101
|
Lei N, Mellem JE, Brockie PJ, Madsen DM, Maricq AV. NRAP-1 Is a Presynaptically Released NMDA Receptor Auxiliary Protein that Modifies Synaptic Strength. Neuron 2017; 96:1303-1316.e6. [DOI: 10.1016/j.neuron.2017.11.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 10/20/2017] [Accepted: 11/14/2017] [Indexed: 12/19/2022]
|
102
|
Prom-Wormley EC, Ebejer J, Dick DM, Bowers MS. The genetic epidemiology of substance use disorder: A review. Drug Alcohol Depend 2017; 180:241-259. [PMID: 28938182 PMCID: PMC5911369 DOI: 10.1016/j.drugalcdep.2017.06.040] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 06/20/2017] [Accepted: 06/23/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND Substance use disorder (SUD) remains a significant public health issue. A greater understanding of how genes and environment interact to regulate phenotypes comprising SUD will facilitate directed treatments and prevention. METHODS The literature studying the neurobiological correlates of SUD with a focus on the genetic and environmental influences underlying these mechanisms was reviewed. Results from twin/family, human genetic association, gene-environment interaction, epigenetic literature, phenome-wide association studies are summarized for alcohol, nicotine, cannabinoids, cocaine, and opioids. RESULTS There are substantial genetic influences on SUD that are expected to influence multiple neurotransmission pathways, and these influences are particularly important within the dopaminergic system. Genetic influences involved in other aspects of SUD etiology including drug processing and metabolism are also identified. Studies of gene-environment interaction emphasize the importance of environmental context in SUD. Epigenetic studies indicate drug-specific changes in gene expression as well as differences in gene expression related to the use of multiple substances. Further, gene expression is expected to differ by stage of SUD such as substance initiation versus chronic substance use. While a substantial literature has developed for alcohol and nicotine use disorders, there is comparatively less information for other commonly abused substances. CONCLUSIONS A better understanding of genetically-mediated mechanisms involved in the neurobiology of SUD provides increased opportunity to develop behavioral and biologically based treatment and prevention of SUD.
Collapse
Affiliation(s)
- Elizabeth C Prom-Wormley
- Dvision of Epidemiology, Department of Family Medicine and Population Health, Virginia Commonwealth University, PO Box 980212, Richmond, VA 23298-0212, USA.
| | - Jane Ebejer
- School of Cognitive Behavioural and Social Sciences, University of New England, Armidale, NSW 2350, Australia
| | - Danielle M Dick
- Department of Psychology, Virginia Commonwealth University, PO Box 842509, Richmond, VA 23284-2509, USA
| | - M Scott Bowers
- Faulk Center for Molecular Therapeutics, Biomedical Engeneering, Northwestern University, Evanston, IL 60201, USA
| |
Collapse
|
103
|
Passlick S, Ellis-Davies GCR. Comparative one- and two-photon uncaging of MNI-glutamate and MNI-kainate on hippocampal CA1 neurons. J Neurosci Methods 2017; 293:321-328. [PMID: 29051090 DOI: 10.1016/j.jneumeth.2017.10.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 10/13/2017] [Accepted: 10/16/2017] [Indexed: 11/25/2022]
Abstract
BACKGROUND The light-induced release of neurotransmitters from caging chromophores provides a powerful means to study the underlying receptors in a physiologically relevant context. Surprisingly, most caged neurotransmitters, including the widely used 4-methoxy-7-nitroindolinyl (MNI)-glutamate, show strong antagonism against GABA-A receptors. Kainate has been shown to exhibit a higher efficacy at glutamate receptors compared to glutamate itself. Thus, uncaging of kainate might allow the application of the caged compound at lower, less antagonistic concentrations. NEW METHODS This study provides a detailed comparison of MNI-glutamate and MNI-kainate uncaging by different modes of one- and two-photon irradiation on hippocampal CA1 pyramidal neurons in acute brain slices. RESULTS/COMPARISON WITH EXISTING METHODS Unexpectedly, the data revealed that currents in response to MNI-glutamate uncaging were larger compared to MNI-kainate with local one-photon laser uncaging at the soma and two-photon uncaging at the same spines. Furthermore, the direct comparison demonstrates the influence of type of caged agonist and light delivery conditions used for uncaging on the amplitude and kinetic properties of the current response. CONCLUSION These findings highlight the importance of experimental design for uncaging experiments and provide a basis for future studies employing one- and two-photon uncaging to understand glutamate-dependent processes. It further provides the first example of two-photon uncaging of kainate at single spines in acute brain slices.
Collapse
Affiliation(s)
- Stefan Passlick
- Department of Neuroscience, Mount Sinai School of Medicine, New York, NY 10029, USA.
| | | |
Collapse
|
104
|
Kato AS, Witkin JM. Auxiliary subunits of AMPA receptors: The discovery of a forebrain-selective antagonist, LY3130481/CERC-611. Biochem Pharmacol 2017; 147:191-200. [PMID: 28987594 DOI: 10.1016/j.bcp.2017.09.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 09/27/2017] [Indexed: 12/11/2022]
Abstract
Drugs originate from the discovery of compounds, natural or synthetic, that bind to proteins (receptors, enzymes, transporters, etc.), the interaction of which modulates biological cascades that have potential therapeutic benefit. Rational strategies for identifying novel drug therapies are typically based on knowledge of the structure of the target proteins and the design of new chemical entities that modulate these proteins in a beneficial manner. The present review discusses a novel approach to drug discovery based on the identification and characterization of auxiliary proteins, the transmembrane AMPA receptor regulatory proteins (TARPs) that are associated with AMPA receptors. Utilizing these auxiliary proteins in compound screening led to the discovery of the TARP-dependent-AMPA forebrain selective receptor antagonist (TDAA), LY3130481/CERC-611 that is currently in clinical development for epilepsy.
Collapse
Affiliation(s)
- Akihiko S Kato
- Neuroscience Discovery Research, Lilly Research Labs, Eli Lilly and Company, Indianapolis, IN 46285-0510, United States.
| | - Jeffrey M Witkin
- Neuroscience Discovery Research, Lilly Research Labs, Eli Lilly and Company, Indianapolis, IN 46285-0510, United States.
| |
Collapse
|
105
|
Rosas-Santiago P, Lagunas-Gomez D, Yáñez-Domínguez C, Vera-Estrella R, Zimmermannová O, Sychrová H, Pantoja O. Plant and yeast cornichon possess a conserved acidic motif required for correct targeting of plasma membrane cargos. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:1809-1818. [DOI: 10.1016/j.bbamcr.2017.07.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 06/27/2017] [Accepted: 07/14/2017] [Indexed: 12/23/2022]
|
106
|
Yamasaki T, Hoyos-Ramirez E, Martenson JS, Morimoto-Tomita M, Tomita S. GARLH Family Proteins Stabilize GABA A Receptors at Synapses. Neuron 2017; 93:1138-1152.e6. [PMID: 28279354 DOI: 10.1016/j.neuron.2017.02.023] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2015] [Revised: 05/26/2016] [Accepted: 02/09/2017] [Indexed: 10/20/2022]
Abstract
Ionotropic neurotransmitter receptors mediate fast synaptic transmission by functioning as ligand-gated ion channels. Fast inhibitory transmission in the brain is mediated mostly by ionotropic GABAA receptors (GABAARs), but their essential components for synaptic localization remain unknown. Here, we identify putative auxiliary subunits of GABAARs, which we term GARLHs, consisting of LH4 and LH3 proteins. LH4 forms a stable tripartite complex with GABAARs and neuroligin-2 in the brain. Moreover, LH4 is required for the synaptic localization of GABAARs and inhibitory synaptic transmission in the hippocampus. Our findings propose GARLHs as the first identified auxiliary subunits for anion channels. These findings provide new insights into the regulation of inhibitory transmission and the molecular constituents of native anion channels in vivo.
Collapse
Affiliation(s)
- Tokiwa Yamasaki
- Department of Cellular and Molecular Physiology, Program in Cellular Neuroscience, Neurodegeneration and Repair, Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Erika Hoyos-Ramirez
- Department of Cellular and Molecular Physiology, Program in Cellular Neuroscience, Neurodegeneration and Repair, Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06520, USA
| | - James S Martenson
- Department of Cellular and Molecular Physiology, Program in Cellular Neuroscience, Neurodegeneration and Repair, Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Megumi Morimoto-Tomita
- Department of Cellular and Molecular Physiology, Program in Cellular Neuroscience, Neurodegeneration and Repair, Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Susumu Tomita
- Department of Cellular and Molecular Physiology, Program in Cellular Neuroscience, Neurodegeneration and Repair, Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
107
|
Hawken NM, Zaika EI, Nakagawa T. Engineering defined membrane-embedded elements of AMPA receptor induces opposing gating modulation by cornichon 3 and stargazin. J Physiol 2017; 595:6517-6539. [PMID: 28815591 DOI: 10.1113/jp274897] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Accepted: 08/04/2017] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS The AMPA-type ionotropic glutamate receptors (AMPARs) mediate the majority of excitatory synaptic transmission and their function impacts learning, cognition and behaviour. The gating of AMPARs occurs in milliseconds, precisely controlled by a variety of auxiliary subunits that are expressed differentially in the brain, but the difference in mechanisms underlying AMPAR gating modulation by auxiliary subunits remains elusive and is investigated. The elements of the AMPAR that are functionally recruited by auxiliary subunits, stargazin and cornichon 3, are located not only in the extracellular domains but also in the lipid-accessible surface of the AMPAR. We reveal that the two auxiliary subunits require a shared surface on the transmembrane domain of the AMPAR for their function, but the gating is influenced by this surface in opposing directions for each auxiliary subunit. Our results provide new insights into the mechanistic difference of AMPAR modulation by auxiliary subunits and a conceptual framework for functional engineering of the complex. ABSTRACT During excitatory synaptic transmission, various structurally unrelated transmembrane auxiliary subunits control the function of AMPA receptors (AMPARs), but the underlying mechanisms remain unclear. We identified lipid-exposed residues in the transmembrane domain (TMD) of the GluA2 subunit of AMPARs that are critical for the function of AMPAR auxiliary subunits, stargazin (Stg) and cornichon 3 (CNIH3). These residues are essential for stabilizing the AMPAR-CNIH3 complex in detergents and overlap with the contacts made between GluA2 TMD and Stg in the cryoEM structures. Mutating these residues had opposite effects on gating modulation and complex stability when Stg- and CNIH3-bound AMPARs were compared. Specifically, in detergent the GluA2-A793F formed an unstable complex with CNIIH3 but in the membrane the GluA2-A793F-CNIH3 complex expressed a gain of function. In contrast, the GluA2-A793F-Stg complex was stable, but had diminished gating modulation. GluA2-C528L destabilized the AMPAR-CNIH3 complex but stabilized the AMPAR-Stg complex, with overall loss of function in gating modulation. Furthermore, loss-of-function mutations in this TMD region cancelled the effects of a gain-of-function Stg carrying mutation in its extracellular loop, demonstrating that both the extracellular and the TMD elements contribute independently to gating modulation. The elements of AMPAR functionally recruited by auxiliary subunits are, therefore, located not only in the extracellular domains but also in the lipid accessible surface of the AMPAR. The TMD surface we defined is a potential target for auxiliary subunit-specific compounds, because engineering of this hotspot induces opposing functional outcomes by Stg and CNIH3. The collection of mutant-phenotype mapping provides a framework for engineering AMPAR gating using auxiliary subunits.
Collapse
Affiliation(s)
- Natalie M Hawken
- Department of Molecular Physiology and Biophysics, Vanderbilt University, School of Medicine, Nashville, TN, 37232, USA
| | - Elena I Zaika
- Department of Molecular Physiology and Biophysics, Vanderbilt University, School of Medicine, Nashville, TN, 37232, USA
| | - Terunaga Nakagawa
- Department of Molecular Physiology and Biophysics, Vanderbilt University, School of Medicine, Nashville, TN, 37232, USA.,Center for Structural Biology, Vanderbilt University, School of Medicine, Nashville, TN, 37232, USA.,Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN, 37232, USA
| |
Collapse
|
108
|
Riva I, Eibl C, Volkmer R, Carbone AL, Plested AJ. Control of AMPA receptor activity by the extracellular loops of auxiliary proteins. eLife 2017; 6:28680. [PMID: 28871958 PMCID: PMC5599240 DOI: 10.7554/elife.28680] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 08/28/2017] [Indexed: 11/13/2022] Open
Abstract
At synapses throughout the mammalian brain, AMPA receptors form complexes with auxiliary proteins, including TARPs. However, how TARPs modulate AMPA receptor gating remains poorly understood. We built structural models of TARP-AMPA receptor complexes for TARPs γ2 and γ8, combining recent structural studies and de novo structure predictions. These models, combined with peptide binding assays, provide evidence for multiple interactions between GluA2 and variable extracellular loops of TARPs. Substitutions and deletions of these loops had surprisingly rich effects on the kinetics of glutamate-activated currents, without any effect on assembly. Critically, by altering the two interacting loops of γ2 and γ8, we could entirely remove all allosteric modulation of GluA2, without affecting formation of AMPA receptor-TARP complexes. Likewise, substitutions in the linker domains of GluA2 completely removed any effect of γ2 on receptor kinetics, indicating a dominant role for this previously overlooked site proximal to the AMPA receptor channel gate.
Collapse
Affiliation(s)
- Irene Riva
- Institute of Biology, Cellular Biophysics, Humboldt Universität zu Berlin, Berlin, Germany.,Molecular Physiology and Cell Biology, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Clarissa Eibl
- Institute of Biology, Cellular Biophysics, Humboldt Universität zu Berlin, Berlin, Germany.,Molecular Physiology and Cell Biology, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Rudolf Volkmer
- Chemical Biology, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Anna L Carbone
- Institute of Biology, Cellular Biophysics, Humboldt Universität zu Berlin, Berlin, Germany.,Molecular Physiology and Cell Biology, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| | - Andrew Jr Plested
- Institute of Biology, Cellular Biophysics, Humboldt Universität zu Berlin, Berlin, Germany.,Molecular Physiology and Cell Biology, Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany
| |
Collapse
|
109
|
Geva Y, Crissman J, Arakel EC, Gómez-Navarro N, Chuartzman SG, Stahmer KR, Schwappach B, Miller EA, Schuldiner M. Two novel effectors of trafficking and maturation of the yeast plasma membrane H + -ATPase. Traffic 2017; 18:672-682. [PMID: 28727280 PMCID: PMC5607100 DOI: 10.1111/tra.12503] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 07/17/2017] [Accepted: 07/17/2017] [Indexed: 11/28/2022]
Abstract
The endoplasmic reticulum (ER) is the entry site of proteins into the endomembrane system. Proteins exit the ER via coat protein II (COPII) vesicles in a selective manner, mediated either by direct interaction with the COPII coat or aided by cargo receptors. Despite the fundamental role of such receptors in protein sorting, only a few have been identified. To further define the machinery that packages secretory cargo and targets proteins from the ER to Golgi membranes, we used multiple systematic approaches, which revealed 2 uncharacterized proteins that mediate the trafficking and maturation of Pma1, the essential yeast plasma membrane proton ATPase. Ydl121c (Exp1) is an ER protein that binds Pma1, is packaged into COPII vesicles, and whose deletion causes ER retention of Pma1. Ykl077w (Psg1) physically interacts with Exp1 and can be found in the Golgi and coat protein I (COPI) vesicles but does not directly bind Pma1. Loss of Psg1 causes enhanced degradation of Pma1 in the vacuole. Our findings suggest that Exp1 is a Pma1 cargo receptor and that Psg1 aids Pma1 maturation in the Golgi or affects its retrieval. More generally our work shows the utility of high content screens in the identification of novel trafficking components.
Collapse
Affiliation(s)
- Yosef Geva
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Jonathan Crissman
- Department of Biological Sciences, Columbia University, New York, NY
| | - Eric C Arakel
- Department of Molecular Biology, Universitätsmedizin Göttingen, Göttingen, Germany
| | | | - Silvia G Chuartzman
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Kyle R Stahmer
- Department of Biological Sciences, Columbia University, New York, NY
| | - Blanche Schwappach
- Department of Molecular Biology, Universitätsmedizin Göttingen, Göttingen, Germany
| | - Elizabeth A Miller
- Department of Biological Sciences, Columbia University, New York, NY.,MRC Laboratory of Molecular Biology, Cell Biology Division, Cambridge, UK
| | - Maya Schuldiner
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
110
|
Diversity in AMPA receptor complexes in the brain. Curr Opin Neurobiol 2017; 45:32-38. [DOI: 10.1016/j.conb.2017.03.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 02/28/2017] [Accepted: 03/03/2017] [Indexed: 11/23/2022]
|
111
|
Frank RA, Grant SG. Supramolecular organization of NMDA receptors and the postsynaptic density. Curr Opin Neurobiol 2017; 45:139-147. [PMID: 28577431 PMCID: PMC5557338 DOI: 10.1016/j.conb.2017.05.019] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 05/02/2017] [Accepted: 05/15/2017] [Indexed: 01/21/2023]
Abstract
The postsynaptic density (PSD) of all vertebrate species share a highly complex proteome with ∼1000 conserved proteins that function as sophisticated molecular computational devices. Here, we review recent studies showing that this complexity can be understood in terms of the supramolecular organization of proteins, which self-assemble within a hierarchy of different length scales, including complexes, supercomplexes and nanodomains. We highlight how genetic and biochemical approaches in mice are being used to uncover the native molecular architecture of the synapse, revealing hitherto unknown molecular structures, including highly selective mechanisms for specifying the assembly of NMDAR-MAGUK supercomplexes. We propose there exists a logical framework that precisely dictates the subunit composition of synaptic complexes, supercomplexes, and nanodomains in vivo.
Collapse
Affiliation(s)
- René Aw Frank
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Seth Gn Grant
- Centre for Clinical Brain Sciences, University of Edinburgh, Chancellor's Building, Edinburgh EH16 4SB, UK.
| |
Collapse
|
112
|
Bettler B, Fakler B. Ionotropic AMPA-type glutamate and metabotropic GABAB receptors: determining cellular physiology by proteomes. Curr Opin Neurobiol 2017; 45:16-23. [DOI: 10.1016/j.conb.2017.02.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Accepted: 02/15/2017] [Indexed: 02/07/2023]
|
113
|
Brechet A, Buchert R, Schwenk J, Boudkkazi S, Zolles G, Siquier-Pernet K, Schaber I, Bildl W, Saadi A, Bole-Feysot C, Nitschke P, Reis A, Sticht H, Al-Sanna'a N, Rolfs A, Kulik A, Schulte U, Colleaux L, Abou Jamra R, Fakler B. AMPA-receptor specific biogenesis complexes control synaptic transmission and intellectual ability. Nat Commun 2017; 8:15910. [PMID: 28675162 PMCID: PMC5500892 DOI: 10.1038/ncomms15910] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 05/11/2017] [Indexed: 11/25/2022] Open
Abstract
AMPA-type glutamate receptors (AMPARs), key elements in excitatory neurotransmission in the brain, are macromolecular complexes whose properties and cellular functions are determined by the co-assembled constituents of their proteome. Here we identify AMPAR complexes that transiently form in the endoplasmic reticulum (ER) and lack the core-subunits typical for AMPARs in the plasma membrane. Central components of these ER AMPARs are the proteome constituents FRRS1l (C9orf4) and CPT1c that specifically and cooperatively bind to the pore-forming GluA1-4 proteins of AMPARs. Bi-allelic mutations in the human FRRS1L gene are shown to cause severe intellectual disability with cognitive impairment, speech delay and epileptic activity. Virus-directed deletion or overexpression of FRRS1l strongly impact synaptic transmission in adult rat brain by decreasing or increasing the number of AMPARs in synapses and extra-synaptic sites. Our results provide insight into the early biogenesis of AMPARs and demonstrate its pronounced impact on synaptic transmission and brain function. The biogenesis of AMPA-type glutamate receptor (AMPAR) complexes is only partially understood. Here the authors identify transient assemblies of GluA1-4 proteins and proteins FRRS1l/CPT1c that drive formation of mature AMPAR complexes in the ER. Mutations in FRRS1l are associated with intellectual disability and epilepsy in three families.
Collapse
Affiliation(s)
- Aline Brechet
- Institute of Physiology, Faculty of Medicine, University of Freiburg, Hermann-Herder-Str. 7, Freiburg 79104, Germany
| | - Rebecca Buchert
- Institute of Human Genetics, University of Erlangen, Schwabachanlage 10, Erlangen 91054, Germany
| | - Jochen Schwenk
- Institute of Physiology, Faculty of Medicine, University of Freiburg, Hermann-Herder-Str. 7, Freiburg 79104, Germany.,Center for Biological Signaling Studies (BIOSS), Schänzlestr. 18, Freiburg 79104, Germany
| | - Sami Boudkkazi
- Institute of Physiology, Faculty of Medicine, University of Freiburg, Hermann-Herder-Str. 7, Freiburg 79104, Germany
| | - Gerd Zolles
- Institute of Physiology, Faculty of Medicine, University of Freiburg, Hermann-Herder-Str. 7, Freiburg 79104, Germany
| | - Karine Siquier-Pernet
- INSERM UMR 1163, Paris-Descartes-Sorbonne Paris Cité University, Institut IMAGINE, Necker-Enfants Malades Hospital, Paris 75015, France
| | - Irene Schaber
- Institute of Physiology, Faculty of Medicine, University of Freiburg, Hermann-Herder-Str. 7, Freiburg 79104, Germany
| | - Wolfgang Bildl
- Institute of Physiology, Faculty of Medicine, University of Freiburg, Hermann-Herder-Str. 7, Freiburg 79104, Germany
| | - Abdelkrim Saadi
- Department de Neurologie, Etablissement Hospitalier Specialisé de Benaknoun, Algers, Algeria
| | - Christine Bole-Feysot
- INSERM UMR 1163, Paris-Descartes-Sorbonne Paris Cité University, Institut IMAGINE, Necker-Enfants Malades Hospital, Paris 75015, France
| | - Patrick Nitschke
- INSERM UMR 1163, Paris-Descartes-Sorbonne Paris Cité University, Institut IMAGINE, Necker-Enfants Malades Hospital, Paris 75015, France
| | - Andre Reis
- Institute of Human Genetics, University of Erlangen, Schwabachanlage 10, Erlangen 91054, Germany
| | - Heinrich Sticht
- Institute of Biochemistry, Emil-Fischer Center, Fahrstraße 17, Erlangen 91054, Germany
| | - Nouriya Al-Sanna'a
- Dharan Health Center, 8131 Medical Access Rd 1, Gharb al Dharan, Dharan 34465, Saudi Arabia
| | - Arndt Rolfs
- Center for Biological Signaling Studies (BIOSS), Schänzlestr. 18, Freiburg 79104, Germany.,Albrecht-Kossel-Institute for Neuroregeneration, Medical University Rostock, Gehlsheimerstr. 20, Rostock 18147, Germany
| | - Akos Kulik
- Institute of Physiology, Faculty of Medicine, University of Freiburg, Hermann-Herder-Str. 7, Freiburg 79104, Germany.,Center for Biological Signaling Studies (BIOSS), Schänzlestr. 18, Freiburg 79104, Germany
| | - Uwe Schulte
- Institute of Physiology, Faculty of Medicine, University of Freiburg, Hermann-Herder-Str. 7, Freiburg 79104, Germany.,Center for Biological Signaling Studies (BIOSS), Schänzlestr. 18, Freiburg 79104, Germany.,Logopharm GmbH, Schlossstr. 14, March-Buchheim 79232, Germany
| | - Laurence Colleaux
- INSERM UMR 1163, Paris-Descartes-Sorbonne Paris Cité University, Institut IMAGINE, Necker-Enfants Malades Hospital, Paris 75015, France
| | - Rami Abou Jamra
- Institute of Human Genetics, University of Erlangen, Schwabachanlage 10, Erlangen 91054, Germany.,Institute of Human Genetics, University of Leipzig Hospitals and Clinics, Philipp-Rosenthal-Str. 55, 04103 Leipzig, Germany
| | - Bernd Fakler
- Institute of Physiology, Faculty of Medicine, University of Freiburg, Hermann-Herder-Str. 7, Freiburg 79104, Germany.,Center for Biological Signaling Studies (BIOSS), Schänzlestr. 18, Freiburg 79104, Germany
| |
Collapse
|
114
|
Greger IH, Watson JF, Cull-Candy SG. Structural and Functional Architecture of AMPA-Type Glutamate Receptors and Their Auxiliary Proteins. Neuron 2017; 94:713-730. [DOI: 10.1016/j.neuron.2017.04.009] [Citation(s) in RCA: 169] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 04/04/2017] [Accepted: 04/05/2017] [Indexed: 12/20/2022]
|
115
|
Azumaya CM, Days EL, Vinson PN, Stauffer S, Sulikowski G, Weaver CD, Nakagawa T. Screening for AMPA receptor auxiliary subunit specific modulators. PLoS One 2017; 12:e0174742. [PMID: 28358902 PMCID: PMC5373622 DOI: 10.1371/journal.pone.0174742] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 03/14/2017] [Indexed: 12/18/2022] Open
Abstract
AMPA receptors (AMPAR) are ligand gated ion channels critical for synaptic transmission and plasticity. Their dysfunction is implicated in a variety of psychiatric and neurological diseases ranging from major depressive disorder to amyotrophic lateral sclerosis. Attempting to potentiate or depress AMPAR activity is an inherently difficult balancing act between effective treatments and debilitating side effects. A newly explored strategy to target subsets of AMPARs in the central nervous system is to identify compounds that affect specific AMPAR-auxiliary subunit complexes. This exploits diverse spatio-temporal expression patterns of known AMPAR auxiliary subunits, providing means for designing brain region-selective compounds. Here we report a high-throughput screening-based pipeline that can identify compounds that are selective for GluA2-CNIH3 and GluA2-stargazin complexes. These compounds will help us build upon the growing library of AMPAR-auxiliary subunit specific inhibitors, which have thus far all been targeted to TARP γ-8. We used a cell-based assay combined with a voltage-sensitive dye (VSD) to identify changes in glutamate-gated cation flow across the membranes of HEK cells co-expressing GluA2 and an auxiliary subunit. We then used a calcium flux assay to further validate hits picked from the VSD assay. VU0612951 and VU0627849 are candidate compounds from the initial screen that were identified as negative and positive allosteric modulators (NAM and PAM), respectively. They both have lower IC50/EC50s on complexes containing stargazin and CNIH3 than GSG1L or the AMPAR alone. We have also identified a candidate compound, VU0539491, that has NAM activity in GluA2(R)-CNIH3 and GluA2(Q) complexes and PAM activity in GluA2(Q)-GSG1L complexes.
Collapse
Affiliation(s)
- Caleigh M Azumaya
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Emily L Days
- Vanderbilt Institute of Chemical Biology High Throughput Screening Core, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Paige N Vinson
- Vanderbilt Institute of Chemical Biology High Throughput Screening Core, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Shaun Stauffer
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee, United States of America.,Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Gary Sulikowski
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee, United States of America
| | - C David Weaver
- Vanderbilt Institute of Chemical Biology High Throughput Screening Core, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America.,Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Terunaga Nakagawa
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America.,Center for Structural Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America.,Vanderbilt Brain Institute, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| |
Collapse
|
116
|
Aviram N, Ast T, Costa EA, Arakel EC, Chuartzman SG, Jan CH, Haßdenteufel S, Dudek J, Jung M, Schorr S, Zimmermann R, Schwappach B, Weissman JS, Schuldiner M. The SND proteins constitute an alternative targeting route to the endoplasmic reticulum. Nature 2017; 540:134-138. [PMID: 27905431 DOI: 10.1038/nature20169] [Citation(s) in RCA: 159] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 10/18/2016] [Indexed: 12/19/2022]
Abstract
In eukaryotes, up to one-third of cellular proteins are targeted to the endoplasmic reticulum, where they undergo folding, processing, sorting and trafficking to subsequent endomembrane compartments. Targeting to the endoplasmic reticulum has been shown to occur co-translationally by the signal recognition particle (SRP) pathway or post-translationally by the mammalian transmembrane recognition complex of 40 kDa (TRC40) and homologous yeast guided entry of tail-anchored proteins (GET) pathways. Despite the range of proteins that can be catered for by these two pathways, many proteins are still known to be independent of both SRP and GET, so there seems to be a critical need for an additional dedicated pathway for endoplasmic reticulum relay. We set out to uncover additional targeting proteins using unbiased high-content screening approaches. To this end, we performed a systematic visual screen using the yeast Saccharomyces cerevisiae, and uncovered three uncharacterized proteins whose loss affected targeting. We suggest that these proteins work together and demonstrate that they function in parallel with SRP and GET to target a broad range of substrates to the endoplasmic reticulum. The three proteins, which we name Snd1, Snd2 and Snd3 (for SRP-independent targeting), can synthetically compensate for the loss of both the SRP and GET pathways, and act as a backup targeting system. This explains why it has previously been difficult to demonstrate complete loss of targeting for some substrates. Our discovery thus puts in place an essential piece of the endoplasmic reticulum targeting puzzle, highlighting how the targeting apparatus of the eukaryotic cell is robust, interlinked and flexible.
Collapse
Affiliation(s)
- Naama Aviram
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Tslil Ast
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Elizabeth A Costa
- Department of Cellular and Molecular Pharmacology, UCSF California Institute for Quantitative Biomedical Research and Howard Hughes Medical Institute, San Francisco, California 94158-2330, USA
| | - Eric C Arakel
- Department of Molecular Biology, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Silvia G Chuartzman
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Calvin H Jan
- Department of Cellular and Molecular Pharmacology, UCSF California Institute for Quantitative Biomedical Research and Howard Hughes Medical Institute, San Francisco, California 94158-2330, USA
| | - Sarah Haßdenteufel
- Department of Medical Biochemistry and Molecular Biology, Saarland University, 66421 Homburg, Germany
| | - Johanna Dudek
- Department of Medical Biochemistry and Molecular Biology, Saarland University, 66421 Homburg, Germany
| | - Martin Jung
- Department of Medical Biochemistry and Molecular Biology, Saarland University, 66421 Homburg, Germany
| | - Stefan Schorr
- Department of Medical Biochemistry and Molecular Biology, Saarland University, 66421 Homburg, Germany
| | - Richard Zimmermann
- Department of Medical Biochemistry and Molecular Biology, Saarland University, 66421 Homburg, Germany
| | - Blanche Schwappach
- Department of Molecular Biology, University Medical Center Göttingen, 37073 Göttingen, Germany.,Max-Planck Institute for Biophysical Chemistry, 37077 Göttingen, Germany
| | - Jonathan S Weissman
- Department of Cellular and Molecular Pharmacology, UCSF California Institute for Quantitative Biomedical Research and Howard Hughes Medical Institute, San Francisco, California 94158-2330, USA
| | - Maya Schuldiner
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| |
Collapse
|
117
|
Wei M, Jia M, Zhang J, Yu L, Zhao Y, Chen Y, Ma Y, Zhang W, Shi YS, Zhang C. The Inhibitory Effect of α/β-Hydrolase Domain-Containing 6 (ABHD6) on the Surface Targeting of GluA2- and GluA3-Containing AMPA Receptors. Front Mol Neurosci 2017; 10:55. [PMID: 28303090 PMCID: PMC5333494 DOI: 10.3389/fnmol.2017.00055] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 02/17/2017] [Indexed: 01/16/2023] Open
Abstract
The α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)-type glutamate receptors (AMPARs) are major excitatory receptors that mediate fast neurotransmission in the mammalian brain. The surface expression of functional AMPARs is crucial for synaptic transmission and plasticity. AMPAR auxiliary subunits control the biosynthesis, membrane trafficking, and synaptic targeting of AMPARs. Our previous report showed that α/β-hydrolase domain-containing 6 (ABHD6), an auxiliary subunit for AMPARs, suppresses the membrane delivery and function of GluA1-containing receptors in both heterologous cells and neurons. However, it remained unclear whether ABHD6 affects the membrane trafficking of glutamate receptor subunits, GluA2 and GluA3. Here, we examine the effects of ABHD6 overexpression in HEK293T cells expressing GluA1, GluA2, GluA3, and stargazin, either alone or in combination. The results show that ABHD6 suppresses the glutamate-induced currents and the membrane expression of AMPARs when expressing GluA2 or GluA3 in the HEK293T cells. We generated a series of GluA2 and GluA3 C-terminal deletion constructs and confirm that the C-terminus of GluAs is required for ABHD6’s inhibitory effects on glutamate-induced currents and surface expression of GluAs. Meanwhile, our pull-down experiments reveal that ABHD6 binds to GluA1–3, and deletion of the C-terminal domain of GluAs abolishes this binding. These findings demonstrate that ABHD6 inhibits the AMPAR-mediated currents and its surface expression, independent of the type of AMPAR subunits, and this inhibitor’s effects are mediated through the binding with the GluAs C-terminal regions.
Collapse
Affiliation(s)
- Mengping Wei
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking UniversityBeijing, China; PKU-IDG (International Digital Group)/McGovern Institute for Brain Research, Peking UniversityBeijing, China
| | - Moye Jia
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking UniversityBeijing, China; PKU-IDG (International Digital Group)/McGovern Institute for Brain Research, Peking UniversityBeijing, China
| | - Jian Zhang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking UniversityBeijing, China; PKU-IDG (International Digital Group)/McGovern Institute for Brain Research, Peking UniversityBeijing, China
| | - Lulu Yu
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking UniversityBeijing, China; PKU-IDG (International Digital Group)/McGovern Institute for Brain Research, Peking UniversityBeijing, China
| | - Yunzhi Zhao
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking UniversityBeijing, China; PKU-IDG (International Digital Group)/McGovern Institute for Brain Research, Peking UniversityBeijing, China
| | - Yingqi Chen
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking UniversityBeijing, China; PKU-IDG (International Digital Group)/McGovern Institute for Brain Research, Peking UniversityBeijing, China
| | - Yimeng Ma
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking UniversityBeijing, China; PKU-IDG (International Digital Group)/McGovern Institute for Brain Research, Peking UniversityBeijing, China
| | - Wei Zhang
- Department of Pharmacology, Institute of Chinese Integrative Medicine, Hebei Medical University Shijiazhuang, China
| | - Yun S Shi
- Ministry of Education (MOE) Key Laboratory of Model Animal for Disease Study, Model Animal Research Center of Nanjing University Nanjing, China
| | - Chen Zhang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Peking UniversityBeijing, China; PKU-IDG (International Digital Group)/McGovern Institute for Brain Research, Peking UniversityBeijing, China
| |
Collapse
|
118
|
Sensitizing exposure to amphetamine increases AMPA receptor phosphorylation without increasing cell surface expression in the rat nucleus accumbens. Neuropharmacology 2017; 117:328-337. [PMID: 28223211 DOI: 10.1016/j.neuropharm.2017.02.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 12/21/2016] [Accepted: 02/17/2017] [Indexed: 12/14/2022]
Abstract
Exposure to psychostimulants like cocaine or amphetamine leads to long-lasting sensitization of their behavioral and neurochemical effects. Here we characterized changes in AMPA receptor distribution and phosphorylation state in the rat nucleus accumbens (NAcc) weeks after amphetamine exposure to assess their potential contribution to sensitization by this drug. Using protein cross-linking, biochemical, subcellular fractionation, and slice electrophysiological approaches in the NAcc, we found that, unlike cocaine, previous exposure to amphetamine did not increase cell surface levels of either GluA1 or GluA2 AMPA receptor subunits, redistribution of these subunits to the synaptic or perisynaptic cellular membrane domains, protein-protein associations required to support the accumulation and retention of AMPA receptors in the PSD, or the peak amplitude of AMPA receptor mediated mEPSCs recorded in NAcc slices. On the other hand, exposure to amphetamine significantly slowed mEPSC decay times and increased levels in the PSD of PKA and CaMKII as well as phosphorylation by these kinases of the GluA1 S845 and S831 residues selectively in this cellular compartment. As the latter effects are known to respectively regulate channel open probability and duration as well as conductance, they provide a novel mechanism that could contribute to the long-lasting AMPA receptor dependent expression of sensitization by amphetamine. Rather than increase the number of surface and synaptic AMPA receptors as with cocaine, this mechanism could increase NAcc medium spiny neuron reactivity to glutamate afferents by increasing the phosphorylation state of critical regulatory sites in the AMPA receptor GluA1 subunit in the PSD.
Collapse
|
119
|
Kroes RA, Nilsson CL. Towards the Molecular Foundations of Glutamatergic-targeted Antidepressants. Curr Neuropharmacol 2017; 15:35-46. [PMID: 26955966 PMCID: PMC5327457 DOI: 10.2174/1570159x14666160309114740] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Revised: 05/08/2015] [Accepted: 01/30/2016] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Depression affects over 120 million individuals of all ages and is the leading cause of disability worldwide. The lack of objective diagnostic criteria, together with the heterogeneity of the depressive disorder itself, makes it challenging to develop effective therapies. The accumulation of preclinical data over the past 20 years derived from a multitude of models using many divergent approaches, has fueled the resurgence of interest in targeting glutamatergic neurotransmission for the treatment of major depression. OBJECTIVE The emergence of mechanistic studies are advancing our understanding of the molecular underpinnings of depression. While clearly far from complete and conclusive, they offer the potential to lead to the rational design of more specific therapeutic strategies and the development of safer and more effective rapid acting, long lasting antidepressants. METHODS The development of comprehensive omics-based approaches to the dysregulation of synaptic transmission and plasticity that underlies the core pathophysiology of MDD are reviewed to illustrate the fundamental elements. RESULTS This review frames the rationale for the conceptualization of depression as a "pathway disease". As such, it culminates in the call for the development of novel state-of-the-art "-omics approaches" and neurosystems biological techniques necessary to advance our understanding of spatiotemporal interactions associated with targeting glutamatergic-triggered signaling in the CNS. CONCLUSION These technologies will enable the development of novel psychiatric medications specifically targeted to impact specific, critical intracellular networks in a more focused manner and have the potential to offer new dimensions in the area of translational neuropsychiatry.
Collapse
Affiliation(s)
- Roger A. Kroes
- Naurex, Inc., 1801 Maple Street, Evanston, Illinois 60201, United States
| | - Carol L. Nilsson
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas, 77555-1074, United States
| |
Collapse
|
120
|
Forebrain-selective AMPA-receptor antagonism guided by TARP γ-8 as an antiepileptic mechanism. Nat Med 2016; 22:1496-1501. [DOI: 10.1038/nm.4221] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 09/30/2016] [Indexed: 12/12/2022]
|
121
|
Rex EB, Shukla N, Gu S, Bredt D, DiSepio D. A Genome-Wide Arrayed cDNA Screen to Identify Functional Modulators of α7 Nicotinic Acetylcholine Receptors. SLAS DISCOVERY 2016; 22:155-165. [PMID: 27789755 DOI: 10.1177/1087057116676086] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Cellular signaling is in part regulated by the composition and subcellular localization of a series of protein interactions that collectively form a signaling complex. Using the α7 nicotinic acetylcholine receptor (α7nAChR) as a proof-of-concept target, we developed a platform to identify functional modulators (or auxiliary proteins) of α7nAChR signaling. The Broad cDNA library was transiently cotransfected with α7nAChR cDNA in HEK293T cells in a high-throughput fashion. Using this approach in combination with a functional assay, we identified positive modulators of α7nAChR activity. We identified known positive modulators/auxiliary proteins present in the cDNA library that regulate α7nAChR signaling, in addition to identifying novel modulators of α7nAChR signaling. These included NACHO, SPDYE11, TCF4, and ZC3H12A, all of which increased PNU-120596-mediated nicotine-dependent calcium flux. Importantly, these auxiliary proteins did not modulate GluR1(o)-mediated Ca flux. To elucidate a possible mechanism of action, we employed an α7nAChR-HA surface staining assay. NACHO enhanced α7nAChR surface expression; however, the mechanism responsible for the SPDYE11-, TCF4-, and ZC3H12A-dependent modulation of α7nAChR has yet to be defined. This report describes the development and validation of a high-throughput, genome-wide cDNA screening platform coupled to FLIPR functional assays in order to identify functional modulators of α7nAChR signaling.
Collapse
Affiliation(s)
- Elizabeth B Rex
- 1 Discovery Sciences, Janssen Research and Development LLC, La Jolla, CA, USA
| | - Nikhil Shukla
- 1 Discovery Sciences, Janssen Research and Development LLC, La Jolla, CA, USA
| | - Shenyan Gu
- 2 Neuroscience, Janssen Research and Development LLC, La Jolla, CA, USA
| | - David Bredt
- 2 Neuroscience, Janssen Research and Development LLC, La Jolla, CA, USA
| | - Daniel DiSepio
- 1 Discovery Sciences, Janssen Research and Development LLC, La Jolla, CA, USA
| |
Collapse
|
122
|
Mao X, Gu X, Lu W. GSG1L regulates the strength of AMPA receptor-mediated synaptic transmission but not AMPA receptor kinetics in hippocampal dentate granule neurons. J Neurophysiol 2016; 117:28-35. [PMID: 27707810 DOI: 10.1152/jn.00307.2016] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 10/03/2016] [Indexed: 11/22/2022] Open
Abstract
GSG1L is an AMPA receptor (AMPAR) auxiliary subunit that regulates AMPAR trafficking and function in hippocampal CA1 pyramidal neurons. However, its physiological roles in other types of neurons remain to be characterized. Here, we investigated the role of GSG1L in hippocampal dentate granule cells and found that GSG1L is important for the regulation of synaptic strength but is not critical for the modulation of AMPAR deactivation and desensitization kinetics. These data demonstrate a neuronal type-specific role of GSG1L and suggest that physiological function of AMPAR auxiliary subunits may vary in different types of neurons. NEW & NOTEWORTHY GSG1L is a newly identified AMPA receptor (AMPAR) auxiliary subunit and plays a unique role in the regulation of AMPAR trafficking and function in hippocampal CA1 pyramidal neurons. However, its role in the regulation of AMPARs in hippocampal dentate granule cells remains to be characterized. The current work reveals that GSG1L regulates strength of AMPAR-mediated synaptic transmission but not the receptor kinetic properties in hippocampal dentate granule neurons.
Collapse
Affiliation(s)
- Xia Mao
- Synapse and Neural Circuit Research Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | - Xinglong Gu
- Synapse and Neural Circuit Research Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| | - Wei Lu
- Synapse and Neural Circuit Research Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
123
|
Compans B, Choquet D, Hosy E. Review on the role of AMPA receptor nano-organization and dynamic in the properties of synaptic transmission. NEUROPHOTONICS 2016; 3:041811. [PMID: 27981061 PMCID: PMC5109202 DOI: 10.1117/1.nph.3.4.041811] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 10/19/2016] [Indexed: 06/06/2023]
Abstract
Receptor trafficking and its regulation have appeared in the last two decades to be a major controller of basal synaptic transmission and its activity-dependent plasticity. More recently, considerable advances in super-resolution microscopy have begun deciphering the subdiffraction organization of synaptic elements and their functional roles. In particular, the dynamic nanoscale organization of neurotransmitter receptors in the postsynaptic membrane has recently been suggested to play a major role in various aspects of synapstic function. We here review the recent advances in our understanding of alpha-amino-3-hydroxy-5-méthyl-4-isoxazolepropionic acid subtype glutamate receptors subsynaptic organization and their role in short- and long-term synaptic plasticity.
Collapse
Affiliation(s)
- Benjamin Compans
- University of Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux F-33000, France
- Interdisciplinary Institute for Neuroscience, CNRS, UMR 5297, Bordeaux F-33000, France
| | - Daniel Choquet
- University of Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux F-33000, France
- Interdisciplinary Institute for Neuroscience, CNRS, UMR 5297, Bordeaux F-33000, France
- University of Bordeaux, Bordeaux Imaging Center, UMS 3420 CNRS, US4 INSERM, France
| | - Eric Hosy
- University of Bordeaux, Interdisciplinary Institute for Neuroscience, UMR 5297, Bordeaux F-33000, France
- Interdisciplinary Institute for Neuroscience, CNRS, UMR 5297, Bordeaux F-33000, France
| |
Collapse
|
124
|
Protein Crowding within the Postsynaptic Density Can Impede the Escape of Membrane Proteins. J Neurosci 2016; 36:4276-95. [PMID: 27076425 DOI: 10.1523/jneurosci.3154-15.2016] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 02/19/2016] [Indexed: 01/03/2023] Open
Abstract
UNLABELLED Mechanisms regulating lateral diffusion and positioning of glutamate receptors within the postsynaptic density (PSD) determine excitatory synaptic strength. Scaffold proteins in the PSD are abundant receptor binding partners, yet electron microscopy suggests that the PSD is highly crowded, potentially restricting the diffusion of receptors regardless of binding. However, the contribution of macromolecular crowding to receptor retention remains poorly understood. We combined experimental and computational approaches to test the effect of synaptic crowding on receptor movement and positioning in Sprague Dawley rat hippocampal neurons. We modeled AMPA receptor diffusion in synapses where the distribution of scaffold proteins was determined from photoactivated localization microscopy experiments, and receptor-scaffold association and dissociation rates were adjusted to fit single-molecule tracking and fluorescence recovery measurements. Simulations predicted that variation of receptor size strongly influences the fractional synaptic area the receptor may traverse, and the proportion that may exchange in and out of the synapse. To test the model experimentally, we designed a set of novel transmembrane (TM) probes. A single-pass TM protein with one PDZ binding motif concentrated in the synapse as do AMPARs yet was more mobile there than the much larger AMPAR. Furthermore, either the single binding motif or an increase in cytoplasmic bulk through addition of a single GFP slowed synaptic movement of a small TM protein. These results suggest that both crowding and binding limit escape of AMPARs from the synapse. Moreover, tight protein packing within the PSD may modulate the synaptic dwell time of many TM proteins important for synaptic function. SIGNIFICANCE STATEMENT Small alterations to the distribution within synapses of key transmembrane proteins, such as receptors, can dramatically change synaptic strength. Indeed, many diseases are thought to unbalance neural circuit function in this manner. Processes that regulate this in healthy synapses are unclear, however. By combining computer simulations with imaging methods that examined protein dynamics at multiple scales in space and time, we showed that both steric effects and protein-protein binding each regulate the mobility of receptors in the synapse. Our findings extend our knowledge of the synapse as a crowded environment that counteracts molecular diffusion, and support the idea that both molecular collisions and biochemical binding can be involved in the regulation of neural circuit performance.
Collapse
|
125
|
GluA1 signal peptide determines the spatial assembly of heteromeric AMPA receptors. Proc Natl Acad Sci U S A 2016; 113:E5645-54. [PMID: 27601647 DOI: 10.1073/pnas.1524358113] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
AMPA-type glutamate receptors (AMPARs) mediate fast excitatory neurotransmission and predominantly assemble as heterotetramers in the brain. Recently, the crystal structures of homotetrameric GluA2 demonstrated that AMPARs are assembled with two pairs of conformationally distinct subunits, in a dimer of dimers formation. However, the structure of heteromeric AMPARs remains unclear. Guided by the GluA2 structure, we performed cysteine mutant cross-linking experiments in full-length GluA1/A2, aiming to draw the heteromeric AMPAR architecture. We found that the amino-terminal domains determine the first level of heterodimer formation. When the dimers further assemble into tetramers, GluA1 and GluA2 subunits have preferred positions, possessing a 1-2-1-2 spatial assembly. By swapping the critical sequences, we surprisingly found that the spatial assembly pattern is controlled by the excisable signal peptides. Replacements with an unrelated GluK2 signal peptide demonstrated that GluA1 signal peptide plays a critical role in determining the spatial priority. Our study thus uncovers the spatial assembly of an important type of glutamate receptors in the brain and reveals a novel function of signal peptides.
Collapse
|
126
|
Li J, Fuchs S, Zhang J, Wellford S, Schuldiner M, Wang X. An unrecognized function for COPII components in recruiting the viral replication protein BMV 1a to the perinuclear ER. J Cell Sci 2016; 129:3597-3608. [PMID: 27539921 DOI: 10.1242/jcs.190082] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 08/13/2016] [Indexed: 01/05/2023] Open
Abstract
Positive-strand RNA viruses invariably assemble their viral replication complexes (VRCs) by remodeling host intracellular membranes. How viral replication proteins are targeted to specific organelle membranes to initiate VRC assembly remains elusive. Brome mosaic virus (BMV), whose replication can be recapitulated in Saccharomyces cerevisiae, assembles its VRCs by invaginating the outer perinuclear endoplasmic reticulum (ER) membrane. Remarkably, BMV replication protein 1a (BMV 1a) is the only viral protein required for such membrane remodeling. We show that ER-vesicle protein of 14 kD (Erv14), a cargo receptor of coat protein complex II (COPII), interacts with BMV 1a. Moreover, the perinuclear ER localization of BMV 1a is disrupted in cells lacking ERV14 or expressing dysfunctional COPII coat components (Sec13, Sec24 or Sec31). The requirement of Erv14 for the localization of BMV 1a is bypassed by addition of a Sec24-recognizable sorting signal to BMV 1a or by overexpressing Sec24, suggesting a coordinated effort by both Erv14 and Sec24 for the proper localization of BMV 1a. The COPII pathway is well known for being involved in protein secretion; our data suggest that a subset of COPII coat proteins have an unrecognized role in targeting proteins to the perinuclear ER membrane.
Collapse
Affiliation(s)
- Jianhui Li
- Department of Plant Pathology, Physiology, and Weed Science, Virginia Tech, Blacksburg, VA 24061, USA
| | - Shai Fuchs
- Department of Molecular Genetics, Weizmann Institute of Sciences, Rehovot 7610001, Israel
| | - Jiantao Zhang
- Department of Plant Pathology, Physiology, and Weed Science, Virginia Tech, Blacksburg, VA 24061, USA
| | - Sebastian Wellford
- Department of Plant Pathology, Physiology, and Weed Science, Virginia Tech, Blacksburg, VA 24061, USA
| | - Maya Schuldiner
- Department of Molecular Genetics, Weizmann Institute of Sciences, Rehovot 7610001, Israel
| | - Xiaofeng Wang
- Department of Plant Pathology, Physiology, and Weed Science, Virginia Tech, Blacksburg, VA 24061, USA
| |
Collapse
|
127
|
Lee FHF, Su P, Xie YF, Wang KE, Wan Q, Liu F. Disrupting GluA2-GAPDH Interaction Affects Axon and Dendrite Development. Sci Rep 2016; 6:30458. [PMID: 27461448 PMCID: PMC4962050 DOI: 10.1038/srep30458] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 07/06/2016] [Indexed: 12/31/2022] Open
Abstract
GluA2-containing AMPA receptors (AMPARs) play a critical role in various aspects of neurodevelopment. However, the molecular mechanisms underlying these processes are largely unknown. We report here that the interaction between GluA2 and glyceraldehyde 3-phosphate dehydrogenase (GAPDH) is necessary for neuron and cortical development. Using an interfering peptide (GluA2-G-Gpep) that specifically disrupts this interaction, we found that primary neuron cultures with peptide treatment displayed growth cone development deficits, impairment of axon formation, less dendritic arborization and lower spine protrusion density. Consistently, in vivo data with mouse brains from pregnant dams injected with GluA2-G-Gpep daily during embryonic day 8 to 19 revealed a reduction of cortical tract axon integrity and neuronal density in post-natal day 1 offspring. Disruption of GluA2-GAPDH interaction also impairs the GluA2-Plexin A4 interaction and reduces p53 acetylation in mice, both of which are possible mechanisms leading to the observed neurodevelopmental abnormalities. Furthermore, electrophysiological experiments indicate altered long-term potentiation (LTP) in hippocampal slices of offspring mice. Our results provide novel evidence that AMPARs, specifically the GluA2 subunit via its interaction with GAPDH, play a critical role in cortical neurodevelopment.
Collapse
Affiliation(s)
- Frankie Hang Fung Lee
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, M5T 1R8 Canada
| | - Ping Su
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, M5T 1R8 Canada
| | - Yu-Feng Xie
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, M5T 1R8 Canada
| | - Kyle Ethan Wang
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, M5T 1R8 Canada
| | - Qi Wan
- Department of Physiology, School of Medicine, Wuhan University, Wuhan 430071, China
| | - Fang Liu
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, M5T 1R8 Canada.,Department of Psychiatry, University of Toronto, Toronto, Ontario, M5T 1R8 Canada
| |
Collapse
|
128
|
Abstract
Transport of newly synthesized proteins from the endoplasmic reticulum (ER) to the Golgi complex is highly selective. As a general rule, such transport is limited to soluble and membrane-associated secretory proteins that have reached properly folded and assembled conformations. To secure the efficiency, fidelity, and control of this crucial transport step, cells use a combination of mechanisms. The mechanisms are based on selective retention of proteins in the ER to prevent uptake into transport vesicles, on selective capture of proteins in COPII carrier vesicles, on inclusion of proteins in these vesicles by default as part of fluid and membrane bulk flow, and on selective retrieval of proteins from post-ER compartments by retrograde vesicle transport.
Collapse
Affiliation(s)
- Charles Barlowe
- Biochemistry Department, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire 03755;
| | - Ari Helenius
- Institute of Biochemistry, ETH Zurich, Zurich CH-8093, Switzerland
| |
Collapse
|
129
|
García-Nafría J, Herguedas B, Watson JF, Greger IH. The dynamic AMPA receptor extracellular region: a platform for synaptic protein interactions. J Physiol 2016; 594:5449-58. [PMID: 26891027 DOI: 10.1113/jp271844] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 01/21/2016] [Indexed: 12/27/2022] Open
Abstract
AMPA receptors (AMPARs) are glutamate-gated cation channels that mediate fast excitatory neurotransmission and synaptic plasticity. Structures of GluA2 homotetramers in distinct functional states, together with simulations, emphasise the loose architecture of the AMPAR extracellular region (ECR). The ECR encompasses ∼80% of the receptor, and consists of the membrane-distal N-terminal domain (NTD) and ligand-binding domain (LBD), which is fused to the ion channel domain. Minimal contacts within and between layers, together with flexible peptide linkers connecting these three domains give rise to an organisation capable of dynamic rearrangements. This building plan is uniquely suited to engage interaction partners in the crowded environment of synapses, permitting the formation of new binding sites and the loss of existing ones. ECR motions are thereby expected to impact signalling as well as synaptic anchorage and may thereby influence AMPAR clustering during synaptic plasticity.
Collapse
Affiliation(s)
- J García-Nafría
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - B Herguedas
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - J F Watson
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - I H Greger
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, UK.
| |
Collapse
|
130
|
NMDA receptors are selectively partitioned into complexes and supercomplexes during synapse maturation. Nat Commun 2016; 7:11264. [PMID: 27117477 PMCID: PMC5227094 DOI: 10.1038/ncomms11264] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 03/04/2016] [Indexed: 01/29/2023] Open
Abstract
How neuronal proteomes self-organize is poorly understood because of their inherent molecular and cellular complexity. Here, focusing on mammalian synapses we use blue-native PAGE and ‘gene-tagging' of GluN1 to report the first biochemical purification of endogenous NMDA receptors (NMDARs) directly from adult mouse brain. We show that NMDARs partition between two discrete populations of receptor complexes and ∼1.5 MDa supercomplexes. We tested the assembly mechanism with six mouse mutants, which indicates a tripartite requirement of GluN2B, PSD93 and PSD95 gate the incorporation of receptors into ∼1.5 MDa supercomplexes, independent of either canonical PDZ-ligands or GluN2A. Supporting the essential role of GluN2B, quantitative gene-tagging revealed a fourfold molar excess of GluN2B over GluN2A in adult forebrain. NMDAR supercomplexes are assembled late in postnatal development and triggered by synapse maturation involving epigenetic and activity-dependent mechanisms. Finally, screening the quaternary organization of 60 native proteins identified numerous discrete supercomplexes that populate the mammalian synapse. NMDARs and MAGUK proteins are capable of forming higher-order protein assemblies, however their organisation in the intact brain is unclear. Here, Frank et al. identify mouse and human supercomplexes and discover their mechanism of assembly using genetic tagging and affinity purification.
Collapse
|
131
|
Abstract
AMPA receptors (AMPARs) are assemblies of four core subunits, GluA1-4, that mediate most fast excitatory neurotransmission. The component subunits determine the functional properties of AMPARs, and the prevailing view is that the subunit composition also determines AMPAR trafficking, which is dynamically regulated during development, synaptic plasticity and in response to neuronal stress in disease. Recently, the subunit dependence of AMPAR trafficking has been questioned, leading to a reappraisal of this field. In this Review, we discuss what is known, uncertain, conjectured and unknown about the roles of the individual subunits, and how they affect AMPAR assembly, trafficking and function under both normal and pathological conditions.
Collapse
|
132
|
Matsuo H, Yamamoto K, Nakaoka H, Nakayama A, Sakiyama M, Chiba T, Takahashi A, Nakamura T, Nakashima H, Takada Y, Danjoh I, Shimizu S, Abe J, Kawamura Y, Terashige S, Ogata H, Tatsukawa S, Yin G, Okada R, Morita E, Naito M, Tokumasu A, Onoue H, Iwaya K, Ito T, Takada T, Inoue K, Kato Y, Nakamura Y, Sakurai Y, Suzuki H, Kanai Y, Hosoya T, Hamajima N, Inoue I, Kubo M, Ichida K, Ooyama H, Shimizu T, Shinomiya N. Genome-wide association study of clinically defined gout identifies multiple risk loci and its association with clinical subtypes. Ann Rheum Dis 2016; 75:652-9. [PMID: 25646370 PMCID: PMC4819613 DOI: 10.1136/annrheumdis-2014-206191] [Citation(s) in RCA: 135] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 12/22/2014] [Accepted: 01/06/2015] [Indexed: 01/28/2023]
Abstract
OBJECTIVE Gout, caused by hyperuricaemia, is a multifactorial disease. Although genome-wide association studies (GWASs) of gout have been reported, they included self-reported gout cases in which clinical information was insufficient. Therefore, the relationship between genetic variation and clinical subtypes of gout remains unclear. Here, we first performed a GWAS of clinically defined gout cases only. METHODS A GWAS was conducted with 945 patients with clinically defined gout and 1213 controls in a Japanese male population, followed by replication study of 1048 clinically defined cases and 1334 controls. RESULTS Five gout susceptibility loci were identified at the genome-wide significance level (p<5.0×10(-8)), which contained well-known urate transporter genes (ABCG2 and SLC2A9) and additional genes: rs1260326 (p=1.9×10(-12); OR=1.36) of GCKR (a gene for glucose and lipid metabolism), rs2188380 (p=1.6×10(-23); OR=1.75) of MYL2-CUX2 (genes associated with cholesterol and diabetes mellitus) and rs4073582 (p=6.4×10(-9); OR=1.66) of CNIH-2 (a gene for regulation of glutamate signalling). The latter two are identified as novel gout loci. Furthermore, among the identified single-nucleotide polymorphisms (SNPs), we demonstrated that the SNPs of ABCG2 and SLC2A9 were differentially associated with types of gout and clinical parameters underlying specific subtypes (renal underexcretion type and renal overload type). The effect of the risk allele of each SNP on clinical parameters showed significant linear relationships with the ratio of the case-control ORs for two distinct types of gout (r=0.96 [p=4.8×10(-4)] for urate clearance and r=0.96 [p=5.0×10(-4)] for urinary urate excretion). CONCLUSIONS Our findings provide clues to better understand the pathogenesis of gout and will be useful for development of companion diagnostics.
Collapse
Affiliation(s)
- Hirotaka Matsuo
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Ken Yamamoto
- Department of Medical Chemistry, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Hirofumi Nakaoka
- Division of Human Genetics, Department of Integrated Genetics, National Institute of Genetics, Mishima, Shizuoka, Japan
| | - Akiyoshi Nakayama
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
- Medical Group, Headquarters, Iwo-to Air Base Group, Japan Air Self-Defense Force, Tokyo, Japan
| | - Masayuki Sakiyama
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
- Department of Dermatology, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Toshinori Chiba
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Atsushi Takahashi
- Laboratory for Statistical Analysis, Center for Integrative Medical Sciences, RIKEN, Yokohama, Kanagawa, Japan
| | - Takahiro Nakamura
- Laboratory for Statistical Analysis, Center for Integrative Medical Sciences, RIKEN, Yokohama, Kanagawa, Japan
- Laboratory for Mathematics, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Hiroshi Nakashima
- Department of Preventive Medicine and Public Health, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Yuzo Takada
- The Central Research Institute, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Inaho Danjoh
- Cell Engineering Division, RIKEN BioResource Center, Tsukuba, Ibaraki, Japan
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Miyagi, Japan
| | - Seiko Shimizu
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Junko Abe
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Yusuke Kawamura
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Sho Terashige
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Hiraku Ogata
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Seishiro Tatsukawa
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Guang Yin
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
- Department of Nutritional Sciences, Faculty of Health and Welfare, Seinan Jo Gakuin University, Fukuoka, Japan
| | - Rieko Okada
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Emi Morita
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Mariko Naito
- Department of Preventive Medicine, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | | | - Hiroyuki Onoue
- Department of Internal Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Keiichi Iwaya
- Department of Pathology, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Toshimitsu Ito
- Department of Internal Medicine, Self-Defense Forces Central Hospital, Tokyo, Japan
| | - Tappei Takada
- Department of Pharmacy, The University of Tokyo Hospital, Tokyo, Japan
| | - Katsuhisa Inoue
- Department of Biopharmaceutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Yukio Kato
- Faculty of Pharmacy, Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Yukio Nakamura
- Cell Engineering Division, RIKEN BioResource Center, Tsukuba, Ibaraki, Japan
| | - Yutaka Sakurai
- Department of Preventive Medicine and Public Health, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Hiroshi Suzuki
- Department of Pharmacy, The University of Tokyo Hospital, Tokyo, Japan
| | - Yoshikatsu Kanai
- Division of Bio-system Pharmacology, Department of Pharmacology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Tatsuo Hosoya
- Division of Kidney and Hypertension, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan
- Department of Pathophysiology and Therapy in Chronic Kidney Disease, Jikei University School of Medicine, Tokyo, Japan
| | - Nobuyuki Hamajima
- Department of Healthcare Administration, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ituro Inoue
- Division of Human Genetics, Department of Integrated Genetics, National Institute of Genetics, Mishima, Shizuoka, Japan
| | - Michiaki Kubo
- Laboratory for Genotyping Development, Center for Integrative Medical Sciences, RIKEN, Yokohama, Kanagawa, Japan
| | - Kimiyoshi Ichida
- Division of Kidney and Hypertension, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan
- Department of Pathophysiology, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | | | | | - Nariyoshi Shinomiya
- Department of Integrative Physiology and Bio-Nano Medicine, National Defense Medical College, Tokorozawa, Saitama, Japan
| |
Collapse
|
133
|
Shisa6 traps AMPA receptors at postsynaptic sites and prevents their desensitization during synaptic activity. Nat Commun 2016; 7:10682. [PMID: 26931375 PMCID: PMC4778035 DOI: 10.1038/ncomms10682] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Accepted: 01/11/2016] [Indexed: 11/08/2022] Open
Abstract
Trafficking and biophysical properties of AMPA receptors (AMPARs) in the brain depend on interactions with associated proteins. We identify Shisa6, a single transmembrane protein, as a stable and directly interacting bona fide AMPAR auxiliary subunit. Shisa6 is enriched at hippocampal postsynaptic membranes and co-localizes with AMPARs. The Shisa6 C-terminus harbours a PDZ domain ligand that binds to PSD-95, constraining mobility of AMPARs in the plasma membrane and confining them to postsynaptic densities. Shisa6 expressed in HEK293 cells alters GluA1- and GluA2-mediated currents by prolonging decay times and decreasing the extent of AMPAR desensitization, while slowing the rate of recovery from desensitization. Using gene deletion, we show that Shisa6 increases rise and decay times of hippocampal CA1 miniature excitatory postsynaptic currents (mEPSCs). Shisa6-containing AMPARs show prominent sustained currents, indicating protection from full desensitization. Accordingly, Shisa6 prevents synaptically trapped AMPARs from depression at high-frequency synaptic transmission. Auxiliary AMPA receptor subunits can affect gating and surface mobility. Here the authors show that Shisa6 traps AMPA receptors at postsynaptic sites via PSD-95, and keeps them in an activated state in the presence of glutamate, preventing full desensitization and consequently synaptic depression.
Collapse
|
134
|
Gu X, Mao X, Lussier MP, Hutchison MA, Zhou L, Hamra FK, Roche KW, Lu W. GSG1L suppresses AMPA receptor-mediated synaptic transmission and uniquely modulates AMPA receptor kinetics in hippocampal neurons. Nat Commun 2016; 7:10873. [PMID: 26932439 PMCID: PMC4778064 DOI: 10.1038/ncomms10873] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Accepted: 01/28/2016] [Indexed: 12/17/2022] Open
Abstract
Regulation of AMPA receptor (AMPAR)-mediated synaptic transmission is a key mechanism
for synaptic plasticity. In the brain, AMPARs assemble with a number of auxiliary
subunits, including TARPs, CNIHs and CKAMP44, which are important for AMPAR forward
trafficking to synapses. Here we report that the membrane protein GSG1L negatively
regulates AMPAR-mediated synaptic transmission. Overexpression of GSG1L strongly
suppresses, and GSG1L knockout (KO) enhances, AMPAR-mediated synaptic transmission.
GSG1L-dependent regulation of AMPAR synaptic transmission relies on the first
extracellular loop domain and its carboxyl-terminus. GSG1L also speeds up AMPAR
deactivation and desensitization in hippocampal CA1 neurons, in contrast to the
effects of TARPs and CNIHs. Furthermore, GSG1L association with AMPARs inhibits
CNIH2-induced slowing of the receptors in heterologous cells. Finally, GSG1L KO rats
have deficits in LTP and show behavioural abnormalities in object recognition tests.
These data demonstrate that GSG1L represents a new class of auxiliary subunit with
distinct functional properties for AMPARs. The molecular mechanism controlling the trafficking and function of
AMPARs at synapses are not fully understood. Here the authors show that GSG1L, a
membrane protein, negatively regulates AMPAR-mediated synaptic transmission and
represents a new class of AMPAR auxiliary subunit.
Collapse
Affiliation(s)
- Xinglong Gu
- Synapse and Neural Circuit Research Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 35 Convent Drive, 3C1000, Bethesda, Maryland 20892, USA
| | - Xia Mao
- Synapse and Neural Circuit Research Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 35 Convent Drive, 3C1000, Bethesda, Maryland 20892, USA
| | - Marc P Lussier
- Receptor Biology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 35 Convent Drive, 2C903, Bethesda, Maryland 20892, USA
| | - Mary Anne Hutchison
- Synapse and Neural Circuit Research Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 35 Convent Drive, 3C1000, Bethesda, Maryland 20892, USA
| | - Liang Zhou
- Synapse and Neural Circuit Research Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 35 Convent Drive, 3C1000, Bethesda, Maryland 20892, USA
| | - F Kent Hamra
- Department of Pharmacology, Cecil H. &Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center in Dallas, Dallas, Texas 75390, USA
| | - Katherine W Roche
- Receptor Biology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 35 Convent Drive, 2C903, Bethesda, Maryland 20892, USA
| | - Wei Lu
- Synapse and Neural Circuit Research Unit, National Institute of Neurological Disorders and Stroke, National Institutes of Health, 35 Convent Drive, 3C1000, Bethesda, Maryland 20892, USA
| |
Collapse
|
135
|
Li B, Rex E, Wang H, Qian Y, Ogden AM, Bleakman D, Johnson KW. Pharmacological Modulation of GluK1 and GluK2 by NETO1, NETO2, and PSD95. Assay Drug Dev Technol 2016; 14:131-43. [PMID: 26991362 DOI: 10.1089/adt.2015.689] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The association between the kainate receptors (KARs) GluK1 and GluK2 and the modifying proteins neuropilin- and tolloid-like 1 (NETO1), neuropilin- and tolloid-like 2 (NETO2), and postsynaptic density protein 95 (PSD95) is likely to produce distinct GluK1 and GluK2 pharmacology in postsynaptic neurons. However, little is known about their corresponding modulatory effects on GluK1 and GluK2 activity in high-throughput assays for cell-based drug discovery. Using heterologous cells that potentially mimic the response in native cells in a fluorescence imaging plate reader (FLIPR) assay, we have investigated assays that incorporate (1) coexpression of GluK1 or GluK2 with their modulatory proteins (NETO1, NETO2, PSD95) and/or (2) enablement of assays with physiological concentration of native GluK1 and GluK2 agonist (glutamate) in the absence of an artificial potentiator (e.g., concanavalin A [Con A]). We found that in the absence of Con A, both NETO1 and NETO2 accessory proteins are able to potentiate kainate- and glutamate-evoked GluK1-mediated Ca(2+) influx. We also noted the striking ability of PSD95 to enhance glutamate-stimulated potentiation effects of NETO2 on GluK1 without the need for Con A and with a robust signal that could be utilized for high-throughput FLIPR assays. These experiments demonstrate the utility of heterologous cells coexpressing PSD95/NETO2 with GluK1 or GluK2 in native cell-mimicking heterologous cell systems for high-throughput assays and represent new avenues into the discovery of KAR modulating therapies.
Collapse
Affiliation(s)
- Baolin Li
- 1 Neuroscience Discovery, Lilly Corporate Center , Eli Lilly and Company, Indianapolis, Indiana
| | - Elizabeth Rex
- 1 Neuroscience Discovery, Lilly Corporate Center , Eli Lilly and Company, Indianapolis, Indiana
| | - He Wang
- 2 TTx-Reagents-Proteins, Lilly Corporate Center , Eli Lilly and Company, Indianapolis, Indiana
| | - Yuewei Qian
- 2 TTx-Reagents-Proteins, Lilly Corporate Center , Eli Lilly and Company, Indianapolis, Indiana
| | - Ann Marie Ogden
- 1 Neuroscience Discovery, Lilly Corporate Center , Eli Lilly and Company, Indianapolis, Indiana
| | - David Bleakman
- 1 Neuroscience Discovery, Lilly Corporate Center , Eli Lilly and Company, Indianapolis, Indiana
| | - Kirk W Johnson
- 1 Neuroscience Discovery, Lilly Corporate Center , Eli Lilly and Company, Indianapolis, Indiana
| |
Collapse
|
136
|
Dawe GB, Musgaard M, Aurousseau MRP, Nayeem N, Green T, Biggin PC, Bowie D. Distinct Structural Pathways Coordinate the Activation of AMPA Receptor-Auxiliary Subunit Complexes. Neuron 2016; 89:1264-1276. [PMID: 26924438 PMCID: PMC4819453 DOI: 10.1016/j.neuron.2016.01.038] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 12/01/2015] [Accepted: 01/13/2016] [Indexed: 11/15/2022]
Abstract
Neurotransmitter-gated ion channels adopt different gating modes to fine-tune signaling at central synapses. At glutamatergic synapses, high and low activity of AMPA receptors (AMPARs) is observed when pore-forming subunits coassemble with or without auxiliary subunits, respectively. Whether a common structural pathway accounts for these different gating modes is unclear. Here, we identify two structural motifs that determine the time course of AMPAR channel activation. A network of electrostatic interactions at the apex of the AMPAR ligand-binding domain (LBD) is essential for gating by pore-forming subunits, whereas a conserved motif on the lower, D2 lobe of the LBD prolongs channel activity when auxiliary subunits are present. Accordingly, channel activity is almost entirely abolished by elimination of the electrostatic network but restored via auxiliary protein interactions at the D2 lobe. In summary, we propose that activation of native AMPAR complexes is coordinated by distinct structural pathways, favored by the association/dissociation of auxiliary subunits. Two distinct structural motifs control the time course of AMPA receptor gating Intraprotein electrostatic interactions govern gating by pore-forming subunits Auxiliary subunits act at a distinct site to prolong channel activity Intra- and interprotein interactions coordinate signaling by AMPA receptor complexes
Collapse
Affiliation(s)
- G Brent Dawe
- Integrated Program in Neuroscience, McGill University, Montréal, QC H3A 2B4, Canada; Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3G 1Y6, Canada
| | - Maria Musgaard
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK
| | - Mark R P Aurousseau
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3G 1Y6, Canada
| | - Naushaba Nayeem
- Department of Pharmacology, University of Liverpool, Liverpool L69 3BX, UK
| | - Tim Green
- Department of Pharmacology, University of Liverpool, Liverpool L69 3BX, UK.
| | - Philip C Biggin
- Department of Biochemistry, University of Oxford, Oxford OX1 3QU, UK.
| | - Derek Bowie
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3G 1Y6, Canada.
| |
Collapse
|
137
|
Erlenhardt N, Yu H, Abiraman K, Yamasaki T, Wadiche JI, Tomita S, Bredt DS. Porcupine Controls Hippocampal AMPAR Levels, Composition, and Synaptic Transmission. Cell Rep 2016; 14:782-794. [PMID: 26776514 DOI: 10.1016/j.celrep.2015.12.078] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2014] [Revised: 11/19/2015] [Accepted: 12/15/2015] [Indexed: 12/17/2022] Open
Abstract
AMPA receptor (AMPAR) complexes contain auxiliary subunits that modulate receptor trafficking and gating. In addition to the transmembrane AMPAR regulatory proteins (TARPs) and cornichons (CNIH-2/3), recent proteomic studies identified a diverse array of additional AMPAR-associated transmembrane and secreted partners. We systematically surveyed these and found that PORCN and ABHD6 increase GluA1 levels in transfected cells. Knockdown of PORCN in rat hippocampal neurons, which express it in high amounts, selectively reduces levels of all tested AMPAR complex components. Regulation of AMPARs is independent of PORCN's membrane-associated O-acyl transferase activity. PORCN knockdown in hippocampal neurons decreases AMPAR currents and accelerates desensitization and leads to depletion of TARP γ-8 from AMPAR complexes. Conditional PORCN knockout mice also exhibit specific changes in AMPAR expression and gating that reduce basal synaptic transmission but leave long-term potentiation intact. These studies define additional roles for PORCN in controlling synaptic transmission by regulating the level and composition of hippocampal AMPAR complexes.
Collapse
Affiliation(s)
- Nadine Erlenhardt
- Neuroscience Discovery, Janssen Pharmaceutical Companies of Johnson & Johnson, 3210 Merryfield Row, San Diego, CA 92121, USA; Institute of Neural and Sensory Physiology, Medical Faculty, University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Hong Yu
- Neuroscience Discovery, Janssen Pharmaceutical Companies of Johnson & Johnson, 3210 Merryfield Row, San Diego, CA 92121, USA
| | - Kavitha Abiraman
- Department of Neurobiology, McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Tokiwa Yamasaki
- CNNR program, Department of Cellular and Molecular Physiology, Yale University School of Medicine, 295 Congress Avenue BCMM441, P.O. Box 208026, New Haven, CT 06510, USA
| | - Jacques I Wadiche
- Department of Neurobiology, McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Susumu Tomita
- CNNR program, Department of Cellular and Molecular Physiology, Yale University School of Medicine, 295 Congress Avenue BCMM441, P.O. Box 208026, New Haven, CT 06510, USA
| | - David S Bredt
- Neuroscience Discovery, Janssen Pharmaceutical Companies of Johnson & Johnson, 3210 Merryfield Row, San Diego, CA 92121, USA.
| |
Collapse
|
138
|
AMPA receptor-positive allosteric modulators for the treatment of schizophrenia: an overview of recent patent applications. Future Med Chem 2016; 7:473-91. [PMID: 25875874 DOI: 10.4155/fmc.15.4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The role of glutamate and its receptors in central nervous system biology and disease has long been of interest to scientists involved in both fundamental research and drug discovery, however the complex pharmacology and lack of highly selective compounds has severely hampered drug discovery efforts in this area. Recent advances in the identification and profiling of positive allosteric modulators of the AMPA receptor offer a potential way forward and the hope of a new treatment for schizophrenia. This article will review recent patent applications published in this area.
Collapse
|
139
|
Jang S, Oh D, Lee Y, Hosy E, Shin H, van Riesen C, Whitcomb D, Warburton JM, Jo J, Kim D, Kim SG, Um SM, Kwon SK, Kim MH, Roh JD, Woo J, Jun H, Lee D, Mah W, Kim H, Kaang BK, Cho K, Rhee JS, Choquet D, Kim E. Synaptic adhesion molecule IgSF11 regulates synaptic transmission and plasticity. Nat Neurosci 2016; 19:84-93. [PMID: 26595655 PMCID: PMC5010778 DOI: 10.1038/nn.4176] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 10/20/2015] [Indexed: 12/11/2022]
Abstract
Synaptic adhesion molecules regulate synapse development and plasticity through mechanisms that include trans-synaptic adhesion and recruitment of diverse synaptic proteins. We found that the immunoglobulin superfamily member 11 (IgSF11), a homophilic adhesion molecule that preferentially expressed in the brain, is a dual-binding partner of the postsynaptic scaffolding protein PSD-95 and AMPA glutamate receptors (AMPARs). IgSF11 required PSD-95 binding for its excitatory synaptic localization. In addition, IgSF11 stabilized synaptic AMPARs, as determined by IgSF11 knockdown-induced suppression of AMPAR-mediated synaptic transmission and increased surface mobility of AMPARs, measured by high-throughput, single-molecule tracking. IgSF11 deletion in mice led to the suppression of AMPAR-mediated synaptic transmission in the dentate gyrus and long-term potentiation in the CA1 region of the hippocampus. IgSF11 did not regulate the functional characteristics of AMPARs, including desensitization, deactivation or recovery. These results suggest that IgSF11 regulates excitatory synaptic transmission and plasticity through its tripartite interactions with PSD-95 and AMPARs.
Collapse
Affiliation(s)
- Seil Jang
- Department of Biological Sciences, Korea Advanced Institute of
Science and Technology (KAIST), Daejeon 305-701, Korea
| | - Daeyoung Oh
- Department of Biomedical Sciences, Korea Advanced Institute of
Science and Technology (KAIST), Daejeon 305-701, Korea
- Department of Psychiatry, CHA Bundang Medical Center, CHA
University, Seoul, Korea
| | - Yeunkum Lee
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science
(IBS), Daejeon 305-701, Korea
| | - Eric Hosy
- University of Bordeaux, Interdisciplinary Institute for
Neuroscience, France; CNRS UMR 5297, F-33000 Bordeaux, France
| | - Hyewon Shin
- Department of Biomedical Sciences, Korea Advanced Institute of
Science and Technology (KAIST), Daejeon 305-701, Korea
| | - Christoph van Riesen
- Department of Molecular Neurobiology, Max Planck Institute of
Experimental Medicine, D-37075 Göttingen, Germany
| | - Daniel Whitcomb
- School of Clinical Sciences, Faculty of Medicine and Dentistry,
University of Bristol, Whitson street, Bristol, UK
- Centre for Synaptic Plasticity, University of Bristol, Whitson
street, Bristol, UK
| | - Julia M. Warburton
- School of Clinical Sciences, Faculty of Medicine and Dentistry,
University of Bristol, Whitson street, Bristol, UK
| | - Jihoon Jo
- School of Clinical Sciences, Faculty of Medicine and Dentistry,
University of Bristol, Whitson street, Bristol, UK
- Department of Biomedical Sciences, Chonnam National University
Medical School, Gwangju, South Korea
| | - Doyoun Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science
(IBS), Daejeon 305-701, Korea
| | - Sun Gyun Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science
(IBS), Daejeon 305-701, Korea
| | - Seung Min Um
- Department of Biological Sciences, Korea Advanced Institute of
Science and Technology (KAIST), Daejeon 305-701, Korea
| | - Seok-kyu Kwon
- Department of Biological Sciences, Korea Advanced Institute of
Science and Technology (KAIST), Daejeon 305-701, Korea
| | - Myoung-Hwan Kim
- Department of Physiology, Seoul National University College of
Medicine, Seoul 110-799, Republic of Korea
- Seoul National University Bundang Hospital, Seongnam, Gyeonggi
463-707, Republic of Korea
| | - Junyeop Daniel Roh
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science
(IBS), Daejeon 305-701, Korea
| | - Jooyeon Woo
- Department of Biological Sciences, Korea Advanced Institute of
Science and Technology (KAIST), Daejeon 305-701, Korea
| | - Heejung Jun
- Brain and Cognitive Sciences, College of Natural Sciences, Seoul
National University, Seoul 151-747, Korea
| | - Dongmin Lee
- Department of Anatomy and Division of Brain Korea 21 Biomedical
Science, College of Medicine, Korea University, 126-1, 5-Ka, Anam-Dong, Seongbuk-Gu,
Seoul 136-705, Korea
| | - Won Mah
- Department of Anatomy and Neurobiology, School of Dentistry,
Kyungpook National University, Daegu 700-412, Korea
| | - Hyun Kim
- Department of Anatomy and Division of Brain Korea 21 Biomedical
Science, College of Medicine, Korea University, 126-1, 5-Ka, Anam-Dong, Seongbuk-Gu,
Seoul 136-705, Korea
| | - Bong-Kiun Kaang
- Brain and Cognitive Sciences, College of Natural Sciences, Seoul
National University, Seoul 151-747, Korea
| | - Kwangwook Cho
- School of Clinical Sciences, Faculty of Medicine and Dentistry,
University of Bristol, Whitson street, Bristol, UK
- Centre for Synaptic Plasticity, University of Bristol, Whitson
street, Bristol, UK
| | - Jeong-Seop Rhee
- Department of Molecular Neurobiology, Max Planck Institute of
Experimental Medicine, D-37075 Göttingen, Germany
| | - Daniel Choquet
- University of Bordeaux, Interdisciplinary Institute for
Neuroscience, France; CNRS UMR 5297, F-33000 Bordeaux, France
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute of
Science and Technology (KAIST), Daejeon 305-701, Korea
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science
(IBS), Daejeon 305-701, Korea
| |
Collapse
|
140
|
Kirk LM, Ti SW, Bishop HI, Orozco-Llamas M, Pham M, Trimmer JS, Díaz E. Distribution of the SynDIG4/proline-rich transmembrane protein 1 in rat brain. J Comp Neurol 2015; 524:2266-80. [PMID: 26660156 DOI: 10.1002/cne.23945] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 11/17/2015] [Accepted: 12/01/2015] [Indexed: 12/22/2022]
Abstract
The modulation of AMPA receptor (AMPAR) content at synapses is thought to be an underlying molecular mechanism of memory and learning. AMPAR content at synapses is highly plastic and is regulated by numerous AMPAR accessory transmembrane proteins such as TARPs, cornichons, and CKAMPs. SynDIG (synapse differentiation-induced gene) defines a family of four genes (SynDIG1-4) expressed in distinct and overlapping patterns in the brain. SynDIG1 was previously identified as a novel transmembrane AMPAR-associated protein that regulates synaptic strength. The related protein SynDIG4 [also known as Prrt1 (proline-rich transmembrane protein 1)] has recently been identified as a component of AMPAR complexes. In this study, we show that SynDIG1 and SynDIG4 have distinct yet overlapping patterns of expression in the central nervous system, with SynDIG4 having especially prominent expression in the hippocampus and particularly within CA1. In contrast to SynDIG1 and other traditional AMPAR auxiliary subunits, SynDIG4 is de-enriched at the postsynaptic density and colocalizes with extrasynaptic GluA1 puncta in primary dissociated neuron culture. These results indicate that, although SynDIG4 shares sequence similarity with SynDIG1, it might act through a unique mechanism as an auxiliary factor for extrasynaptic GluA1-containing AMPARs. J. Comp. Neurol. 524:2266-2280, 2016. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Lyndsey M Kirk
- Department of Pharmacology, University of California Davis School of Medicine, Davis, California, 95616
| | - Shu W Ti
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, California, 95616
| | - Hannah I Bishop
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, California, 95616
| | - Mayra Orozco-Llamas
- Department of Pharmacology, University of California Davis School of Medicine, Davis, California, 95616
| | - Michelle Pham
- Department of Pharmacology, University of California Davis School of Medicine, Davis, California, 95616
| | - James S Trimmer
- Department of Neurobiology, Physiology and Behavior, University of California, Davis, California, 95616.,Department of Physiology and Membrane Biology, University of California Davis School of Medicine, Davis, California, 95616
| | - Elva Díaz
- Department of Pharmacology, University of California Davis School of Medicine, Davis, California, 95616
| |
Collapse
|
141
|
Modular composition and dynamics of native GABAB receptors identified by high-resolution proteomics. Nat Neurosci 2015; 19:233-42. [PMID: 26691831 DOI: 10.1038/nn.4198] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 11/16/2015] [Indexed: 01/04/2023]
Abstract
GABAB receptors, the most abundant inhibitory G protein-coupled receptors in the mammalian brain, display pronounced diversity in functional properties, cellular signaling and subcellular distribution. We used high-resolution functional proteomics to identify the building blocks of these receptors in the rodent brain. Our analyses revealed that native GABAB receptors are macromolecular complexes with defined architecture, but marked diversity in subunit composition: the receptor core is assembled from GABAB1a/b, GABAB2, four KCTD proteins and a distinct set of G-protein subunits, whereas the receptor's periphery is mostly formed by transmembrane proteins of different classes. In particular, the periphery-forming constituents include signaling effectors, such as Cav2 and HCN channels, and the proteins AJAP1 and amyloid-β A4, both of which tightly associate with the sushi domains of GABAB1a. Our results unravel the molecular diversity of GABAB receptors and their postnatal assembly dynamics and provide a roadmap for studying the cellular signaling of this inhibitory neurotransmitter receptor.
Collapse
|
142
|
Farrow P, Khodosevich K, Sapir Y, Schulmann A, Aslam M, Stern-Bach Y, Monyer H, von Engelhardt J. Auxiliary subunits of the CKAMP family differentially modulate AMPA receptor properties. eLife 2015; 4:e09693. [PMID: 26623514 PMCID: PMC4733035 DOI: 10.7554/elife.09693] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 11/30/2015] [Indexed: 01/01/2023] Open
Abstract
AMPA receptor (AMPAR) function is modulated by auxiliary subunits. Here, we report on three AMPAR interacting proteins—namely CKAMP39, CKAMP52 and CKAMP59—that, together with the previously characterized CKAMP44, constitute a novel family of auxiliary subunits distinct from other families of AMPAR interacting proteins. The new members of the CKAMP family display distinct regional and developmental expression profiles in the mouse brain. Notably, despite their structural similarities they exert diverse modulation on AMPAR gating by influencing deactivation, desensitization and recovery from desensitization, as well as glutamate and cyclothiazide potency to AMPARs. This study indicates that AMPAR function is very precisely controlled by the cell-type specific expression of the CKAMP family members. The brain processes and transmits information through large networks of cells called neurons. A neuron can pass the information it receives to other neurons by releasing chemicals called neurotransmitters across junctions known as synapses. These chemicals bind to receptor proteins on the surface of the neighboring neuron, which triggers changes that affect the activity of this neuron. Glutamate is the most commonly used neurotransmitter in the brain and binds to receptor proteins called AMPA receptors. If a neuron frequently sends glutamate across a particular synapse, the number of AMPA receptors in the second neuron will increase in response. This makes signaling across the synapse easier – a process known as synaptic strengthening. The ability to change the strength of synapses is important for learning and memory. Proteins called auxiliary subunits also bind to AMPA receptors and regulate their properties, and hence also affect the strength of the synapse. For instance, some auxiliary subunits increase the number of AMPA receptors at the synapse, while others have an effect on how the receptor protein works. In 2010, researchers identified a new auxiliary protein called CKAMP44 that modifies AMPA receptor activity. Now, Farrow, Khodosevich, Sapir, Schulmann et al. – including some of the researchers involved in the 2010 study – have identified three other auxiliary proteins that are similar to CKAMP44. Collectively, these four proteins are termed the CKAMP family. The sequences of all four proteins were found to share many common features, especially in the regions that bind to the AMPA receptors. Like CKAMP44, the new members of the CKAMP family are only present in the brain, although each protein is produced in different brain regions. Further investigation revealed that each member of the CKAMP family affects the AMPA receptor channels in a different way. Taken together, Farrow et al.’s results suggest that the different CKAMP family members allow the activity of the AMPA receptors to be precisely controlled. The next challenge is to understand in more detail how each CKAMP family member influences how AMPA receptors work.
Collapse
Affiliation(s)
- Paul Farrow
- Synaptic Signalling and Neurodegeneration, German Cancer Research Center, Heidelberg, Germany.,Synaptic Signalling and Neurodegeneration, German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Konstantin Khodosevich
- Department of Clinical Neurobiology, Medical Faculty of Heidelberg University, Heidelberg, Germany.,German Cancer Research Center, Heidelberg, Germany
| | - Yechiam Sapir
- Department of Biochemistry and Molecular Biology, Institute for Medical Research - Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Anton Schulmann
- Department of Clinical Neurobiology, Medical Faculty of Heidelberg University, Heidelberg, Germany.,German Cancer Research Center, Heidelberg, Germany
| | - Muhammad Aslam
- Synaptic Signalling and Neurodegeneration, German Cancer Research Center, Heidelberg, Germany.,Synaptic Signalling and Neurodegeneration, German Center for Neurodegenerative Diseases, Bonn, Germany
| | - Yael Stern-Bach
- Department of Biochemistry and Molecular Biology, Institute for Medical Research - Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Hannah Monyer
- Department of Clinical Neurobiology, Medical Faculty of Heidelberg University, Heidelberg, Germany
| | - Jakob von Engelhardt
- Synaptic Signalling and Neurodegeneration, German Cancer Research Center, Heidelberg, Germany.,Synaptic Signalling and Neurodegeneration, German Center for Neurodegenerative Diseases, Bonn, Germany
| |
Collapse
|
143
|
Falsafi SK, Ghafari M, Miklósi AG, Engidawork E, Gröger M, Höger H, Lubec G. Mouse hippocampal GABAB1 but not GABAB2 subunit-containing receptor complex levels are paralleling retrieval in the multiple-T-maze. Front Behav Neurosci 2015; 9:276. [PMID: 26539091 PMCID: PMC4609755 DOI: 10.3389/fnbeh.2015.00276] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 09/28/2015] [Indexed: 11/13/2022] Open
Abstract
GABAB receptors are heterodimeric G-protein coupled receptors known to be involved in learning and memory. Although a role for GABAB receptors in cognitive processes is evident, there is no information on hippocampal GABAB receptor complexes in a multiple T maze (MTM) task, a robust paradigm for evaluation of spatial learning. Trained or untrained (yoked control) C57BL/6J male mice (n = 10/group) were subjected to the MTM task and sacrificed 6 h following their performance. Hippocampi were taken, membrane proteins extracted and run on blue native PAGE followed by immunoblotting with specific antibodies against GABAB1, GABAB1a, and GABAB2. Immunoprecipitation with subsequent mass spectrometric identification of co-precipitates was carried out to show if GABAB1 and GABAB2 as well as other interacting proteins co-precipitate. An antibody shift assay (ASA) and a proximity ligation assay (PLA) were also used to see if the two GABAB subunits are present in the receptor complex. Single bands were observed on Western blots, each representing GABAB1, GABAB1a, or GABAB2 at an apparent molecular weight of approximately 100 kDa. Subsequently, densitometric analysis revealed that levels of GABAB1 and GABAB1a but not GABAB2- containing receptor complexes were significantly higher in trained than untrained groups. Immunoprecipitation followed by mass spectrometric studies confirmed the presence of GABAB1, GABAB2, calcium calmodulin kinases I and II, GluA1 and GluA2 as constituents of the complex. ASA and PLA also showed the presence of the two subunits of GABAB receptor within the complex. It is shown that increased levels of GABAB1 subunit-containing complexes are paralleling performance in a land maze.
Collapse
Affiliation(s)
- Soheil K Falsafi
- Department of Pediatrics, Medical University of Vienna Vienna, Austria
| | - Maryam Ghafari
- Department of Pediatrics, Medical University of Vienna Vienna, Austria
| | - András G Miklósi
- Department of Pediatrics, Medical University of Vienna Vienna, Austria
| | - Ephrem Engidawork
- Department of Pharmacology and Clinical Pharmacy, School of Pharmacy, College of Health Sciences, Addis Ababa University Addis Ababa, Ethiopia
| | - Marion Gröger
- Skin and Endothelium Research Division, Department of Dermatology, Medical University Vienna, Austria
| | - Harald Höger
- Core Unit of Biomedical Research, Division of Laboratory Animal Science and Genetics, Medical University of Vienna Vienna, Austria
| | - Gert Lubec
- Department of Pediatrics, Medical University of Vienna Vienna, Austria
| |
Collapse
|
144
|
Schroeter A, Wen S, Mölders A, Erlenhardt N, Stein V, Klöcker N. Depletion of the AMPAR reserve pool impairs synaptic plasticity in a model of hepatic encephalopathy. Mol Cell Neurosci 2015; 68:331-9. [DOI: 10.1016/j.mcn.2015.09.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 07/22/2015] [Accepted: 09/06/2015] [Indexed: 02/07/2023] Open
|
145
|
Advances in the pharmacology of lGICs auxiliary subunits. Pharmacol Res 2015; 101:65-73. [PMID: 26255765 DOI: 10.1016/j.phrs.2015.07.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 07/25/2015] [Accepted: 07/26/2015] [Indexed: 11/21/2022]
Abstract
Ligand-gated ion channels (LGICs) are cell surface integral proteins that mediate the fast neurotransmission in the nervous system. LGICs require auxiliary subunits for their trafficking, assembly and pharmacological modulation. Auxiliary subunits do not form functional homomeric receptors, but are reported to assemble with the principal subunits in order to modulate their pharmacological profiles. For example, nACh receptors are built at least by co-assemble of α and β subunits, and the neuronal auxiliary subunits β3 and α5 and muscle type β, δ, γ, and ϵ determine the agonist affinity of these receptors. Serotonergic 5-HT3B, 5-HT3C, 5-HT3D and 5-HT3E are reported to assemble with the 5-HT3A subunit to modulate its pharmacological profile. Functional studies evaluating the role of γ2 and δ auxiliary subunits of GABAA receptors have made important advances in the understanding of the action of benzodiazepines, ethanol and neurosteroids. Glycine receptors are composed principally by α1-3 subunits and the auxiliary subunit β determines their synaptic location and their pharmacological response to propofol and ethanol. NMDA receptors appear to be functional as heterotetrameric channels. So far, the existence of NMDA auxiliary subunits is controversial. On the other hand, Kainate receptors are modulated by NETO 1 and 2. AMPA receptors are modulated by TARPs, Shisa 9, CKAMP44, CNIH2-3 auxiliary proteins reported that controls their trafficking, conductance and gating of channels. P2X receptors are able to associate with auxiliary Pannexin-1 protein to modulate P2X7 receptors. Considering the pharmacological relevance of different LGICs auxiliary subunits in the present work we will highlight the therapeutic potential of these modulator proteins.
Collapse
|
146
|
MacLean DM, Ramaswamy SS, Du M, Howe JR, Jayaraman V. Stargazin promotes closure of the AMPA receptor ligand-binding domain. ACTA ACUST UNITED AC 2015; 144:503-12. [PMID: 25422502 PMCID: PMC4242809 DOI: 10.1085/jgp.201411287] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Stargazin enhances closure of the AMPA receptor ligand-binding domain, thereby facilitating channel activation. Transmembrane AMPA receptor (AMPAR) regulatory proteins (TARPs) markedly enhance AMPAR function, altering ligand efficacy and receptor gating kinetics and thereby shaping the postsynaptic response. The structural mechanism underlying TARP effects on gating, however, is unknown. Here we find that the prototypical member of the TARP family, stargazin or γ-2, rescues gating deficits in AMPARs carrying mutations that destabilize the closed-cleft states of the ligand-binding domain (LBD), suggesting that stargazin reverses the effects of these mutations and likely stabilizes closed LBD states. Furthermore, stargazin promotes a more closed conformation of the LBD, as indicated by reduced accessibility to the large antagonist NBQX. Consistent with the functional studies, luminescence resonance energy transfer experiments directly demonstrate that the AMPAR LBD is on average more closed in the presence of stargazin, in both the apo and agonist-bound states. The additional cleft closure and/or stabilization of the more closed-cleft states of the LBD is expected to translate to higher agonist efficacy and could contribute to the structural mechanism for stargazin modulation of AMPAR function.
Collapse
Affiliation(s)
- David M MacLean
- Department of Biochemistry and Molecular Biology, Center for Membrane Biology, University of Texas Health Science Center, Houston, TX 77030
| | - Swarna S Ramaswamy
- Department of Biochemistry and Molecular Biology, Center for Membrane Biology, University of Texas Health Science Center, Houston, TX 77030
| | - Mei Du
- Department of Biochemistry and Molecular Biology, Center for Membrane Biology, University of Texas Health Science Center, Houston, TX 77030
| | - James R Howe
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06530
| | - Vasanthi Jayaraman
- Department of Biochemistry and Molecular Biology, Center for Membrane Biology, University of Texas Health Science Center, Houston, TX 77030
| |
Collapse
|
147
|
|
148
|
Transmembrane AMPAR regulatory protein γ-2 is required for the modulation of GABA release by presynaptic AMPARs. J Neurosci 2015; 35:4203-14. [PMID: 25762667 DOI: 10.1523/jneurosci.4075-14.2015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Presynaptic ionotropic glutamate receptors (iGluRs) play important roles in the control of synaptogenesis and neurotransmitter release, yet their regulation is poorly understood. In particular, the contribution of transmembrane auxiliary proteins, which profoundly shape the trafficking and gating of somatodendritic iGluRs, is unknown. Here we examined the influence of transmembrane AMPAR regulatory proteins (TARPs) on presynaptic AMPARs in cerebellar molecular layer interneurons (MLIs). 6-cyano-7-nitroquinoxaline-2,3-dione (CNQX), a partial agonist at TARP-associated AMPARs, enhanced spontaneous GABA release in wild-type mice but not in stargazer mice that lack the prototypical TARP stargazin (γ-2). These findings were replicated in mechanically dissociated Purkinje cells with functional adherent synaptic boutons, demonstrating the presynaptic locus of modulation. In dissociated Purkinje cells from stargazer mice, AMPA was able to enhance mIPSC frequency, but only in the presence of the positive allosteric modulator cyclothiazide. Thus, ordinarily, presynaptic AMPARs are unable to enhance spontaneous release without γ-2, which is required predominantly for its effects on channel gating. Presynaptic AMPARs are known to reduce action potential-driven GABA release from MLIs. Although a G-protein-dependent non-ionotropic mechanism has been suggested to underlie this inhibition, paradoxically we found that γ-2, and thus AMPAR gating, was required. Following glutamate spillover from climbing fibers or application of CNQX, evoked GABA release was reduced; in stargazer mice such effects were markedly attenuated in acute slices and abolished in the dissociated Purkinje cell-nerve bouton preparation. We suggest that γ-2 association, by increasing charge transfer, allows presynaptic AMPARs to depolarize the bouton membrane sufficiently to modulate both phasic and spontaneous release.
Collapse
|
149
|
Rosas-Santiago P, Lagunas-Gómez D, Barkla BJ, Vera-Estrella R, Lalonde S, Jones A, Frommer WB, Zimmermannova O, Sychrová H, Pantoja O. Identification of rice cornichon as a possible cargo receptor for the Golgi-localized sodium transporter OsHKT1;3. JOURNAL OF EXPERIMENTAL BOTANY 2015; 66:2733-48. [PMID: 25750424 PMCID: PMC4986874 DOI: 10.1093/jxb/erv069] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Membrane proteins are synthesized and folded in the endoplasmic reticulum (ER), and continue their path to their site of residence along the secretory pathway. The COPII system has been identified as a key player for selecting and directing the fate of membrane and secretory cargo proteins. Selection of cargo proteins within the COPII vesicles is achieved by cargo receptors. The cornichon cargo receptor belongs to a conserved protein family found in eukaryotes that has been demonstrated to participate in the selection of integral membrane proteins as cargo for their correct targeting. Here it is demonstrated at the cellular level that rice cornichon OsCNIH1 interacts with OsHKT1;3 and, in yeast cells, enables the expression of the sodium transporter to the Golgi apparatus. Physical and functional HKT-cornichon interactions are confirmed by the mating-based split ubiquitin system, bimolecular fluorescence complementation, and Xenopus oocyte and yeast expression systems. The interaction between the two proteins occurs in the ER of plant cells and their co-expression in oocytes leads to the sequestration of the transporter in the ER. In the yeast cornichon mutant erv14, OsHKT1;3 is mistargeted, preventing the toxic effects of sodium transport in the cell observed in wild-type cells or in the erv14 mutant that co-expressed OsHKT1;3 with either OsCNIH1 or Erv14p. Identification and characterization of rice cornichon as a possible cargo receptor opens up the opportunity to improve our knowledge on membrane protein targeting in plant cells.
Collapse
Affiliation(s)
- Paul Rosas-Santiago
- Instituto de Biotecnología, Universidad Nacional de Autónoma de México, Cuernavaca, Morelos 62250, México Department of Plant Biology, Carnegie Institution for Science, Stanford, CA 94305, USA
| | - Daniel Lagunas-Gómez
- Instituto de Biotecnología, Universidad Nacional de Autónoma de México, Cuernavaca, Morelos 62250, México
| | - Bronwyn J Barkla
- Southern Cross Plant Science, Southern Cross University, Lismore, Australia
| | - Rosario Vera-Estrella
- Instituto de Biotecnología, Universidad Nacional de Autónoma de México, Cuernavaca, Morelos 62250, México
| | - Sylvie Lalonde
- Department of Plant Biology, Carnegie Institution for Science, Stanford, CA 94305, USA
| | - Alexander Jones
- Department of Plant Biology, Carnegie Institution for Science, Stanford, CA 94305, USA
| | - Wolf B Frommer
- Department of Plant Biology, Carnegie Institution for Science, Stanford, CA 94305, USA
| | - Olga Zimmermannova
- Southern Cross Plant Science, Southern Cross University, Lismore, Australia
| | - Hana Sychrová
- Department of Membrane Transport, Institute of Physiology, Academy of Sciences of the Czech Republic, v.v.i., 142 20 Prague 4, Czech Republic
| | - Omar Pantoja
- Instituto de Biotecnología, Universidad Nacional de Autónoma de México, Cuernavaca, Morelos 62250, México
| |
Collapse
|
150
|
Abstract
TARP [transmembrane AMPA (α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) receptor regulatory protein] γ-8 is an auxiliary subunit of AMPA receptors that is widely distributed in the hippocampus. It has been shown that TARP γ-8 promotes surface expression of AMPA receptors; however, how TARP γ-8 regulates the expression of AMPA receptors remains unclear. In the present study, we examined the effect of TARP glycosylation on AMPA receptor trafficking. We first showed that TARP γ-8 is an N-glycosylated protein, which contains two glycosylation sites, Asn53 and Asn56, and compared this with the glycosylation of TARP γ-2 and the AMPA receptor auxiliary protein CNIH-2 (cornichon homologue 2). We next examine the effect of TARP glycosylation on TARP trafficking and also on AMPA receptor surface expression. We find that TARP γ-8 glycosylation is critical for surface expression of both TARP γ-8 and GluA1 in heterologous cells and neurons. Specifically, knockdown of TARP γ-8 causes a decrease in both total and surface AMPA receptors. We find that the expression of unglycosylated TARP γ-8 in cultured neurons is unable to restore GluA1 expression fully. Furthermore, when the maturation of TARP γ-8 is impaired, a large pool of immature GluA1 is retained intracellularly. Taken together, our data reveal an important role for the maturation of TARP γ-8 in the trafficking and function of the AMPA receptor complex.
Collapse
|