institute of Defense Technology, Beijing 100071, China
| | - Kaijie Wu
- Innovation Laboratory of Terahertz Biophysics,
National Innovation Institute of Defense Technology, Beijing 100071, China
| | - Xiao Yang
- Innovation Laboratory of Terahertz Biophysics,
National Innovation Institute of Defense Technology, Beijing 100071, China
| | - Jiangang Long
- School of Life Science and Technology,
Xi’an Jiaotong University, Xi’an 710049, China
| | - Chao Chang
- Innovation Laboratory of Terahertz Biophysics,
National Innovation Institute of Defense Technology, Beijing 100071, China
- School of Physics,
Peking University, Beijing 100871, China
| |
Collapse
|
64
|
Ritter N, Disse P, Aymanns I, Mücher L, Schreiber JA, Brenker C, Strünker T, Schepmann D, Budde T, Strutz-Seebohm N, Ametamey SM, Wünsch B, Seebohm G. Downstream Allosteric Modulation of NMDA Receptors by 3-Benzazepine Derivatives. Mol Neurobiol 2023; 60:7238-7252. [PMID: 37542648 PMCID: PMC10657792 DOI: 10.1007/s12035-023-03526-1] [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: 10/19/2022] [Accepted: 07/20/2023] [Indexed: 08/07/2023]
Abstract
N-Methyl-D-aspartate receptors (NMDARs) composed of different splice variants display distinct pH sensitivities and are crucial for learning and memory, as well as for inflammatory or injury processes. Dysregulation of the NMDAR has been linked to diseases like Parkinson's, Alzheimer's, schizophrenia, and drug addiction. The development of selective receptor modulators, therefore, constitutes a promising approach for numerous therapeutical applications. Here, we identified (R)-OF-NB1 as a promising splice variant selective NMDAR antagonist. We investigated the interaction of (R)-OF-NB1 and NMDAR from a biochemical, bioinformatical, and electrophysiological perspective to characterize the downstream allosteric modulation of NMDAR by 3-benzazepine derivatives. The allosteric modulatory pathway starts at the ifenprodil binding pocket in the amino terminal domain and immobilizes the connecting α5-helix to the ligand binding domain, resulting in inhibition. In contrast, the exon 5 splice variant GluN1-1b elevates the NMDARs flexibility and promotes the open state of its ligand binding domain.
Collapse
Affiliation(s)
- Nadine Ritter
- Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, 48149, Münster, Germany.
- Chembion, University of Münster, 48149, Münster, Germany.
| | - Paul Disse
- Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, 48149, Münster, Germany
- Chembion, University of Münster, 48149, Münster, Germany
| | - Isabel Aymanns
- Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, 48149, Münster, Germany
| | - Lena Mücher
- Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, 48149, Münster, Germany
| | - Julian A Schreiber
- Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, 48149, Münster, Germany
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Corrensstr. 48, 48149, Münster, Germany
| | - Christoph Brenker
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, University of Münster, Domagkstr. 11, 48149, Münster, Germany
| | - Timo Strünker
- Chembion, University of Münster, 48149, Münster, Germany
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, University of Münster, Domagkstr. 11, 48149, Münster, Germany
| | - Dirk Schepmann
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Corrensstr. 48, 48149, Münster, Germany
| | - Thomas Budde
- Institute of Physiology I, University of Münster, 48149, Münster, Germany
| | - Nathalie Strutz-Seebohm
- Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, 48149, Münster, Germany
| | - Simon M Ametamey
- Institute of Pharmaceutical Sciences, ETH Zürich, Zürich, Switzerland
| | - Bernhard Wünsch
- Chembion, University of Münster, 48149, Münster, Germany
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Corrensstr. 48, 48149, Münster, Germany
| | - Guiscard Seebohm
- Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, 48149, Münster, Germany
- Chembion, University of Münster, 48149, Münster, Germany
| |
Collapse
|
65
|
Mony L, Paoletti P. Mechanisms of NMDA receptor regulation. Curr Opin Neurobiol 2023; 83:102815. [PMID: 37988826 DOI: 10.1016/j.conb.2023.102815] [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: 09/13/2023] [Revised: 10/31/2023] [Accepted: 10/31/2023] [Indexed: 11/23/2023]
Abstract
N-methyl-D-aspartate receptors (NMDARs) are glutamate-gated ion channels widely expressed in the central nervous system that play key role in brain development and plasticity. On the downside, NMDAR dysfunction, be it hyperactivity or hypofunction, is harmful to neuronal function and has emerged as a common theme in various neuropsychiatric disorders including autism spectrum disorders, epilepsy, intellectual disability, and schizophrenia. Not surprisingly, NMDAR signaling is under a complex set of regulatory mechanisms that maintain NMDAR-mediated transmission in check. These include an unusual large number of endogenous agents that directly bind NMDARs and tune their activity in a subunit-dependent manner. Here, we review current knowledge on the regulation of NMDAR signaling. We focus on the regulation of the receptor by its microenvironment as well as by external (i.e. pharmacological) factors and their underlying molecular and cellular mechanisms. Recent developments showing how NMDAR dysregulation participate to disease mechanisms are also highlighted.
Collapse
Affiliation(s)
- Laetitia Mony
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole Normale Supérieure, Université PSL, CNRS, INSERM, F-75005 Paris, France.
| | - Pierre Paoletti
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole Normale Supérieure, Université PSL, CNRS, INSERM, F-75005 Paris, France.
| |
Collapse
|
66
|
Wu E, Zhang J, Zhang J, Zhu S. Structural insights into gating mechanism and allosteric regulation of NMDA receptors. Curr Opin Neurobiol 2023; 83:102806. [PMID: 37950957 DOI: 10.1016/j.conb.2023.102806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 09/28/2023] [Accepted: 10/13/2023] [Indexed: 11/13/2023]
Abstract
N-methyl-d-aspartate receptors (NMDARs) belong to the ionotropic glutamate receptors (iGluRs) superfamily and act as coincidence detectors that are crucial to neuronal development and synaptic plasticity. They typically assemble as heterotetramers of two obligatory GluN1 subunits and two alternative GluN2 (from 2A to 2D) and/or GluN3 (3A and 3B) subunits. These alternative subunits mainly determine the diverse biophysical and pharmacological properties of different NMDAR subtypes. Over the past decade, the unprecedented advances in structure elucidation of these tetrameric NMDARs have provided atomic insights into channel gating, allosteric modulation and the action of therapeutic drugs. A wealth of structural and functional information would accelerate the artificial intelligence-based drug design to exploit more NMDAR subtype-specific molecules for the treatment of neurological and psychiatric disorders.
Collapse
Affiliation(s)
- Enjiang Wu
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, China; Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China. https://twitter.com/DuDaDa_Flower
| | - Jilin Zhang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing, China
| | - Jiwei Zhang
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education Key Laboratory for Standardization of Chinese Medicines, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai, 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 200031, China; University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
67
|
He M, Wang Y, Zhang X, Zhang L. Exploration of the potential neuroprotective compounds targeting GluN1-GluN2B NMDA receptors. J Biomol Struct Dyn 2023; 41:10900-10908. [PMID: 36591642 DOI: 10.1080/07391102.2022.2159527] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 12/10/2022] [Indexed: 01/03/2023]
Abstract
The N-methyl-d-aspartic acid (NMDA) receptors belongs to the family of ionotropic glutamate receptors, which could mediate most excitatory synaptic transmission in the brain. It is interesting to know if some available drugs have regulatory effects on the NMDARs. Herein, the present study reports the discovery of drugs targeting NMDAR using virtual screening. In this study, talniflumate with the EC50 value at 61.49 nM was successfully screened. The interaction analysis of this compound was further explored through molecular dynamics simulation. It is indicated that talniflumate could form stable interactions with GluN1-GluN2B NMDA receptors. In particular, H-bond interactions with high occupancies between GluN1-GluN2B NMDA receptors and talniflumate were observed. Compared to de novo drug discovery, this approach could be an alternative choice for development of safety and efficiency NMDAR inhibitors from available drugs.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Meixi He
- CAS Key Laboratory of Separation Sciences of Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yi Wang
- CAS Key Laboratory of Separation Sciences of Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiaozhe Zhang
- CAS Key Laboratory of Separation Sciences of Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
- Jiangsu Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, JS, China
| | - Lihua Zhang
- CAS Key Laboratory of Separation Sciences of Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| |
Collapse
|
68
|
Janus A, Lustyk K, Pytka K. MK-801 and cognitive functions: Investigating the behavioral effects of a non-competitive NMDA receptor antagonist. Psychopharmacology (Berl) 2023; 240:2435-2457. [PMID: 37725119 PMCID: PMC10640442 DOI: 10.1007/s00213-023-06454-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 08/21/2023] [Indexed: 09/21/2023]
Abstract
RATIONALE MK-801 (dizocilpine) is a non-competitive NMDA receptor antagonist originally explored for anticonvulsant potential. Despite its original purpose, its amnestic properties led to the development of pivotal models of various cognitive impairments widely employed in research and greatly impacting scientific progress. MK-801 offers several advantages; however, it also presents drawbacks, including inducing dose-dependent hyperlocomotion or ambiguous effects on anxiety, which can impact the interpretation of behavioral research results. OBJECTIVES The present review attempts to summarize and discuss the effects of MK-801 on different types of memory and cognitive functions in animal studies. RESULTS A plethora of behavioral research suggests that MK-801 can detrimentally impact cognitive functions. The specific effect of this compound is influenced by variables including developmental stage, gender, species, strain, and, crucially, the administered dose. Notably, when considering the undesirable effects of MK-801, doses up to 0.1 mg/kg were found not to induce stereotypy or hyperlocomotion. CONCLUSION Dizocilpine continues to be of significant importance in preclinical research, facilitating the exploration of various procognitive therapeutic agents. However, given its potential undesirable effects, it is imperative to meticulously determine the appropriate dosages and conduct supplementary evaluations for any undesirable outcomes, which could complicate the interpretation of the findings.
Collapse
Affiliation(s)
- Anna Janus
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Krakow, Poland
| | - Klaudia Lustyk
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Krakow, Poland
| | - Karolina Pytka
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Krakow, Poland.
| |
Collapse
|
69
|
Korff M, Lüken J, Schmidt J, Schepmann D, Goerges G, Ritter N, Disse P, Schreiber JA, Seebohm G, Wünsch B. Negative allosteric modulators of NMDA receptors with GluN2B subunit: synthesis of β-aminoalcohols by epoxide opening and subsequent rearrangement. Org Biomol Chem 2023; 21:7616-7638. [PMID: 37682049 DOI: 10.1039/d3ob01208e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/09/2023]
Abstract
In order to obtain novel antagonists of GluN2B subunit containing NMDA receptors, aryloxiranes were opened with benzylpiperidines. Phenyloxiranes 6 and (indazolyl)oxirane 15 were opened regioselectively at the position bearing the aryl moiety. Reaction of the resulting β-aminoalcohols 7 and 16 with carboxylic acids under Mitsunobu conditions (DIAD, PPh3) led to rearrangement and after ester hydrolysis to the regioisomeric β-aminoalcohols 9 and 18. This strategy allows the synthesis of amino-ifenprodil 12 as well using phthalimide in the Mitsunobu reaction. Unexpectedly, the isomeric (indazolyl)oxirane 21 reacted with benzylpiperidines to afford both regioisomeric β-aminoalcohols 22 and 23. In radioligand receptor binding studies, the indazolyl derivative 18a, which can be regarded as indazole bioisostere of ifenprodil, showed high GluN2B affinity (Ki = 31 nM). Replacement of the benzylic OH moiety of ifenprodil by the NH2 moiety in amino-ifenprodil 12 also resulted in low nanomolar GluN2B affinity (Ki = 72 nM). In TEVC experiments, 18a inhibited the ion flux to the same extent as ifenprodil proving that the phenol of ifenprodil can be replaced bioisosterically by an indazole ring maintaining affinity and inhibitory activity. Whereas 10-fold selectivity was found for the ifenprodil binding site over σ1 receptors, only low preference for the GluN2B receptor over σ2 receptors was detected. The log D7.4 value of 18a (log D7.4 = 2.08) indicates promising bioavailability.
Collapse
Affiliation(s)
- Marvin Korff
- Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstr. 48, D-48149 Münster, Germany.
- Universität Münster, GRK 2515, Chemical biology of ion channels (Chembion), Corrensstraße 48, D-48149 Münster, Germany
| | - Judith Lüken
- Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstr. 48, D-48149 Münster, Germany.
| | - Judith Schmidt
- Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstr. 48, D-48149 Münster, Germany.
| | - Dirk Schepmann
- Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstr. 48, D-48149 Münster, Germany.
| | - Gunnar Goerges
- University Hospital Münster, Cellular Electrophysiology and Molecular Biology, Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, Robert-Koch-Str. 45, D-48149 Münster, Germany
| | - Nadine Ritter
- University Hospital Münster, Cellular Electrophysiology and Molecular Biology, Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, Robert-Koch-Str. 45, D-48149 Münster, Germany
- Universität Münster, GRK 2515, Chemical biology of ion channels (Chembion), Corrensstraße 48, D-48149 Münster, Germany
| | - Paul Disse
- University Hospital Münster, Cellular Electrophysiology and Molecular Biology, Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, Robert-Koch-Str. 45, D-48149 Münster, Germany
- Universität Münster, GRK 2515, Chemical biology of ion channels (Chembion), Corrensstraße 48, D-48149 Münster, Germany
| | - Julian A Schreiber
- Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstr. 48, D-48149 Münster, Germany.
- University Hospital Münster, Cellular Electrophysiology and Molecular Biology, Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, Robert-Koch-Str. 45, D-48149 Münster, Germany
| | - Guiscard Seebohm
- University Hospital Münster, Cellular Electrophysiology and Molecular Biology, Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, Robert-Koch-Str. 45, D-48149 Münster, Germany
- Universität Münster, GRK 2515, Chemical biology of ion channels (Chembion), Corrensstraße 48, D-48149 Münster, Germany
| | - Bernhard Wünsch
- Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstr. 48, D-48149 Münster, Germany.
- Universität Münster, GRK 2515, Chemical biology of ion channels (Chembion), Corrensstraße 48, D-48149 Münster, Germany
| |
Collapse
|
70
|
He S, Sun C, Zhu Q, Li L, Huang J, Wu G, Cao Y, Liao J, Lu Y, Su Q, Lin S, Ma X, Zhong C. A juvenile mouse model of anti-N-methyl-D-aspartate receptor encephalitis by active immunization. Front Mol Neurosci 2023; 16:1211119. [PMID: 37790883 PMCID: PMC10544982 DOI: 10.3389/fnmol.2023.1211119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 08/25/2023] [Indexed: 10/05/2023] Open
Abstract
INTRODUCTION Anti-N-methyl-D-aspartate receptor (NMDAR) encephalitis is a common autoimmune encephalitis, and it is associated with psychosis, dyskinesia, and seizures. Anti-NMDAR encephalitis (NMDARE) in juveniles and adults presents different clinical charactreistics. However, the pathogenesis of juvenile anti-NMDAR encephalitis remains unclear, partly because of a lack of suitable animal models. METHODS We developed a model of juvenile anti-NMDAR encephalitis using active immunization with an amino terminal domain peptide from the GluN1 subunit (GluN1356 - 385) against NMDARs in 3-week-old female C57BL/6J mice. RESULTS Immunofluorescence staining suggested that autoantibody levels in the hippocampus increased, and HEK-293T cells staining identified the target of the autoantibodies as GluN1, suggesting that GluN1-specific immunoglobulin G was successfully induced. Behavior assessment showed that the mice suffered significant cognition impairment and sociability reduction, which is similar to what is observed in patients affected by anti-NMDAR encephalitis. The mice also exhibited impaired long-term potentiation in hippocampal CA1. Pilocarpine-induced epilepsy was more severe and had a longer duration, while no spontaneous seizures were observed. CONCLUSION The juvenile mouse model for anti-NMDAR encephalitis is of great importance to investigate the pathological mechanism and therapeutic strategies for the disease, and could accelerate the study of autoimmune encephalitis.
Collapse
Affiliation(s)
- Shuyu He
- Shenzhen Key Laboratory of Precision Diagnosis and Treatment of Depression, CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institution, Shenzhen, China
- Department of Clinical Research, Department of Neurology, Surgery Division, Epilepsy Center, Shenzhen Children's Hospital, Shenzhen, China
- Shenzhen Children's Hospital of China Medical University, Shenzhen, China
| | - Chongyang Sun
- Shenzhen Key Laboratory of Precision Diagnosis and Treatment of Depression, CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institution, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qian Zhu
- Shenzhen Key Laboratory of Precision Diagnosis and Treatment of Depression, CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institution, Shenzhen, China
- Department of Clinical Research, Department of Neurology, Surgery Division, Epilepsy Center, Shenzhen Children's Hospital, Shenzhen, China
| | - Lin Li
- Department of Clinical Research, Department of Neurology, Surgery Division, Epilepsy Center, Shenzhen Children's Hospital, Shenzhen, China
| | - Jianyu Huang
- Shenzhen Key Laboratory of Precision Diagnosis and Treatment of Depression, CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institution, Shenzhen, China
| | - Ge Wu
- Shenzhen Key Laboratory of Precision Diagnosis and Treatment of Depression, CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institution, Shenzhen, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yi Cao
- Shenzhen Key Laboratory of Precision Diagnosis and Treatment of Depression, CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institution, Shenzhen, China
| | - Jianxiang Liao
- Department of Clinical Research, Department of Neurology, Surgery Division, Epilepsy Center, Shenzhen Children's Hospital, Shenzhen, China
| | - Yi Lu
- Shenzhen Key Laboratory of Precision Diagnosis and Treatment of Depression, CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institution, Shenzhen, China
| | - Qiru Su
- Department of Clinical Research, Department of Neurology, Surgery Division, Epilepsy Center, Shenzhen Children's Hospital, Shenzhen, China
| | - Sufang Lin
- Department of Clinical Research, Department of Neurology, Surgery Division, Epilepsy Center, Shenzhen Children's Hospital, Shenzhen, China
| | - Xiaopeng Ma
- Department of Clinical Research, Department of Neurology, Surgery Division, Epilepsy Center, Shenzhen Children's Hospital, Shenzhen, China
| | - Cheng Zhong
- Shenzhen Key Laboratory of Precision Diagnosis and Treatment of Depression, CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institution, Shenzhen, China
| |
Collapse
|
71
|
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
|
72
|
Zhou JS, Peng GF, Liang WD, Chen Z, Liu YY, Wang BY, Guo ML, Deng YL, Ye JM, Zhong ML, Wang LF. Recent advances in the study of anesthesia-and analgesia-related mechanisms of S-ketamine. Front Pharmacol 2023; 14:1228895. [PMID: 37781698 PMCID: PMC10539608 DOI: 10.3389/fphar.2023.1228895] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 09/04/2023] [Indexed: 10/03/2023] Open
Abstract
Ketamine is a racemic mixture of equal amounts of R-ketamine and S-ketamine and is well known to anesthesiologists for its unique dissociative anesthetic properties. The pharmacological properties of ketamine, namely, its sympathetic excitation, mild respiratory depression, and potent analgesia, are still highly valued in its use as an anesthetic for some patients. In particular, since its advent, S-ketamine has been widely used as an anesthetic in many countries due to its increased affinity for NMDA receptors and its enhanced anesthetic and analgesic effects. However, the anesthetic and analgesic mechanisms of S-ketamine are not fully understood. In addition to antagonizing NMDA receptors, a variety of other receptors or channels may be involved, but there are no relevant mechanistic summaries in the literature. Therefore, the purpose of this paper is to review the mechanisms of action of S-ketamine on relevant receptors and systems in the body that result in its pharmacological properties, such as anesthesia and analgesia, with the aim of providing a reference for its clinical applications and research.
Collapse
Affiliation(s)
- Jian-shun Zhou
- The First Clinical Medical College of Gannan Medical University, Ganzhou, China
| | - Guan-fa Peng
- The First Clinical Medical College of Gannan Medical University, Ganzhou, China
| | - Wei-dong Liang
- Department of Anesthesiology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Ganzhou Key Laboratory of Anesthesiology, Ganzhou, China
| | - Zhen Chen
- The First Clinical Medical College of Gannan Medical University, Ganzhou, China
| | - Ying-ying Liu
- The First Clinical Medical College of Gannan Medical University, Ganzhou, China
| | - Bing-yu Wang
- The First Clinical Medical College of Gannan Medical University, Ganzhou, China
| | - Ming-ling Guo
- The First Clinical Medical College of Gannan Medical University, Ganzhou, China
| | - Yun-ling Deng
- Department of Anesthesiology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Ganzhou Key Laboratory of Anesthesiology, Ganzhou, China
| | - Jun-ming Ye
- Department of Anesthesiology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Ganzhou Key Laboratory of Anesthesiology, Ganzhou, China
| | - Mao-lin Zhong
- Department of Anesthesiology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Ganzhou Key Laboratory of Anesthesiology, Ganzhou, China
| | - Li-feng Wang
- Department of Anesthesiology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Ganzhou Key Laboratory of Anesthesiology, Ganzhou, China
| |
Collapse
|
73
|
Zhang Z, Le GNT, Ge Y, Tang X, Chen X, Ejim L, Bordeleau E, Wright GD, Burns DC, Tran S, Axerio-Cilies P, Wang YT, Dong M, Woolley GA. Isomerization of bioactive acylhydrazones triggered by light or thiols. Nat Chem 2023; 15:1285-1295. [PMID: 37308709 DOI: 10.1038/s41557-023-01239-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 05/12/2023] [Indexed: 06/14/2023]
Abstract
The acylhydrazone unit is well represented in screening databases used to find ligands for biological targets, and numerous bioactive acylhydrazones have been reported. However, potential E/Z isomerization of the C=N bond in these compounds is rarely examined when bioactivity is assayed. Here we analysed two ortho-hydroxylated acylhydrazones discovered in a virtual drug screen for modulators of N-methyl-D-aspartate receptors and other bioactive hydroxylated acylhydrazones with structurally defined targets reported in the Protein Data Bank. We found that ionized forms of these compounds, which are populated under laboratory conditions, photoisomerize readily and the isomeric forms have markedly different bioactivity. Furthermore, we show that glutathione, a tripeptide involved with cellular redox balance, catalyses dynamic E⇄Z isomerization of acylhydrazones. The ratio of E to Z isomers in cells is determined by the relative stabilities of the isomers regardless of which isomer was applied. We conclude that E/Z isomerization may be a common feature of the bioactivity observed with acylhydrazones and should be routinely analysed.
Collapse
Affiliation(s)
- Zhiwei Zhang
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
- Key Laboratory for Advanced Materials, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, China
| | - Giang N T Le
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Yang Ge
- Department of Medicine, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Xiaowen Tang
- Department of Medicinal Chemistry, School of Pharmacy, Qingdao University, Qingdao, China
| | - Xin Chen
- Department of Medicinal Chemistry, School of Pharmacy, Qingdao University, Qingdao, China
| | - Linda Ejim
- David Braley Centre for Antibiotics Discovery M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Emily Bordeleau
- David Braley Centre for Antibiotics Discovery M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Gerard D Wright
- David Braley Centre for Antibiotics Discovery M.G. DeGroote Institute for Infectious Disease Research, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Darcy C Burns
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Susannah Tran
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Peter Axerio-Cilies
- Department of Medicine, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada.
| | - Yu Tian Wang
- Department of Medicine, Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada.
| | - Mingxin Dong
- Department of Medicinal Chemistry, School of Pharmacy, Qingdao University, Qingdao, China.
| | - G Andrew Woolley
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
74
|
Zhou C, Tajima N. Structural insights into NMDA receptor pharmacology. Biochem Soc Trans 2023; 51:1713-1731. [PMID: 37431773 PMCID: PMC10586783 DOI: 10.1042/bst20230122] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/12/2023] [Accepted: 06/16/2023] [Indexed: 07/12/2023]
Abstract
N-methyl-d-aspartate receptors (NMDARs) comprise a subfamily of ionotropic glutamate receptors that form heterotetrameric ligand-gated ion channels and play fundamental roles in neuronal processes such as synaptic signaling and plasticity. Given their critical roles in brain function and their therapeutic importance, enormous research efforts have been devoted to elucidating the structure and function of these receptors and developing novel therapeutics. Recent studies have resolved the structures of NMDARs in multiple functional states, and have revealed the detailed gating mechanism, which was found to be distinct from that of other ionotropic glutamate receptors. This review provides a brief overview of the recent progress in understanding the structures of NMDARs and the mechanisms underlying their function, focusing on subtype-specific, ligand-induced conformational dynamics.
Collapse
Affiliation(s)
- Changping Zhou
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH, U.S.A
| | - Nami Tajima
- Department of Physiology and Biophysics, Case Western Reserve University, Cleveland, OH, U.S.A
| |
Collapse
|
75
|
Lüken J, Goerges G, Ritter N, Disse P, Schreiber JA, Schmidt J, Frehland B, Schepmann D, Seebohm G, Wünsch B. Indazole as a Phenol Bioisostere: Structure-Affinity Relationships of GluN2B-Selective NMDA Receptor Antagonists. J Med Chem 2023; 66:11573-11588. [PMID: 37580890 DOI: 10.1021/acs.jmedchem.3c01161] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2023]
Abstract
Negative allosteric modulation of GluN2B subunit-containing NMDA receptors prevents overstimulation, resulting in neuroprotective effects. Since the phenol of prominent negative allosteric modulators is prone to rapid glucuronidation, its bioisosteric replacement by an indazole was envisaged. The key step in the synthesis was a Sonogashira reaction of non-protected iodoindazoles with propargylpiperidine derivatives. Modification of the alkynyl moiety allowed the introduction of several functional groups. The synthesized indazoles showed very high GluN2B affinity but limited selectivity over σ receptors. Molecular dynamics simulations revealed the same molecular interactions with the ifenprodil binding site as the analogous phenols. In two-electrode voltage-clamp experiments, enantiomeric 3-(4-benzylpiperidin-1-yl)-1-(1H-indazol-5-yl)propan-1-ols (S)-10a and (R)-10a displayed higher inhibitory activity than ifenprodil. In contrast to phenolic GluN2B antagonists, the indazoles were not conjugated with glucuronic acid. It can be concluded that the phenol of potent GluN2B antagonists can be replaced bioisosterically by an indazole, retaining the high GluN2B affinity and activity but inhibiting glucuronidation.
Collapse
Affiliation(s)
- Judith Lüken
- Institut für Pharmazeutische und Medizinische Chemie, Universität Münster, Corrensstraße 48, D-48149 Münster, Germany
| | - Gunnar Goerges
- Cellular Electrophysiology and Molecular Biology, Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, Robert-Koch-Str. 45, D-48149 Münster, Germany
| | - Nadine Ritter
- Cellular Electrophysiology and Molecular Biology, Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, Robert-Koch-Str. 45, D-48149 Münster, Germany
- GRK 2515, Chemical Biology of Ion Channels (Chembion), Universität Münster, Corrensstraße 48, D-48149 Münster, Germany
| | - Paul Disse
- Cellular Electrophysiology and Molecular Biology, Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, Robert-Koch-Str. 45, D-48149 Münster, Germany
- GRK 2515, Chemical Biology of Ion Channels (Chembion), Universität Münster, Corrensstraße 48, D-48149 Münster, Germany
| | - Julian A Schreiber
- Institut für Pharmazeutische und Medizinische Chemie, Universität Münster, Corrensstraße 48, D-48149 Münster, Germany
- Cellular Electrophysiology and Molecular Biology, Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, Robert-Koch-Str. 45, D-48149 Münster, Germany
| | - Judith Schmidt
- Institut für Pharmazeutische und Medizinische Chemie, Universität Münster, Corrensstraße 48, D-48149 Münster, Germany
| | - Bastian Frehland
- Institut für Pharmazeutische und Medizinische Chemie, Universität Münster, Corrensstraße 48, D-48149 Münster, Germany
| | - Dirk Schepmann
- Institut für Pharmazeutische und Medizinische Chemie, Universität Münster, Corrensstraße 48, D-48149 Münster, Germany
| | - Guiscard Seebohm
- Cellular Electrophysiology and Molecular Biology, Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, Robert-Koch-Str. 45, D-48149 Münster, Germany
- GRK 2515, Chemical Biology of Ion Channels (Chembion), Universität Münster, Corrensstraße 48, D-48149 Münster, Germany
| | - Bernhard Wünsch
- Institut für Pharmazeutische und Medizinische Chemie, Universität Münster, Corrensstraße 48, D-48149 Münster, Germany
- GRK 2515, Chemical Biology of Ion Channels (Chembion), Universität Münster, Corrensstraße 48, D-48149 Münster, Germany
| |
Collapse
|
76
|
Vinnakota C, Hudson MR, Jones NC, Sundram S, Hill RA. Potential Roles for the GluN2D NMDA Receptor Subunit in Schizophrenia. Int J Mol Sci 2023; 24:11835. [PMID: 37511595 PMCID: PMC10380280 DOI: 10.3390/ijms241411835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/19/2023] [Accepted: 07/22/2023] [Indexed: 07/30/2023] Open
Abstract
Glutamate N-methyl-D-aspartate receptor (NMDAR) hypofunction has been proposed to underlie schizophrenia symptoms. This theory arose from the observation that administration of NMDAR antagonists, which are compounds that inhibit NMDAR activity, reproduces behavioural and molecular schizophrenia-like phenotypes, including hallucinations, delusions and cognitive impairments in healthy humans and animal models. However, the role of specific NMDAR subunits in these schizophrenia-relevant phenotypes is largely unknown. Mounting evidence implicates the GluN2D subunit of NMDAR in some of these symptoms and pathology. Firstly, genetic and post-mortem studies show changes in the GluN2D subunit in people with schizophrenia. Secondly, the psychosis-inducing effects of NMDAR antagonists are blunted in GluN2D-knockout mice, suggesting that the GluN2D subunit mediates NMDAR-antagonist-induced psychotomimetic effects. Thirdly, in the mature brain, the GluN2D subunit is relatively enriched in parvalbumin (PV)-containing interneurons, a cell type hypothesized to underlie the cognitive symptoms of schizophrenia. Lastly, the GluN2D subunit is widely and abundantly expressed early in development, which could be of importance considering schizophrenia is a disorder that has its origins in early neurodevelopment. The limitations of currently available therapies warrant further research into novel therapeutic targets such as the GluN2D subunit, which may help us better understand underlying disease mechanisms and develop novel and more effective treatment options.
Collapse
Affiliation(s)
- Chitra Vinnakota
- Department of Psychiatry, School of Clinical Sciences, Faculty of Medical, Nursing and Health Sciences, Monash University, Clayton, VIC 3168, Australia
| | - Matthew R Hudson
- Department of Neuroscience, Faculty of Medical, Nursing and Health Sciences, Monash University, Melbourne, VIC 3004, Australia
| | - Nigel C Jones
- Department of Neuroscience, Faculty of Medical, Nursing and Health Sciences, Monash University, Melbourne, VIC 3004, Australia
| | - Suresh Sundram
- Department of Psychiatry, School of Clinical Sciences, Faculty of Medical, Nursing and Health Sciences, Monash University, Clayton, VIC 3168, Australia
- Mental Health Program, Monash Health, Clayton, VIC 3168, Australia
| | - Rachel A Hill
- Department of Psychiatry, School of Clinical Sciences, Faculty of Medical, Nursing and Health Sciences, Monash University, Clayton, VIC 3168, Australia
| |
Collapse
|
77
|
Façanha Wendel C, Hapuque Oliveira Alencar Q, Viana Vieira R, Teixeira KN. In silico insight into Amurensinine - an N-Methyl-D-Aspartate receptor antagonist. World J Pharmacol 2023; 12:25-34. [DOI: 10.5497/wjp.v12.i3.25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/05/2023] [Accepted: 05/31/2023] [Indexed: 06/15/2023] Open
Abstract
BACKGROUND Some isopavines can exhibit important biological activity in the treatment of neurological disorders since it is considered an antagonist of the specific N-methyl-D-Aspartate (NMDA) receptor. Amurensinine is an isopavine which still has few studies. In view of the potential of isopavines as NMDA receptor antagonists, theoretical studies using bioinformatics were carried out in order to investigate whether Amurensinine binds to the NMDA receptor and to analyze the receptor/Ligand complex. This data can contribute to understanding of the onset of neurological diseases and contribute to the planning of drugs for the treatment of neurological diseases involving the NMDA receptor.
AIM To investigate the interaction of the antagonist Amurensinine on the GluN1A/ GluN2B isoform of the NMDA receptor using bioinformatics.
METHODS The three-dimen-sional structure of the GluN1A/GluN2B NMDA receptor was selected from the Protein Data Bank (PDB) - PDB: 4PE5, and the three-dimen-sional structure of Amurensinine (ligand) was designed and optimized using ACD/SchemsketchTM software. Prediction of the protonation state of Amur-ensinine at physiological pH was performed using MarvinSketch software (ChemAxon). Protonated and non-protonated Amurensin were prepared using AutoDock Tools 4 software and simulations were performed using Autodock Vina v.1.2.0. The receptor/Ligand complexes were analyzed using PyMol (Schrödinger, Inc) and BIOVIA Discovery Studio (Dassault Systemes) software. To evaluate the NMDA receptor/Amurensinine complex and validate the molecular docking, simulations using NMDA receptor and Ifenprodil antagonist were performed under the same conditions. Ifenprodil was also designed, optimized and protonated, under the same conditions as Amurensinine.
RESULTS Molecular docking simulations showed that both non-protonated and protonated Amurensinine bind to the amino terminal domain (ATD) domain of the GluN1A/GluN2B NMDA receptor with significant affinity energy, -7.9 Kcal/mol and -8.1 Kcal/mol, respectively. The NMDA receptor/non-protonated Amurensinine complex was stabilized by 15 bonds, while the NMDA receptor/protonated Amurensinine complex was stabilized by less than half, 6 bonds. Despite the difference in the number of bonds, the variation in bond length and the average bond length values are similar in both complexes. The complex formed by the NMDA receptor and Ifenprodil showed an affinity energy of -8.2 Kcal/mol, a value very close to that obtained for the NMDA receptor/Amurensinine complex. Molecular docking between Ifenprodil and the GluN1A /GluN2B NMDA receptor demonstrated that this antagonist interacts with the ATD of the receptor, which validates the simulations performed with Amurensinine.
CONCLUSION Amurensinine binds to the NMDA receptor on ATD, similar to Ifenprodil, and the affinity energy is closer. These data suggest that Amurensinine could behave as a receptor inhibitor, indicating that this compound may have a potential biological application, which should be evaluated by in vitro and preclinical assays.
Collapse
Affiliation(s)
- Cinthia Façanha Wendel
- Campus Toledo, Universidade Federal do Paraná, Toledo 85.919-899, Paraná, Brazil
- Programa Multicêntrico de Pós-graduação em Bioquímica e Biologia Molecular - Setor Palotina, Universidade Federal do Paraná, Palotina 85.950-000, Paraná, Brazil
| | | | - Rafaela Viana Vieira
- Campus Toledo, Universidade Federal do Paraná, Toledo 85.919-899, Paraná, Brazil
| | - Kádima Nayara Teixeira
- Campus Toledo, Universidade Federal do Paraná, Toledo 85.919-899, Paraná, Brazil
- Programa Multicêntrico de Pós-graduação em Bioquímica e Biologia Molecular - Setor Palotina, Universidade Federal do Paraná, Palotina 85.950-000, Paraná, Brazil
| |
Collapse
|
78
|
Rouzbeh N, Rau AR, Benton AJ, Yi F, Anderson CM, Johns MR, Jensen L, Lotti JS, Holley DC, Hansen KB. Allosteric modulation of GluN1/GluN3 NMDA receptors by GluN1-selective competitive antagonists. J Gen Physiol 2023; 155:e202313340. [PMID: 37078900 PMCID: PMC10125900 DOI: 10.1085/jgp.202313340] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/07/2023] [Accepted: 03/29/2023] [Indexed: 04/21/2023] Open
Abstract
NMDA-type ionotropic glutamate receptors are critical for normal brain function and are implicated in central nervous system disorders. Structure and function of NMDA receptors composed of GluN1 and GluN3 subunits are less understood compared to those composed of GluN1 and GluN2 subunits. GluN1/3 receptors display unusual activation properties in which binding of glycine to GluN1 elicits strong desensitization, while glycine binding to GluN3 alone is sufficient for activation. Here, we explore mechanisms by which GluN1-selective competitive antagonists, CGP-78608 and L-689,560, potentiate GluN1/3A and GluN1/3B receptors by preventing glycine binding to GluN1. We show that both CGP-78608 and L-689,560 prevent desensitization of GluN1/3 receptors, but CGP-78608-bound receptors display higher glycine potency and efficacy at GluN3 subunits compared to L-689,560-bound receptors. Furthermore, we demonstrate that L-689,560 is a potent antagonist of GluN1FA+TL/3A receptors, which are mutated to abolish glycine binding to GluN1, and that this inhibition is mediated by a non-competitive mechanism involving binding to the mutated GluN1 agonist binding domain (ABD) to negatively modulate glycine potency at GluN3A. Molecular dynamics simulations reveal that CGP-78608 and L-689,560 binding or mutations in the GluN1 glycine binding site promote distinct conformations of the GluN1 ABD, suggesting that the GluN1 ABD conformation influences agonist potency and efficacy at GluN3 subunits. These results uncover the mechanism that enables activation of native GluN1/3A receptors by application of glycine in the presence of CGP-78608, but not L-689,560, and demonstrate strong intra-subunit allosteric interactions in GluN1/3 receptors that may be relevant to neuronal signaling in brain function and disease.
Collapse
Affiliation(s)
- Nirvan Rouzbeh
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT, USA
| | - Andrew R. Rau
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT, USA
| | - Avery J. Benton
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT, USA
- Department of Biomedical and Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Montana, Missoula, MT, USA
| | - Feng Yi
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT, USA
| | - Carly M. Anderson
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT, USA
| | - Mia R. Johns
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT, USA
| | - Loren Jensen
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT, USA
- Department of Biomedical and Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Montana, Missoula, MT, USA
| | - James S. Lotti
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT, USA
- Department of Biomedical and Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Montana, Missoula, MT, USA
| | - David C. Holley
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT, USA
- Department of Biomedical and Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Montana, Missoula, MT, USA
| | - Kasper B. Hansen
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT, USA
| |
Collapse
|
79
|
Bej A, Ames JB. Chemical shift assignments of calmodulin bound to a cytosolic domain of GluN2A (residues 1004-1024) from the NMDA receptor. BIOMOLECULAR NMR ASSIGNMENTS 2023; 17:89-93. [PMID: 37029330 DOI: 10.1007/s12104-023-10125-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 03/30/2023] [Indexed: 05/21/2023]
Abstract
N-methyl-D-aspartate receptors (NMDARs) consist of glycine-binding GluN1 and glutamate-binding GluN2 subunits that form tetrameric ion channels. NMDARs in the neuronal post-synaptic membrane are important for controlling neuroplasticity and synaptic transmission in the brain. Calmodulin (CaM) binds to the cytosolic C0 domains of both GluN1 (residues 841-865) and GluN2 (residues 1004-1024) that may play a role in the Ca2+-dependent desensitization of NMDAR channels. Mutations that disrupt Ca2+-dependent desensitization of NMDARs are linked to Alzheimer's disease, depression, stroke, epilepsy, and schizophrenia. NMR chemical shift assignments are reported here for Ca2+-saturated CaM bound to the GluN2A C0 domain of NMDAR (BMRB no. 51821).
Collapse
Affiliation(s)
- Aritra Bej
- Department of Chemistry, University of California, Davis, CA, 95616, USA
| | - James B Ames
- Department of Chemistry, University of California, Davis, CA, 95616, USA.
| |
Collapse
|
80
|
Bej A, Ames JB. Chemical shift assignments of calmodulin bound to the GluN1 C0 domain (residues 841-865) of the NMDA receptor. BIOMOLECULAR NMR ASSIGNMENTS 2023; 17:61-65. [PMID: 36739573 PMCID: PMC10232624 DOI: 10.1007/s12104-023-10121-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 01/26/2023] [Indexed: 05/31/2023]
Abstract
Neuroplasticity and synaptic transmission in the brain are regulated by N-methyl-D-aspartate receptors (NMDARs) that consist of hetero-tetrameric combinations of the glycine-binding GluN1 and glutamate-binding GluN2 subunits. Calmodulin (CaM) binds to the cytosolic C0 domain of GluN1 (residues 841-865) that may play a role in the Ca2+-dependent inactivation (CDI) of NMDAR channel activity. Dysregulation of NMDARs are linked to various neurological disorders, including Alzheimer's disease, depression, stroke, epilepsy, and schizophrenia. Here, we report complete NMR chemical shift assignments of Ca2+-saturated CaM bound to the GluN1 C0 domain of the human NMDAR (BMRB no. 51715).
Collapse
Affiliation(s)
- Aritra Bej
- Department of Chemistry, University of California, Davis, CA, 95616, USA
| | - James B Ames
- Department of Chemistry, University of California, Davis, CA, 95616, USA.
| |
Collapse
|
81
|
Bossi S, Pizzamiglio L, Paoletti P. Excitatory GluN1/GluN3A glycine receptors (eGlyRs) in brain signaling. Trends Neurosci 2023:S0166-2236(23)00127-3. [PMID: 37248111 DOI: 10.1016/j.tins.2023.05.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 04/28/2023] [Accepted: 05/04/2023] [Indexed: 05/31/2023]
Abstract
GluN3A is a glycine-binding subunit belonging to the NMDA receptor (NMDAR) family that can assemble with GluN1 subunits to form unconventional NMDARs insensitive to glutamate and activated by glycine only. The existence of such excitatory glycine receptors (eGlyRs) in the central nervous system (CNS) has long remained elusive. Recently, eGlyRs have been identified in specific brain regions, where they represent a novel neuronal signaling modality by which extracellular glycine tunes neuronal excitability, circuit function, and behavior. In this review, we summarize the emerging knowledge regarding these underappreciated receptors. The existence of eGlyRs reshapes current understanding of NMDAR diversity and of glycinergic signaling, previously thought to be primarily inhibitory. Given that GluN3A expression is concentrated in brain regions regulating emotional responses, eGlyRs are potential new targets of therapeutic interest in neuropsychiatry.
Collapse
Affiliation(s)
- Simon Bossi
- Institut de Biologie de l'École Normale Supérieure (IBENS), Ecole Normale Supérieure, Université PSL, CNRS, INSERM, F-75005 Paris, France
| | - Lara Pizzamiglio
- Institut de Biologie de l'École Normale Supérieure (IBENS), Ecole Normale Supérieure, Université PSL, CNRS, INSERM, F-75005 Paris, France
| | - Pierre Paoletti
- Institut de Biologie de l'École Normale Supérieure (IBENS), Ecole Normale Supérieure, Université PSL, CNRS, INSERM, F-75005 Paris, France.
| |
Collapse
|
82
|
Ye S, Han Y, Wei Z, Li J. Binding Affinity and Mechanisms of Potential Antidepressants Targeting Human NMDA Receptors. Molecules 2023; 28:4346. [PMID: 37298821 PMCID: PMC10254814 DOI: 10.3390/molecules28114346] [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: 04/17/2023] [Revised: 05/12/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
Depression, a mental disorder that plagues the world, is a burden on many families. There is a great need for new, fast-acting antidepressants to be developed. N-methyl-D-aspartic acid (NMDA) is an ionotropic glutamate receptor that plays an important role in learning and memory processes and its TMD region is considered as a potential target to treat depression. However, due to the unclear binding sites and pathways, the mechanism of drug binding lacks basic explanation, which brings great complexity to the development of new drugs. In this study, we investigated the binding affinity and mechanisms of an FDA-approved antidepressant (S-ketamine) and seven potential antidepressants (R-ketamine, memantine, lanicemine, dextromethorphan, Ro 25-6981, ifenprodil, and traxoprodil) targeting the NMDA receptor by ligand-protein docking and molecular dynamics simulations. The results indicated that Ro 25-6981 has the strongest binding affinity to the TMD region of the NMDA receptor among the eight selected drugs, suggesting its potential effective inhibitory effect. We also calculated the critical binding-site residues at the active site and found that residues Leu124 and Met63 contributed the most to the binding energy by decomposing the free energy contributions on a per-residue basis. We further compared S-ketamine and its chiral molecule, R-ketamine, and found that R-ketamine had a stronger binding capacity to the NMDA receptor. This study provides a computational reference for the treatment of depression targeting NMDA receptors, and the proposed results will provide potential strategies for further antidepressant development and is a useful resource for the future discovery of fast-acting antidepressant candidates.
Collapse
Affiliation(s)
- Simin Ye
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China;
- Key Laboratory for Thin Film and Microfabrication of Ministry of Education, Department of Micro/Nano-Electronics, Shanghai Jiao Tong University, Shanghai 200240, China;
| | - Yanqiang Han
- Key Laboratory for Thin Film and Microfabrication of Ministry of Education, Department of Micro/Nano-Electronics, Shanghai Jiao Tong University, Shanghai 200240, China;
| | - Zhiyun Wei
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China;
| | - Jinjin Li
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai 200092, China;
- Key Laboratory for Thin Film and Microfabrication of Ministry of Education, Department of Micro/Nano-Electronics, Shanghai Jiao Tong University, Shanghai 200240, China;
| |
Collapse
|
83
|
Chen JZ, Church WB, Bastard K, Duff AP, Balle T. Binding and Dynamics Demonstrate the Destabilization of Ligand Binding for the S688Y Mutation in the NMDA Receptor GluN1 Subunit. Molecules 2023; 28:molecules28104108. [PMID: 37241849 DOI: 10.3390/molecules28104108] [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: 04/05/2023] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Encephalopathies are brain dysfunctions that lead to cognitive, sensory, and motor development impairments. Recently, the identification of several mutations within the N-methyl-D-aspartate receptor (NMDAR) have been identified as significant in the etiology of this group of conditions. However, a complete understanding of the underlying molecular mechanism and changes to the receptor due to these mutations has been elusive. We studied the molecular mechanisms by which one of the first mutations within the NMDAR GluN1 ligand binding domain, Ser688Tyr, causes encephalopathies. We performed molecular docking, randomly seeded molecular dynamics simulations, and binding free energy calculations to determine the behavior of the two major co-agonists: glycine and D-serine, in both the wild-type and S688Y receptors. We observed that the Ser688Tyr mutation leads to the instability of both ligands within the ligand binding site due to structural changes associated with the mutation. The binding free energy for both ligands was significantly more unfavorable in the mutated receptor. These results explain previously observed in vitro electrophysiological data and provide detailed aspects of ligand association and its effects on receptor activity. Our study provides valuable insight into the consequences of mutations within the NMDAR GluN1 ligand binding domain.
Collapse
Affiliation(s)
- Jake Zheng Chen
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
- Brain and Mind Centre, The University of Sydney, Camperdown, NSW 2050, Australia
| | - William Bret Church
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Karine Bastard
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Anthony P Duff
- National Deuteration Facility, Australian Nuclear Science and Technology Organization, New Illawarra Road, Lucas Heights, NSW 2234, Australia
| | - Thomas Balle
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
- Brain and Mind Centre, The University of Sydney, Camperdown, NSW 2050, Australia
| |
Collapse
|
84
|
Ritter N, Disse P, Wünsch B, Seebohm G, Strutz-Seebohm N. Pharmacological Potential of 3-Benzazepines in NMDAR-Linked Pathophysiological Processes. Biomedicines 2023; 11:1367. [PMID: 37239037 PMCID: PMC10216354 DOI: 10.3390/biomedicines11051367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/22/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
The number of N-Methyl-D-aspartate receptor (NMDAR) linked neurodegenerative diseases such as Alzheimer's disease and dementia is constantly increasing. This is partly due to demographic change and presents new challenges to societies. To date, there are no effective treatment options. Current medications are nonselective and can lead to unwanted side effects in patients. A promising therapeutic approach is the targeted inhibition of NMDARs in the brain. NMDARs containing different subunits and splice variants display different physiological properties and play a crucial role in learning and memory, as well as in inflammatory or injury processes. They become overactivated during the course of the disease, leading to nerve cell death. Until now, there has been a lack of understanding of the general functions of the receptor and the mechanism of inhibition, which need to be understood in order to develop inhibitors. Ideal compounds should be highly targeted and even splice-variant-selective. However, a potent and splice-variant-selective NMDAR-targeting drug has yet to be developed. Recently developed 3-benzazepines are promising inhibitors for further drug development. The NMDAR splice variants GluN1-1b-4b carry a 21-amino-acid-long, flexible exon 5. Exon 5 lowers the NMDAR's sensitivity to allosteric modulators by probably acting as an NMDAR modulator itself. The role of exon 5 in NMDAR modulation is still poorly understood. In this review, we summarize the structure and pharmacological relevance of tetrahydro-3-benzazepines.
Collapse
Affiliation(s)
- Nadine Ritter
- Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, D-48149 Münster, Germany; (P.D.); (G.S.); (N.S.-S.)
- Chembion, University of Münster, D-48149 Münster, Germany;
| | - Paul Disse
- Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, D-48149 Münster, Germany; (P.D.); (G.S.); (N.S.-S.)
- Chembion, University of Münster, D-48149 Münster, Germany;
| | - Bernhard Wünsch
- Chembion, University of Münster, D-48149 Münster, Germany;
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Corrensstr. 48, D-48149 Münster, Germany
| | - Guiscard Seebohm
- Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, D-48149 Münster, Germany; (P.D.); (G.S.); (N.S.-S.)
- Chembion, University of Münster, D-48149 Münster, Germany;
| | - Nathalie Strutz-Seebohm
- Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, D-48149 Münster, Germany; (P.D.); (G.S.); (N.S.-S.)
| |
Collapse
|
85
|
Engin AB, Engin A, Engin ED, Memis L. Does lithium attenuate the liver damage due to oxidative stress and liver glycogen depletion in experimental common bile duct obstruction? Toxicol Appl Pharmacol 2023; 466:116489. [PMID: 36963521 DOI: 10.1016/j.taap.2023.116489] [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: 12/08/2022] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 03/26/2023]
Abstract
In extrahepatic cholestasis, the molecular mechanisms of liver damage due to bile acid accumulation remain elusive. In this study, the activation of glutamatergic receptors was hypothesized to be responsible for bile acid-induced oxidative stress and liver damage. Recent evidence showed that lithium, as an N-methyl-d-aspartate receptor (NMDAR) GluN2B subunit inhibitor, may act on the glutamate/NMDAR signaling axis. Guinea pigs were assigned to four groups, as sham laparotomy (SL), bile duct ligated (BDL), lithium-treated SL (SL + Li) and lithium-treated BDL (BDL + Li) groups. Cholestasis-induced liver injury was evaluated by aspartate aminotransferase (AST), alanine transaminase (ALT), interleukin-6 (IL-6), tissue malondialdehyde (MDA), copper‑zinc superoxide dismutase and reduced glutathione levels. The liability of glutamate/NMDAR signaling axis was clarified by glutamate levels in both plasma and liver samples, with the production of nitric oxide (NO), as well as with the serum calcium concentrations. Blood glucose, glucagon, insulin levels and glucose consumption rates, in addition to tissue glycogen were measured to evaluate the liver glucose-glycogen metabolism. A high liver damage index (AST/ALT) was calculated in BDL animals in comparison to SL group. In the BDL animals, lithium reduced plasma NO and glutamate in addition to tissue glutamate concentrations, while serum calcium increased. The antioxidant capacities and liver glycogen contents significantly increased, whereas blood glucose levels unchanged and tissue MDA levels decreased 3-fold in lithium-treated cholestatic animals. It was concluded that lithium largely protects the cholestatic hepatocyte from bile acid-mediated damage by blocking the NMDAR-GluN2B subunit.
Collapse
Affiliation(s)
- Ayse Basak Engin
- Gazi University, Faculty of Pharmacy, Department of Toxicology, Ankara, Turkey.
| | - Atilla Engin
- Gazi University, Faculty of Medicine, Department of General Surgery, Ankara, Turkey
| | - Evren Doruk Engin
- Ankara University, Biotechnology Institute, Gumusdere Campus, Kecioren, Ankara, Turkey
| | - Leyla Memis
- Gazi University, Faculty of Medicine, Department of Pathology, Ankara, Turkey
| |
Collapse
|
86
|
Xie L, McDaniel MJ, Perszyk RE, Kim S, Cappuccio G, Shapiro KA, Muñoz-Cabello B, Sanchez-Lara PA, Grand K, Zhang J, Nocilla KA, Sheikh R, Armengol L, Romano R, Pierson TM, Yuan H, Myers SJ, Traynelis SF. Functional effects of disease-associated variants reveal that the S1-M1 linker of the NMDA receptor critically controls channel opening. Cell Mol Life Sci 2023; 80:110. [PMID: 37000222 PMCID: PMC10641759 DOI: 10.1007/s00018-023-04705-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/12/2023] [Accepted: 01/20/2023] [Indexed: 04/01/2023]
Abstract
The short pre-M1 helix within the S1-M1 linker (also referred to as the pre-M1 linker) between the agonist-binding domain (ABD, S1) and the M1 transmembrane helix of the NMDA receptor (NMDAR) is devoid of missense variants within the healthy population but is a locus for de novo pathogenic variants associated with neurological disorders. Several de novo variants within this helix have been identified in patients presenting early in life with intellectual disability, developmental delay, and/or epilepsy. In this study, we evaluated functional properties for twenty variants within the pre-M1 linker in GRIN1, GRIN2A, and GRIN2B genes, including six novel missense variants. The effects of pre-M1 variants on agonist potency, sensitivity to endogenous allosteric modulators, response time course, channel open probability, and surface expression were assessed. Our data indicated that virtually all of the variants evaluated altered channel function, and multiple variants had profound functional consequences, which may contribute to the neurological conditions in the patients harboring the variants in this region. These data strongly suggest that the residues within the pre-M1 helix play a key role in channel gating and are highly intolerant to genetic variation.
Collapse
Affiliation(s)
- Lingling Xie
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 1510 Clifton Road, NE, Atlanta, GA, 30322, USA
- Department of Neurology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Miranda J McDaniel
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 1510 Clifton Road, NE, Atlanta, GA, 30322, USA
| | - Riley E Perszyk
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 1510 Clifton Road, NE, Atlanta, GA, 30322, USA
| | - Sukhan Kim
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 1510 Clifton Road, NE, Atlanta, GA, 30322, USA
- Center for Functional Evaluation of Rare Variants (CFERV), Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Gerarda Cappuccio
- Section of Pediatrics, Department of Translational Medicine, 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
| | - Kevin A Shapiro
- Department of Neurology, University of California, UCSF Memory and Aging Center, Sandler Neurosciences Center, San Francisco, CA, USA
| | | | - Pedro A Sanchez-Lara
- Division of Medical Genetics, Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, USA
| | - Katheryn Grand
- Division of Medical Genetics, Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, USA
| | - Jing Zhang
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 1510 Clifton Road, NE, Atlanta, GA, 30322, USA
| | - Kelsey A Nocilla
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 1510 Clifton Road, NE, Atlanta, GA, 30322, USA
| | - Rehan Sheikh
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 1510 Clifton Road, NE, Atlanta, GA, 30322, USA
| | - Lluis Armengol
- Quantitative Genomic Medicine Laboratories, SL (qGenomics), Barcelona, Spain
| | - Roberta Romano
- Section of Pediatrics, Department of Translational Medicine, Federico II University, Via Pansini 5, 80131, Naples, Italy
| | - Tyler Mark Pierson
- Division of Pediatric Neurology, Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, USA
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, USA
- Center for the Undiagnosed Patient, Cedars-Sinai Medical Center, Los Angeles, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, USA
| | - Hongjie Yuan
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 1510 Clifton Road, NE, Atlanta, GA, 30322, USA
- Center for Functional Evaluation of Rare Variants (CFERV), Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Scott J Myers
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, 1510 Clifton Road, NE, 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, 1510 Clifton Road, NE, 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, Atlanta, GA, 30322, USA.
| |
Collapse
|
87
|
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
|
88
|
Disse P, Aymanns I, Ritter N, Peischard S, Korn L, Wiendl H, Pawlowski M, Kovac S, Meuth SG, Budde T, Strutz-Seebohm N, Wünsch B, Seebohm G. A novel NMDA receptor test model based on hiPSC-derived neural cells. Biol Chem 2023; 404:267-277. [PMID: 36630596 DOI: 10.1515/hsz-2022-0216] [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: 06/30/2022] [Accepted: 12/12/2022] [Indexed: 01/13/2023]
Abstract
N-Methyl-D-aspartate receptors (NMDARs) are central for learning and information processing in the brain. Dysfunction of NMDARs can play a key role in the pathogenesis of neurodegeneration and drug addiction. The development of selective NMDAR modulators represents a promising strategy to target these diseases. Among such modulating compounds are ifenprodil and its 3-benzazepine derivatives. Classically, the effects of these NMDAR modulators have been tested by techniques like two-electrode voltage clamp (TEVC), patch clamp, or fluorescence-based assays. However, testing their functional effects in complex human systems requires more advanced approaches. Here, we established a human induced pluripotent stem cell-derived (hiPSC-derived) neural cell system and proved its eligibility as a test system for investigating NMDAR modulators and pharmaceutical effects on human neurons.
Collapse
Affiliation(s)
- Paul Disse
- Institut für Genetik von Herzerkrankungen (IfGH), Zelluläre Elektrophysiologie, Universitätsklinikum Münster, D-48149 Münster, Germany
- Chembion, GRK 2515, Westfälische Wilhelms-Universität Münster, D-48149 Münster, Germany
| | - Isabel Aymanns
- Institut für Genetik von Herzerkrankungen (IfGH), Zelluläre Elektrophysiologie, Universitätsklinikum Münster, D-48149 Münster, Germany
| | - Nadine Ritter
- Institut für Genetik von Herzerkrankungen (IfGH), Zelluläre Elektrophysiologie, Universitätsklinikum Münster, D-48149 Münster, Germany
- Chembion, GRK 2515, Westfälische Wilhelms-Universität Münster, D-48149 Münster, Germany
| | - Stefan Peischard
- Institut für Genetik von Herzerkrankungen (IfGH), Zelluläre Elektrophysiologie, Universitätsklinikum Münster, D-48149 Münster, Germany
| | - Lisanne Korn
- Klinik für Neurologie mit Institut für Translationale Neurologie, Universitätsklinikum Münster, D-48149 Münster, Germany
| | - Heinz Wiendl
- Klinik für Neurologie mit Institut für Translationale Neurologie, Universitätsklinikum Münster, D-48149 Münster, Germany
| | - Matthias Pawlowski
- Klinik für Neurologie mit Institut für Translationale Neurologie, Universitätsklinikum Münster, D-48149 Münster, Germany
| | - Stjepana Kovac
- Klinik für Neurologie mit Institut für Translationale Neurologie, Universitätsklinikum Münster, D-48149 Münster, Germany
| | - Sven G Meuth
- Neurologische Klinik, Heinrich-Heine-Universität Düsseldorf, D-40225 Düsseldorf, Germany
| | - Thomas Budde
- Institut für Physiologie I, Westfälische Wilhelms-Universität, D-48149 Münster, Germany
| | - Nathalie Strutz-Seebohm
- Institut für Genetik von Herzerkrankungen (IfGH), Zelluläre Elektrophysiologie, Universitätsklinikum Münster, D-48149 Münster, Germany
| | - Bernhard Wünsch
- Institut für Pharmazeutische and Medizinische Chemie, Westfälische Wilhelms-Universität Münster, D-48149 Münster, Germany
| | - Guiscard Seebohm
- Institut für Genetik von Herzerkrankungen (IfGH), Zelluläre Elektrophysiologie, Universitätsklinikum Münster, D-48149 Münster, Germany
| |
Collapse
|
89
|
Bechthold E, Grey L, Diamant E, Schmidt J, Steigerwald R, Zhao F, Hansen KB, Bunch L, Clausen RP, Wünsch B. In vitro ADME characterization of a very potent 3-acylamino-2-aminopropionic acid-derived GluN2C-NMDA receptor agonist and its ester prodrugs. Biol Chem 2023; 404:255-265. [PMID: 36427206 PMCID: PMC10012426 DOI: 10.1515/hsz-2022-0229] [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: 07/12/2022] [Accepted: 11/01/2022] [Indexed: 11/26/2022]
Abstract
The GluN2C subunit exists predominantly, but not exclusively in NMDA receptors within the cerebellum. Antagonists such as UBP1700 and positive allosteric modulators including PYD-106 and 3-acylamino-2-aminopropionic acid derivatives such as UA3-10 ((R)-2-amino-3-{[5-(2-bromophenyl)thiophen-2-yl]carboxamido}propionic acid) represent promising tool compounds to investigate the role of GluN2C-containing NMDA receptors in the signal transduction in the brain. However, due to its high polarity the bioavailability and CNS penetration of the amino acid UA3-10 are expected to be rather low. Herein, three ester prodrugs 12a-c of the NMDA receptor glycine site agonist UA3-10 were prepared and pharmacokinetically characterized. The esters 12a-c showed higher lipophilicity (higher logD 7.4 values) than the acid UA3-10 but almost the same binding at human serum albumin. The acid UA3-10 was rather stable upon incubation with mouse liver microsomes and NADPH, but the esters 12a-c were fast hydrolyzed to afford the acid UA3-10. Incubation with pig liver esterase and mouse serum led to rapid hydrolysis of the esters 12a-c. The isopropyl ester 12c showed a promising logD 7.4 value of 3.57 and the highest stability in the presence of pig liver esterase and mouse serum. These results demonstrate that ester prodrugs of UA3-10 can potentially afford improved bioavailability and CNS penetration.
Collapse
Affiliation(s)
- Elena Bechthold
- Westfälische Wilhelms-Universität Münster, GRK 2515, Chemical Biology of Ion Channels (Chembion), Corrensstraße 48, D-48149Münster, Germany
- Westfälische Wilhelms-Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstraße 48, D-48149Münster, Germany
| | - Lucie Grey
- Westfälische Wilhelms-Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstraße 48, D-48149Münster, Germany
| | - Emil Diamant
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100Copenhagen, Denmark
| | - Judith Schmidt
- Westfälische Wilhelms-Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstraße 48, D-48149Münster, Germany
| | - Ruben Steigerwald
- Westfälische Wilhelms-Universität Münster, GRK 2515, Chemical Biology of Ion Channels (Chembion), Corrensstraße 48, D-48149Münster, Germany
- Westfälische Wilhelms-Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstraße 48, D-48149Münster, Germany
| | - Fabao Zhao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, No. 44, Wenhua West Road, Lixia District, Ji’nan, Shandong, 250012, China
| | - Kasper B. Hansen
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, 32 Campus Drive, Missoula, MT59812, USA
| | - Lennart Bunch
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100Copenhagen, Denmark
| | - Rasmus P. Clausen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100Copenhagen, Denmark
| | - Bernhard Wünsch
- Westfälische Wilhelms-Universität Münster, GRK 2515, Chemical Biology of Ion Channels (Chembion), Corrensstraße 48, D-48149Münster, Germany
- Westfälische Wilhelms-Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstraße 48, D-48149Münster, Germany
| |
Collapse
|
90
|
Korff M, Steigerwald R, Bechthold E, Schepmann D, Schreiber JA, Meuth SG, Seebohm G, Wünsch B. Chemical, pharmacodynamic and pharmacokinetic characterization of the GluN2B receptor antagonist 3-(4-phenylbutyl)-2,3,4,5-tetrahydro-1 H-3-benzazepine-1,7-diol - starting point for PET tracer development. Biol Chem 2023; 404:279-289. [PMID: 36215695 DOI: 10.1515/hsz-2022-0222] [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: 07/06/2022] [Accepted: 09/22/2022] [Indexed: 11/15/2022]
Abstract
GluN2B-NMDA receptors play a key role in several neurological and neurodegenerative disorders. In order to develop novel negative allosteric GluN2B-NMDA receptor modulators, the concept of conformational restriction was pursued, i.e. the flexible aminoethanol substructure of ifenprodil was embedded into a more rigid tetrahydro-3-benzazepine system. The resulting tetrahydro-3-benzazepine-1,7-diol (±)-2 (WMS-1410) showed promising receptor affinity in receptor binding studies (K i = 84 nM) as well as pharmacological activity in two-electrode-voltage-clamp experiments (IC 50 = 116 nM) and in cytoprotective assays (IC 50 = 18.5 nM). The interactions of (R)-2 with the ifenprodil binding site of GluN2B-NMDA receptors were analyzed on the molecular level and the "foot-in-the-door" mechanism was developed. Due to promising pharmacokinetic parameters (logD7.4 = 1.68, plasma protein binding of 76-77%, sufficient metabolic stability) F-substituted analogs were prepared and evaluated as tracers for positron emission tomography (PET). Both fluorine-18-labeled PET tracers [18F]11 and [18F]15 showed high brain uptake, specific accumulation in regions known for high GluN2B-NMDA receptor expression, but no interactions with σ 1 receptors. Radiometabolites were not observed in the brain. Both PET tracers might be suitable for application in humans.
Collapse
Affiliation(s)
- Marvin Korff
- Chemical Biology of Ion Channels (Chembion), Westfälische Wilhelms-Universität Münster, GRK 2515 Munster, Germany
- Westfälische Wilhelms-Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstraße 48, D-48149 Münster, Germany
| | - Ruben Steigerwald
- Chemical Biology of Ion Channels (Chembion), Westfälische Wilhelms-Universität Münster, GRK 2515 Munster, Germany
- Westfälische Wilhelms-Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstraße 48, D-48149 Münster, Germany
| | - Elena Bechthold
- Chemical Biology of Ion Channels (Chembion), Westfälische Wilhelms-Universität Münster, GRK 2515 Munster, Germany
- Westfälische Wilhelms-Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstraße 48, D-48149 Münster, Germany
| | - Dirk Schepmann
- Westfälische Wilhelms-Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstraße 48, D-48149 Münster, Germany
| | - Julian A Schreiber
- Westfälische Wilhelms-Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstraße 48, D-48149 Münster, Germany
- Department of Cardiovascular Medicine, Westfälische Wilhelms-Universität Münster, Cellular Electrophysiology and Molecular Biology, Institute for Genetics of Heart Diseases (IfGH), University Hospital Münster, Robert-Koch-Str. 45, D-48149 Münster, Germany
| | - Sven G Meuth
- Chemical Biology of Ion Channels (Chembion), Westfälische Wilhelms-Universität Münster, GRK 2515 Munster, Germany
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Guiscard Seebohm
- Chemical Biology of Ion Channels (Chembion), Westfälische Wilhelms-Universität Münster, GRK 2515 Munster, Germany
- Department of Cardiovascular Medicine, Westfälische Wilhelms-Universität Münster, Cellular Electrophysiology and Molecular Biology, Institute for Genetics of Heart Diseases (IfGH), University Hospital Münster, Robert-Koch-Str. 45, D-48149 Münster, Germany
| | - Bernhard Wünsch
- Chemical Biology of Ion Channels (Chembion), Westfälische Wilhelms-Universität Münster, GRK 2515 Munster, Germany
- Westfälische Wilhelms-Universität Münster, Institut für Pharmazeutische und Medizinische Chemie, Corrensstraße 48, D-48149 Münster, Germany
| |
Collapse
|
91
|
Zhang J, Zhang M, Wang Q, Wen H, Liu Z, Wang F, Wang Y, Yao F, Song N, Kou Z, Li Y, Guo F, Zhu S. Distinct structure and gating mechanism in diverse NMDA receptors with GluN2C and GluN2D subunits. Nat Struct Mol Biol 2023; 30:629-639. [PMID: 36959261 DOI: 10.1038/s41594-023-00959-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 03/01/2023] [Indexed: 03/25/2023]
Abstract
N-methyl-D-aspartate (NMDA) receptors are heterotetramers comprising two GluN1 and two alternate GluN2 (N2A-N2D) subunits. Here we report full-length cryo-EM structures of the human N1-N2D di-heterotetramer (di-receptor), rat N1-N2C di-receptor and N1-N2A-N2C tri-heterotetramer (tri-receptor) at a best resolution of 3.0 Å. The bilobate N-terminal domain (NTD) in N2D intrinsically adopts a closed conformation, leading to a compact NTD tetramer in the N1-N2D receptor. Additionally, crosslinking the ligand-binding domain (LBD) of two N1 protomers significantly elevated the channel open probability (Po) in N1-N2D di-receptors. Surprisingly, the N1-N2C di-receptor adopted both symmetric (minor) and asymmetric (major) conformations, the latter further locked by an allosteric potentiator, PYD-106, binding to a pocket between the NTD and LBD in only one N2C protomer. Finally, the N2A and N2C subunits in the N1-N2A-N2C tri-receptor display a conformation close to one protomer in the N1-N2A and N1-N2C di-receptors, respectively. These findings provide a comprehensive structural understanding of diverse function in major NMDA receptor subtypes.
Collapse
Affiliation(s)
- Jilin Zhang
- 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
| | - Ming Zhang
- University of Chinese Academy of Sciences, Beijing, China
- Center for Neurological and Psychiatric Research and Drug Discovery, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | | | - Han Wen
- DP Technology, Beijing, China
| | - Zheyi Liu
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | - Fangjun Wang
- CAS Key Laboratory of Separation Sciences for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, China
| | | | - Fenyong Yao
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Nan Song
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Zengwei Kou
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Yang Li
- University of Chinese Academy of Sciences, Beijing, China
- Center for Neurological and Psychiatric Research and Drug Discovery, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Fei Guo
- University of Chinese Academy of Sciences, Beijing, China
- Center for Neurological and Psychiatric Research and Drug Discovery, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 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
|
92
|
Amin JB, He M, Prasad R, Leng X, Zhou HX, Wollmuth LP. Two gates mediate NMDA receptor activity and are under subunit-specific regulation. Nat Commun 2023; 14:1623. [PMID: 36959168 PMCID: PMC10036335 DOI: 10.1038/s41467-023-37260-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 03/09/2023] [Indexed: 03/25/2023] Open
Abstract
Kinetics of NMDA receptor (NMDAR) ion channel opening and closing contribute to their unique role in synaptic signaling. Agonist binding generates free energy to open a canonical gate at the M3 helix bundle crossing. Single channel activity is characterized by clusters, or periods of rapid opening and closing, that are separated by long silent periods. A conserved glycine in the outer most transmembrane helices, the M4 helices, regulates NMDAR function. Here we find that the GluN1 glycine mainly regulates single channel events within a cluster, whereas the GluN2 glycine mainly regulates entry and exit from clusters. Molecular dynamics simulations suggest that, whereas the GluN2 M4 (along with GluN2 pre-M1) regulates the gate at the M3 helix bundle crossing, the GluN1 glycine regulates a 'gate' at the M2 loop. Subsequent functional experiments support this interpretation. Thus, the distinct kinetics of NMDARs are mediated by two gates that are under subunit-specific regulation.
Collapse
Affiliation(s)
- Johansen B Amin
- Graduate Program in Cellular and Molecular Pharmacology, Stony Brook University, Stony Brook, NY, 11794-5230, USA
- Medical Scientist Training Program (MSTP), Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Miaomiao He
- Graduate Program in Biochemistry and Structural Biology, Stony Brook University, Stony Brook, NY, 11794-5230, USA
| | - Ramesh Prasad
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Xiaoling Leng
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Huan-Xiang Zhou
- Department of Chemistry, University of Illinois at Chicago, Chicago, IL, 60607, USA
- Department of Physics, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Lonnie P Wollmuth
- Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY, 11794-5230, USA.
- Department of Neurobiology & Behavior, Stony Brook University, Stony Brook, NY, 11794-5230, USA.
- Department of Biochemistry & Cell Biology, Stony Brook University, Stony Brook, NY, 11794-5230, USA.
| |
Collapse
|
93
|
Harris L, Regan MC, Myers SJ, Nocilla KA, Akins NS, Tahirovic YA, Wilson LJ, Dingledine R, Furukawa H, Traynelis SF, Liotta DC. Novel GluN2B-Selective NMDA Receptor Negative Allosteric Modulator Possesses Intrinsic Analgesic Properties and Enhances Analgesia of Morphine in a Rodent Tail Flick Pain Model. ACS Chem Neurosci 2023; 14:917-935. [PMID: 36779874 PMCID: PMC9983021 DOI: 10.1021/acschemneuro.2c00779] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 01/24/2023] [Indexed: 02/14/2023] Open
Abstract
Many cases of accidental death associated with drug overdose are due to chronic opioid use, tolerance, and addiction. Analgesic tolerance is characterized by a decreased response to the analgesic effects of opioids, requiring increasingly higher doses to maintain the desired level of pain relief. Overactivation of GluN2B-containing N-methyl-d-Aspartate receptors is thought to play a key role in mechanisms underlying cellular adaptation that takes place in the development of analgesic tolerance. Herein, we describe a novel GluN2B-selective negative allosteric modulator, EU93-108, that shows high potency and brain penetrance. We describe the structural basis for binding at atomic resolution. This compound possesses intrinsic analgesic properties in the rodent tail immersion test. EU93-108 has an acute and significant anodyne effect, whereby morphine when combined with EU93-108 produces a higher tail flick latency compared to that of morphine alone. These data suggest that engagement of GluN2B as a target has utility in the treatment of pain, and EU93-108 could serve as an appropriate tool compound to interrogate this hypothesis. Future structure-activity relationship work around this scaffold could give rise to compounds that can be co-administered with opioids to diminish the onset of tolerance due to chronic opioid use, thereby modifying their utility.
Collapse
Affiliation(s)
- Lynnea
D. Harris
- Department
of Chemistry, Emory University, Atlanta, Georgia30322, United States
| | - Michael C. Regan
- W.M.
Keck Structural Biology Laboratory, Cold
Spring Harbor Laboratory, New York, New York11724, United States
- RADD
Pharmaceuticals, Westport, Connecticut06880, United States
| | - Scott J. Myers
- Department
of Pharmacology and Chemical Biology, Emory
University, Atlanta, Georgia30322, United States
| | - Kelsey A. Nocilla
- Department
of Pharmacology and Chemical Biology, Emory
University, Atlanta, Georgia30322, United States
| | - Nicholas S. Akins
- Department
of Chemistry, Emory University, Atlanta, Georgia30322, United States
| | - Yesim A. Tahirovic
- Department
of Chemistry, Emory University, Atlanta, Georgia30322, United States
| | - Lawrence J. Wilson
- Department
of Chemistry, Emory University, Atlanta, Georgia30322, United States
| | - Ray Dingledine
- Department
of Pharmacology and Chemical Biology, Emory
University, Atlanta, Georgia30322, United States
| | - Hiro Furukawa
- W.M.
Keck Structural Biology Laboratory, Cold
Spring Harbor Laboratory, New York, New York11724, United States
| | - Stephen F. Traynelis
- Department
of Pharmacology and Chemical Biology, Emory
University, Atlanta, Georgia30322, United States
| | - Dennis C. Liotta
- Department
of Chemistry, Emory University, Atlanta, Georgia30322, United States
| |
Collapse
|
94
|
Tang W, Beckley JT, Zhang J, Song R, Xu Y, Kim S, Quirk MC, Robichaud AJ, Diaz ES, Myers SJ, Doherty JJ, Ackley MA, Traynelis SF, Yuan H. Novel neuroactive steroids as positive allosteric modulators of NMDA receptors: mechanism, site of action, and rescue pharmacology on GRIN variants associated with neurological conditions. Cell Mol Life Sci 2023; 80:42. [PMID: 36645496 PMCID: PMC10644378 DOI: 10.1007/s00018-022-04667-7] [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: 08/19/2022] [Revised: 11/18/2022] [Accepted: 12/11/2022] [Indexed: 01/17/2023]
Abstract
N-methyl-D-aspartate receptors (NMDARs) play vital roles in normal brain functions (i.e., learning, memory, and neuronal development) and various neuropathological conditions, such as epilepsy, autism, Parkinson's disease, Alzheimer's disease, and traumatic brain injury. Endogenous neuroactive steroids such as 24(S)-hydroxycholesterol (24(S)-HC) have been shown to influence NMDAR activity, and positive allosteric modulators (PAMs) derived from 24(S)-hydroxycholesterol scaffold can also enhance NMDAR function. This study describes the structural determinants and mechanism of action for 24(S)-hydroxycholesterol and two novel synthetic analogs (SGE-550 and SGE-301) on NMDAR function. We also show that these agents can mitigate the altered function caused by a set of loss-of-function missense variants in NMDAR GluN subunit-encoding GRIN genes associated with neurological and neuropsychiatric disorders. We anticipate that the evaluation of novel neuroactive steroid NMDAR PAMs may catalyze the development of new treatment strategies for GRIN-related neuropsychiatric conditions.
Collapse
Affiliation(s)
- Weiting Tang
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | | | - Jin Zhang
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Department of Neurology, The First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, 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, 250012, Shandong, China
| | - 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, 325000, Zhejiang, 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
| | | | | | - Eva Sarai Diaz
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - 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
| | | | | | - 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.
| | - 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.
| |
Collapse
|
95
|
Inoue R, Ni X, Mori H. Blockade of D-serine signaling and adult hippocampal neurogenesis attenuates remote contextual fear memory following multiple memory retrievals in male mice. Front Neurosci 2023; 16:1030702. [PMID: 36685240 PMCID: PMC9845639 DOI: 10.3389/fnins.2022.1030702] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 12/06/2022] [Indexed: 01/06/2023] Open
Abstract
The retrieval of fear memories induces two opposing processes, reconsolidation, and extinction. The memory reconsolidation is an active process that involves gene expression and updates an existing memory. It is hypothesized that blockade of reconsolidation by manipulating the neurobiological factors, which are mechanistically involved in the process, could weaken or disrupt the original fear memory. The N-methyl-D-aspartate (NMDA) receptor and hippocampal neurogenesis play crucial roles in hippocampus-dependent memory processes, including reconsolidation. Using contextual fear conditioning paradigm with multiple retrievals, we attempted to weaken the original contextual fear memory by repeatedly disrupting retrieval-induced reconsolidation via downregulation of NMDA receptor signaling and inhibition of neurogenesis. In the first experiment, prior to fear conditioning, NMDA receptor signaling was downregulated by the genetic reduction of its co-agonist, D-serine, and the neurogenesis was dampened by focal X-ray irradiation on the hippocampus. We found that simultaneous D-serine reduction and neurogenesis dampening resulted in a progressive decrease in freezing following each retrieval, leading to an attenuation of remote contextual fear memory on day 28. In the second experiment using the same behavioral protocols, after conditioning, pharmacological approaches were conducted to simultaneously block D-serine signaling and neurogenesis, resulting in a similar suppressive effect on the remote fear memory. The present findings provide insights for understanding the role of D-serine-mediated NMDA receptor signaling and neurogenesis in memory retrieval and the maintenance of remote fear memory, and improving the efficacy of exposure-based therapy for the treatment of post-traumatic stress disorder (PTSD).
Collapse
Affiliation(s)
- Ran Inoue
- Department of Molecular Neuroscience, Faculty of Medicine, University of Toyama, Toyama, Japan,Research Center for Idling Brain Science, University of Toyama, Toyama, Japan
| | - Xiance Ni
- Department of Molecular Neuroscience, Faculty of Medicine, University of Toyama, Toyama, Japan,Graduate School of Innovative Life Science, University of Toyama, Toyama, Japan
| | - Hisashi Mori
- Department of Molecular Neuroscience, Faculty of Medicine, University of Toyama, Toyama, Japan,Research Center for Idling Brain Science, University of Toyama, Toyama, Japan,*Correspondence: Hisashi Mori,
| |
Collapse
|
96
|
Chen H, Dong Y, Wu Y, Yi F. Targeting NMDA receptor signaling for therapeutic intervention in brain disorders. Rev Neurosci 2023:revneuro-2022-0096. [PMID: 36586105 DOI: 10.1515/revneuro-2022-0096] [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: 07/31/2022] [Accepted: 12/03/2022] [Indexed: 01/01/2023]
Abstract
N-Methyl-d-aspartate (NMDA) receptor hyperfunction plays a key role in the pathological processes of depression and neurodegenerative diseases, whereas NMDA receptor hypofunction is implicated in schizophrenia. Considerable efforts have been made to target NMDA receptor function for the therapeutic intervention in those brain disorders. In this mini-review, we first discuss ion flux-dependent NMDA receptor signaling and ion flux-independent NMDA receptor signaling that result from structural rearrangement upon binding of endogenous agonists. Then, we review current strategies for exploring druggable targets of the NMDA receptor signaling and promising future directions, which are poised to result in new therapeutic agents for several brain disorders.
Collapse
Affiliation(s)
- He Chen
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou 510515, P. R. China
| | - Yuanping Dong
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou 510515, P. R. China
| | - Yun Wu
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou 510515, P. R. China
| | - Feng Yi
- Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou 510515, P. R. China
| |
Collapse
|
97
|
Washburn HR, Chander P, Srikanth KD, Dalva MB. Transsynaptic Signaling of Ephs in Synaptic Development, Plasticity, and Disease. Neuroscience 2023; 508:137-152. [PMID: 36460219 DOI: 10.1016/j.neuroscience.2022.11.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 11/22/2022] [Accepted: 11/23/2022] [Indexed: 12/03/2022]
Abstract
Synapse formation between neurons is critical for proper circuit and brain function. Prior to activity-dependent refinement of connections between neurons, activity-independent cues regulate the contact and recognition of potential synaptic partners. Formation of a synapse results in molecular recognition events that initiate the process of synaptogenesis. Synaptogenesis requires contact between axon and dendrite, selection of correct and rejection of incorrect partners, and recruitment of appropriate pre- and postsynaptic proteins needed for the establishment of functional synaptic contact. Key regulators of these events are families of transsynaptic proteins, where one protein is found on the presynaptic neuron and the other is found on the postsynaptic neuron. Of these families, the EphBs and ephrin-Bs are required during each phase of synaptic development from target selection, recruitment of synaptic proteins, and formation of spines to regulation of synaptic plasticity at glutamatergic spine synapses in the mature brain. These roles also place EphBs and ephrin-Bs as important regulators of human neurological diseases. This review will focus on the role of EphBs and ephrin-Bs at synapses.
Collapse
Affiliation(s)
- Halley R Washburn
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA; Department of Neuroscience, Jefferson Synaptic Biology Center, Sidney Kimmel Medical College at Thomas Jefferson University, 233 South 10th Street, Bluemle Life Sciences Building, Room 324, Philadelphia, PA 19107, USA
| | - Praveen Chander
- Department of Neuroscience, Jefferson Synaptic Biology Center, Sidney Kimmel Medical College at Thomas Jefferson University, 233 South 10th Street, Bluemle Life Sciences Building, Room 324, Philadelphia, PA 19107, USA
| | - Kolluru D Srikanth
- Department of Neuroscience, Jefferson Synaptic Biology Center, Sidney Kimmel Medical College at Thomas Jefferson University, 233 South 10th Street, Bluemle Life Sciences Building, Room 324, Philadelphia, PA 19107, USA
| | - Matthew B Dalva
- Department of Neuroscience, Jefferson Synaptic Biology Center, Sidney Kimmel Medical College at Thomas Jefferson University, 233 South 10th Street, Bluemle Life Sciences Building, Room 324, Philadelphia, PA 19107, USA.
| |
Collapse
|
98
|
Kolcheva M, Ladislav M, Netolicky J, Kortus S, Rehakova K, Krausova BH, Hemelikova K, Misiachna A, Kadkova A, Klima M, Chalupska D, Horak M. The pathogenic N650K variant in the GluN1 subunit regulates the trafficking, conductance, and pharmacological properties of NMDA receptors. Neuropharmacology 2023; 222:109297. [PMID: 36341805 DOI: 10.1016/j.neuropharm.2022.109297] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 10/07/2022] [Accepted: 10/18/2022] [Indexed: 11/05/2022]
Abstract
N-methyl-D-aspartate receptors (NMDARs) play an essential role in excitatory neurotransmission in the mammalian brain, and their physiological importance is underscored by the large number of pathogenic mutations that have been identified in the receptor's GluN subunits and associated with a wide range of diseases and disorders. Here, we characterized the functional and pharmacological effects of the pathogenic N650K variant in the GluN1 subunit, which is associated with developmental delay and seizures. Our microscopy experiments showed that when expressed in HEK293 cells (from ATCC®), the GluN1-N650K subunit increases the surface expression of both GluN1/GluN2A and GluN1/GluN2B receptors, but not GluN1/GluN3A receptors, consistent with increased surface expression of the GluN1-N650K subunit expressed in hippocampal neurons (from embryonic day 18 of Wistar rats of both sexes). Using electrophysiology, we found that the GluN1-N650K variant increases the potency of GluN1/GluN2A receptors to both glutamate and glycine but decreases the receptor's conductance and open probability. In addition, the GluN1-N650K subunit does not form functional GluN1/GluN2B receptors but does form fully functional GluN1/GluN3A receptors. Moreover, in the presence of extracellular Mg2+, GluN1-N650K/GluN2A receptors have a similar and increased response to ketamine and memantine, respectively, while the effect of both drugs had markedly slower onset and offset compared to wild-type GluN1/GluN2A receptors. Finally, we found that expressing the GluN1-N650K subunit in hippocampal neurons reduces excitotoxicity, and memantine shows promising neuroprotective effects in neurons expressing either wild-type GluN1 or the GluN1-N650K subunit. This study provides the functional and pharmacological characterization of NMDARs containing the GluN1-N650K variant.
Collapse
Affiliation(s)
- Marharyta Kolcheva
- Department of Neurochemistry, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 14220, Prague 4, Czech Republic; Department of Physiology, Faculty of Science, Charles University in Prague, Albertov 6, 12843, Prague 2, Czech Republic; Laboratory of Cellular Neurophysiology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 14220, Prague 4, Czech Republic
| | - Marek Ladislav
- Department of Neurochemistry, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 14220, Prague 4, Czech Republic
| | - Jakub Netolicky
- Department of Neurochemistry, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 14220, Prague 4, Czech Republic; Department of Physiology, Faculty of Science, Charles University in Prague, Albertov 6, 12843, Prague 2, Czech Republic
| | - Stepan Kortus
- Department of Neurochemistry, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 14220, Prague 4, Czech Republic
| | - Kristyna Rehakova
- Department of Neurochemistry, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 14220, Prague 4, Czech Republic
| | - Barbora Hrcka Krausova
- Department of Neurochemistry, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 14220, Prague 4, Czech Republic
| | - Katarina Hemelikova
- Department of Neurochemistry, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 14220, Prague 4, Czech Republic
| | - Anna Misiachna
- Department of Neurochemistry, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 14220, Prague 4, Czech Republic
| | - Anna Kadkova
- Department of Neurochemistry, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 14220, Prague 4, Czech Republic
| | - Martin Klima
- Department of Molecular Biology and Biochemistry, Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo namesti 542/2, P.O. Box:16000, Prague 6, Czech Republic
| | - Dominika Chalupska
- Department of Molecular Biology and Biochemistry, Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo namesti 542/2, P.O. Box:16000, Prague 6, Czech Republic
| | - Martin Horak
- Department of Neurochemistry, Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, 14220, Prague 4, Czech Republic.
| |
Collapse
|
99
|
Zhao T, Zhong R, Zhang X, Li G, Zhou C, Fang S, Ding Y, Lin W. Efavirenz restored NMDA receptor dysfunction and inhibited epileptic seizures in GluN2A/Grin2a mutant mice. Front Neurosci 2023; 17:1086462. [PMID: 36937661 PMCID: PMC10017539 DOI: 10.3389/fnins.2023.1086462] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 02/02/2023] [Indexed: 03/06/2023] Open
Abstract
Introduction N-methyl-D-aspartate receptor (NMDAR) is one of the main receptor of the excitatory neurotransmitter glutamate in the brain, which is the key determinant of the excitatory/inhibitory balance of neural network. GluN2A/GRIN2A is one of the subunits of NMDAR and plays an important role in epilepsy. Approximately 78% of patients with GluN2A/Grin2a mutations have epilepsy, and the underlying mechanism of this association is not well characterized. Methods We constructed a mouse model of hyperthermic seizure, and conducted in vitro and in vivo electrophysiological and behavioral studies to clarify the pathogenic characteristics and mechanism of GluN2A/GRIN2A-V685G mutation. In addition, the drug efavirenz (EFV), which is used to treat HIV infection, was administrated to mutant animals to assess whether it can restore the loss of function. Results Mutant mice showed no significant change in the mRNA or protein expressions of NMDAR compared with wild type (WT) mice. Mice with GluN2A/GRIN2A-V685G mutation exhibited shorter latency to seizure, increased frequency of seizure-like events, decreased peak current and current area of NMDAR excitatory postsynaptic current, and decreased event frequency of micro-inhibitory postsynaptic current, compared to WT mice. They also exhibited decreased threshold, increased amplitude, increased input resistance, and increased root number of action potential. EFV administration reversed these changes. The loss-of-function (LoF) mutation of NMDAR changed the excitatory/inhibitory balance of neural network, rendering animal more prone to seizures. Discussion EFV was indicated to hold its potential in the treatment of inherited epilepsy.
Collapse
Affiliation(s)
- Teng Zhao
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Rui Zhong
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Xinyue Zhang
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Guangjian Li
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Chunkui Zhou
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Shaokuan Fang
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
- Shaokuan Fang,
| | - Ying Ding
- Department of Radiology, The First Hospital of Jilin University, Changchun, China
- Ying Ding,
| | - Weihong Lin
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
- *Correspondence: Weihong Lin,
| |
Collapse
|
100
|
Basak S, Saikia N, Kwun D, Choi UB, Ding F, Bowen ME. Different Forms of Disorder in NMDA-Sensitive Glutamate Receptor Cytoplasmic Domains Are Associated with Differences in Condensate Formation. Biomolecules 2022; 13:4. [PMID: 36671389 PMCID: PMC9855357 DOI: 10.3390/biom13010004] [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: 10/21/2022] [Revised: 12/05/2022] [Accepted: 12/10/2022] [Indexed: 12/24/2022] Open
Abstract
The N-methyl-D-aspartate (NMDA)-sensitive glutamate receptor (NMDAR) helps assemble downstream signaling pathways through protein interactions within the postsynaptic density (PSD), which are mediated by its intracellular C-terminal domain (CTD). The most abundant NMDAR subunits in the brain are GluN2A and GluN2B, which are associated with a developmental switch in NMDAR composition. Previously, we used single molecule fluorescence resonance energy transfer (smFRET) to show that the GluN2B CTD contained an intrinsically disordered region with slow, hop-like conformational dynamics. The CTD from GluN2B also undergoes liquid-liquid phase separation (LLPS) with synaptic proteins. Here, we extend these observations to the GluN2A CTD. Sequence analysis showed that both subunits contain a form of intrinsic disorder classified as weak polyampholytes. However, only GluN2B contained matched patterning of arginine and aromatic residues, which are linked to LLPS. To examine the conformational distribution, we used discrete molecular dynamics (DMD), which revealed that GluN2A favors extended disordered states containing secondary structures while GluN2B favors disordered globular states. In contrast to GluN2B, smFRET measurements found that GluN2A lacked slow conformational dynamics. Thus, simulation and experiments found differences in the form of disorder. To understand how this affects protein interactions, we compared the ability of these two NMDAR isoforms to undergo LLPS. We found that GluN2B readily formed condensates with PSD-95 and SynGAP, while GluN2A failed to support LLPS and instead showed a propensity for colloidal aggregation. That GluN2A fails to support this same condensate formation suggests a developmental switch in LLPS propensity.
Collapse
Affiliation(s)
- Sujit Basak
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY 11794, USA
| | - Nabanita Saikia
- Department of Chemistry, Navajo Technical University, Crownpoint, NM 87313, USA
| | - David Kwun
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY 11794, USA
| | | | - Feng Ding
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634-0978, USA
| | - Mark E. Bowen
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY 11794, USA
| |
Collapse
|