151
|
Essiz S, Gencel M, Aktolun M, Demir A, Carpenter TS, Servili B. Correlated conformational dynamics of the human GluN1-GluN2A type N-methyl-D-aspartate (NMDA) receptor. J Mol Model 2021; 27:162. [PMID: 33969428 DOI: 10.1007/s00894-021-04755-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 04/15/2021] [Indexed: 11/30/2022]
Abstract
N-Methyl-D-aspartate receptors (NMDARs) are glutamate-gated ion channels found in the nerve cell membranes. As a result of overexcitation of NMDARs, neuronal death occurs and may lead to diseases such as epilepsy, stroke, Alzheimer's disease, and Parkinson's disease. In this study, human GluN1- GluN2A type NMDAR structure is modeled based on the X-ray structure of the Xenopus laevis template and missing loops are added by ab-initio loop modeling. The final structure is chosen according to two different model assessment scores. To be able to observe the structural changes upon ligand binding, glycine and glutamate molecules are docked into the corresponding binding sites of the receptor. Subsequently, molecular dynamics simulations of 1.3 μs are performed for both apo and ligand-bound structures. Structural parameters, which have been considered to show functionally important changes in previous NMDAR studies, are monitored as conformational rulers to understand the dynamics of the conformational changes. Moreover, principal component analysis (PCA) is performed for the equilibrated part of the simulations. From these analyses, the differences in between apo and ligand-bound simulations can be summarized as the following: The girdle right at the beginning of the pore loop, which connects M2 and M3 helices of the ion channel, partially opens. Ligands act like an adhesive for the ligand-binding domain (LBD) by keeping the bi-lobed structure together and consequently this is reflected to the overall dynamics of the protein as an increased correlation of the LBD with especially the amino-terminal domain (ATD) of the protein.
Collapse
Affiliation(s)
- Sebnem Essiz
- Bioinformatics and Genetics Department, Faculty of Engineering and Natural Sciences, Kadir Has University, 34083 Fatih, Istanbul, Turkey.
| | - Melis Gencel
- Bioinformatics and Genetics Department, Faculty of Engineering and Natural Sciences, Kadir Has University, 34083 Fatih, Istanbul, Turkey
| | - Muhammed Aktolun
- Bioinformatics and Genetics Department, Faculty of Engineering and Natural Sciences, Kadir Has University, 34083 Fatih, Istanbul, Turkey
| | - Ayhan Demir
- Bioinformatics and Genetics Department, Faculty of Engineering and Natural Sciences, Kadir Has University, 34083 Fatih, Istanbul, Turkey
| | - Timothy S Carpenter
- Biosciences and Biotechnology Division, Physical and Life Sciences Directorate, Lawrence Livermore National Laboratory, Livermore, CA, USA
| | - Burak Servili
- Bioinformatics and Genetics Department, Faculty of Engineering and Natural Sciences, Kadir Has University, 34083 Fatih, Istanbul, Turkey
| |
Collapse
|
152
|
Li Z, Cai G, Fang F, Li W, Fan M, Lian J, Qiu Y, Xu X, Lv X, Li Y, Zheng R, Wang Y, Li Z, Zhang G, Liu Z, Huang Z, Zhang L. Discovery of Novel and Potent N-Methyl-d-aspartate Receptor Positive Allosteric Modulators with Antidepressant-like Activity in Rodent Models. J Med Chem 2021; 64:5551-5576. [PMID: 33934604 DOI: 10.1021/acs.jmedchem.0c02018] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
N-Methyl-d-aspartate receptors (NMDARs) are glutamate-gated Na+ and Ca2+-permeable ion channels involved in excitatory synaptic transmission and synaptic plasticity. NMDAR hypofunction has long been implicated in the pathophysiology including major depressive disorders (MDDs). Herein, we report a series of furan-2-carboxamide analogues as novel NMDAR-positive allosteric modulators (PAMs). Through structure-based virtual screen and electrophysiological tests, FS2921 was identified as a novel NMDAR PAM with potential antidepressant effects. Further structure-activity relationship studies led to the discovery of novel analogues with increased potentiation. Compound 32h caused a significant increase in NMDAR excitability in vitro and impressive activity in the forced swimming test. Moreover, compound 32h showed no significant inhibition of hERG or cell viability and possessed a favorable PK/PD profile. Our study presented a series of novel NMDAR PAMs and provided potential opportunities for discovering of new antidepressants.
Collapse
Affiliation(s)
- Zhongtang Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Guanxing Cai
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Fan Fang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Wenchao Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Minghua Fan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Jingjing Lian
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yinli Qiu
- Jiangsu Nhwa Pharmaceutical Co., Ltd. 69 Democratic South Road, Xuzhou, Jiangsu 221116, China
| | - Xiangqing Xu
- Jiangsu Nhwa Pharmaceutical Co., Ltd. 69 Democratic South Road, Xuzhou, Jiangsu 221116, China
| | - Xuehui Lv
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yiyan Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Ruqiu Zheng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yuxi Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Zhongjun Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Guisen Zhang
- Jiangsu Key Laboratory of Marine Biological Resources and Environment, Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, School of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, China
| | - Zhenming Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Zhuo Huang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Liangren Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
153
|
Thapaliya ER, Mony L, Sanchez R, Serraz B, Paoletti P, Ellis-Davies GCR. Photochemical control of drug efficacy - a comparison of uncaging and photoswitching ifenprodil on NMDA receptors. CHEMPHOTOCHEM 2021; 5:445-454. [PMID: 36540756 PMCID: PMC9762817 DOI: 10.1002/cptc.202000240] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Indexed: 09/29/2023]
Abstract
Ifenprodil is an important negative allosteric modulator of the N-methyl-D-aspartate (NMDA) receptors. We have synthesized caged and photoswitchable derivatives of this small molecule drug. Caged ifenprodil was biologically inert before photolysis, UV irradiation efficiently released the drug allowing selective inhibition of GluN2B-containing NMDA receptors. Azobenzene-modified ifenprodil, on the other hand, is inert in both its trans and cis configurations, although in silico modeling predicted the trans form to be able to bind to the receptor. The disparity in effectiveness between the two compounds reflects, in part, the inherent ability of each method in manipulating the binding properties of drugs. With appropriate structure-activity relationship uncaging enables binary control of effector binding, whereas photoswitching using feely diffusable chromophores shifts the dose-response curve of drug-receptor interaction. Our data suggest that the efficacy of pharmacophores having a confined binding site such as ifenprodil can be controlled more easily by uncaging in comparison to photoswitching.
Collapse
Affiliation(s)
- Ek Raj Thapaliya
- Department of Neuroscience, Mount Sinai School of Medicine, New York, NY 10029, USA
- Equal contribution
| | - Laetitia Mony
- Institut de Biologie de l’Ecole Normale Supérieure (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, Paris 75005, France
- Equal contribution
| | - Roberto Sanchez
- Department of Pharmacological Sciences and Drug Discovery Institute, Mount Sinai School of Medicine, New York, NY 10029, USA
| | - Benjamin Serraz
- Institut de Biologie de l’Ecole Normale Supérieure (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, Paris 75005, France
| | - Pierre Paoletti
- Institut de Biologie de l’Ecole Normale Supérieure (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, Paris 75005, France
| | | |
Collapse
|
154
|
Tseng YS, Liao CH, Wu WB, Ma MC. N-methyl-d-aspartate receptor hyperfunction contributes to d-serine-mediated renal insufficiency. Am J Physiol Renal Physiol 2021; 320:F799-F813. [PMID: 33749324 DOI: 10.1152/ajprenal.00461.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 03/18/2021] [Indexed: 01/07/2023] Open
Abstract
Glutamate N-methyl-d-aspartate receptor (NMDAR) hyperfunction is known to contribute to acute renal failure due to ischemia-reperfusion and endotoxemia. d-Serine is a coagonist for NMDAR activation, but whether NMDARs play a role in d-serine-mediated nephrotoxicity remains unclear. Here, we demonstrate that NMDAR blockade ameliorated d-serine-induced renal injury. In NMDAR-expressing LLC-PK1 cells, which were used as a proximal tubule model, d-serine but not l-serine induced cytotoxicity in a dose-dependent manner, which was abrogated by the selective NMDAR blockers MK-801 and AP-5. Time-dependent oxidative stress, evidenced by gradually increased superoxide and H2O2 production, was associated with d-serine-mediated cytotoxicity; these reactive oxygen species could be alleviated not only after NMDAR inhibition but also by NADPH oxidase (NOX) inhibition. Activation of protein kinase C (PKC)-δ and PKC-ζ is a downstream signal for NMDAR-mediated NOX activation because PKC inhibition diminishes the NOX activity that is induced by d-serine. Renal injury was further confirmed in male Wistar rats that intraperitoneally received d-serine but not l-serine. Peak changes in glucosuria, proteinuria, and urinary excretion of lactate dehydrogenase and malondialdehyde were found after 24 h of treatment. Persistent tubular damage was observed after 7 days of treatment. Cotreatment with the NMDAR blocker MK-801 for 24 h abolished d-serine-induced functional insufficiency and tubular damage. MK-801 attenuated renal superoxide formation by lowering NOX activity and protein upregulation of NOX4 but not NOX2. These results reveal that NMDAR hyperfunction underlies d-serine-induced renal injury via the effects of NOX4 on triggering oxidative stress.NEW & NOTEWORTHY Ionotropic N-methyl-d-aspartate receptors (NMDARs) are not only present in the nervous system but also expressed in the kidney. Overstimulation of renal NMDARs leads to oxidative stress via the signal pathway of calcium/protein kinase C/NADPH oxidase in d-serine-mediated tubular cell damage. Intervention of NMDAR blockade may prevent acute renal injury caused by d-serine.
Collapse
Affiliation(s)
- Yi-Shiou Tseng
- Division of Urology, Department of Surgery, Far Eastern Memorial Hospital, New Taipei, Taiwan
| | - Chun-Hou Liao
- Divisions of Urology, Department of Surgery, Cardinal Tien Hospital, New Taipei, Taiwan
- School of Medicine, Fu Jen Catholic University, New Taipei, Taiwan
| | - Wen-Bin Wu
- School of Medicine, Fu Jen Catholic University, New Taipei, Taiwan
| | - Ming-Chieh Ma
- School of Medicine, Fu Jen Catholic University, New Taipei, Taiwan
| |
Collapse
|
155
|
Dixit S, Thakur N, Shukla A, Upadhyay SK, C Verma P. Molecular characterization of N-methyl-d-aspartate receptor from Bemisia tabaci. INSECT MOLECULAR BIOLOGY 2021; 30:231-240. [PMID: 33368750 DOI: 10.1111/imb.12690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/13/2020] [Accepted: 12/23/2020] [Indexed: 06/12/2023]
Abstract
The N-methyl-d-aspartate receptors (NMDARs) are ionotropic ligand gated channels that are highly permeable to calcium ions. In insects, NMDARs are associated with glutamatergic neurotransmission governing diverse physiological and biological processes like vitellogenesis and ovarian development. Therefore, NMDAR may act as attractive target for insect pest control. In present study, we performed structural and functional characterization of NMDARs in Bemisia tabaci, a highly invasive crop pest and potent virus vector. We identified that NMDAR consists of three subunits each encoded by single gene in whiteflies which are highly conserved among different insect orders. Expression analysis suggests that subunit 1 (BtNR1) and subunit 2 (BtNR2) are the main functional units. External supplementation of NMDAR ligand or BtNRs silencing was lethal to insects, which suggested that NMDAR function is highly balanced in whiteflies.
Collapse
Affiliation(s)
- S Dixit
- Molecular Biology and Biotechnology, CSIR-National Botanical Research Institute, (Council of Scientific and Industrial Research), Rana Pratap Marg, Lucknow, India
- Department of Biology, University of Western Ontario, London, Ontario, Canada
| | - N Thakur
- Molecular Biology and Biotechnology, CSIR-National Botanical Research Institute, (Council of Scientific and Industrial Research), Rana Pratap Marg, Lucknow, India
- DST-Centre for Policy Research, IIT-Delhi, New Delhi, India
| | - A Shukla
- Department of Biology, University of Western Ontario, London, Ontario, Canada
| | - S K Upadhyay
- Department of Botany, Panjab University, Chandigarh, India
| | - P C Verma
- Molecular Biology and Biotechnology, CSIR-National Botanical Research Institute, (Council of Scientific and Industrial Research), Rana Pratap Marg, Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| |
Collapse
|
156
|
Khanra N, Brown PMGE, Perozzo AM, Bowie D, Meyerson JR. Architecture and structural dynamics of the heteromeric GluK2/K5 kainate receptor. eLife 2021; 10:e66097. [PMID: 33724189 PMCID: PMC7997659 DOI: 10.7554/elife.66097] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 03/05/2021] [Indexed: 12/15/2022] Open
Abstract
Kainate receptors (KARs) are L-glutamate-gated ion channels that regulate synaptic transmission and modulate neuronal circuits. KARs have strict assembly rules and primarily function as heteromeric receptors in the brain. A longstanding question is how KAR heteromer subunits organize and coordinate together to fulfill their signature physiological roles. Here we report structures of the GluK2/GluK5 heteromer in apo, antagonist-bound, and desensitized states. The receptor assembles with two copies of each subunit, ligand binding domains arranged as two heterodimers and GluK5 subunits proximal to the channel. Strikingly, during desensitization, GluK2, but not GluK5, subunits undergo major structural rearrangements to facilitate channel closure. We show how the large conformational differences between antagonist-bound and desensitized states are mediated by the linkers connecting the pore helices to the ligand binding domains. This work presents the first KAR heteromer structure, reveals how its subunits are organized, and resolves how the heteromer can accommodate functionally distinct closed channel structures.
Collapse
Affiliation(s)
- Nandish Khanra
- Department of Physiology and Biophysics, Weill Cornell Medical CollegeNew YorkUnited States
| | - Patricia MGE Brown
- Department of Pharmacology and Therapeutics, McGill UniversityMontréalCanada
| | - Amanda M Perozzo
- Department of Pharmacology and Therapeutics, McGill UniversityMontréalCanada
| | - Derek Bowie
- Department of Pharmacology and Therapeutics, McGill UniversityMontréalCanada
| | - Joel R Meyerson
- Department of Physiology and Biophysics, Weill Cornell Medical CollegeNew YorkUnited States
| |
Collapse
|
157
|
Horak M, Barackova P, Langore E, Netolicky J, Rivas-Ramirez P, Rehakova K. The Extracellular Domains of GluN Subunits Play an Essential Role in Processing NMDA Receptors in the ER. Front Neurosci 2021; 15:603715. [PMID: 33796003 PMCID: PMC8007919 DOI: 10.3389/fnins.2021.603715] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 02/19/2021] [Indexed: 12/31/2022] Open
Abstract
N-methyl-D-aspartate receptors (NMDARs) belong to a family of ionotropic glutamate receptors that play essential roles in excitatory neurotransmission and synaptic plasticity in the mammalian central nervous system (CNS). Functional NMDARs consist of heterotetramers comprised of GluN1, GluN2A-D, and/or GluN3A-B subunits, each of which contains four membrane domains (M1 through M4), an intracellular C-terminal domain, a large extracellular N-terminal domain composed of the amino-terminal domain and the S1 segment of the ligand-binding domain (LBD), and an extracellular loop between M3 and M4, which contains the S2 segment of the LBD. Both the number and type of NMDARs expressed at the cell surface are regulated at several levels, including their translation and posttranslational maturation in the endoplasmic reticulum (ER), intracellular trafficking via the Golgi apparatus, lateral diffusion in the plasma membrane, and internalization and degradation. This review focuses on the roles played by the extracellular regions of GluN subunits in ER processing. Specifically, we discuss the presence of ER retention signals, the integrity of the LBD, and critical N-glycosylated sites and disulfide bridges within the NMDAR subunits, each of these steps must pass quality control in the ER in order to ensure that only correctly assembled NMDARs are released from the ER for subsequent processing and trafficking to the surface. Finally, we discuss the effect of pathogenic missense mutations within the extracellular domains of GluN subunits with respect to ER processing of NMDARs.
Collapse
Affiliation(s)
- Martin Horak
- Department of Neurochemistry, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| | - Petra Barackova
- Department of Neurochemistry, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| | - Emily Langore
- Department of Neurochemistry, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| | - Jakub Netolicky
- Department of Neurochemistry, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| | - Paula Rivas-Ramirez
- Department of Neurochemistry, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| | - Kristyna Rehakova
- Department of Neurochemistry, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
158
|
Tikhonov DB. Channel Blockers of Ionotropic Glutamate
Receptors. J EVOL BIOCHEM PHYS+ 2021. [DOI: 10.1134/s0022093021020149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
159
|
Palmitoylation Controls NMDA Receptor Function and Steroid Sensitivity. J Neurosci 2021; 41:2119-2134. [PMID: 33526476 DOI: 10.1523/jneurosci.2654-20.2021] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/08/2020] [Accepted: 01/06/2021] [Indexed: 11/21/2022] Open
Abstract
NMDARs are ligand-gated ion channels that cause an influx of Na+ and Ca2+ into postsynaptic neurons. The resulting intracellular Ca2+ transient triggers synaptic plasticity. When prolonged, it may induce excitotoxicity, but it may also activate negative feedback to control the activity of NMDARs. Here, we report that a transient rise in intracellular Ca2+ (Ca2+ challenge) increases the sensitivity of NMDARs but not AMPARs/kainate receptors to the endogenous inhibitory neurosteroid 20-oxo-5β-pregnan-3α-yl 3-sulfate and to its synthetic analogs, such as 20-oxo-5β-pregnan-3α-yl 3-hemipimelate (PAhPim). In cultured hippocampal neurons, 30 μm PAhPim had virtually no effect on NMDAR responses; however, following the Ca2+ challenge, it inhibited the responses by 62%; similarly, the Ca2+ challenge induced a 3.7-fold decrease in the steroid IC50 on recombinant GluN1/GluN2B receptors. The increase in the NMDAR sensitivity to PAhPim was dependent on three cysteines (C849, C854, and C871) located in the carboxy-terminal domain of the GluN2B subunit, previously identified to be palmitoylated (Hayashi et al., 2009). Our experiments suggested that the Ca2+ challenge induced receptor depalmitoylation, and single-channel analysis revealed that this was accompanied by a 55% reduction in the probability of channel opening. Results of in silico modeling indicate that receptor palmitoylation promotes anchoring of the GluN2B subunit carboxy-terminal domain to the plasma membrane and facilitates channel opening. Depalmitoylation-induced changes in the NMDAR pharmacology explain the neuroprotective effect of PAhPim on NMDA-induced excitotoxicity. We propose that palmitoylation-dependent changes in the NMDAR sensitivity to steroids serve as an acute endogenous mechanism that controls NMDAR activity.SIGNIFICANCE STATEMENT There is considerable interest in negative allosteric modulators of NMDARs that could compensate for receptor overactivation by glutamate or de novo gain-of-function mutations in neurodevelopmental disorders. By a combination of electrophysiological, pharmacological, and computational techniques we describe a novel feedback mechanism regulating NMDAR activity. We find that a transient rise in intracellular Ca2+ increases NMDAR sensitivity to inhibitory neurosteroids in a process dependent on GluN2B subunit depalmitoylation. These results improve our understanding of the molecular mechanisms of steroid action at the NMDAR and indeed of the basic properties of this important glutamate-gated ion channel and may aid in the development of therapeutics for treating neurologic and psychiatric diseases related to overactivation of NMDARs without affecting normal physiological functions.
Collapse
|
160
|
Furukawa H, Simorowski N, Michalski K. Effective production of oligomeric membrane proteins by EarlyBac-insect cell system. Methods Enzymol 2021; 653:3-19. [PMID: 34099177 DOI: 10.1016/bs.mie.2020.12.019] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Despite major advances in methodologies for membrane protein production over the last two decades, there remain challenging protein complexes that are technically difficult to yield by conventional recombinant expression methods. A large number of these proteins are multimeric membrane proteins from eukaryotic species, which are required to pass through stringent quality control mechanisms of host cells for proper folding and complex assembly. Here, we describe the development procedure to improve the production efficiency of multi-oligomeric membrane protein complexes in insect cells and recombinant baculovirus, which involves screening of promoters, enhancers, and untranslated regions for expression levels, using calcium homeostasis modulator (CALHM) and N-methyl-d-aspartate receptor (NMDAR) proteins as examples. We demonstrate that our insect cell expression strategy is effective in expression of both multi-homomeric CALHM proteins and multi-heteromeric NMDARs.
Collapse
Affiliation(s)
- Hiro Furukawa
- WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, United States.
| | - Noriko Simorowski
- WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, United States
| | - Kevin Michalski
- WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, United States
| |
Collapse
|
161
|
Iacobucci GJ, Wen H, Helou M, Liu B, Zheng W, Popescu GK. Cross-subunit interactions that stabilize open states mediate gating in NMDA receptors. Proc Natl Acad Sci U S A 2021; 118:e2007511118. [PMID: 33384330 PMCID: PMC7812756 DOI: 10.1073/pnas.2007511118] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
NMDA receptors are excitatory channels with critical functions in the physiology of central synapses. Their activation reaction proceeds as a series of kinetically distinguishable, reversible steps, whose structural bases are currently under investigation. Very likely, the earliest steps include glutamate binding to glycine-bound receptors and subsequent constriction of the ligand-binding domain. Later, three short linkers transduce this movement to open the gate by mechanical pulling on transmembrane helices. Here, we used molecular and kinetic simulations and double-mutant cycle analyses to show that a direct chemical interaction between GluN1-I642 (on M3 helix) and GluN2A-L550 (on L1-M1 linker) stabilizes receptors after they have opened and thus represents one of the structural changes that occur late in the activation reaction. This native interaction extends the current decay, and its absence causes deficits in charge transfer by GluN1-I642L, a pathogenic human variant.
Collapse
Affiliation(s)
- Gary J Iacobucci
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, SUNY, Buffalo, NY 14203
| | - Han Wen
- Department of Physics, College of Arts and Sciences, University at Buffalo, SUNY, Buffalo, NY 14260
| | - Matthew Helou
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, SUNY, Buffalo, NY 14203
| | - Beiying Liu
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, SUNY, Buffalo, NY 14203
| | - Wenjun Zheng
- Department of Physics, College of Arts and Sciences, University at Buffalo, SUNY, Buffalo, NY 14260
| | - Gabriela K Popescu
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, SUNY, Buffalo, NY 14203;
| |
Collapse
|
162
|
Bechthold E, Schreiber JA, Lehmkuhl K, Frehland B, Schepmann D, Bernal FA, Daniliuc C, Álvarez I, Garcia CV, Schmidt TJ, Seebohm G, Wünsch B. Ifenprodil Stereoisomers: Synthesis, Absolute Configuration, and Correlation with Biological Activity. J Med Chem 2021; 64:1170-1179. [PMID: 33426889 DOI: 10.1021/acs.jmedchem.0c01912] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Ifenprodil (1) is a potent GluN2B-selective N-methyl-d-aspartate (NMDA) receptor antagonist that is used as a cerebral vasodilator and has been examined in clinical trials for the treatment of drug addiction, idiopathic pulmonary fibrosis, and COVID-19. To correlate biological data with configuration, all four ifenprodil stereoisomers were prepared by diastereoselective reduction and subsequent separation of enantiomers by chiral HPLC. The absolute configuration of ifenprodil stereoisomers was determined by X-ray crystal structure analysis of (1R,2S)-1a and (1S,2S)-1d. GluN2B affinity, ion channel inhibitory activity, and selectivity over α, σ, and 5-HT receptors were evaluated. (1R,2R)-Ifenprodil ((1R,2R)-1c) showed the highest affinity toward GluN2B-NMDA receptors (Ki = 5.8 nM) and high inhibition of ion flux in two-electrode voltage clamp experiments (IC50 = 223 nM). Whereas the configuration did not influence considerably the GluN2B-NMDA receptor binding, (1R)-configuration is crucial for elevated inhibitory activity. (1R,2R)-Configured ifenprodil (1R,2R)-1c exhibited high selectivity for GluN2B-NMDA receptors over adrenergic, serotonergic, and σ1 receptors.
Collapse
Affiliation(s)
- Elena Bechthold
- GRK 2515, Chemical Biology of Ion Channels (Chembion), Westfälische Wilhelms-Universität Münster, D-48149 Münster, Germany.,Institut für Pharmazeutische und Medizinische Chemie, Westfälische Wilhelms-Universität Münster, Corrensstraße 48, D-48149 Münster, Germany
| | - Julian A Schreiber
- Institut für Pharmazeutische und Medizinische Chemie, Westfälische Wilhelms-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-Strasse 45, D-48149 Münster, Germany
| | - Kirstin Lehmkuhl
- Institut für Pharmazeutische und Medizinische Chemie, Westfälische Wilhelms-Universität Münster, Corrensstraße 48, D-48149 Münster, Germany
| | - Bastian Frehland
- Institut für Pharmazeutische und Medizinische Chemie, Westfälische Wilhelms-Universität Münster, Corrensstraße 48, D-48149 Münster, Germany
| | - Dirk Schepmann
- Institut für Pharmazeutische und Medizinische Chemie, Westfälische Wilhelms-Universität Münster, Corrensstraße 48, D-48149 Münster, Germany
| | - Freddy A Bernal
- Institut für Pharmazeutische Biologie und Phytochemie, Westfälische Wilhelms-Universität Münster, Corrensstraße 48, D-48149 Münster, Germany
| | - Constantin Daniliuc
- Organisch-chemisches Institut, Westfälische Wilhelms-Universität Münster, Corrensstraße 40, D-48149 Münster, Germany
| | - Inés Álvarez
- In Vitro Pharmacology, WeLab, Parc Cientific de Barcelona, Baldiri Reixac 4-8, 08028 Barcelona, Spain
| | - Cristina Val Garcia
- Grupo de Investigación Biofarma. Departamento de Farmacología, Farmacia y Tecnología Farmacéutica. Centro de Investigación CIMUS. Universidad de Santiago de Compostela, 15782 Santiago de Compostella, Spain
| | - Thomas J Schmidt
- Institut für Pharmazeutische Biologie und Phytochemie, Westfälische Wilhelms-Universität Münster, Corrensstraße 48, D-48149 Münster, Germany
| | - Guiscard Seebohm
- GRK 2515, Chemical Biology of Ion Channels (Chembion), Westfälische Wilhelms-Universität Münster, 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-Strasse 45, D-48149 Münster, Germany.,Grupo de Investigación Biofarma. Departamento de Farmacología, Farmacia y Tecnología Farmacéutica. Centro de Investigación CIMUS. Universidad de Santiago de Compostela, 15782 Santiago de Compostella, Spain
| | - Bernhard Wünsch
- GRK 2515, Chemical Biology of Ion Channels (Chembion), Westfälische Wilhelms-Universität Münster, D-48149 Münster, Germany.,Institut für Pharmazeutische und Medizinische Chemie, Westfälische Wilhelms-Universität Münster, Corrensstraße 48, D-48149 Münster, Germany
| |
Collapse
|
163
|
Strong KL, Epplin MP, Ogden KK, Burger PB, Kaiser TM, Wilding TJ, Kusumoto H, Camp CR, Shaulsky G, Bhattacharya S, Perszyk RE, Menaldino DS, McDaniel MJ, Zhang J, Le P, Banke TG, Hansen KB, Huettner JE, Liotta DC, Traynelis SF. Distinct GluN1 and GluN2 Structural Determinants for Subunit-Selective Positive Allosteric Modulation of N-Methyl-d-aspartate Receptors. ACS Chem Neurosci 2021; 12:79-98. [PMID: 33326224 DOI: 10.1021/acschemneuro.0c00561] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
N-Methyl-d-aspartate receptors (NMDARs) are ionotropic ligand-gated glutamate receptors that mediate fast excitatory synaptic transmission in the central nervous system (CNS). Several neurological disorders may involve NMDAR hypofunction, which has driven therapeutic interest in positive allosteric modulators (PAMs) of NMDAR function. Here we describe modest changes to the tetrahydroisoquinoline scaffold of GluN2C/GluN2D-selective PAMs that expands activity to include GluN2A- and GluN2B-containing recombinant and synaptic NMDARs. These new analogues are distinct from GluN2C/GluN2D-selective compounds like (+)-(3-chlorophenyl)(6,7-dimethoxy-1-((4-methoxyphenoxy)methyl)-3,4-dihydroisoquinolin-2(1H)-yl)methanone (CIQ) by virtue of their subunit selectivity, molecular determinants of action, and allosteric regulation of agonist potency. The (S)-enantiomers of two analogues (EU1180-55, EU1180-154) showed activity at NMDARs containing all subunits (GluN2A, GluN2B, GluN2C, GluN2D), whereas the (R)-enantiomers were primarily active at GluN2C- and GluN2D-containing NMDARs. Determination of the actions of enantiomers on triheteromeric receptors confirms their unique pharmacology, with greater activity of (S) enantiomers at GluN2A/GluN2D and GluN2B/GluN2D subunit combinations than (R) enantiomers. Evaluation of the (S)-EU1180-55 and EU1180-154 response of chimeric kainate/NMDA receptors revealed structural determinants of action within the pore-forming region and associated linkers. Scanning mutagenesis identified structural determinants within the GluN1 pre-M1 and M1 regions that alter the activity of (S)-EU1180-55 but not (R)-EU1180-55. By contrast, mutations in pre-M1 and M1 regions of GluN2D perturb the actions of only the (R)-EU1180-55 but not the (S) enantiomer. Molecular modeling supports the idea that the (S) and (R) enantiomers interact distinctly with GluN1 and GluN2 pre-M1 regions, suggesting that two distinct sites exist for these NMDAR PAMs, each of which has different functional effects.
Collapse
Affiliation(s)
- Katie L. Strong
- Department of Pharmacology, Emory University, 1510 Clifton Road, Atlanta, Georgia 30322, United States
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Matthew P. Epplin
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Kevin K. Ogden
- Department of Pharmacology, Emory University, 1510 Clifton Road, Atlanta, Georgia 30322, United States
| | - Pieter B. Burger
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Thomas M. Kaiser
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Timothy J. Wilding
- Department of Cell Biology and Physiology, Washington University, St Louis, Missouri 63110, United States
| | - Hiro Kusumoto
- Department of Pharmacology, Emory University, 1510 Clifton Road, Atlanta, Georgia 30322, United States
| | - Chad R. Camp
- Department of Pharmacology, Emory University, 1510 Clifton Road, Atlanta, Georgia 30322, United States
| | - Gil Shaulsky
- Department of Pharmacology, Emory University, 1510 Clifton Road, Atlanta, Georgia 30322, United States
| | - Subhrajit Bhattacharya
- Department of Drug Discovery and Development, Auburn University, Auburn, Alabama 36849, United States
| | - Riley E. Perszyk
- Department of Pharmacology, Emory University, 1510 Clifton Road, Atlanta, Georgia 30322, United States
| | - David S. Menaldino
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Miranda J. McDaniel
- Department of Pharmacology, Emory University, 1510 Clifton Road, Atlanta, Georgia 30322, United States
| | - Jing Zhang
- Department of Pharmacology, Emory University, 1510 Clifton Road, Atlanta, Georgia 30322, United States
| | - Phuong Le
- Department of Pharmacology, Emory University, 1510 Clifton Road, Atlanta, Georgia 30322, United States
| | - Tue G. Banke
- Department of Pharmacology, Emory University, 1510 Clifton Road, Atlanta, Georgia 30322, United States
| | - Kasper B. Hansen
- Department of Pharmacology, Emory University, 1510 Clifton Road, Atlanta, Georgia 30322, United States
- Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, Division for Biological Sciences, University of Montana, 32 Campus Drive, Missoula, Montana 59812, United States
| | - James E. Huettner
- Department of Cell Biology and Physiology, Washington University, St Louis, Missouri 63110, United States
| | - Dennis C. Liotta
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Stephen F. Traynelis
- Department of Pharmacology, Emory University, 1510 Clifton Road, Atlanta, Georgia 30322, United States
| |
Collapse
|
164
|
Jiao J, Li L, Sun M, Fang J, Meng L, Zhang Y, Jia C, Ma L. Identification of a novel GRIN2D variant in a neonate with intractable epileptic encephalopathy-a case report. BMC Pediatr 2021; 21:5. [PMID: 33397303 PMCID: PMC7780707 DOI: 10.1186/s12887-020-02462-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 12/10/2020] [Indexed: 11/19/2022] Open
Abstract
Background N-methyl-D-aspartate (NMDA) receptors are ligand-gated ion channels that mediate excitatory synaptic transmission in the central nervous system. The functional NMDA receptors are heterotetramers consisting mainly of two GluN1 and two GluN2 subunits. GluN2 is encoded by the GRIN2D gene. A few case series have shown that GRIN2D variants are linked to developmental and epileptic encephalopathy. In this article, we report a novel GRIN2D variant, namely c.2021C > A (p.T674K) in a neonate with intractable epileptic encephalopathy. Case presentation A 12-day-old boy who had stiffness of the lower and upper extremities since birth was transferred from a local hospital to our department. On admission, the patient presented with head tilting backwards, staring, apnea and hypertonia of limbs. Video electroencephalogram showed continuous, generalized or multi-focal spike-wave and spike-and-slow wave discharges and hypsarrhythmia. A treatment regimen composed of phenobarbital, midazolam, levetiracetam and clonazepam was administered, which however led to only partial control of the seizure. Whole-exome sequencing identified c.2021C > A (p.T674K) in GRIN2D in the patient while such a mutation was not detected in the parents. The patient was hospitalized for 1 month and died of sudden cardio-respiratory arrest 2 weeks after discharge. Conclusions A novel variant of GRIN2D was identified in a neonate with epileptic encephalopathy. Epilepsy associated with this GRIN2D mutation is refractory to conventional anti-epileptic medications.
Collapse
Affiliation(s)
- Jiancheng Jiao
- Neonatology Department, Children's Hospital of Hebei Province, 133 Jianhua South Street, 050031, Shijiazhuang, Hebei Province, China
| | - Li Li
- Neonatology Department, Children's Hospital of Hebei Province, 133 Jianhua South Street, 050031, Shijiazhuang, Hebei Province, China
| | - Min Sun
- Neonatology Department, Children's Hospital of Hebei Province, 133 Jianhua South Street, 050031, Shijiazhuang, Hebei Province, China
| | - Junchen Fang
- Neonatology Department, Children's Hospital of Hebei Province, 133 Jianhua South Street, 050031, Shijiazhuang, Hebei Province, China
| | - Lingzhi Meng
- Neonatology Department, Children's Hospital of Hebei Province, 133 Jianhua South Street, 050031, Shijiazhuang, Hebei Province, China
| | - Yudong Zhang
- Neonatology Department, Children's Hospital of Hebei Province, 133 Jianhua South Street, 050031, Shijiazhuang, Hebei Province, China
| | - Chao Jia
- Neonatology Department, Children's Hospital of Hebei Province, 133 Jianhua South Street, 050031, Shijiazhuang, Hebei Province, China
| | - Li Ma
- Neonatology Department, Children's Hospital of Hebei Province, 133 Jianhua South Street, 050031, Shijiazhuang, Hebei Province, China.
| |
Collapse
|
165
|
Zhou W, Guan Z. Ion Channels in Anesthesia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1349:401-413. [DOI: 10.1007/978-981-16-4254-8_19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
166
|
Das M, Mahler F, Hariharan P, Wang H, Du Y, Mortensen JS, Patallo EP, Ghani L, Glück D, Lee HJ, Byrne B, Loland CJ, Guan L, Kobilka BK, Keller S, Chae PS. Diastereomeric Cyclopentane-Based Maltosides (CPMs) as Tools for Membrane Protein Study. J Am Chem Soc 2020; 142:21382-21392. [PMID: 33315387 PMCID: PMC8015409 DOI: 10.1021/jacs.0c09629] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Amphiphilic agents, called detergents, are invaluable tools for studying membrane proteins. However, membrane proteins encapsulated by conventional head-to-tail detergents tend to denature or aggregate, necessitating the development of structurally distinct molecules with improved efficacy. Here, a novel class of diastereomeric detergents with a cyclopentane core unit, designated cyclopentane-based maltosides (CPMs), were prepared and evaluated for their ability to solubilize and stabilize several model membrane proteins. A couple of CPMs displayed enhanced behavior compared with the benchmark conventional detergent, n-dodecyl-β-d-maltoside (DDM), for all the tested membrane proteins including two G-protein-coupled receptors (GPCRs). Furthermore, CPM-C12 was notable for its ability to confer enhanced membrane protein stability compared with the previously developed conformationally rigid NBMs [J. Am. Chem. Soc. 2017, 139, 3072] and LMNG. The effect of the individual CPMs on protein stability varied depending on both the detergent configuration (cis/trans) and alkyl chain length, allowing us draw conclusions on the detergent structure-property-efficacy relationship. Thus, this study not only provides novel detergent tools useful for membrane protein research but also reports on structural features of the detergents critical for detergent efficacy in stabilizing membrane proteins.
Collapse
Affiliation(s)
- Manabendra Das
- Department of Bionanotechnology, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 155-88, Korea
- Technische Universität Kaiserslautern (TUK), Erwin-Schrödinger-Str. 13, 67663 Kaiserslautern, Germany
| | - Florian Mahler
- Technische Universität Kaiserslautern (TUK), Erwin-Schrödinger-Str. 13, 67663 Kaiserslautern, Germany
| | - Parameswaran Hariharan
- Department of Cell Physiology and Molecular Biophysics, Center for Membrane Protein Research, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas 79430, United States
| | - Haoqing Wang
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, California 94305, United States
| | - Yang Du
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, California 94305, United States
| | - Jonas S Mortensen
- Department of Neuroscience, University of Copenhagen, Copenhagen DK-2200, Denmark
| | - Eugenio Pérez Patallo
- Technische Universität Kaiserslautern (TUK), Erwin-Schrödinger-Str. 13, 67663 Kaiserslautern, Germany
| | - Lubna Ghani
- Department of Bionanotechnology, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 155-88, Korea
| | - David Glück
- Technische Universität Kaiserslautern (TUK), Erwin-Schrödinger-Str. 13, 67663 Kaiserslautern, Germany
| | - Ho Jin Lee
- Department of Bionanotechnology, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 155-88, Korea
| | - Bernadette Byrne
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| | - Claus J Loland
- Department of Neuroscience, University of Copenhagen, Copenhagen DK-2200, Denmark
| | - Lan Guan
- Department of Cell Physiology and Molecular Biophysics, Center for Membrane Protein Research, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas 79430, United States
| | - Brian K Kobilka
- Department of Molecular and Cellular Physiology, Stanford University, Stanford, California 94305, United States
| | - Sandro Keller
- Technische Universität Kaiserslautern (TUK), Erwin-Schrödinger-Str. 13, 67663 Kaiserslautern, Germany
- Institute of Molecular Biosciences (IMB), NAWI Graz, University of Graz, Humboldtstr. 50/III, 8010 Graz, Austria
- Field of Excellence BioHealth, University of Graz, 8010 Graz, Austria
- BioTechMed-Graz, Graz, Austria
| | - Pil Seok Chae
- Department of Bionanotechnology, Center for Bionano Intelligence Education and Research, Hanyang University, Ansan 155-88, Korea
| |
Collapse
|
167
|
Wong HHW, Rannio S, Jones V, Thomazeau A, Sjöström PJ. NMDA receptors in axons: there's no coincidence. J Physiol 2020; 599:367-387. [PMID: 33141440 DOI: 10.1113/jp280059] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 10/27/2020] [Indexed: 12/16/2022] Open
Abstract
In the textbook view, N-methyl-d-aspartate (NMDA) receptors are postsynaptically located detectors of coincident activity in Hebbian learning. However, controversial presynaptically located NMDA receptors (preNMDARs) have for decades been repeatedly reported in the literature. These preNMDARs have typically been implicated in the regulation of short-term and long-term plasticity, but precisely how they signal and what their functional roles are have been poorly understood. The functional roles of preNMDARs across several brain regions and different forms of plasticity can differ vastly, with recent discoveries showing key involvement of unusual subunit composition. Increasing evidence shows preNMDAR can signal through both ionotropic action by fluxing calcium and in metabotropic mode even in the presence of magnesium blockade. We argue that these unusual properties may explain why controversy has surrounded this receptor type. In addition, the expression of preNMDARs at some synapse types but not others can underlie synapse-type-specific plasticity. Last but not least, preNMDARs are emerging therapeutic targets in disease states such as neuropathic pain. We conclude that axonally located preNMDARs are required for specific purposes and do not end up there by accident.
Collapse
Affiliation(s)
- Hovy Ho-Wai Wong
- Department of Medicine, Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montreal General Hospital, 1650 Cedar Ave, Montreal, Quebec, H3G 1A4, Canada
| | - Sabine Rannio
- Department of Medicine, Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montreal General Hospital, 1650 Cedar Ave, Montreal, Quebec, H3G 1A4, Canada.,Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
| | - Victoria Jones
- Department of Medicine, Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montreal General Hospital, 1650 Cedar Ave, Montreal, Quebec, H3G 1A4, Canada.,Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
| | - Aurore Thomazeau
- Department of Medicine, Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montreal General Hospital, 1650 Cedar Ave, Montreal, Quebec, H3G 1A4, Canada
| | - P Jesper Sjöström
- Department of Medicine, Department of Neurology and Neurosurgery, Centre for Research in Neuroscience, Brain Repair and Integrative Neuroscience Program, The Research Institute of the McGill University Health Centre, Montreal General Hospital, 1650 Cedar Ave, Montreal, Quebec, H3G 1A4, Canada
| |
Collapse
|
168
|
Burada AP, Vinnakota R, Bharti P, Dutta P, Dubey N, Kumar J. Emerging insights into the structure and function of ionotropic glutamate delta receptors. Br J Pharmacol 2020; 179:3612-3627. [DOI: 10.1111/bph.15313] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 10/26/2020] [Accepted: 10/27/2020] [Indexed: 12/22/2022] Open
Affiliation(s)
- Ananth Prasad Burada
- Laboratory of Membrane Protein Biology National Centre for Cell Science, NCCS Complex, S. P. Pune University Pune India
| | - Rajesh Vinnakota
- Laboratory of Membrane Protein Biology National Centre for Cell Science, NCCS Complex, S. P. Pune University Pune India
| | - Pratibha Bharti
- Laboratory of Membrane Protein Biology National Centre for Cell Science, NCCS Complex, S. P. Pune University Pune India
| | - Priyanka Dutta
- Laboratory of Membrane Protein Biology National Centre for Cell Science, NCCS Complex, S. P. Pune University Pune India
| | - Neelima Dubey
- Molecular Neuroscience Research Lab Dr. D. Y. Patil Biotechnology and Bioinformatics Institute, Dr. D. Y. Patil Vidyapeeth Tathawade Pune 411033 India
| | - Janesh Kumar
- Laboratory of Membrane Protein Biology National Centre for Cell Science, NCCS Complex, S. P. Pune University Pune India
| |
Collapse
|
169
|
Amin JB, Gochman A, He M, Certain N, Wollmuth LP. NMDA Receptors Require Multiple Pre-opening Gating Steps for Efficient Synaptic Activity. Neuron 2020; 109:488-501.e4. [PMID: 33264592 DOI: 10.1016/j.neuron.2020.11.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/06/2020] [Accepted: 11/11/2020] [Indexed: 12/27/2022]
Abstract
NMDA receptors (NMDARs) are glutamate-gated ion channels that mediate fast excitatory synaptic transmission in the nervous system. Applying glutamate to outside-out patches containing a single NMDAR, we find that agonist-bound receptors transition to the open state via two conformations, an "unconstrained pre-active" state that contributes to fast synaptic events and a "constrained pre-active" state that does not. To define how glutamate drives these conformations, we decoupled the ligand-binding domains from specific transmembrane segments for GluN1 and GluN2A. Displacements of the pore-forming M3 segments define the energy of fast opening. However, to enter the unconstrained conformation and contribute to fast signaling, the GluN2 pre-M1 helix must be displaced before the M3 segments move. This pre-M1 displacement is facilitated by the flexibility of the S2-M4 of GluN1 and GluN2A. Thus, outer structures-pre-M1 and S2-M4-work in concert to remove constraints and prime the channel for rapid opening, facilitating fast synaptic transmission.
Collapse
Affiliation(s)
- Johansen B Amin
- Graduate Program in Molecular & Cellular Pharmacology, Stony Brook University, Stony Brook, NY 11794-5230, USA; Medical Scientist Training Program (MSTP), Stony Brook University, Stony Brook, NY 11794-5230, USA
| | - Aaron Gochman
- Department of Neurobiology & Behavior, Stony Brook University, Stony Brook, NY 11794-5230, USA
| | - Miaomiao He
- Graduate Program in Biochemistry & Structural Biology, Stony Brook University, Stony Brook, NY 11794-5230, USA
| | - Noele Certain
- Graduate Program in Molecular & Cellular Pharmacology, Stony Brook University, Stony Brook, NY 11794-5230, USA
| | - Lonnie P Wollmuth
- 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; Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY 11794-5230, USA.
| |
Collapse
|
170
|
García-Recio A, Santos-Gómez A, Soto D, Julia-Palacios N, García-Cazorla À, Altafaj X, Olivella M. GRIN database: A unified and manually curated repertoire of GRIN variants. Hum Mutat 2020; 42:8-18. [PMID: 33252190 DOI: 10.1002/humu.24141] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 10/13/2020] [Accepted: 11/16/2020] [Indexed: 12/22/2022]
Abstract
Glutamatergic neurotransmission is crucial for brain development, wiring neuronal function, and synaptic plasticity mechanisms. Recent genetic studies showed the existence of autosomal dominant de novo GRIN gene variants associated with GRIN-related disorders (GRDs), a rare pediatric neurological disorder caused by N-methyl- d-aspartate receptor (NMDAR) dysfunction. Notwithstanding, GRIN variants identification is exponentially growing and their clinical, genetic, and functional annotations remain highly fragmented, representing a bottleneck in GRD patient's stratification. To shorten the gap between GRIN variant identification and patient stratification, we present the GRIN database (GRINdb), a publicly available, nonredundant, updated, and curated database gathering all available genetic, functional, and clinical data from more than 4000 GRIN variants. The manually curated GRINdb outputs on a web server, allowing query and retrieval of reported GRIN variants, and thus representing a fast and reliable bioinformatics resource for molecular clinical advice. Furthermore, the comprehensive mapping of GRIN variants' genetic and clinical information along NMDAR structure revealed important differences in GRIN variants' pathogenicity and clinical phenotypes, shedding light on GRIN-specific fingerprints. Overall, the GRINdb and web server is a resource for molecular stratification of GRIN variants, delivering clinical and investigational insights into GRDs. GRINdb is accessible at http://lmc.uab.es/grindb.
Collapse
Affiliation(s)
- Adrián García-Recio
- Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.,Laboratori de Medicina Computacional, Unitat de Bioestadística, Facultat de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Ana Santos-Gómez
- Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.,Neurophysiology Laboratory, Department of Biomedicine, Institute of Neurosciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - David Soto
- Neurophysiology Laboratory, Department of Biomedicine, Institute of Neurosciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain.,August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Hospital Clínic, University of Barcelona, Barcelona, Spain
| | - Natalia Julia-Palacios
- Neurometabolic Unit, Department of Neurology, Hospital Sant Joan de Déu and CIBERER, Barcelona, Spain
| | - Àngels García-Cazorla
- Neurometabolic Unit, Department of Neurology, Hospital Sant Joan de Déu and CIBERER, Barcelona, Spain
| | - Xavier Altafaj
- Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain.,Neurophysiology Laboratory, Department of Biomedicine, Institute of Neurosciences, Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Mireia Olivella
- School of International Studies, ESCI-UPF, Barcelona, Spain.,Bioinfomatics and Medical Statistics Group, University of Vic-Central University of Catalonia, Vic, Spain
| |
Collapse
|
171
|
Loureiro CM, Fachim HA, Corsi-Zuelli F, Shuhama R, Joca S, Menezes PR, Dalton CF, Del-Ben CM, Louzada-Junior P, Reynolds GP. Epigenetic-mediated N-methyl-D-aspartate receptor changes in the brain of isolated reared rats. Epigenomics 2020; 12:1983-1997. [PMID: 33242253 DOI: 10.2217/epi-2020-0151] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Aim: We investigated: Grin1, Grin2a, Grin2b DNA methylation; NR1 and NR2 mRNA/protein in the prefrontal cortex (PFC); and hippocampus of male Wistar rats exposed to isolation rearing. Materials & methods: Animals were kept isolated or grouped (n = 10/group) from weaning for 10 weeks. Tissues were dissected for RNA/DNA extraction and N-methyl-D-aspartate receptor subunits were analyzed using quantitative reverse transcription (RT)-PCR, ELISA and pyrosequencing. Results: Isolated-reared animals had: decreased mRNA in PFC for all markers, increased NR1 protein in hippocampus and hypermethylation of Grin1 in PFC and Grin2b in hippocampus, compared with grouped rats. Associations between mRNA/protein and DNA methylation were found for both brain areas. Conclusion: This study indicates that epigenetic DNA methylation may underlie N-methyl-D-aspartate receptor mRNA/protein expression alterations caused by isolation rearing.
Collapse
Affiliation(s)
- Camila Marcelino Loureiro
- Department of Internal Medicine, Division of Clinical Immunology. Ribeirão Preto Medical School, University of São Paulo, Brazil.,Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, UK
| | - Helene Aparecida Fachim
- Department of Endocrinology & Metabolism, Salford Royal Foundation Trust, Salford, UK.,Department of Neurosciences & Behaviour, Ribeirão Preto Medical School, University of São Paulo, Brazil
| | - Fabiana Corsi-Zuelli
- Department of Neurosciences & Behaviour, Ribeirão Preto Medical School, University of São Paulo, Brazil
| | - Rosana Shuhama
- Department of Neurosciences & Behaviour, Ribeirão Preto Medical School, University of São Paulo, Brazil
| | - Sâmia Joca
- Department of Biomolecular Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Brazil.,Aarhus Institute of Advanced Studies (AIAS), Aarhus University, Denmark
| | - Paulo Rossi Menezes
- Department of Preventive Medicine, Faculty of Medicine, University of São Paulo, Brazil
| | - Caroline F Dalton
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, UK
| | - Cristina Marta Del-Ben
- Department of Neurosciences & Behaviour, Ribeirão Preto Medical School, University of São Paulo, Brazil
| | - Paulo Louzada-Junior
- Department of Internal Medicine, Division of Clinical Immunology. Ribeirão Preto Medical School, University of São Paulo, Brazil
| | - Gavin P Reynolds
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, UK
| |
Collapse
|
172
|
Bhatia NK, Carrillo E, Durham RJ, Berka V, Jayaraman V. Allosteric Changes in the NMDA Receptor Associated with Calcium-Dependent Inactivation. Biophys J 2020; 119:2349-2359. [PMID: 33098865 DOI: 10.1016/j.bpj.2020.08.045] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/14/2020] [Accepted: 08/20/2020] [Indexed: 01/07/2023] Open
Abstract
N-methyl-D-aspartate (NMDA) receptors mediate synaptic excitatory signaling in the mammalian central nervous system by forming calcium-permeable transmembrane channels upon binding glutamate and coagonist glycine. Ca2+ influx through NMDA receptors leads to channel inactivation through a process mediated by resident calmodulin bound to the intracellular C-terminal segment of the GluN1 subunit of the receptor. Using single-molecule FRET investigations, we show that in the presence of calcium-calmodulin, the distance across the two GluN1 subunits at the entrance of the first transmembrane segment is shorter and the bilobed cleft of the glycine-binding domain in GluN1 is more closed when bound to glycine and glutamate relative to what is observed in the presence of barium-calmodulin. Consistent with these observations, the glycine deactivation rate is slower in the presence of calcium-calmodulin. Taken together, these results show that the binding of calcium-calmodulin to the C-terminus has long-range allosteric effects on the extracellular segments of the receptor that may contribute to the calcium-dependent inactivation.
Collapse
Affiliation(s)
- Nidhi Kaur Bhatia
- Center for Membrane Biology, Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston, Houston, Texas
| | - Elisa Carrillo
- Center for Membrane Biology, Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston, Houston, Texas
| | - Ryan J Durham
- Center for Membrane Biology, Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston, Houston, Texas; MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, Texas
| | - Vladimir Berka
- Center for Membrane Biology, Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston, Houston, Texas
| | - Vasanthi Jayaraman
- Center for Membrane Biology, Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston, Houston, Texas; MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, Texas.
| |
Collapse
|
173
|
Tikhonov DB, Zhorov BS. The pore domain in glutamate-gated ion channels: Structure, drug binding and similarity with potassium channels. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2020; 1862:183401. [PMID: 32562696 DOI: 10.1016/j.bbamem.2020.183401] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 06/11/2020] [Accepted: 06/15/2020] [Indexed: 01/28/2023]
Abstract
Ionotropic glutamate receptors in the CNS excitatory synapses of vertebrates are involved in numerous physiological and pathological processes. Decades of intensive studies greatly advanced our understanding of molecular organization of these transmembrane proteins. Here we focus on the channel pore domain, its selectivity filter and the activation gate, and the pore block by organic ligands. We compare findings from indirect experimental approaches, including site-directed mutagenesis, with recent crystal and cryo-EM structures of different channels in different functional states and complexed with different ligands. We summarize remaining uncertainties and unresolved problems related to the channel structure, function and pharmacology.
Collapse
Affiliation(s)
- Denis B Tikhonov
- I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 44 Thorez Prospect, St. Petersburg 194223, Russia.
| | - Boris S Zhorov
- I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, 44 Thorez Prospect, St. Petersburg 194223, Russia; Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West, Hamilton, Ontario L8N 4K1, Canada
| |
Collapse
|
174
|
Sengar AS, Li H, Zhang W, Leung C, Ramani AK, Saw NM, Wang Y, Tu Y, Ross PJ, Scherer SW, Ellis J, Brudno M, Jia Z, Salter MW. Control of Long-Term Synaptic Potentiation and Learning by Alternative Splicing of the NMDA Receptor Subunit GluN1. Cell Rep 2020; 29:4285-4294.e5. [PMID: 31875540 DOI: 10.1016/j.celrep.2019.11.087] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 10/05/2019] [Accepted: 11/21/2019] [Indexed: 01/09/2023] Open
Abstract
NMDA receptors (NMDARs) are critical for physiological synaptic plasticity, learning, and memory and for pathological plasticity and neuronal death. The GluN1 subunit is encoded by a single gene, GRIN1, with 8 splice variants, but whether the diversity generated by this splicing has physiological consequences remains enigmatic. Here, we generate mice lacking from the GluN1 exon 5-encoded N1 cassette (GluN1a mice) or compulsorily expressing this exon (GluN1b mice). Despite no differences in basal synaptic transmission, long-term potentiation in the hippocampus is significantly enhanced in GluN1a mice compared with that in GluN1b mice. Furthermore, GluN1a mice learn more quickly and have significantly better spatial memory performance than do GluN1b mice. In addition, in human iPSC-derived neurons in autism spectrum disorder NMDARs show characteristics of N1-lacking GluN1. Our findings indicate that alternative splicing of GluN1 is a mechanism for controlling physiological long-lasting synaptic potentiation, learning, and memory.
Collapse
Affiliation(s)
- Ameet S Sengar
- Program in Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Hongbin Li
- Program in Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Wenbo Zhang
- Program in Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Celeste Leung
- Program in Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Arun K Ramani
- Centre for Computational Medicine, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Ner Mu Saw
- Program in Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Yongqian Wang
- Program in Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - YuShan Tu
- Program in Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - P Joel Ross
- Biology Department, University of Prince Edward Island, Charlottetown, PE C1A 4P3, Canada
| | - Stephen W Scherer
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; McLaughlin Centre, University of Toronto, Toronto, ON M5G 0A4, Canada
| | - James Ellis
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Michael Brudno
- Centre for Computational Medicine, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; Department of Computer Science, University of Toronto, Toronto, ON M5T 3A1, Canada
| | - Zhengping Jia
- Program in Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Michael W Salter
- Program in Neurosciences & Mental Health, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
175
|
Stroebel D, Paoletti P. Architecture and function of NMDA receptors: an evolutionary perspective. J Physiol 2020; 599:2615-2638. [PMID: 32786006 DOI: 10.1113/jp279028] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 07/21/2020] [Indexed: 12/20/2022] Open
Abstract
Ionotropic glutamate receptors (iGluRs) are a major class of ligand-gated ion channels that are widespread in the living kingdom. Their critical role in excitatory neurotransmission and brain function of arthropods and vertebrates has made them a compelling subject of interest for neurophysiologists and pharmacologists. This is particularly true for NMDA receptor (NMDARs), a subclass of iGluRs that act as central drivers of synaptic plasticity in the CNS. How and when the unique properties of NMDARs arose during evolution, and how they relate to the evolution of the nervous system, remain open questions. Recent years have witnessed a boom in both genomic and structural data, such that it is now possible to analyse the evolution of iGluR genes on an unprecedented scale and within a solid molecular framework. In this review, combining insights from phylogeny, atomic structure and physiological and mechanistic data, we discuss how evolution of NMDAR motifs and sequences shaped their architecture and functionalities. We trace differences and commonalities between NMDARs and other iGluRs, emphasizing a few distinctive properties of the former regarding ligand binding and gating, permeation, allosteric modulation and intracellular signalling. Finally, we speculate on how specific molecular properties of iGuRs arose to supply new functions to the evolving structure of the nervous system, from early metazoan to present mammals.
Collapse
Affiliation(s)
- David Stroebel
- Ecole Normale Supérieure, CNRS, INSERM, Institute de Biologie de l'Ecole Normale Supérieure (IBENS), Université PSL, Paris, France
| | - Pierre Paoletti
- Ecole Normale Supérieure, CNRS, INSERM, Institute de Biologie de l'Ecole Normale Supérieure (IBENS), Université PSL, Paris, France
| |
Collapse
|
176
|
Callahan PM, Terry AV, Nelson FR, Volkmann RA, Vinod AB, Zainuddin M, Menniti FS. Modulating inhibitory response control through potentiation of GluN2D subunit-containing NMDA receptors. Neuropharmacology 2020; 173:107994. [PMID: 32057801 DOI: 10.1016/j.neuropharm.2020.107994] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 01/16/2020] [Accepted: 02/05/2020] [Indexed: 01/15/2023]
Abstract
NMDA receptors containing GluN2D subunits are expressed in the subthalamic nucleus and external globus pallidus, key nuclei of the indirect and hyperdirect pathways of the basal ganglia. This circuitry integrates cortical input with dopaminergic signaling to select advantageous behaviors among available choices. In the experiments described here, we characterized the effects of PTC-174, a novel positive allosteric modulator (PAM) of GluN2D subunit-containing NMDA receptors, on response control regulated by this circuitry. The indirect pathway suppresses less advantageous behavioral choices, a manifestation of which is suppression of locomotor activity in rats. Systemic administration of PTC-174 produced a dose-dependent reduction in activity in rats placed in a novel open field or administered the stimulants MK-801 or amphetamine. The hyperdirect pathway controls release of decisions from the basal ganglia to the cortex to optimize choice processing. Such response control was modeled in rats as premature responding in the 5-choice serial reaction time (5-CSRT) task. PTC-174 produced a dose-dependent reduction in premature responding in this task. These data suggest that potentiation of GluN2D receptor activity by PTC-174 facilitates the complex basal ganglia information processing that underlies response control. The behavioral effects occurred at estimated free PTC-174 brain concentrations predicted to induce 10-50% increases in GluN2D activity. The present findings suggest the potential of GluN2D PAMs to modulate basal ganglia function and to treat neurological disorders related to dysfunctional response control.
Collapse
Affiliation(s)
- Patrick M Callahan
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA; Small Animal Behavior Core, Augusta University, Augusta, GA, 30912, USA
| | - Alvin V Terry
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA; Small Animal Behavior Core, Augusta University, Augusta, GA, 30912, USA
| | | | | | - A B Vinod
- Jubilant Biosys Ltd, Yeshwantpur, Bangalore, 560022, Karnataka, India
| | - Mohd Zainuddin
- Jubilant Biosys Ltd, Yeshwantpur, Bangalore, 560022, Karnataka, India
| | - Frank S Menniti
- The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, 02881, USA.
| |
Collapse
|
177
|
A comprehensive description of GluN2B-selective N-methyl-D-aspartate (NMDA) receptor antagonists. Eur J Med Chem 2020; 200:112447. [DOI: 10.1016/j.ejmech.2020.112447] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 05/06/2020] [Accepted: 05/08/2020] [Indexed: 12/17/2022]
|
178
|
Bae HE, Cecchetti C, Du Y, Katsube S, Mortensen JS, Huang W, Rehan S, Lee HJ, Loland CJ, Guan L, Kobilka BK, Byrne B, Chae PS. Pendant-bearing glucose-neopentyl glycol (P-GNG) amphiphiles for membrane protein manipulation: Importance of detergent pendant chain for protein stabilization. Acta Biomater 2020; 112:250-261. [PMID: 32522715 PMCID: PMC7366829 DOI: 10.1016/j.actbio.2020.06.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 05/31/2020] [Accepted: 06/02/2020] [Indexed: 12/15/2022]
Abstract
Glucoside detergents are successfully used for membrane protein crystallization mainly because of their ability to form small protein-detergent complexes. In a previous study, we introduced glucose neopentyl glycol (GNG) amphiphiles with a branched diglucoside structure that has facilitated high resolution crystallographic structure determination of several membrane proteins. Like other glucoside detergents, however, these GNGs were less successful than DDM in stabilizing membrane proteins, limiting their wide use in protein structural study. As a strategy to improve GNG efficacy for protein stabilization, we introduced two different alkyl chains (i.e., main and pendant chains) into the GNG scaffold while maintaining the branched diglucoside head group. Of these pendant-bearing GNGs (P-GNGs), three detergents (GNG-2,14, GNG-3,13 and GNG-3,14) were not only notably better than both DDM (a gold standard detergent) and the previously described GNGs at stabilizing all six membrane proteins tested here, but were also as efficient as DDM at membrane protein extraction. The results suggest that the C14 main chain of the P-GNGs is highly compatible with the hydrophobic widths of membrane proteins, while the C2/C3 pendant chain is effective at strengthening detergent hydrophobic interactions. Based on the marked effect on protein stability and solubility, these glucoside detergents hold significant potential for membrane protein structural study. Furthermore, the independent roles of the detergent two alkyl chains first introduced in this study have shed light on new amphiphile design for membrane protein study. STATEMENT OF SIGNIFICANCE: Detergent efficacy for protein stabilization tends to be protein-specific, thus it is challenging to find a detergent that is effective at stabilizing multiple membrane proteins. By incorporating a pendant chain into our previous GNG scaffold, we prepared pendant chain-bearing GNGs (P-GNGs) and identified three P-GNGs that were highly effective at stabilizing all membrane proteins tested here including two GPCRs. In addition, the new detergents were as efficient as DDM at extracting membrane proteins, enabling use of these detergents over the multiple steps of protein isolation. The key difference between the P-GNGs and other glucoside detergents, the presence of a pendant chain, is likely to be responsible for their markedly enhanced protein stabilization behavior.
Collapse
Affiliation(s)
- Hyoung Eun Bae
- Department of Bionanotechnology, Hanyang University, Ansan, 15588 (Korea)
| | - Cristina Cecchetti
- Department of Life Sciences, Imperial College London, London, SW7 2AZ (UK)
| | - Yang Du
- Department of Molecular and Cellular Physiology, Stanford University, CA 94305 (USA)
| | - Satoshi Katsube
- Department of Cell Physiology and Molecular Biophysics, Center for Membrane Protein Research, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430 (USA)
| | - Jonas S Mortensen
- Department of Neuroscience, University of Copenhagen, Copenhagen, DK-2200 (Denmark)
| | - Weijiao Huang
- Department of Molecular and Cellular Physiology, Stanford University, CA 94305 (USA)
| | - Shahid Rehan
- Institute of Biotechnology, University of Helsinki, Helsinki (Finland); HiLIFE, University of Helsinki, Helsinki (Finland)
| | - Ho Jin Lee
- Department of Bionanotechnology, Hanyang University, Ansan, 15588 (Korea)
| | - Claus J Loland
- Department of Neuroscience, University of Copenhagen, Copenhagen, DK-2200 (Denmark)
| | - Lan Guan
- Department of Cell Physiology and Molecular Biophysics, Center for Membrane Protein Research, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430 (USA)
| | - Brian K Kobilka
- Department of Molecular and Cellular Physiology, Stanford University, CA 94305 (USA)
| | - Bernadette Byrne
- Department of Life Sciences, Imperial College London, London, SW7 2AZ (UK)
| | - Pil Seok Chae
- Department of Bionanotechnology, Hanyang University, Ansan, 15588 (Korea).
| |
Collapse
|
179
|
Cheng H, Cheng Q, Bao X, Luo Y, Zhou Y, Li Y, Hua Q, Liu W, Tang S, Feng D, Luo Z. Over-activation of NMDA receptors promotes ABCA1 degradation and foam cell formation. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158778. [PMID: 32745694 DOI: 10.1016/j.bbalip.2020.158778] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 07/24/2020] [Accepted: 07/27/2020] [Indexed: 11/17/2022]
Abstract
ATP-binding cassette transporter A1 (ABCA1) is an essential regulator of intracellular cholesterol efflux. Secreted cholesterol binds to lipid-free apolipoprotein A-I (apoA-I) in peripheral blood to constitute high-density lipoprotein cholesterol (HDL) complexes. ABCA1 protein on the surface of macrophages acts as a crucial controller in preventing cholesterol accumulation. Importantly, ABCA1 is unstable and easily degraded via a series of biochemical activities, including but not limited to calpain-mediated and ubiquitin-proteasome system-mediated processes. How accelerated ABCA1 degradation impacts disordered lipid metabolism in macrophages and foam cell formation is unclear. N-methyl d-aspartate receptors (NMDARs) are ionotropic glutamate receptors with high calcium permeability. Calcium influx via NMDARs activates downstream signaling pathways. Over-activation of NMDARs stimulated by NMDA contributes to dysfunctional lipid metabolism in macrophages and foam cell formation via promotion of calpain-mediated ABCA1 proteolysis. However, increased NMDAR activity does not affect liver X receptor expression or ABCA1 mRNA levels. Following NMDA receptor silencing or calpain inhibition, NMDA treatment did not reduce ABCA1 protein levels, nor caused lipid accumulation in macrophages. In addition, NMDAR over-activation activates NF-κB signaling to promote IL-1β and IL-6 macrophage marker expression. However, NMDAR silencing and calpain inhibition reduce inflammatory macrophage responses. In summary, our study suggests that NMDAR activation reduces surface ABCA1 protein, promotes lipid accumulation, and induces the production and secretion of many inflammatory mediators in macrophages, possibly through enhanced calpain-mediated ABCA1 protein degradation. Thus, the NMDAR receptor may be a novel pharmacologic target for atherosclerosis therapy.
Collapse
Affiliation(s)
- Haipeng Cheng
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Qingmei Cheng
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xingwen Bao
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yongyu Luo
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yan Zhou
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Yanghang Li
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Qingzhong Hua
- Xiangya Nursing School, Central South University, Changsha, Hunan, China
| | - Wei Liu
- Xiangya Nursing School, Central South University, Changsha, Hunan, China
| | - Siyuan Tang
- Xiangya Nursing School, Central South University, Changsha, Hunan, China
| | - Dandan Feng
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China.
| | - Ziqiang Luo
- Department of Physiology, Xiangya School of Medicine, Central South University, Changsha, Hunan, China.
| |
Collapse
|
180
|
Tang W, Liu D, Traynelis SF, Yuan H. Positive allosteric modulators that target NMDA receptors rectify loss-of-function GRIN variants associated with neurological and neuropsychiatric disorders. Neuropharmacology 2020; 177:108247. [PMID: 32712275 DOI: 10.1016/j.neuropharm.2020.108247] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 05/20/2020] [Accepted: 07/20/2020] [Indexed: 12/23/2022]
Abstract
N-methyl-d-aspartate receptors (NMDARs) mediate a slow component of excitatory synaptic transmission that plays important roles in normal brain function and development. A large number of disease-associated variants in the GRIN gene family encoding NMDAR GluN subunits have been identified in patients with various neurological and neuropsychiatric disorders. Many of these variants reduce the function of NMDARs by a range of different mechanisms, including reduced glutamate potency, reduced glycine potency, accelerated deactivation time course, decreased surface expression, and/or reduced open probability. We have evaluated whether three positive allosteric modulators of NMDAR receptor function (24(S)-hydroxycholesterol, pregnenolone sulfate, tobramycin) and three co-agonists (d-serine, l-serine, and d-cycloserine) can mitigate the diminished function of NMDARs harboring GRIN variants. We examined the effects of these modulators on NMDARs that contained 21 different loss-of-function variants in GRIN1, GRIN2A, or GRIN2B, identified in patients with epilepsy, intellectual disability, autism, and/or movement disorders. For all variants, some aspect of the reduced function was partially restored. Moreover, some variants showed enhanced sensitivity to positive allosteric modulators compared to wild type receptors. These results raise the possibility that enhancement of NMDAR function by positive allosteric modulators may be a useful therapeutic strategy.
Collapse
Affiliation(s)
- Weiting Tang
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Ding Liu
- Department of Pharmacology and Chemical Biology, 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
|
181
|
Chou TH, Tajima N, Romero-Hernandez A, Furukawa H. Structural Basis of Functional Transitions in Mammalian NMDA Receptors. Cell 2020; 182:357-371.e13. [PMID: 32610085 PMCID: PMC8278726 DOI: 10.1016/j.cell.2020.05.052] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 04/22/2020] [Accepted: 05/27/2020] [Indexed: 12/18/2022]
Abstract
Excitatory neurotransmission meditated by glutamate receptors including N-methyl-D-aspartate receptors (NMDARs) is pivotal to brain development and function. NMDARs are heterotetramers composed of GluN1 and GluN2 subunits, which bind glycine and glutamate, respectively, to activate their ion channels. Despite importance in brain physiology, the precise mechanisms by which activation and inhibition occur via subunit-specific binding of agonists and antagonists remain largely unknown. Here, we show the detailed patterns of conformational changes and inter-subunit and -domain reorientation leading to agonist-gating and subunit-dependent competitive inhibition by providing multiple structures in distinct ligand states at 4 Å or better. The structures reveal that activation and competitive inhibition by both GluN1 and GluN2 antagonists occur by controlling the tension of the linker between the ligand-binding domain and the transmembrane ion channel of the GluN2 subunit. Our results provide detailed mechanistic insights into NMDAR pharmacology, activation, and inhibition, which are fundamental to the brain physiology.
Collapse
Affiliation(s)
- Tsung-Han Chou
- WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Nami Tajima
- WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Annabel Romero-Hernandez
- WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; Watson School of Biological Sciences, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Hiro Furukawa
- WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; Watson School of Biological Sciences, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
182
|
Taylor CP, Harris EW. Analgesia with Gabapentin and Pregabalin May Involve N-Methyl-d-Aspartate Receptors, Neurexins, and Thrombospondins. J Pharmacol Exp Ther 2020; 374:161-174. [PMID: 32321743 DOI: 10.1124/jpet.120.266056] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 04/17/2020] [Indexed: 03/08/2025] Open
Abstract
The gabapentinoid drugs gabapentin and pregabalin (Neurontin and Lyrica) are mainstay treatments for neuropathic pain and preventing focal seizures. Both drugs have similar effects to each other in animal models and clinically. Studies have shown that a protein first identified as an auxiliary subunit of voltage-gated calcium channels (the α 2 δ-subunit type 1 [α 2 δ-1], or Ca V a2d1) is the high-affinity binding site for gabapentin and pregabalin and is required for the efficacy of these drugs. The α 2 δ-1 protein is required for the ability of gabapentin and pregabalin to reduce neurotransmitter release in neuronal tissue, consistent with a therapeutic mechanism of action via voltage-gated calcium channels. However, recent studies have revealed that α 2 δ-1 interacts with several proteins in addition to voltage-gated calcium channels, and these additional proteins could be involved in gabapentinoid pharmacology. Furthermore, gabapentin and pregabalin have been shown to modify the action of a subset of N-methyl-d-aspartate-sensitive glutamate receptors, neurexin-1α, and thrombospondin proteins by binding to α 2 δ-1. Thus, these effects may contribute substantially to gabapentinoid therapeutic mechanism of action. SIGNIFICANCE STATEMENT: It is widely believed that gabapentin and pregabalin act by modestly reducing the membrane localization and activation of voltage-gated calcium channels at synaptic endings in spinal cord and neocortex via binding to the α 2 δ-1 protein. However, recent findings show that the α 2 δ-1 protein also interacts with N-methyl-d-aspartate-sensitive glutamate receptors, neurexin-1α, thrombospondins (adhesion molecules), and other presynaptic proteins. These newly discovered interactions, in addition to actions at calcium channels, may be important mediators of gabapentin and pregabalin therapeutic effects.
Collapse
Affiliation(s)
- Charles P Taylor
- CP Taylor Consulting, Chelsea, Michigan (C.P.T.) and Cambrium Group, Raleigh, North Carolina (E.W.H.)
| | - Eric W Harris
- CP Taylor Consulting, Chelsea, Michigan (C.P.T.) and Cambrium Group, Raleigh, North Carolina (E.W.H.)
| |
Collapse
|
183
|
Site of Action of Brain Neurosteroid Pregnenolone Sulfate at the N-Methyl-D-Aspartate Receptor. J Neurosci 2020; 40:5922-5936. [PMID: 32611707 DOI: 10.1523/jneurosci.3010-19.2020] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 06/09/2020] [Accepted: 06/17/2020] [Indexed: 12/19/2022] Open
Abstract
N-methyl-D-aspartate receptor (NMDAR) hypofunction has been implicated in several neurodevelopmental disorders. NMDAR function can be augmented by positive allosteric modulators, including endogenous compounds, such as cholesterol and neurosteroid pregnenolone sulfate (PES). Here we report that PES accesses the receptor via the membrane, and its binding site is different from that of cholesterol. Alanine mutagenesis has identified residues that disrupt the steroid potentiating effect at the rat GluN1 (G638; I642) and GluN2B (W559; M562; Y823; M824) subunit. Molecular dynamics simulation indicates that, in the absence of PES, the GluN2B M1 helix residue W559 interacts with the M4 helix residue M824. In the presence of PES, the M1 and M4 helices of agonist-activated receptor rearrange, forming a tighter interaction with the GluN1 M3 helix residues G638 and I642. This stabilizes the open-state position of the GluN1 M3 helices. Together, our data identify a likely binding site for the NMDAR-positive allosteric modulator PES and describe a novel molecular mechanism by which NMDAR activity can be augmented.SIGNIFICANCE STATEMENT There is considerable interest in drugs that enhance NMDAR function and could compensate for receptor hypofunction associated with certain neuropsychiatric disorders. Positive allosteric modulators of NMDARs include an endogenous neurosteroid pregnenolone sulfate (PES), but the binding site of PES on the NMDAR and the molecular mechanism of potentiation are unknown. We use patch-clamp electrophysiology in combination with mutagenesis and in silico modeling to describe the interaction of PES with the NMDAR. Our data indicate that PES binds to the transmembrane domain of the receptor at a discrete group of residues at the GluN2B membrane helices M1 and M4 and the GluN1 helix M3, and that PES potentiates NMDAR function by stabilizing the open-state position of the GluN1 M3 helices.
Collapse
|
184
|
Valente P, Kiryushko D, Sacchetti S, Machado P, Cobley CM, Mangini V, Porter AE, Spatz JP, Fleck RA, Benfenati F, Fiammengo R. Conopeptide-Functionalized Nanoparticles Selectively Antagonize Extrasynaptic N-Methyl-d-aspartate Receptors and Protect Hippocampal Neurons from Excitotoxicity In Vitro. ACS NANO 2020; 14:6866-6877. [PMID: 32510204 DOI: 10.1021/acsnano.0c00866] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
N-methyl-d-aspartate receptors (NMDARs) are ionotropic glutamate receptors controlling fundamental physiological processes in the central nervous system, such as learning and memory. Excessive activation of NMDARs causes excitotoxicity and results in neurodegeneration, which is observed in a number of pathological conditions. Because of their dichotomous role, therapeutic targeting of NMDAR is difficult. However, several lines of evidence suggest that excitotoxicity is predominantly linked to extrasynaptically located NMDARs. Here, we report on a nanoparticle-based strategy to inhibit extrasynaptic NMDARs exclusively and subtype selectively, while allowing synaptic NMDARs activity. We designed gold nanoparticles (AuNPs) carrying conopeptide derivatives conjugated on their poly(ethylene glycol) coating as allosteric NMDAR inhibitors and show that these nanoparticles antagonize exclusively extrasynaptic NMDAR-mediated currents in cultured hippocampal neurons. Additionally, we show that conopeptide-functionalized AuNPs are neuroprotective in an in vitro model of excitotoxicity. By using AuNPs carrying different allosteric inhibitors with distinct NMDAR subtype selectivity such as peptide conantokin-G or peptide conantokin-R, we suggest activation of extrasynaptic GluN2B-containing diheteromeric NMDARs as the main cause of excitotoxicity.
Collapse
Affiliation(s)
- Pierluigi Valente
- Department of Experimental Medicine, University of Genoa, Genoa 16132, Italy
- IRCSS Ospedale Policlinico San Martino, Genoa 16132, Italy
| | - Darya Kiryushko
- Department of Materials and London Center for Nanotechnology, Imperial College London, London SW7 2AZ, United Kingdom
- Center for Neuroinflammation and Neurodegeneration, Imperial College London, London W12 0NN, United Kingdom
| | - Silvio Sacchetti
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genoa 16132, Italy
| | - Pedro Machado
- Centre for Ultrastructural Imaging, Kings College London, London SE1 1UL, United Kingdom
| | - Claire M Cobley
- Department of Physical Chemistry, University of Heidelberg, Heidelberg 69120, Germany
- Max Planck Institute for Intelligent Systems, Stuttgart 70569, Germany
| | - Vincenzo Mangini
- Center for Biomolecular Nanotechnologies@UniLe, Istituto Italiano di Tecnologia, Arnesano, Lecce 73010, Italy
| | - Alexandra E Porter
- Department of Materials and London Center for Nanotechnology, Imperial College London, London SW7 2AZ, United Kingdom
| | - Joachim P Spatz
- Department of Physical Chemistry, University of Heidelberg, Heidelberg 69120, Germany
- Max Planck Institute for Medical Research, Heidelberg 69120, Germany
| | - Roland A Fleck
- Centre for Ultrastructural Imaging, Kings College London, London SE1 1UL, United Kingdom
| | - Fabio Benfenati
- IRCSS Ospedale Policlinico San Martino, Genoa 16132, Italy
- Center for Synaptic Neuroscience and Technology, Istituto Italiano di Tecnologia, Genoa 16132, Italy
| | - Roberto Fiammengo
- Center for Biomolecular Nanotechnologies@UniLe, Istituto Italiano di Tecnologia, Arnesano, Lecce 73010, Italy
| |
Collapse
|
185
|
Ehsan M, Katsube S, Cecchetti C, Du Y, Mortensen JS, Wang H, Nygaard A, Ghani L, Loland CJ, Kobilka BK, Byrne B, Guan L, Chae PS. New Malonate-Derived Tetraglucoside Detergents for Membrane Protein Stability. ACS Chem Biol 2020; 15:1697-1707. [PMID: 32501004 DOI: 10.1021/acschembio.0c00316] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Membrane proteins are widely studied in detergent micelles, a membrane-mimetic system formed by amphiphilic compounds. However, classical detergents have serious limitations in their utility, particularly for unstable proteins such as eukaryotic membrane proteins and membrane protein complexes, and thus, there is an unmet need for novel amphiphiles with enhanced ability to stabilize membrane proteins. Here, we developed a new class of malonate-derived detergents with four glucosides, designated malonate-derived tetra-glucosides (MTGs), and compared these new detergents with previously reported octyl glucose neopentyl glycol (OGNG) and n-dodecyl-β-d-maltoside (DDM). When tested with two G-protein coupled receptors (GPCRs) and three transporters, a couple of MTGs consistently conferred enhanced stability to all tested proteins compared to DDM and OGNG. As a result of favorable behaviors for a range of membrane proteins, these MTGs have substantial potential for membrane protein research. This study additionally provides a new detergent design principle based on the effect of a polar functional group (i.e., ether) on protein stability depending on its position in the detergent scaffold.
Collapse
Affiliation(s)
- Muhammad Ehsan
- Department of Bionanotechnology, Hanyang University, Ansan, 15588, Korea
| | - Satoshi Katsube
- Department of Cell Physiology and Molecular Biophysics, Center for Membrane Protein Research, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas 79430, United States
| | - Cristina Cecchetti
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, United Kingdom
| | - Yang Du
- School of Life and Health Sciences, Kobilka Institute of Innovative Drug Discovery, Chinese University of Hong Kong, 2001 Longxiang Avenue, Shenzhen, Guangdong 518172, China
| | - Jonas S. Mortensen
- Department of Neuroscience, University of Copenhagen, Copenhagen, DK-2200, Denmark
| | - Haoqing Wang
- Molecular and Cellular Physiology, Stanford University, Stanford, California 94305, United States
| | - Andreas Nygaard
- Department of Neuroscience, University of Copenhagen, Copenhagen, DK-2200, Denmark
| | - Lubna Ghani
- Department of Bionanotechnology, Hanyang University, Ansan, 15588, Korea
| | - Claus J. Loland
- Department of Neuroscience, University of Copenhagen, Copenhagen, DK-2200, Denmark
| | - Brian K. Kobilka
- Molecular and Cellular Physiology, Stanford University, Stanford, California 94305, United States
| | - Bernadette Byrne
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, United Kingdom
| | - Lan Guan
- Department of Cell Physiology and Molecular Biophysics, Center for Membrane Protein Research, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas 79430, United States
| | - Pil Seok Chae
- Department of Bionanotechnology, Hanyang University, Ansan, 15588, Korea
| |
Collapse
|
186
|
Perszyk RE, Myers SJ, Yuan H, Gibb AJ, Furukawa H, Sobolevsky AI, Traynelis SF. Hodgkin-Huxley-Katz Prize Lecture: Genetic and pharmacological control of glutamate receptor channel through a highly conserved gating motif. J Physiol 2020; 598:3071-3083. [PMID: 32468591 DOI: 10.1113/jp278086] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 05/18/2020] [Indexed: 12/15/2022] Open
Abstract
Glutamate receptors are essential ligand-gated ion channels in the central nervous system that mediate excitatory synaptic transmission in response to the release of glutamate from presynaptic terminals. The structural and biophysical basis underlying the function of these receptors has been studied for decades by a wide range of approaches. However recent structural, pharmacological and genetic studies have provided new insight into the regions of this protein that are critical determinants of receptor function. Lack of variation in specific areas of the protein amino acid sequences in the human population has defined three regions in each receptor subunit that are under selective pressure, which has focused research efforts and driven new hypotheses. In addition, these three closely positioned elements reside near a cavity that is shown by multiple studies to be a likely site of action for allosteric modulators, one of which is currently in use as an FDA-approved anticonvulsant. These structural elements are capable of controlling gating of the pore, and appear to permit some modulators bound within the cavity to also alter permeation properties. This creates a new precedent whereby features of the channel pore can be modulated by exogenous drugs that bind outside the pore. The convergence of structural, genetic, biophysical and pharmacological approaches is a powerful means to gain insight into the complex biological processes defined by neurotransmitter receptor function.
Collapse
Affiliation(s)
- Riley E Perszyk
- 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
| | - 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
| | - Alasdair J Gibb
- Department of Neuroscience, Physiology & Pharmacology, University College London, London, UK
| | - Hiro Furukawa
- WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, New York, NY, 11724, USA
| | - Alexander I Sobolevsky
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, 10032, 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
| |
Collapse
|
187
|
Warnet XL, Bakke Krog H, Sevillano-Quispe OG, Poulsen H, Kjaergaard M. The C-terminal domains of the NMDA receptor: How intrinsically disordered tails affect signalling, plasticity and disease. Eur J Neurosci 2020; 54:6713-6739. [PMID: 32464691 DOI: 10.1111/ejn.14842] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 04/16/2020] [Accepted: 05/18/2020] [Indexed: 01/14/2023]
Abstract
NMDA receptors are part of the ionotropic glutamate receptor family, and are crucial for neurotransmission and memory. At the cellular level, the effects of activating these receptors include long-term potentiation (LTP) or depression (LTD). The NMDA receptor is a stringently gated cation channel permeable to Ca2+ , and it shares the molecular architecture of a tetrameric ligand-gated ion channel with the other family members. Its subunits, however, have uniquely long cytoplasmic C-terminal domains (CTDs). While the molecular gymnastics of the extracellular domains have been described in exquisite detail, much less is known about the structure and function of these CTDs. The CTDs vary dramatically in length and sequence between receptor subunits, but they all have a composition characteristic of intrinsically disordered proteins. The CTDs affect channel properties, trafficking and downstream signalling output from the receptor, and these functions are regulated by alternative splicing, protein-protein interactions, and post-translational modifications such as phosphorylation and palmitoylation. Here, we review the roles of the CTDs in synaptic plasticity with a focus on biochemical mechanisms. In total, the CTDs play a multifaceted role as a modifier of channel function, a regulator of cellular location and abundance, and signalling scaffold control the downstream signalling output.
Collapse
Affiliation(s)
- Xavier L Warnet
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark.,The Danish Research Institute for Translational Neuroscience (DANDRITE), Aarhus University, Aarhus, Denmark.,The Center for Proteins in Memory (PROMEMO), Aarhus University, Aarhus, Denmark
| | - Helle Bakke Krog
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark.,The Danish Research Institute for Translational Neuroscience (DANDRITE), Aarhus University, Aarhus, Denmark.,The Center for Proteins in Memory (PROMEMO), Aarhus University, Aarhus, Denmark
| | - Oscar G Sevillano-Quispe
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark.,The Danish Research Institute for Translational Neuroscience (DANDRITE), Aarhus University, Aarhus, Denmark.,The Center for Proteins in Memory (PROMEMO), Aarhus University, Aarhus, Denmark
| | - Hanne Poulsen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark.,The Danish Research Institute for Translational Neuroscience (DANDRITE), Aarhus University, Aarhus, Denmark.,The Center for Proteins in Memory (PROMEMO), Aarhus University, Aarhus, Denmark
| | - Magnus Kjaergaard
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark.,The Danish Research Institute for Translational Neuroscience (DANDRITE), Aarhus University, Aarhus, Denmark.,The Center for Proteins in Memory (PROMEMO), Aarhus University, Aarhus, Denmark
| |
Collapse
|
188
|
Shin W, Kim K, Serraz B, Cho YS, Kim D, Kang M, Lee EJ, Lee H, Bae YC, Paoletti P, Kim E. Early correction of synaptic long-term depression improves abnormal anxiety-like behavior in adult GluN2B-C456Y-mutant mice. PLoS Biol 2020; 18:e3000717. [PMID: 32353004 PMCID: PMC7217483 DOI: 10.1371/journal.pbio.3000717] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 05/12/2020] [Accepted: 04/15/2020] [Indexed: 01/28/2023] Open
Abstract
Extensive evidence links Glutamate receptor, ionotropic, NMDA2B (GRIN2B), encoding the GluN2B/NR2B subunit of N-methyl-D-aspartate receptors (NMDARs), with various neurodevelopmental disorders, including autism spectrum disorders (ASDs), but the underlying mechanisms remain unclear. In addition, it remains unknown whether mutations in GluN2B, which starts to be expressed early in development, induces early pathophysiology that can be corrected by early treatments for long-lasting effects. We generated and characterized Grin2b-mutant mice that carry a heterozygous, ASD-risk C456Y mutation (Grin2b+/C456Y). In Grin2b+/C456Y mice, GluN2B protein levels were strongly reduced in association with decreased hippocampal NMDAR currents and NMDAR-dependent long-term depression (LTD) but unaltered long-term potentiation, indicative of mutation-induced protein degradation and LTD sensitivity. Behaviorally, Grin2b+/C456Y mice showed normal social interaction but exhibited abnormal anxiolytic-like behavior. Importantly, early, but not late, treatment of young Grin2b+/C456Y mice with the NMDAR agonist D-cycloserine rescued NMDAR currents and LTD in juvenile mice and improved anxiolytic-like behavior in adult mice. Therefore, GluN2B-C456Y haploinsufficiency decreases GluN2B protein levels, NMDAR-dependent LTD, and anxiety-like behavior, and early activation of NMDAR function has long-lasting effects on adult mouse behavior.
Collapse
Affiliation(s)
- Wangyong Shin
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Kyungdeok Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Benjamin Serraz
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole Normale Supérieure, Université PSL, CNRS, INSERM, Paris, France
| | - Yi Sul Cho
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu, Korea
| | - Doyoun Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, Korea
| | - Muwon Kang
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Eun-Jae Lee
- Department of Neurology, Asan Medical Center, University of Ulsan, College of Medicine, Seoul, Korea
| | - Hyejin Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Yong Chul Bae
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu, Korea
| | - Pierre Paoletti
- Institut de Biologie de l'Ecole Normale Supérieure (IBENS), Ecole Normale Supérieure, Université PSL, CNRS, INSERM, Paris, France
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, Korea
- * E-mail:
| |
Collapse
|
189
|
Thum S, Schepmann D, Reinoso RF, Alvarez I, Ametamey SM, Wünsch B. Synthesis and pharmacological evaluation of fluorinated benzo[7]annulen-7-amines as GluN2B-selective NMDA receptor antagonists. J Labelled Comp Radiopharm 2020; 62:354-379. [PMID: 30850991 DOI: 10.1002/jlcr.3718] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/24/2019] [Accepted: 02/28/2019] [Indexed: 01/30/2023]
Abstract
Because of their neuroprotective potential, GluN2B-selective ligands are of great interest for the treatment of various neurological and neurodegenerative disorders. Fluorinated benzo[7]annulen-7-amines, capable for PET, were synthesized by combining fluorinated phenylalkylamines with differently substituted ketones. Relationships between substitution pattern and GluN2B affinity as well as selectivity towards σ1 and σ2 receptors were investigated. Two promising ligands (18a and 20c) were selected for further pharmacological evaluation. Besides a slight serotonin transporter (SERT), norepinephrine transporter (NET), and hERG affinity, they did not show interaction with other targets. Furthermore, the pKa value of a set fluorinated ligands, bearing the fluorine atom in different positions, was determined.
Collapse
Affiliation(s)
- Simone Thum
- Institute of Pharmaceutical and Medicinal Chemistry, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Dirk Schepmann
- Institute of Pharmaceutical and Medicinal Chemistry, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | | | | | - Simon M Ametamey
- Center for Radiopharmaceutical Science of ETH, PSI, and USZ, ETH-Hönggerberg, Zürich, Switzerland
| | - Bernhard Wünsch
- Institute of Pharmaceutical and Medicinal Chemistry, Westfälische Wilhelms-Universität Münster, Münster, Germany.,Cells-in-Motion Cluster of Excellence (EXC 1003-CiM), Westfälische Wilhelms-Universität Münster, Germany
| |
Collapse
|
190
|
Amin JB, Moody GR, Wollmuth LP. From bedside-to-bench: What disease-associated variants are teaching us about the NMDA receptor. J Physiol 2020; 599:397-416. [PMID: 32144935 DOI: 10.1113/jp278705] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 01/21/2020] [Indexed: 12/25/2022] Open
Abstract
NMDA receptors (NMDARs) are glutamate-gated ion channels that contribute to nearly all brain processes. Not surprisingly then, genetic variations in the genes encoding NMDAR subunits can be associated with neurodevelopmental, neurological and psychiatric disorders. These disease-associated variants (DAVs) present challenges, such as defining how DAV-induced alterations in receptor function contribute to disease progression and how to treat the affected individual clinically. As a starting point to overcome these challenges, we need to refine our understanding of the complexity of NMDAR structure function. In this regard, DAVs have expanded our knowledge of NMDARs because they do not just target well-known structure-function motifs, but rather give an unbiased view of structural elements that are important to the biology of NMDARs. Indeed, established NMDAR structure-function motifs have been validated by the appearance of disorders in patients where these motifs have been altered, and DAVs have identified novel structural features in NMDARs such as gating triads and hinges in the gating machinery. Still, the majority of DAVs remain unexplored and occur at sites in the protein with unidentified function or alter receptor properties in multiple and unanticipated ways. Detailed mechanistic and structural investigations are required of both established and novel motifs to develop a highly refined pathomechanistic model that accounts for the complex machinery that regulates NMDARs. Such a model would provide a template for rational drug design and a starting point for personalized medicine.
Collapse
Affiliation(s)
- Johansen B Amin
- Medical Scientist Training Program (MSTP), Stony Brook University, Stony Brook, NY, 11794-5230.,Graduate Program in Molecular and Cellular Pharmacology, Stony Brook University, Stony Brook, NY, 11794-5230
| | - Gabrielle R Moody
- Graduate Program in Molecular and Cellular Pharmacology, Stony Brook University, Stony Brook, NY, 11794-5230
| | - Lonnie P Wollmuth
- Department of Neurobiology & Behavior, Stony Brook University, Stony Brook, NY, 11794-5230.,Department of Biochemistry & Cell Biology, Stony Brook University, Stony Brook, NY, 11794-5230.,Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY, 11794-5230
| |
Collapse
|
191
|
Durham RJ, Paudyal N, Carrillo E, Bhatia NK, Maclean DM, Berka V, Dolino DM, Gorfe AA, Jayaraman V. Conformational spread and dynamics in allostery of NMDA receptors. Proc Natl Acad Sci U S A 2020; 117:3839-3847. [PMID: 32015122 PMCID: PMC7035515 DOI: 10.1073/pnas.1910950117] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Allostery can be manifested as a combination of repression and activation in multidomain proteins allowing for fine tuning of regulatory mechanisms. Here we have used single molecule fluorescence resonance energy transfer (smFRET) and molecular dynamics simulations to study the mechanism of allostery underlying negative cooperativity between the two agonists glutamate and glycine in the NMDA receptor. These data show that binding of one agonist leads to conformational flexibility and an increase in conformational spread at the second agonist site. Mutational and cross-linking studies show that the dimer-dimer interface at the agonist-binding domain mediates the allostery underlying the negative cooperativity. smFRET on the transmembrane segments shows that they are tightly coupled in the unliganded and single agonist-bound form and only upon binding both agonists the transmembrane domain explores looser packing which would facilitate activation.
Collapse
Affiliation(s)
- Ryan J Durham
- Center for Membrane Biology, Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston, Houston, TX 77030
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Nabina Paudyal
- Center for Membrane Biology, Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston, Houston, TX 77030
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Elisa Carrillo
- Center for Membrane Biology, Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Nidhi Kaur Bhatia
- Center for Membrane Biology, Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - David M Maclean
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY 14642
| | - Vladimir Berka
- Center for Membrane Biology, Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Drew M Dolino
- Center for Membrane Biology, Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston, Houston, TX 77030
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Alemayehu A Gorfe
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center at Houston, Houston, TX 77030
| | - Vasanthi Jayaraman
- Center for Membrane Biology, Department of Biochemistry and Molecular Biology, University of Texas Health Science Center at Houston, Houston, TX 77030;
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX 77030
| |
Collapse
|
192
|
Rajani V, Sengar AS, Salter MW. Tripartite signalling by NMDA receptors. Mol Brain 2020; 13:23. [PMID: 32070387 PMCID: PMC7029596 DOI: 10.1186/s13041-020-0563-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 02/03/2020] [Indexed: 12/31/2022] Open
Abstract
N-methyl-d-aspartate receptors (NMDARs) are excitatory glutamatergic receptors that are fundamental for many neuronal processes, including synaptic plasticity. NMDARs are comprised of four subunits derived from heterogeneous subunit families, yielding a complex diversity in NMDAR form and function. The quadruply-liganded state of binding of two glutamate and two glycine molecules to the receptor drives channel gating, allowing for monovalent cation flux, Ca2+ entry and the initiation of Ca2+-dependent signalling. In addition to this ionotropic function, non-ionotropic signalling can be initiated through the exclusive binding of glycine or of glutamate to the NMDAR. This binding may trigger a transmembrane conformational change of the receptor, inducing intracellular protein-protein signalling between the cytoplasmic domain and secondary messengers. In this review, we outline signalling cascades that can be activated by NMDARs and propose that the receptor transduces signalling through three parallel streams: (i) signalling via both glycine and glutamate binding, (ii) signalling via glycine binding, and (iii) signalling via glutamate binding. This variety in signal transduction mechanisms and downstream signalling cascades complements the widespread prevalence and rich diversity of NMDAR activity throughout the central nervous system and in disease pathology.
Collapse
Affiliation(s)
- Vishaal Rajani
- Program in Neurosciences & Mental Health, Hospital for Sick Children, Toronto, Ontario, M5G 1X8, Canada
| | - Ameet S Sengar
- Program in Neurosciences & Mental Health, Hospital for Sick Children, Toronto, Ontario, M5G 1X8, Canada
| | - Michael W Salter
- Program in Neurosciences & Mental Health, Hospital for Sick Children, Toronto, Ontario, M5G 1X8, Canada. .,Department of Physiology, University of Toronto, Toronto, Ontario, M5S 1A8, Canada.
| |
Collapse
|
193
|
Molecular docking utilising the OliveNet™ library reveals novel phenolic compounds which may potentially target key proteins associated with major depressive disorder. Comput Biol Chem 2020; 86:107234. [PMID: 32220809 DOI: 10.1016/j.compbiolchem.2020.107234] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 01/31/2020] [Accepted: 02/10/2020] [Indexed: 11/21/2022]
Abstract
The antidepressant medications that are currently prescribed to patients suffering from major depressive disorder (MDD) have limitations and as a result, there is an urgent need to increase the options that are available. A number of studies have found that natural polyphenols have neuroprotective properties and there is evidence to suggest that they modulate neurotransmitter systems. There are more than 200 phenolic compounds that have been identified in Olea europaea, many of which have not yet been investigated for their potential biological effects. In this study, in silico methods were used to screen the phenolic library from the OliveNet™ database and identify novel lead compounds for proteins implicated in the pathophysiology of MDD. The molecular docking results revealed that the monoamine oxidase enzyme isoforms (MAO-A/MAO-B) had binding specificities for certain phenolic subclasses. The lead ligands that were identified from these subclasses were positioned near the flavin adenine dinucleotide (FAD) cofactor, interacting in a similar manner as known inhibitors. In addition to the MAO enzymes, several phenolic compounds were docked to neurotransmitter transporters and postsynaptic receptors, as well as proteins involved in neuroinflammation, oxidative stress and the endocannabinoid system. Based on the binding affinity, position, orientation and interactions of the lead phenolic compounds identified in this study, it is predicted that they may have antidepressant properties. The results should be validated further using molecular dynamics (MD) simulations, as well as in vivo and in vitro techniques.
Collapse
|
194
|
Perszyk RE, Swanger SA, Shelley C, Khatri A, Fernandez-Cuervo G, Epplin MP, Zhang J, Le P, Bülow P, Garnier-Amblard E, Gangireddy PKR, Bassell GJ, Yuan H, Menaldino DS, Liotta DC, Liebeskind LS, Traynelis SF. Biased modulators of NMDA receptors control channel opening and ion selectivity. Nat Chem Biol 2020; 16:188-196. [PMID: 31959964 PMCID: PMC6986806 DOI: 10.1038/s41589-019-0449-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 11/08/2019] [Accepted: 12/04/2019] [Indexed: 12/26/2022]
Abstract
Allosteric modulators of ion channels typically alter the transitions rates between conformational states without changing the properties of the open pore. Here we describe a new class of positive allosteric modulators of N-methyl D-aspartate receptors (NMDARs) that mediate a calcium-permeable component of glutamatergic synaptic transmission and play essential roles in learning, memory and cognition, as well as neurological disease. EU1622-14 increases agonist potency and channel-open probability, slows receptor deactivation and decreases both single-channel conductance and calcium permeability. The unique functional selectivity of this chemical probe reveals a mechanism for enhancing NMDAR function while limiting excess calcium influx, and shows that allosteric modulators can act as biased modulators of ion-channel permeation.
Collapse
Affiliation(s)
- Riley E. Perszyk
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322
| | - Sharon A. Swanger
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322,Virginia Tech Carilion Research Institute, Roanoke, VA 24016; Department of Internal Medicine, Virginia Tech Carilion School of Medicine, Roanoke, VA 24016; Department of Biomedical Sciences and Pathobiology, Virginia-Maryland School of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061
| | - Chris Shelley
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322,Department of Biology, University of the South, Sewanee, TN 37383
| | - Alpa Khatri
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322
| | | | | | - Jing Zhang
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322
| | - Phuong Le
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322
| | - Pernille Bülow
- Department of Physiology, Emory University, Atlanta, GA 30322,Department of Cell Biology, Emory University, Atlanta, GA 30322
| | | | | | - Gary J. Bassell
- Department of Cell Biology, Emory University, Atlanta, GA 30322
| | - Hongjie Yuan
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322
| | | | | | | | - Stephen F. Traynelis
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA 30322,Correspondence to: Stephen F. Traynelis, 1510 Clifton Rd NE, Atlanta, GA, 30322, , 404-727-0357
| |
Collapse
|
195
|
Washburn HR, Xia NL, Zhou W, Mao YT, Dalva MB. Positive surface charge of GluN1 N-terminus mediates the direct interaction with EphB2 and NMDAR mobility. Nat Commun 2020; 11:570. [PMID: 31996679 PMCID: PMC6989673 DOI: 10.1038/s41467-020-14345-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 12/19/2019] [Indexed: 12/31/2022] Open
Abstract
Localization of the N-methyl-D-aspartate type glutamate receptor (NMDAR) to dendritic spines is essential for excitatory synaptic transmission and plasticity. Rather than remaining trapped at synaptic sites, NMDA receptors undergo constant cycling into and out of the postsynaptic density. Receptor movement is constrained by protein-protein interactions with both the intracellular and extracellular domains of the NMDAR. The role of extracellular interactions on the mobility of the NMDAR is poorly understood. Here we demonstrate that the positive surface charge of the hinge region of the N-terminal domain in the GluN1 subunit of the NMDAR is required to maintain NMDARs at dendritic spine synapses and mediates the direct extracellular interaction with a negatively charged phospho-tyrosine on the receptor tyrosine kinase EphB2. Loss of the EphB-NMDAR interaction by either mutating GluN1 or knocking down endogenous EphB2 increases NMDAR mobility. These findings begin to define a mechanism for extracellular interactions mediated by charged domains.
Collapse
Affiliation(s)
- Halley R Washburn
- Department of Neuroscience and Jefferson Center for Synaptic Biology, Thomas Jefferson University, 233 South 10th Street, Bluemle Life Sciences Building, Room 324, Philadelphia, PA, 19107, USA
| | - Nan L Xia
- Department of Neuroscience and Jefferson Center for Synaptic Biology, Thomas Jefferson University, 233 South 10th Street, Bluemle Life Sciences Building, Room 324, Philadelphia, PA, 19107, USA
- Department of Neurobiology, University of Chicago, 924 East 57th street, JFKR212, Chicago, IL, 60637, USA
| | - Wei Zhou
- Department of Neuroscience and Jefferson Center for Synaptic Biology, Thomas Jefferson University, 233 South 10th Street, Bluemle Life Sciences Building, Room 324, Philadelphia, PA, 19107, USA
| | - Yu-Ting Mao
- Department of Neuroscience and Jefferson Center for Synaptic Biology, Thomas Jefferson University, 233 South 10th Street, Bluemle Life Sciences Building, Room 324, Philadelphia, PA, 19107, USA
| | - Matthew B Dalva
- Department of Neuroscience and Jefferson Center for Synaptic Biology, Thomas Jefferson University, 233 South 10th Street, Bluemle Life Sciences Building, Room 324, Philadelphia, PA, 19107, USA.
| |
Collapse
|
196
|
Structure and assembly of calcium homeostasis modulator proteins. Nat Struct Mol Biol 2020; 27:150-159. [PMID: 31988524 PMCID: PMC7015811 DOI: 10.1038/s41594-019-0369-9] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 12/23/2019] [Indexed: 01/17/2023]
Abstract
The biological membranes of many cell types contain large-pore channels through which a wide variety of ions and metabolites permeate. Examples include connexin, innexin and pannexin, which form gap junctions and/or bona fide cell surface channels. The most recently identified large-pore channels are the calcium homeostasis modulators (CALHMs), through which ions and ATP permeate in a voltage-dependent manner to control neuronal excitability, taste signaling and pathologies of depression and Alzheimer's disease. Despite such critical biological roles, the structures and patterns of their oligomeric assembly remain unclear. Here, we reveal the structures of two CALHMs, chicken CALHM1 and human CALHM2, by single-particle cryo-electron microscopy (cryo-EM), which show novel assembly of the four transmembrane helices into channels of octamers and undecamers, respectively. Furthermore, molecular dynamics simulations suggest that lipids can favorably assemble into a bilayer within the larger CALHM2 pore, but not within CALHM1, demonstrating the potential correlation between pore size, lipid accommodation and channel activity.
Collapse
|
197
|
Yi F, Rouzbeh N, Hansen KB, Xu Y, Fanger CM, Gordon E, Paschetto K, Menniti FS, Volkmann RA. PTC-174, a positive allosteric modulator of NMDA receptors containing GluN2C or GluN2D subunits. Neuropharmacology 2020; 173:107971. [PMID: 31987864 DOI: 10.1016/j.neuropharm.2020.107971] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 01/14/2020] [Accepted: 01/16/2020] [Indexed: 01/14/2023]
Abstract
NMDA receptors are ionotropic glutamate receptors that mediate excitatory neurotransmission. The diverse functions of these receptors are tuned by deploying different combinations of GluN1 and GluN2 subunits (GluN2A-D) to form either diheteromeric NMDA receptors, which contain two GluN1 and two identical GluN2 subunits, or triheteromeric NMDA receptors, which contain two GluN1 and two distinct GluN2 subunits. Here, we characterize PTC-174, a novel positive allosteric modulator (PAM) of receptors containing GluN2C or GluN2D subunits. PTC-174 potentiates maximal current amplitudes by 1.8-fold for diheteromeric GluN1/2B receptors and by > 10-fold for GluN1/2C and GluN1/2D receptors. PTC-174 also potentiates responses from triheteromeric GluN1/2B/2D and GluN1/2A/2C receptors by 4.5-fold and 1.7-fold, respectively. By contrast, PTC-174 produces partial inhibition of responses from diheteromeric GluN1/2A and triheteromeric GluN1/2A/2B receptors. PTC-174 increases potencies of co-agonists glutamate and glycine by 2- to 5-fold at GluN1/2C and GluN1/2D receptors, and NMDA receptor activation facilitates allosteric modulation by PTC-174. At native NMDA receptors in GluN2D-expressing subthalamic nucleus neurons, PTC-174 increases the amplitude of responses to NMDA application and slows the decay of excitatory postsynaptic currents (EPSCs) evoked by internal capsule stimulation. Furthermore, PTC-174 increases the amplitude and slows the decay of EPSCs in hippocampal interneurons, but has not effect on the amplitudes of NMDA receptor-mediated EPSCs in hippocampal CA1 pyramidal neurons. Thus, PTC-174 provides a useful new pharmacological tool to investigate the molecular pharmacology and physiology of GluN2C- and GluN2D-containing NMDA receptors.
Collapse
Affiliation(s)
- Feng Yi
- Department of Biomedical and Pharmaceutical Sciences, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, University of Montana, Missoula, MT, 59812, USA
| | - Nirvan Rouzbeh
- Department of Biomedical and Pharmaceutical Sciences, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, University of Montana, Missoula, MT, 59812, USA
| | - Kasper B Hansen
- Department of Biomedical and Pharmaceutical Sciences, Center for Biomolecular Structure and Dynamics, Center for Structural and Functional Neuroscience, University of Montana, Missoula, MT, 59812, USA
| | - Yuelian Xu
- Chinglu Pharmaceutical Research LLC, Newington, CT, 06111, USA
| | | | - Earl Gordon
- Reaction Biology Corporation, Malvern, PA, 19355, USA
| | - Kathy Paschetto
- Jubilant Discovery Services, Inc. 365 Phoenixville Pike, Malvern, PA, 19355, USA
| | - Frank S Menniti
- The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, 02881, USA.
| | | |
Collapse
|
198
|
Wang JX, Irvine MW, Burnell ES, Sapkota K, Thatcher RJ, Li M, Simorowski N, Volianskis A, Collingridge GL, Monaghan DT, Jane DE, Furukawa H. Structural basis of subtype-selective competitive antagonism for GluN2C/2D-containing NMDA receptors. Nat Commun 2020; 11:423. [PMID: 31969570 PMCID: PMC6976569 DOI: 10.1038/s41467-020-14321-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 12/18/2019] [Indexed: 12/15/2022] Open
Abstract
N-Methyl-D-aspartate receptors (NMDARs) play critical roles in the central nervous system. Their heterotetrameric composition generates subtypes with distinct functional properties and spatio-temporal distribution in the brain, raising the possibility for subtype-specific targeting by pharmacological means for treatment of neurological diseases. While specific compounds for GluN2A and GluN2B-containing NMDARs are well established, those that target GluN2C and GluN2D are currently underdeveloped with low potency and uncharacterized binding modes. Here, using electrophysiology and X-ray crystallography, we show that UBP791 ((2S*,3R*)-1-(7-(2-carboxyethyl)phenanthrene-2-carbonyl)piperazine-2,3-dicarboxylic acid) inhibits GluN2C/2D with 40-fold selectivity over GluN2A-containing receptors, and that a methionine and a lysine residue in the ligand binding pocket (GluN2D-Met763/Lys766, GluN2C-Met736/Lys739) are the critical molecular elements for the subtype-specific binding. These findings led to development of UBP1700 ((2S*,3R*)-1-(7-(2-carboxyvinyl)phenanthrene-2-carbonyl)piperazine-2,3-dicarboxylic acid) which shows over 50-fold GluN2C/2D-selectivity over GluN2A with potencies in the low nanomolar range. Our study shows that the L-glutamate binding site can be targeted for GluN2C/2D-specific inhibition.
Collapse
Affiliation(s)
- Jue Xiang Wang
- WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
- Watson School of Biological Sciences, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| | - Mark W Irvine
- Glutamate Research Group, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK
| | - Erica S Burnell
- Glutamate Research Group, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK
- School of Chemistry, National University of Ireland Galway, Galway, H91TK33, Ireland
| | - Kiran Sapkota
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5800, USA
| | - Robert J Thatcher
- Glutamate Research Group, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK
| | - Minjun Li
- WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| | - Noriko Simorowski
- WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| | - Arturas Volianskis
- Centre for Neuroscience and Trauma, Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Graham L Collingridge
- Glutamate Research Group, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK
- Tanz Centre for Research in Neurodegenerative Diseases, Department of Physiology, University of Toronto, Krembil Discovery Tower, 60 Leonard Avenue, Toronto, ON, M5T 0S8, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, 600 University Avenue, Toronto, ON, M5G 1X5, Canada
| | - Daniel T Monaghan
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5800, USA
| | - David E Jane
- Glutamate Research Group, School of Physiology, Pharmacology and Neuroscience, University of Bristol, Biomedical Sciences Building, University Walk, Bristol, BS8 1TD, UK.
| | - Hiro Furukawa
- WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA.
- Watson School of Biological Sciences, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA.
| |
Collapse
|
199
|
Template-free detection and classification of membrane-bound complexes in cryo-electron tomograms. Nat Methods 2020; 17:209-216. [PMID: 31907446 DOI: 10.1038/s41592-019-0675-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 11/11/2019] [Indexed: 01/12/2023]
Abstract
With faithful sample preservation and direct imaging of fully hydrated biological material, cryo-electron tomography provides an accurate representation of molecular architecture of cells. However, detection and precise localization of macromolecular complexes within cellular environments is aggravated by the presence of many molecular species and molecular crowding. We developed a template-free image processing procedure for accurate tracing of complex networks of densities in cryo-electron tomograms, a comprehensive and automated detection of heterogeneous membrane-bound complexes and an unsupervised classification (PySeg). Applications to intact cells and isolated endoplasmic reticulum (ER) allowed us to detect and classify small protein complexes. This classification provided sufficiently homogeneous particle sets and initial references to allow subsequent de novo subtomogram averaging. Spatial distribution analysis showed that ER complexes have different localization patterns forming nanodomains. Therefore, this procedure allows a comprehensive detection and structural analysis of complexes in situ.
Collapse
|
200
|
GRIN2D/GluN2D NMDA receptor: Unique features and its contribution to pediatric developmental and epileptic encephalopathy. Eur J Paediatr Neurol 2020; 24:89-99. [PMID: 31918992 PMCID: PMC7035963 DOI: 10.1016/j.ejpn.2019.12.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 12/06/2019] [Indexed: 12/14/2022]
Abstract
N-methyl-d-aspartate receptors (NMDARs), a subset of ligand-gated ionotropic glutamate receptors, are critical for learning, memory, and neuronal development. However, when NMDAR subunits are mutated, a host of neuropathological conditions can occur, including epilepsy. Recently, genetic variation within the GRIN2D gene, which encodes the GluN2D subunit of the NMDAR, has been associated with a set of early-onset neurological diseases, notably developmental and epileptic encephalopathy (DEE). Importantly, patients with GRIN2D variants are largely refractory to conventional anti-epileptic drug (AED) treatment, highlighting the need to further understand the distinctive characteristics of GluN2D in neurological and pathological functions. In this review, we first summarize GluN2D's unique spatial and temporal expression patterns, electrophysiological profiles, and contributions to both pre- and postsynaptic signaling. Next, we review thirteen unique case studies from DEE patients harboring ten different causal GRIN2D variants. These patients are highly heterogenous, manifesting multiple seizure types, electroencephalographic recordings, and neurological and developmental outcomes. Lastly, this review concludes by highlighting the difficulty in treating patients with DEE-associated GRIN2D variants, and stresses the need for selective therapeutic agents delivered within a precise time window.
Collapse
|