201
|
Twomey EC, Yelshanskaya MV, Grassucci RA, Frank J, Sobolevsky AI. Structural Bases of Desensitization in AMPA Receptor-Auxiliary Subunit Complexes. Neuron 2017; 94:569-580.e5. [PMID: 28472657 PMCID: PMC5492975 DOI: 10.1016/j.neuron.2017.04.025] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 04/10/2017] [Accepted: 04/17/2017] [Indexed: 02/09/2023]
Abstract
Fast excitatory neurotransmission is mediated by AMPA-subtype ionotropic glutamate receptors (AMPARs). AMPARs, localized at post-synaptic densities, are regulated by transmembrane auxiliary subunits that modulate AMPAR assembly, trafficking, gating, and pharmacology. Aberrancies in AMPAR-mediated signaling are associated with numerous neurological disorders. Here, we report cryo-EM structures of an AMPAR in complex with the auxiliary subunit GSG1L in the closed and desensitized states. GSG1L favors the AMPAR desensitized state, where channel closure is facilitated by profound structural rearrangements in the AMPAR extracellular domain, with ligand-binding domain dimers losing their local 2-fold rotational symmetry. Our structural and functional experiments suggest that AMPAR auxiliary subunits share a modular architecture and use a common transmembrane scaffold for distinct extracellular modules to differentially regulate AMPAR gating. By comparing the AMPAR-GSG1L complex structures, we map conformational changes accompanying AMPAR recovery from desensitization and reveal structural bases for regulation of synaptic transmission by auxiliary subunits.
Collapse
Affiliation(s)
- Edward C Twomey
- Department of Biochemistry and Molecular Biophysics, Columbia University, 650 West 168(th) Street, New York, NY 10032, USA; Integrated Program in Cellular, Molecular, and Biomedical Studies, Columbia University, 650 West 168(th) Street, New York, NY 10032, USA
| | - Maria V Yelshanskaya
- Department of Biochemistry and Molecular Biophysics, Columbia University, 650 West 168(th) Street, New York, NY 10032, USA
| | - Robert A Grassucci
- Department of Biochemistry and Molecular Biophysics, Columbia University, 650 West 168(th) Street, New York, NY 10032, USA; Howard Hughes Medical Institute, 650 West 168(th) Street, New York, NY 10032, USA
| | - Joachim Frank
- Department of Biochemistry and Molecular Biophysics, Columbia University, 650 West 168(th) Street, New York, NY 10032, USA; Department of Biological Sciences, Columbia University, 650 West 168(th) Street, New York, NY 10032, USA; Howard Hughes Medical Institute, 650 West 168(th) Street, New York, NY 10032, USA.
| | - Alexander I Sobolevsky
- Department of Biochemistry and Molecular Biophysics, Columbia University, 650 West 168(th) Street, New York, NY 10032, USA.
| |
Collapse
|
202
|
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]
|
203
|
Kucharz K, Søndergaard Rasmussen I, Bach A, Strømgaard K, Lauritzen M. PSD-95 uncoupling from NMDA receptors by Tat- N-dimer ameliorates neuronal depolarization in cortical spreading depression. J Cereb Blood Flow Metab 2017; 37:1820-1828. [PMID: 27107027 PMCID: PMC5435282 DOI: 10.1177/0271678x16645595] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Cortical spreading depression is associated with activation of NMDA receptors, which interact with the postsynaptic density protein 95 (PSD-95) that binds to nitric oxide synthase (nNOS). Here, we tested whether inhibition of the nNOS/PSD-95/NMDA receptor complex formation by anti-ischemic compound, UCCB01-144 (Tat- N-dimer) ameliorates the persistent effects of cortical spreading depression on cortical function. Using in vivo two-photon microscopy in somatosensory cortex in mice, we show that fluorescently labelled Tat- N-dimer readily crosses blood-brain barrier and accumulates in nerve cells during the first hour after i.v. injection. The Tat- N-dimer suppressed stimulation-evoked synaptic activity by 2-20%, while cortical blood flow and cerebral oxygen metabolic (CMRO2) responses were preserved. During cortical spreading depression, the Tat- N-dimer reduced the average amplitude of the negative shift in direct current potential by 33% (4.1 mV). Furthermore, the compound diminished the average depression of spontaneous electrocorticographic activity by 11% during first 40 min of post-cortical spreading depression recovery, but did not mitigate the suppressing effect of cortical spreading depression on cortical blood flow and CMRO2. We suggest that uncoupling of PSD-95 from NMDA receptors reduces overall neuronal excitability and the amplitude of the spreading depolarization wave. These findings may be of interest for understanding the neuroprotective effects of the nNOS/PSD-95 uncoupling in stroke.
Collapse
Affiliation(s)
- Krzysztof Kucharz
- 1 Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen N, Denmark
| | | | - Anders Bach
- 2 Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen Ø, Denmark
| | - Kristian Strømgaard
- 2 Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen Ø, Denmark
| | - Martin Lauritzen
- 1 Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen N, Denmark.,3 Department of Clinical Neurophysiology, Glostrup Hospital, Glostrup, Denmark
| |
Collapse
|
204
|
Baucum AJ. Proteomic Analysis of Postsynaptic Protein Complexes Underlying Neuronal Plasticity. ACS Chem Neurosci 2017; 8:689-701. [PMID: 28211672 DOI: 10.1021/acschemneuro.7b00008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Normal neuronal communication and synaptic plasticity at glutamatergic synapses requires dynamic regulation of postsynaptic molecules. Protein expression and protein post-translational modifications regulate protein interactions that underlie this organization. In this Review, we highlight data obtained over the last 20 years that have used qualitative and quantitative proteomics-based approaches to identify postsynaptic protein complexes. Herein, we describe how these proteomics studies have helped lay the foundation for understanding synaptic physiology and perturbations in synaptic signaling observed in different pathologies. We also describe emerging technologies that can be useful in these analyses. We focus on protein complexes associated with the highly abundant and functionally critical proteins: calcium/calmodulin-dependent protein kinase II, the N-methyl-d-aspartate, and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid glutamate receptors, and postsynaptic density protein of 95 kDa.
Collapse
Affiliation(s)
- Anthony J. Baucum
- Department of Biology, Stark Neurosciences
Research Institute, Indiana University-Purdue University Indianapolis, 723 W. Michigan St., Indianapolis, Indiana 46202, United States
| |
Collapse
|
205
|
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
|
206
|
Ben-Yaacov A, Gillor M, Haham T, Parsai A, Qneibi M, Stern-Bach Y. Molecular Mechanism of AMPA Receptor Modulation by TARP/Stargazin. Neuron 2017; 93:1126-1137.e4. [PMID: 28238551 DOI: 10.1016/j.neuron.2017.01.032] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 12/28/2016] [Accepted: 01/30/2017] [Indexed: 12/19/2022]
Abstract
AMPA receptors (AMPARs) mediate the majority of fast excitatory transmission in the brain and critically contribute to synaptic plasticity and pathology. AMPAR trafficking and gating are tightly controlled by auxiliary transmembrane AMPAR regulatory proteins (TARPs). Here, using systematic domain swaps with the TARP-insensitive kainate receptor GluK2, we show that AMPAR interaction with the prototypical TARP stargazin/γ2 primarily involves the AMPAR membrane domains M1 and M4 of neighboring subunits, initiated or stabilized by the AMPAR C-tail, and that these interactions are sufficient to enable full receptor modulation. Moreover, employing TARP chimeras disclosed a key role in this process also for the TARP transmembrane domains TM3 and TM4 and extracellular loop 2. Mechanistically, our data support a two-step action in which binding of TARP to the AMPAR membrane domains destabilizes the channel closed state, thereby enabling an efficient opening upon agonist binding, which then stabilizes the open state via subsequent interactions.
Collapse
Affiliation(s)
- Anat Ben-Yaacov
- Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Moshe Gillor
- Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Tomer Haham
- Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Alon Parsai
- Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Mohammad Qneibi
- Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Yael Stern-Bach
- Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada (IMRIC), Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91120, Israel.
| |
Collapse
|
207
|
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
|
208
|
Fukata Y, Yokoi N, Miyazaki Y, Fukata M. The LGI1–ADAM22 protein complex in synaptic transmission and synaptic disorders. Neurosci Res 2017; 116:39-45. [DOI: 10.1016/j.neures.2016.09.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 09/18/2016] [Accepted: 09/22/2016] [Indexed: 12/21/2022]
|
209
|
Won S, Levy JM, Nicoll RA, Roche KW. MAGUKs: multifaceted synaptic organizers. Curr Opin Neurobiol 2017; 43:94-101. [PMID: 28236779 DOI: 10.1016/j.conb.2017.01.006] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 01/25/2017] [Accepted: 01/30/2017] [Indexed: 11/19/2022]
Abstract
The PSD-95 family of proteins, known as MAGUKs, have long been recognized to be central building blocks of the PSD. They are categorized as scaffolding proteins, which link surface-expressed receptors to the intracellular signaling molecules. Although the four members of the PSD-95 family (PSD-95, PSD-93, SAP102, and SAP97) have many shared roles in regulating synaptic function, recent studies have begun to delineate specific binding partners and roles in plasticity. In the current review, we will highlight the conserved and unique roles of these proteins.
Collapse
Affiliation(s)
- Sehoon Won
- Receptor Biology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, United States
| | - Jon M Levy
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, United States
| | - Roger A Nicoll
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, CA 94158, United States; Department of Physiology, University of California, San Francisco, CA 94158, United States
| | - Katherine W Roche
- Receptor Biology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, United States.
| |
Collapse
|
210
|
Barad Z, Grattan DR, Leitch B. NMDA Receptor Expression in the Thalamus of the Stargazer Model of Absence Epilepsy. Sci Rep 2017; 7:42926. [PMID: 28220891 PMCID: PMC5318904 DOI: 10.1038/srep42926] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 01/16/2017] [Indexed: 11/09/2022] Open
Abstract
In the stargazer mouse model of absence epilepsy, altered corticothalamic excitation of reticular thalamic nucleus (RTN) neurons has been suggested to contribute to abnormal synchronicity in the corticothalamic-thalamocortical circuit, leading to spike-wave discharges, the hallmark of absence seizures. AMPA receptor expression and function are decreased in stargazer RTN, due to a mutation of AMPAR auxiliary subunit stargazin. It is unresolved and debated, however, if decreased excitation of RTN is compatible with epileptogenesis. We tested the hypothesis that relative NMDAR expression may be increased in RTN and/or thalamic synapses in stargazers using Western blot on dissected thalamic nuclei and biochemically isolated synapses, as well as immunogold cytochemistry in RTN. Expression of main NMDAR subunits was variable in stargazer RTN and relay thalamus; however, mean expression values were not statistically significantly different compared to controls. Furthermore, no systematic changes in synaptic NMDAR levels could be detected in stargazer thalamus. In contrast, AMPAR subunits were markedly decreased in both nucleus-specific and synaptic preparations. Thus, defective AMPAR trafficking in stargazer thalamus does not appear to lead to a ubiquitous compensatory increase in total and synaptic NMDAR expression, suggesting that elevated NMDAR function is not mediated by changes in protein expression in stargazer mice.
Collapse
Affiliation(s)
- Z Barad
- Department of Anatomy, Otago School of Biomedical Sciences, University of Otago, Dunedin, New Zealand.,Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| | - D R Grattan
- Department of Anatomy, Otago School of Biomedical Sciences, University of Otago, Dunedin, New Zealand.,Centre for Neuroendocrinology, Dunedin, New Zealand
| | - B Leitch
- Department of Anatomy, Otago School of Biomedical Sciences, University of Otago, Dunedin, New Zealand.,Brain Health Research Centre, University of Otago, Dunedin, New Zealand
| |
Collapse
|
211
|
Trafficking of neuronal calcium channels. Neuronal Signal 2017; 1:NS20160003. [PMID: 32714572 PMCID: PMC7373241 DOI: 10.1042/ns20160003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Revised: 01/20/2017] [Accepted: 01/19/2017] [Indexed: 12/18/2022] Open
Abstract
Neuronal voltage-gated calcium channels (VGCCs) serve complex yet essential physiological functions via their pivotal role in translating electrical signals into intracellular calcium elevations and associated downstream signalling pathways. There are a number of regulatory mechanisms to ensure a dynamic control of the number of channels embedded in the plasma membrane, whereas alteration of the surface expression of VGCCs has been linked to various disease conditions. Here, we provide an overview of the mechanisms that control the trafficking of VGCCs to and from the plasma membrane, and discuss their implication in pathophysiological conditions and their potential as therapeutic targets.
Collapse
|
212
|
|
213
|
Nicoll RA. A Brief History of Long-Term Potentiation. Neuron 2017; 93:281-290. [DOI: 10.1016/j.neuron.2016.12.015] [Citation(s) in RCA: 334] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 12/08/2016] [Accepted: 12/08/2016] [Indexed: 12/19/2022]
|
214
|
Presynaptic Neuronal Pentraxin Receptor Organizes Excitatory and Inhibitory Synapses. J Neurosci 2016; 37:1062-1080. [PMID: 27986928 DOI: 10.1523/jneurosci.2768-16.2016] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 11/23/2016] [Accepted: 12/12/2016] [Indexed: 01/05/2023] Open
Abstract
Three neuronal pentraxins are expressed in brain, the membrane-bound "neuronal pentraxin receptor" (NPR) and the secreted proteins NP1 and NARP (i.e., NP2). Neuronal pentraxins bind to AMPARs at excitatory synapses and play important, well-documented roles in the activity-dependent regulation of neural circuits via this binding activity. However, it is unknown whether neuronal pentraxins perform roles in synapses beyond modulating postsynaptic AMPAR-dependent plasticity, and whether they may even act in inhibitory synapses. Here, we show that NPR expressed in non-neuronal cells potently induces formation of both excitatory and inhibitory postsynaptic specializations in cocultured hippocampal neurons. Knockdown of NPR in hippocampal neurons, conversely, dramatically decreased assembly and function of both excitatory and inhibitory postsynaptic specializations. Overexpression of NPR rescued the NPR knockdown phenotype but did not in itself change synapse numbers or properties. However, the NPR knockdown decreased the levels of NARP, whereas NPR overexpression produced a dramatic increase in the levels of NP1 and NARP, suggesting that NPR recruits and stabilizes NP1 and NARP on the presynaptic plasma membrane. Mechanistically, NPR acted in excitatory synapse assembly by binding to the N-terminal domain of AMPARs; antagonists of AMPA and GABA receptors selectively inhibited NPR-induced heterologous excitatory and inhibitory synapse assembly, respectively, but did not affect neurexin-1β-induced synapse assembly as a control. Our data suggest that neuronal pentraxins act as signaling complexes that function as general trans-synaptic organizers of both excitatory and inhibitory synapses by a mechanism that depends, at least in part, on the activity of the neurotransmitter receptors at these synapses. SIGNIFICANCE STATEMENT Neuronal pentraxins comprise three neuronal proteins, neuronal pentraxin receptor (NPR) which is a type-II transmembrane protein on the neuronal surface, and secreted neuronal pentraxin-1 and NARP. The general functions of neuronal pentraxins at synapses have not been explored, except for their basic AMPAR binding properties. Here, we examined the functional role of NPR at synapses because it is the only neuronal pentraxin that is anchored to the neuronal cell-surface membrane. We find that NPR is a potent inducer of both excitatory and inhibitory heterologous synapses, and that knockdown of NPR in cultured neurons decreases the density of both excitatory and inhibitory synapses. Our data suggest that NPR performs a general, previously unrecognized function as a universal organizer of synapses.
Collapse
|
215
|
Palmitoylation regulates glutamate receptor distributions in postsynaptic densities through control of PSD95 conformation and orientation. Proc Natl Acad Sci U S A 2016; 113:E8482-E8491. [PMID: 27956638 DOI: 10.1073/pnas.1612963113] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Postsynaptic density protein 95 (PSD95) and synapse-associated protein 97 (SAP97) are homologous scaffold proteins with different N-terminal domains, possessing either a palmitoylation site (PSD95) or an L27 domain (SAP97). Here, we measured PSD95 and SAP97 conformation in vitro and in postsynaptic densities (PSDs) using FRET and EM, and examined how conformation regulated interactions with AMPA-type and NMDA-type glutamate receptors (AMPARs/NMDARs). Palmitoylation of PSD95 changed its conformation from a compact to an extended configuration. PSD95 associated with AMPARs (via transmembrane AMPAR regulatory protein subunits) or NMDARs [via glutamate ionotropic receptor NMDA-type subunit 2B (GluN2B) subunits] only in its palmitoylated and extended conformation. In contrast, in its extended conformation, SAP97 associates with NMDARs, but not with AMPARs. Within PSDs, PSD95 and SAP97 were largely in the extended conformation, but had different orientations. PSD95 oriented perpendicular to the PSD membrane, with its palmitoylated, N-terminal domain at the membrane. SAP97 oriented parallel to the PSD membrane, likely as a dimer through interactions of its N-terminal L27 domain. Changing PSD95 palmitoylation in PSDs altered PSD95 and AMPAR levels but did not affect NMDAR levels. These results indicate that in PSDs, PSD95 palmitoylation, conformation, and its interactions are dynamic when associated with AMPARs and more stable when associated with NMDARs. Altogether, our results are consistent with differential regulation of PSD95 palmitoylation in PSDs resulting from the clustering of palmitoylating and depalmitoylating enzymes into AMPAR nanodomains segregated away from NMDAR nanodomains.
Collapse
|
216
|
Pratt KG, Hiramoto M, Cline HT. An Evolutionarily Conserved Mechanism for Activity-Dependent Visual Circuit Development. Front Neural Circuits 2016; 10:79. [PMID: 27818623 PMCID: PMC5073143 DOI: 10.3389/fncir.2016.00079] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 09/26/2016] [Indexed: 12/01/2022] Open
Abstract
Neural circuit development is an activity-dependent process. This activity can be spontaneous, such as the retinal waves that course across the mammalian embryonic retina, or it can be sensory-driven, such as the activation of retinal ganglion cells (RGCs) by visual stimuli. Whichever the source, neural activity provides essential instruction to the developing circuit. Indeed, experimentally altering activity has been shown to impact circuit development and function in many different ways and in many different model systems. In this review, we contemplate the idea that retinal waves in amniotes, the animals that develop either in ovo or utero (namely reptiles, birds and mammals) could be an evolutionary adaptation to life on land, and that the anamniotes, animals whose development is entirely external (namely the aquatic amphibians and fish), do not display retinal waves, most likely because they simply don’t need them. We then review what is known about the function of both retinal waves and visual stimuli on their respective downstream targets, and predict that the experience-dependent development of the tadpole visual system is a blueprint of what will be found in future studies of the effects of spontaneous retinal waves on instructing development of retinorecipient targets such as the superior colliculus (SC) and the lateral geniculate nucleus.
Collapse
Affiliation(s)
- Kara G Pratt
- Program in Neuroscience, Department of Zoology and Physiology, University of Wyoming Laramie, WY, USA
| | - Masaki Hiramoto
- Department of Molecular and Cellular Neuroscience and The Dorris Neuroscience Center, The Scripps Research Institute La Jolla, CA, USA
| | - Hollis T Cline
- Department of Molecular and Cellular Neuroscience and The Dorris Neuroscience Center, The Scripps Research Institute La Jolla, CA, USA
| |
Collapse
|
217
|
Rademacher N, Schmerl B, Lardong JA, Wahl MC, Shoichet SA. MPP2 is a postsynaptic MAGUK scaffold protein that links SynCAM1 cell adhesion molecules to core components of the postsynaptic density. Sci Rep 2016; 6:35283. [PMID: 27756895 PMCID: PMC5069480 DOI: 10.1038/srep35283] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 09/15/2016] [Indexed: 02/07/2023] Open
Abstract
At neuronal synapses, multiprotein complexes of trans-synaptic adhesion molecules, scaffold proteins and neurotransmitter receptors assemble to essential building blocks required for synapse formation and maintenance. Here we describe a novel role for the membrane-associated guanylate kinase (MAGUK) protein MPP2 (MAGUK p55 subfamily member 2) at synapses of rat central neurons. Through interactions mediated by its C-terminal SH3-GK domain module, MPP2 binds to the abundant postsynaptic scaffold proteins PSD-95 and GKAP and localises to postsynaptic sites in hippocampal neurons. MPP2 also colocalises with the synaptic adhesion molecule SynCAM1. We demonstrate that the SynCAM1 C-terminus interacts directly with the MPP2 PDZ domain and that MPP2 does not interact in this manner with other highly abundant postsynaptic transmembrane proteins. Our results highlight a previously unexplored role for MPP2 at postsynaptic sites as a scaffold that links SynCAM1 cell adhesion molecules to core proteins of the postsynaptic density.
Collapse
Affiliation(s)
- Nils Rademacher
- Neuroscience Research Center and Cluster of Excellence NeuroCure, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Bettina Schmerl
- Neuroscience Research Center and Cluster of Excellence NeuroCure, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Jennifer A. Lardong
- Institute of Chemistry and Biochemistry, Structural Biochemistry, Freie Universität Berlin, Takustr. 6, 14195 Berlin, Germany
| | - Markus C. Wahl
- Institute of Chemistry and Biochemistry, Structural Biochemistry, Freie Universität Berlin, Takustr. 6, 14195 Berlin, Germany
| | - Sarah A. Shoichet
- Neuroscience Research Center and Cluster of Excellence NeuroCure, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
218
|
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
|
219
|
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
|
220
|
TARP γ-2 and γ-8 Differentially Control AMPAR Density Across Schaffer Collateral/Commissural Synapses in the Hippocampal CA1 Area. J Neurosci 2016; 36:4296-312. [PMID: 27076426 DOI: 10.1523/jneurosci.4178-15.2016] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 02/19/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED The number of AMPA-type glutamate receptors (AMPARs) at synapses is the major determinant of synaptic strength and varies from synapse to synapse. To clarify the underlying molecular mechanisms, the density of AMPARs, PSD-95, and transmembrane AMPAR regulatory proteins (TARPs) were compared at Schaffer collateral/commissural (SCC) synapses in the adult mouse hippocampal CA1 by quantitative immunogold electron microscopy using serial sections. We examined four types of SCC synapses: perforated and nonperforated synapses on pyramidal cells and axodendritic synapses on parvalbumin-positive (PV synapse) and pravalbumin-negative interneurons (non-PV synapse). SCC synapses were categorized into those expressing high-density (perforated and PV synapses) or low-density (nonperforated and non-PV synapses) AMPARs. Although the density of PSD-95 labeling was fairly constant, the density and composition of TARP isoforms was highly variable depending on the synapse type. Of the three TARPs expressed in hippocampal neurons, the disparity in TARP γ-2 labeling was closely related to that of AMPAR labeling. Importantly, AMPAR density was significantly reduced at perforated and PV synapses in TARP γ-2-knock-out (KO) mice, resulting in a virtual loss of AMPAR disparity among SCC synapses. In comparison, TARP γ-8 was the only TARP expressed at nonperforated synapses, where AMPAR labeling further decreased to a background level in TARP γ-8-KO mice. These results show that synaptic inclusion of TARP γ-2 potently increases AMPAR expression and transforms low-density synapses into high-density ones, whereas TARP γ-8 is essential for low-density or basal expression of AMPARs at nonperforated synapses. Therefore, these TARPs are critically involved in AMPAR density control at SCC synapses. SIGNIFICANCE STATEMENT Although converging evidence implicates the importance of transmembrane AMPA-type glutamate receptor (AMPAR) regulatory proteins (TARPs) in AMPAR stabilization during basal transmission and synaptic plasticity, how they control large disparities in AMPAR numbers or densities across central synapses remains largely unknown. We compared the density of AMPARs with that of TARPs among four types of Schaffer collateral/commissural (SCC) hippocampal synapses in wild-type and TARP-knock-out mice. We show that the density of AMPARs correlates with that of TARP γ-2 across SCC synapses and its high expression is linked to high-density AMPAR expression at perforated type of pyramidal cell synapses and synapses on parvalbumin-positive interneurons. In comparison, TARP γ-8 is the only TARP expressed at nonperforated type of pyramidal cell synapses, playing an essential role in low-density or basal AMPAR expression.
Collapse
|
221
|
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: 43] [Impact Index Per Article: 4.8] [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
|
222
|
Twomey EC, Yelshanskaya MV, Grassucci RA, Frank J, Sobolevsky AI. Elucidation of AMPA receptor-stargazin complexes by cryo-electron microscopy. Science 2016; 353:83-6. [PMID: 27365450 PMCID: PMC5125255 DOI: 10.1126/science.aaf8411] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 06/08/2016] [Indexed: 11/02/2022]
Abstract
AMPA-subtype ionotropic glutamate receptors (AMPARs) mediate fast excitatory neurotransmission and contribute to high cognitive processes such as learning and memory. In the brain, AMPAR trafficking, gating, and pharmacology is tightly controlled by transmembrane AMPAR regulatory proteins (TARPs). Here, we used cryo-electron microscopy to elucidate the structural basis of AMPAR regulation by one of these auxiliary proteins, TARP γ2, or stargazin (STZ). Our structures illuminate the variable interaction stoichiometry of the AMPAR-TARP complex, with one or two TARP molecules binding one tetrameric AMPAR. Analysis of the AMPAR-STZ binding interfaces suggests that electrostatic interactions between the extracellular domains of AMPAR and STZ play an important role in modulating AMPAR function through contact surfaces that are conserved across AMPARs and TARPs. We propose a model explaining how TARPs stabilize the activated state of AMPARs and how the interactions between AMPARs and their auxiliary proteins control fast excitatory synaptic transmission.
Collapse
Affiliation(s)
- Edward C Twomey
- Department of Biochemistry and Molecular Biophysics, Columbia University, 650 West 168th Street, New York, NY 10032, USA. Integrated Program in Cellular, Molecular, and Biomedical Studies, Columbia University, 650 West 168th Street, New York, NY 10032, USA
| | - Maria V Yelshanskaya
- Department of Biochemistry and Molecular Biophysics, Columbia University, 650 West 168th Street, New York, NY 10032, USA
| | - Robert A Grassucci
- Department of Biochemistry and Molecular Biophysics, Columbia University, 650 West 168th Street, New York, NY 10032, USA. Howard Hughes Medical Institute, 650 West 168th Street, New York, NY 10032, USA
| | - Joachim Frank
- Department of Biochemistry and Molecular Biophysics, Columbia University, 650 West 168th Street, New York, NY 10032, USA. Department of Biological Sciences, Columbia University, 650 West 168th Street, New York, NY 10032, USA. Howard Hughes Medical Institute, 650 West 168th Street, New York, NY 10032, USA.
| | - Alexander I Sobolevsky
- Department of Biochemistry and Molecular Biophysics, Columbia University, 650 West 168th Street, New York, NY 10032, USA.
| |
Collapse
|
223
|
Zhao Y, Chen S, Yoshioka C, Baconguis I, Gouaux E. Architecture of fully occupied GluA2 AMPA receptor-TARP complex elucidated by cryo-EM. Nature 2016; 536:108-11. [PMID: 27368053 DOI: 10.1038/nature18961] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 06/24/2016] [Indexed: 11/09/2022]
Abstract
Fast excitatory neurotransmission in the mammalian central nervous system is largely carried out by AMPA-sensitive ionotropic glutamate receptors. Localized within the postsynaptic density of glutamatergic spines, AMPA receptors are composed of heterotetrameric receptor assemblies associated with auxiliary subunits, the most common of which are transmembrane AMPA receptor regulatory proteins (TARPs). The association of TARPs with AMPA receptors modulates receptor trafficking and the kinetics of receptor gating and pharmacology. Here we report the cryo-electron microscopy (cryo-EM) structure of the homomeric rat GluA2 AMPA receptor saturated with TARP γ2 subunits, which shows how the TARPs are arranged with four-fold symmetry around the ion channel domain and make extensive interactions with the M1, M2 and M4 transmembrane helices. Poised like partially opened ‘hands’ underneath the two-fold symmetric ligand-binding domain (LBD) 'clamshells', one pair of TARPs is juxtaposed near the LBD dimer interface, whereas the other pair is near the LBD dimer-dimer interface. The extracellular ‘domains’ of TARP are positioned to not only modulate LBD clamshell closure, but also affect conformational rearrangements of the LBD layer associated with receptor activation and desensitization, while the TARP transmembrane domains buttress the ion channel pore.
Collapse
|
224
|
Straub C, Noam Y, Nomura T, Yamasaki M, Yan D, Fernandes HB, Zhang P, Howe JR, Watanabe M, Contractor A, Tomita S. Distinct Subunit Domains Govern Synaptic Stability and Specificity of the Kainate Receptor. Cell Rep 2016; 16:531-544. [PMID: 27346345 DOI: 10.1016/j.celrep.2016.05.093] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 04/28/2016] [Accepted: 05/26/2016] [Indexed: 12/01/2022] Open
Abstract
Synaptic communication between neurons requires the precise localization of neurotransmitter receptors to the correct synapse type. Kainate-type glutamate receptors restrict synaptic localization that is determined by the afferent presynaptic connection. The mechanisms that govern this input-specific synaptic localization remain unclear. Here, we examine how subunit composition and specific subunit domains contribute to synaptic localization of kainate receptors. The cytoplasmic domain of the GluK2 low-affinity subunit stabilizes kainate receptors at synapses. In contrast, the extracellular domain of the GluK4/5 high-affinity subunit synergistically controls the synaptic specificity of kainate receptors through interaction with C1q-like proteins. Thus, the input-specific synaptic localization of the native kainate receptor complex involves two mechanisms that underlie specificity and stabilization of the receptor at synapses.
Collapse
Affiliation(s)
- Christoph Straub
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520, USA; CNNR Program, Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Yoav Noam
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520, USA; CNNR Program, Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Toshihiro Nomura
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Miwako Yamasaki
- Department of Anatomy, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Dan Yan
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520, USA; CNNR Program, Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Herman B Fernandes
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Ping Zhang
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - James R Howe
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Masahiko Watanabe
- Department of Anatomy, Hokkaido University Graduate School of Medicine, Sapporo 060-8638, Japan
| | - Anis Contractor
- Department of Physiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Susumu Tomita
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520, USA; CNNR Program, Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06520, USA.
| |
Collapse
|
225
|
García-Nafría J, Watson JF, Greger IH. IVA cloning: A single-tube universal cloning system exploiting bacterial In Vivo Assembly. Sci Rep 2016; 6:27459. [PMID: 27264908 PMCID: PMC4893743 DOI: 10.1038/srep27459] [Citation(s) in RCA: 171] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 05/13/2016] [Indexed: 12/11/2022] Open
Abstract
In vivo homologous recombination holds the potential for optimal molecular cloning, however, current strategies require specialised bacterial strains or laborious protocols. Here, we exploit a recA-independent recombination pathway, present in widespread laboratory E.coli strains, to develop IVA (In Vivo Assembly) cloning. This system eliminates the need for enzymatic assembly and reduces all molecular cloning procedures to a single-tube, single-step PCR, performed in <2 hours from setup to transformation. Unlike other methods, IVA is a complete system, and offers significant advantages over alternative methods for all cloning procedures (insertions, deletions, site-directed mutagenesis and sub-cloning). Significantly, IVA allows unprecedented simplification of complex cloning procedures: five simultaneous modifications of any kind, multi-fragment assembly and library construction are performed in approximately half the time of current protocols, still in a single-step fashion. This system is efficient, seamless and sequence-independent, and requires no special kits, enzymes or proprietary bacteria, which will allow its immediate adoption by the academic and industrial molecular biology community.
Collapse
Affiliation(s)
- Javier García-Nafría
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, CB2 0QH, UK
| | - Jake F. Watson
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, CB2 0QH, UK
| | - Ingo H. Greger
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, CB2 0QH, UK
| |
Collapse
|
226
|
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
|
227
|
α/β-Hydrolase domain-containing 6 (ABHD6) negatively regulates the surface delivery and synaptic function of AMPA receptors. Proc Natl Acad Sci U S A 2016; 113:E2695-704. [PMID: 27114538 DOI: 10.1073/pnas.1524589113] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
In the brain, AMPA-type glutamate receptors are major postsynaptic receptors at excitatory synapses that mediate fast neurotransmission and synaptic plasticity. α/β-Hydrolase domain-containing 6 (ABHD6), a monoacylglycerol lipase, was previously found to be a component of AMPA receptor macromolecular complexes, but its physiological significance in the function of AMPA receptors (AMPARs) has remained unclear. The present study shows that overexpression of ABHD6 in neurons drastically reduced excitatory neurotransmission mediated by AMPA but not by NMDA receptors at excitatory synapses. Inactivation of ABHD6 expression in neurons by either CRISPR/Cas9 or shRNA knockdown methods significantly increased excitatory neurotransmission at excitatory synapses. Interestingly, overexpression of ABHD6 reduced glutamate-induced currents and the surface expression of GluA1 in HEK293T cells expressing GluA1 and stargazin, suggesting a direct functional interaction between these two proteins. The C-terminal tail of GluA1 was required for the binding between of ABHD6 and GluA1. Mutagenesis analysis revealed a GFCLIPQ sequence in the GluA1 C terminus that was essential for the inhibitory effect of ABHD6. The hydrolase activity of ABHD6 was not required for the effects of ABHD6 on AMPAR function in either neurons or transfected HEK293T cells. Thus, these findings reveal a novel and unexpected mechanism governing AMPAR trafficking at synapses through ABHD6.
Collapse
|
228
|
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
|
229
|
Wang J, Li L, Shao SS, He Z, Chen YL, Kong R, Zhang XH, Gong JH, Song RR. Association analysis of genetic variant of rs13331 in PSD95 gene with autism spectrum disorders: A case-control study in a Chinese population. ACTA ACUST UNITED AC 2016; 36:285-288. [PMID: 27072977 DOI: 10.1007/s11596-016-1581-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 10/27/2015] [Indexed: 11/26/2022]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder characterized by high heritability. Recently, autism, the most profound form of ASD, has been increasingly attributed to synaptic abnormalities. Postsynaptic density 95 (PSD95), encoding PSD protein-95, was found essential for synaptic formation, maturation and plasticity at a PSD of excitatory synapse. It is possibly a crucial candidate gene for the pathogenesis of ASD. To identify the relationship between the rs13331 of PSD95 gene and ASD, we performed a case-control study in 212 patients and 636 controls in a Chinese population by using a polymerase chain reaction-restriction fragment length polymerase (PCR-RFLP) assay. The results showed that in genetic analysis of the heterozygous model, an association between the T allele of the rs13331 and ASD was found in the dominant model (OR=1.709, 95% CI 1.227-2.382, P=0.002) and the additive model (OR=1.409, 95% CI=1.104-1.800, P=0.006). Our data indicate that the genetic mutation C>T at the rs13331 in the PSD95 gene is strikingly associated with an increased risk of ASD.
Collapse
Affiliation(s)
- Jia Wang
- Department of Maternal and Child Health and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Li Li
- Maternity and Children Health Care Hospital of Luohu District, Shenzhen, 518019, China
| | - Shan-Shan Shao
- Department of Maternal and Child Health and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhen He
- Central Hospital of Longhua New District, Shenzhen, 518000, China
| | - Yan-Lin Chen
- Maternity and Children Health Care Hospital of Luohu District, Shenzhen, 518019, China
| | - Rui Kong
- Department of Maternal and Child Health and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiao-Hui Zhang
- Department of Maternal and Child Health and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jian-Hua Gong
- Department of Maternal and Child Health and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- Maternity and Children Health Care Hospital of Luohu District, Shenzhen, 518019, China.
| | - Ran-Ran Song
- Department of Maternal and Child Health and MOE Key Lab of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
230
|
Brown PMGE, Aurousseau MRP, Musgaard M, Biggin PC, Bowie D. Kainate receptor pore-forming and auxiliary subunits regulate channel block by a novel mechanism. J Physiol 2016; 594:1821-40. [PMID: 26682513 PMCID: PMC4818602 DOI: 10.1113/jp271690] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Accepted: 12/07/2015] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Kainate receptor heteromerization and auxiliary subunits, Neto1 and Neto2, attenuate polyamine ion-channel block by facilitating blocker permeation. Relief of polyamine block in GluK2/GluK5 heteromers results from a key proline residue that produces architectural changes in the channel pore α-helical region. Auxiliary subunits exert an additive effect to heteromerization, and thus relief of polyamine block is due to a different mechanism. Our findings have broad implications for work on polyamine block of other cation-selective ion channels. ABSTRACT Channel block and permeation by cytoplasmic polyamines is a common feature of many cation-selective ion channels. Although the channel block mechanism has been studied extensively, polyamine permeation has been considered less significant as it occurs at extreme positive membrane potentials. Here, we show that kainate receptor (KAR) heteromerization and association with auxiliary proteins, Neto1 and Neto2, attenuate polyamine block by enhancing blocker permeation. Consequently, polyamine permeation and unblock occur at more negative and physiologically relevant membrane potentials. In GluK2/GluK5 heteromers, enhanced permeation is due to a single proline residue in GluK5 that alters the dynamics of the α-helical region of the selectivity filter. The effect of auxiliary proteins is additive, and therefore the structural basis of polyamine permeation and unblock is through a different mechanism. As native receptors are thought to assemble as heteromers in complex with auxiliary proteins, our data identify an unappreciated impact of polyamine permeation in shaping the signalling properties of neuronal KARs and point to a structural mechanism that may be shared amongst other cation-selective ion channels.
Collapse
Affiliation(s)
- Patricia M G E Brown
- Integrated Program in Neurosciences, McGill University, Montréal, Québec, Canada, H3G 0B1
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada, H3G 0B1
| | - Mark R P Aurousseau
- Graduate Program in Pharmacology, McGill University, Montréal, Québec, Canada, H3G 0B1
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada, H3G 0B1
| | - Maria Musgaard
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK
| | - Philip C Biggin
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK
| | - Derek Bowie
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada, H3G 0B1
| |
Collapse
|
231
|
CNQX facilitates inhibitory synaptic transmission in rat hypoglossal nucleus. Brain Res 2016; 1637:71-80. [DOI: 10.1016/j.brainres.2016.02.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 01/26/2016] [Accepted: 02/11/2016] [Indexed: 11/21/2022]
|
232
|
Lee AG, Capanzana R, Brockhurst J, Cheng MY, Buckmaster CL, Absher D, Schatzberg AF, Lyons DM. Learning to cope with stress modulates anterior cingulate cortex stargazin expression in monkeys and mice. Neurobiol Learn Mem 2016; 131:95-100. [PMID: 27003116 DOI: 10.1016/j.nlm.2016.03.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 03/16/2016] [Accepted: 03/18/2016] [Indexed: 11/29/2022]
Abstract
Intermittent mildly stressful situations provide opportunities to learn, practice, and improve coping with gains in subsequent emotion regulation. Here we investigate the effects of learning to cope with stress on anterior cingulate cortex gene expression in monkeys and mice. Anterior cingulate cortex is involved in learning, memory, cognitive control, and emotion regulation. Monkeys and mice were randomized to either stress coping or no-stress treatment conditions. Profiles of gene expression were acquired with HumanHT-12v4.0 Expression BeadChip arrays adapted for monkeys. Three genes identified in monkeys by arrays were then assessed in mice by quantitative real-time polymerase chain reaction. Expression of a key gene (PEMT) involved in acetylcholine biosynthesis was increased in monkeys by coping but this result was not verified in mice. Another gene (SPRY2) that encodes a negative regulator of neurotrophic factor signaling was decreased in monkeys by coping but this result was only partly verified in mice. The CACNG2 gene that encodes stargazin (also called TARP gamma-2) was increased by coping in monkeys as well as mice randomized to coping with or without subsequent behavioral tests of emotionality. As evidence of coping effects distinct from repeated stress exposures per se, increased stargazin expression induced by coping correlated with diminished emotionality in mice. Stargazin modulates glutamate receptor signaling and plays a role in synaptic plasticity. Molecular mechanisms of synaptic plasticity that mediate learning and memory in the context of coping with stress may provide novel targets for new treatments of disorders in human mental health.
Collapse
Affiliation(s)
- Alex G Lee
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA
| | - Roxanne Capanzana
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA
| | | | | | | | - Devin Absher
- HudsonAlpha Institute for Biotechnology, Huntsville, AL
| | - Alan F Schatzberg
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA
| | - David M Lyons
- Department of Psychiatry and Behavioral Sciences, Stanford University, Stanford, CA
| |
Collapse
|
233
|
Maher MP, Wu N, Ravula S, Ameriks MK, Savall BM, Liu C, Lord B, Wyatt RM, Matta JA, Dugovic C, Yun S, Ver Donck L, Steckler T, Wickenden AD, Carruthers NI, Lovenberg TW. Discovery and Characterization of AMPA Receptor Modulators Selective for TARP- 8. ACTA ACUST UNITED AC 2016; 357:394-414. [DOI: 10.1124/jpet.115.231712] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 03/11/2016] [Indexed: 01/14/2023]
|
234
|
Guntupalli S, Widagdo J, Anggono V. Amyloid-β-Induced Dysregulation of AMPA Receptor Trafficking. Neural Plast 2016; 2016:3204519. [PMID: 27073700 PMCID: PMC4814684 DOI: 10.1155/2016/3204519] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 02/28/2016] [Indexed: 01/22/2023] Open
Abstract
Evidence from neuropathological, genetic, animal model, and biochemical studies has indicated that the accumulation of amyloid-beta (Aβ) is associated with, and probably induces, profound neuronal changes in brain regions critical for memory and cognition in the development of Alzheimer's disease (AD). There is considerable evidence that synapses are particularly vulnerable to AD, establishing synaptic dysfunction as one of the earliest events in pathogenesis, prior to neuronal loss. It is clear that excessive Aβ levels can disrupt excitatory synaptic transmission and plasticity, mainly due to dysregulation of the AMPA and NMDA glutamate receptors in the brain. Importantly, AMPA receptors are the principal glutamate receptors that mediate fast excitatory neurotransmission. This is essential for synaptic plasticity, a cellular correlate of learning and memory, which are the cognitive functions that are most disrupted in AD. Here we review recent advances in the field and provide insights into the molecular mechanisms that underlie Aβ-induced dysfunction of AMPA receptor trafficking. This review focuses primarily on NMDA receptor- and metabotropic glutamate receptor-mediated signaling. In particular, we highlight several mechanisms that underlie synaptic long-term depression as common signaling pathways that are hijacked by the neurotoxic effects of Aβ.
Collapse
Affiliation(s)
- Sumasri Guntupalli
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Jocelyn Widagdo
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Victor Anggono
- Clem Jones Centre for Ageing Dementia Research, Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| |
Collapse
|
235
|
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: 61] [Impact Index Per Article: 6.8] [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
|
236
|
Wang JG, Wang YL, Xu F, Zhao JX, Zhou SY, Yu Y, Chazot PL, Wang XF, Lu CB. Activity- and development-dependent down-regulation of TARPγ8 and GluA1 in cultured rat hippocampal neurons. Acta Pharmacol Sin 2016; 37:303-11. [PMID: 26725511 DOI: 10.1038/aps.2015.112] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 10/26/2015] [Indexed: 01/16/2023] Open
Abstract
AIM Transmembrane AMPA receptor regulatory proteins (TARPs) regulate the trafficking and expression of AMPA receptors that are essential for the fast excitatory synaptic transmission and plasticity in the brain. This study aimed to investigate the activity-dependent regulation of TARPγ8 in cultured rat hippocampal neurons. METHODS Rat hippocampal neurons cultured for 7-8 DIV or 17-18 DIV were exposed to the AMPA receptor agonist AMPA at a non-toxic concentration (100 μmol/L) for 4 h. The protein levels of TARPγ8 and AMPA receptor subunits (GluA1 and GluA2) were measured using Western blotting analysis. AMPA-induced currents were recorded in the neurons using a whole-cell recording method. RESULTS Four-hour exposure to AMPA significantly decreased the protein levels of TARPγ8 and GluA1 in the neurons at 17-18 DIV, but did not change the protein level of TARPγ8 in the neurons cultured at 7-8 DIV. AMPA-induced down-regulation of TARPγ8 and GluA1 was largely blocked by the calpain inhibitor calpeptin (50 μmol/L), but not affected by the caspase inhibitor zVAD (50 μmol/L). Four-hour exposure to AMPA significantly decreased AMPA-induced currents in the neurons at 17-18 DIV, which was blocked by co-exposure to calpeptin (50 μmol/L). CONCLUSION The down-regulation of TARPγ8 and GluA1 protein levels and AMPA-induced currents in cultured rat hippocampal neurons is activity- and development-dependent, and mediated by endogenous calpain.
Collapse
|
237
|
Emerging Link between Alzheimer's Disease and Homeostatic Synaptic Plasticity. Neural Plast 2016; 2016:7969272. [PMID: 27019755 PMCID: PMC4785275 DOI: 10.1155/2016/7969272] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 01/31/2016] [Indexed: 01/14/2023] Open
Abstract
Alzheimer's disease (AD) is an irreversible brain disorder characterized by progressive cognitive decline and neurodegeneration of brain regions that are crucial for learning and memory. Although intracellular neurofibrillary tangles and extracellular senile plaques, composed of insoluble amyloid-β (Aβ) peptides, have been the hallmarks of postmortem AD brains, memory impairment in early AD correlates better with pathological accumulation of soluble Aβ oligomers and persistent weakening of excitatory synaptic strength, which is demonstrated by inhibition of long-term potentiation, enhancement of long-term depression, and loss of synapses. However, current, approved interventions aiming to reduce Aβ levels have failed to retard disease progression; this has led to a pressing need to identify and target alternative pathogenic mechanisms of AD. Recently, it has been suggested that the disruption of Hebbian synaptic plasticity in AD is due to aberrant metaplasticity, which is a form of homeostatic plasticity that tunes the magnitude and direction of future synaptic plasticity based on previous neuronal or synaptic activity. This review examines emerging evidence for aberrant metaplasticity in AD. Putative mechanisms underlying aberrant metaplasticity in AD will also be discussed. We hope this review inspires future studies to test the extent to which these mechanisms contribute to the etiology of AD and offer therapeutic targets.
Collapse
|
238
|
Hagena H, Hansen N, Manahan-Vaughan D. β-Adrenergic Control of Hippocampal Function: Subserving the Choreography of Synaptic Information Storage and Memory. Cereb Cortex 2016; 26:1349-64. [PMID: 26804338 PMCID: PMC4785955 DOI: 10.1093/cercor/bhv330] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Noradrenaline (NA) is a key neuromodulator for the regulation of behavioral state and cognition. It supports learning by increasing arousal and vigilance, whereby new experiences are “earmarked” for encoding. Within the hippocampus, experience-dependent information storage occurs by means of synaptic plasticity. Furthermore, novel spatial, contextual, or associative learning drives changes in synaptic strength, reflected by the strengthening of long-term potentiation (LTP) or long-term depression (LTD). NA acting on β-adrenergic receptors (β-AR) is a key determinant as to whether new experiences result in persistent hippocampal synaptic plasticity. This can even dictate the direction of change of synaptic strength. The different hippocampal subfields play different roles in encoding components of a spatial representation through LTP and LTD. Strikingly, the sensitivity of synaptic plasticity in these subfields to β-adrenergic control is very distinct (dentate gyrus > CA3 > CA1). Moreover, NA released from the locus coeruleus that acts on β-AR leads to hippocampal LTD and an enhancement of LTD-related memory processing. We propose that NA acting on hippocampal β-AR, that is graded according to the novelty or saliency of the experience, determines the content and persistency of synaptic information storage in the hippocampal subfields and therefore of spatial memories.
Collapse
Affiliation(s)
- Hardy Hagena
- Department of Neurophysiology, Medical Faculty, Ruhr University Bochum, Bochum, Germany
| | - Niels Hansen
- Department of Neurophysiology, Medical Faculty, Ruhr University Bochum, Bochum, Germany
| | | |
Collapse
|
239
|
Wang J, Sun K, Shen Y, Xu Y, Xie J, Huang R, Zhang Y, Xu C, Zhang X, Wang R, Lin Y. DNA methylation is critical for tooth agenesis: implications for sporadic non-syndromic anodontia and hypodontia. Sci Rep 2016; 6:19162. [PMID: 26759063 PMCID: PMC4725352 DOI: 10.1038/srep19162] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 12/02/2015] [Indexed: 02/05/2023] Open
Abstract
Hypodontia is caused by interactions among genetic, epigenetic, and environmental factors during tooth development, but the actual mechanism is unknown. DNA methylation now appears to play a significant role in abnormal developments, flawed phenotypes, and acquired diseases. Methylated DNA immunoprecipitation (MeDIP) has been developed as a new method of scanning large-scale DNA-methylation profiles within particular regions or in the entire genome. Here, we performed a genome-wide scan of paired DNA samples obtained from 4 patients lacking two mandibular incisors and 4 healthy controls with normal dentition. We scanned another female with non-syndromic anodontia and her younger brother with the same gene mutations of the PAX9,MSX1,AXIN2 and EDA, but without developmental abnormalities in the dentition. Results showed significant differences in the methylation level of the whole genome between the hypodontia and the normal groups. Nine genes were spotted, some of which have not been associated with dental development; these genes were related mainly to the development of cartilage, bone, teeth, and neural transduction, which implied a potential gene cascade network in hypodontia at the methylation level. This pilot study reveals the critical role of DNA methylation in hypodontia and might provide insights into developmental biology and the pathobiology of acquired diseases.
Collapse
Affiliation(s)
- Jing Wang
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No.301, Middle Yanchang Road, Shanghai 200072, P.R. China
| | - Ke Sun
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, No.14., 3rd Sec, Ren Min Nan Road, Chengdu 610041, P.R. China
| | - Yun Shen
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No.301, Middle Yanchang Road, Shanghai 200072, P.R. China
| | - Yuanzhi Xu
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No.301, Middle Yanchang Road, Shanghai 200072, P.R. China
| | - Jing Xie
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, No.14., 3rd Sec, Ren Min Nan Road, Chengdu 610041, P.R. China
| | - Renhuan Huang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, No.14., 3rd Sec, Ren Min Nan Road, Chengdu 610041, P.R. China
| | - Yiming Zhang
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No.301, Middle Yanchang Road, Shanghai 200072, P.R. China
| | - Chenyuan Xu
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No.301, Middle Yanchang Road, Shanghai 200072, P.R. China
| | - Xu Zhang
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No.301, Middle Yanchang Road, Shanghai 200072, P.R. China
| | - Raorao Wang
- Department of Stomatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No.301, Middle Yanchang Road, Shanghai 200072, P.R. China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, No.14., 3rd Sec, Ren Min Nan Road, Chengdu 610041, P.R. China
| |
Collapse
|
240
|
Superactivation of AMPA receptors by auxiliary proteins. Nat Commun 2016; 7:10178. [PMID: 26744192 PMCID: PMC4729862 DOI: 10.1038/ncomms10178] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 11/11/2015] [Indexed: 12/15/2022] Open
Abstract
Glutamate receptors form complexes in the brain with auxiliary proteins, which control their activity during fast synaptic transmission through a seemingly bewildering array of effects. Here we devise a way to isolate the activation of complexes using polyamines, which enables us to show that transmembrane AMPA receptor regulatory proteins (TARPs) exert their effects principally on the channel opening reaction. A thermodynamic argument suggests that because TARPs promote channel opening, receptor activation promotes AMPAR-TARP complexes into a superactive state with high open probability. A simple model based on this idea predicts all known effects of TARPs on AMPA receptor function. This model also predicts unexpected phenomena including massive potentiation in the absence of desensitization and supramaximal recovery that we subsequently detected in electrophysiological recordings. This transient positive feedback mechanism has implications for information processing in the brain, because it should allow activity-dependent facilitation of excitatory synaptic transmission through a postsynaptic mechanism. Transmembrane AMPA receptor regulatory proteins (TARPs) exert a number of effects on fast glutamatergic synaptic transmission. Here, the authors model the interactions of two such proteins, Stargazin and γ-8, and propose a simple kinetic mechanism by which TARPs modulate channel opening reaction.
Collapse
|
241
|
Lee K, Goodman L, Fourie C, Schenk S, Leitch B, Montgomery JM. AMPA Receptors as Therapeutic Targets for Neurological Disorders. ION CHANNELS AS THERAPEUTIC TARGETS, PART A 2016; 103:203-61. [DOI: 10.1016/bs.apcsb.2015.10.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
242
|
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
|
243
|
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
|
244
|
Berry RH, Qu J, John SWM, Howell GR, Jakobs TC. Synapse Loss and Dendrite Remodeling in a Mouse Model of Glaucoma. PLoS One 2015; 10:e0144341. [PMID: 26637126 PMCID: PMC4670161 DOI: 10.1371/journal.pone.0144341] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 11/17/2015] [Indexed: 01/05/2023] Open
Abstract
It has been hypothesized that synaptic pruning precedes retinal ganglion cell degeneration in glaucoma, causing early dysfunction to retinal ganglion cells. To begin to assess this, we studied the excitatory synaptic inputs to individual ganglion cells in normal mouse retinas and in retinas with ganglion cell degeneration from glaucoma (DBA/2J), or following an optic nerve crush. Excitatory synapses were labeled by AAV2-mediated transfection of ganglion cells with PSD-95-GFP. After both insults the linear density of synaptic inputs to ganglion cells decreased. In parallel, the dendritic arbors lost complexity. We did not observe any cells that had lost dendritic synaptic input while preserving a normal or near-normal morphology. Within the temporal limits of these observations, dendritic remodeling and synapse pruning thus appear to occur near-simultaneously.
Collapse
Affiliation(s)
- Ryan H. Berry
- Harvard Medical School, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Boston, MA 02114, United States of America
| | - Juan Qu
- Harvard Medical School, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Boston, MA 02114, United States of America
| | - Simon W. M. John
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, United States of America
- The Howard Hughes Medical Institute, The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, United States of America
- Department of Ophthalmology, Tufts University School of Medicine, Boston, MA, United States of America
| | - Gareth R. Howell
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, United States of America
- * E-mail: (TJ); (GH)
| | - Tatjana C. Jakobs
- Harvard Medical School, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Boston, MA 02114, United States of America
- * E-mail: (TJ); (GH)
| |
Collapse
|
245
|
PSD-95 family MAGUKs are essential for anchoring AMPA and NMDA receptor complexes at the postsynaptic density. Proc Natl Acad Sci U S A 2015; 112:E6983-92. [PMID: 26604311 DOI: 10.1073/pnas.1517045112] [Citation(s) in RCA: 202] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The postsynaptic density (PSD)-95 family of membrane-associated guanylate kinases (MAGUKs) are major scaffolding proteins at the PSD in glutamatergic excitatory synapses, where they maintain and modulate synaptic strength. How MAGUKs underlie synaptic strength at the molecular level is still not well understood. Here, we explore the structural and functional roles of MAGUKs at hippocampal excitatory synapses by simultaneous knocking down PSD-95, PSD-93, and synapse-associated protein (SAP)102 and combining electrophysiology and transmission electron microscopic (TEM) tomography imaging to analyze the resulting changes. Acute MAGUK knockdown greatly reduces synaptic transmission mediated by α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate receptors (AMPARs) and N-methyl-d-aspartate receptors (NMDARs). This knockdown leads to a significant rise in the number of silent synapses, diminishes the size of PSDs without changes in pre- or postsynaptic membrane, and depletes the number of membrane-associated PSD-95-like vertical filaments and transmembrane structures, identified as AMPARs and NMDARs by EM tomography. The differential distribution of these receptor-like structures and dependence of their abundance on PSD size matches that of AMPARs and NMDARs in the hippocampal synapses. The loss of these structures following MAGUK knockdown tracks the reduction in postsynaptic AMPAR and NMDAR transmission, confirming the structural identities of these two types of receptors. These results demonstrate that MAGUKs are required for anchoring both types of glutamate receptors at the PSD and are consistent with a structural model where MAGUKs, corresponding to membrane-associated vertical filaments, are the essential structural proteins that anchor and organize both types of glutamate receptors and govern the overall molecular organization of the PSD.
Collapse
|
246
|
Qin X, Jiang Y, Tse YC, Wang Y, Wong TP, Paudel HK. Early Growth Response 1 (Egr-1) Regulates N-Methyl-d-aspartate Receptor (NMDAR)-dependent Transcription of PSD-95 and α-Amino-3-hydroxy-5-methyl-4-isoxazole Propionic Acid Receptor (AMPAR) Trafficking in Hippocampal Primary Neurons. J Biol Chem 2015; 290:29603-16. [PMID: 26475861 DOI: 10.1074/jbc.m115.668889] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Indexed: 01/07/2023] Open
Abstract
The N-methyl-d-aspartate receptor (NMDAR) controls synaptic plasticity and memory function and is one of the major inducers of transcription factor Egr-1 in the hippocampus. However, how Egr-1 mediates the NMDAR signal in neurons has remained unclear. Here, we show that the hippocampus of mice lacking Egr-1 displays electrophysiology properties and ultrastructure that are similar to mice overexpressing PSD-95, a major scaffolding protein of postsynaptic density involved in synapse formation, synaptic plasticity, and synaptic targeting of AMPA receptors (AMPARs), which mediate the vast majority of excitatory transmission in the CNS. We demonstrate that Egr-1 is a transcription repressor of the PSD-95 gene and is recruited to the PSD-95 promoter in response to NMDAR activation. Knockdown of Egr-1 in rat hippocampal primary neurons blocks NMDAR-induced PSD-95 down-regulation and AMPAR endocytosis. Likewise, overexpression of Egr-1 in rat hippocampal primary neurons causes reduction in PSD-95 protein level and promotes AMPAR endocytosis. Our data indicate that Egr-1 is involved in NMDAR-mediated PSD-95 down-regulation and AMPAR endocytosis, a process important in the expression of long term depression.
Collapse
Affiliation(s)
- Xike Qin
- From The Bloomfield Center for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital, and
| | - Yongjun Jiang
- From The Bloomfield Center for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital, and Department of Neurology and Neurosurgery
| | - Yiu Chung Tse
- Douglas Mental Health University Institute, and Department of Psychiatry, McGill University, Montréal, Quebec H4H 1R3, Canada
| | - Yunling Wang
- From The Bloomfield Center for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital, and
| | - Tak Pan Wong
- Douglas Mental Health University Institute, and Department of Psychiatry, McGill University, Montréal, Quebec H4H 1R3, Canada
| | - Hemant K Paudel
- From The Bloomfield Center for Research in Aging, Lady Davis Institute for Medical Research, Jewish General Hospital, and Department of Neurology and Neurosurgery,
| |
Collapse
|
247
|
PDZ interaction of Vangl2 links PSD-95 and Prickle2 but plays only a limited role in the synaptic localisation of Vangl2. Sci Rep 2015; 5:12916. [PMID: 26257100 PMCID: PMC4530445 DOI: 10.1038/srep12916] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 07/14/2015] [Indexed: 02/02/2023] Open
Abstract
Postsynaptic density-95/Discs large/Zonula occludens-1 (PDZ) domain-mediated protein interactions play pivotal roles in various molecular biological events, including protein localisation, assembly, and signal transduction. Although the vertebrate regulator of planar cell polarity Van Gogh-like 2 (Vangl2) was recently described as a postsynaptic molecule with a PDZ-binding motif, the role of its PDZ interaction at the synapse is unknown. In this report, we demonstrate that the PDZ interaction was dispensable for the normal cluster formation of Vangl2 and not absolutely required for the synapse-associated localisation of Vangl2 in cultured hippocampal neurons. We further showed that the synaptic localisation of Vangl2 was categorised into two types: overlapping co-localisation with postsynaptic density (PSD)-95 or highly correlated but complementary pattern of association with PSD-95. Only the former was significantly sensitive to deletion of the PDZ-binding motif. In addition, the PDZ interaction enhanced the protein interactions between PSD-95 and Prickle2, which is another planar cell polarity factor that is localised at the postsynaptic density. Taken together with our recent report that the density of PSD-95 clusters was reduced in Vangl2-silenced neurons, these results suggest that Vangl2 determines the complex formation and clustering of postsynaptic molecules for synaptogenesis in mammalian brains.
Collapse
|
248
|
Roy B, Ahmed KT, Cunningham ME, Ferdous J, Mukherjee R, Zheng W, Chen XZ, Ali DW. Zebrafish TARP Cacng2 is required for the expression and normal development of AMPA receptors at excitatory synapses. Dev Neurobiol 2015; 76:487-506. [PMID: 26178704 DOI: 10.1002/dneu.22327] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 07/01/2015] [Accepted: 07/14/2015] [Indexed: 01/03/2023]
Abstract
Fast excitatory synaptic transmission in the CNS is mediated by the neurotransmitter glutamate, binding to and activating AMPA receptors (AMPARs). AMPARs are known to interact with auxiliary proteins that modulate their behavior. One such family of proteins is the transmembrane AMPA receptor-related proteins, known as TARPs. Little is known about the role of TARPs during development, or about their function in non-mammalian organisms. Here we report the presence of TARPs, specifically the prototypical TARP, stargazin, in developing zebrafish. We find that zebrafish express two forms of stargazin, Cacng2a and Cacng2b from as early as 12-h post fertilization (hpf). Knockdown of Cacng2a and Cacng2b via splice-blocking morpholinos resulted in embryos that exhibited deficits in C-start escape responses, showing reduced C-bend angles, smaller tail velocities and aberrant C-bend turning directions. Injection of the morphants with Cacng2a or 2b mRNA rescued the morphological phenotype and the synaptic deficits. To investigate the effect of reduced Cacng2a and 2b levels on synaptic physiology, we performed whole cell patch clamp recordings of AMPA mEPSCs from zebrafish Mauthner cells. Knockdown of Cacng2a results in reduced AMPA currents and lower mEPSC frequencies, whereas knockdown of Cacng2b displayed no significant change in mEPSC amplitude or frequency. Non-stationary fluctuation analysis confirmed a reduction in the number of active synaptic receptors in the Cacng2a but not in the Cacng2b morphants. Together, these results suggest that Cacng2a is required for normal trafficking and function of synaptic AMPARs, while Cacng2b is largely non-functional with respect to the development of AMPA synaptic transmission.
Collapse
Affiliation(s)
- Birbickram Roy
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada, T6G 2E9
| | - Kazi T Ahmed
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada, T6G 2E9
| | - Marcus E Cunningham
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada, T6G 2E9
| | - Jannatul Ferdous
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada, T6G 2E9
| | - Rajarshi Mukherjee
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada, T6G 2E9
| | - Wang Zheng
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada, T6G 2H7
| | - Xing-Zhen Chen
- Department of Physiology, University of Alberta, Edmonton, Alberta, Canada, T6G 2H7
| | - Declan W Ali
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada, T6G 2E9.,Department of Physiology, University of Alberta, Edmonton, Alberta, Canada, T6G 2H7.,Centre for Neuroscience, University of Alberta, Edmonton, Alberta, Canada, T6G 2E1
| |
Collapse
|
249
|
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
|
250
|
Dunn HA, Ferguson SSG. PDZ Protein Regulation of G Protein–Coupled Receptor Trafficking and Signaling Pathways. Mol Pharmacol 2015; 88:624-39. [DOI: 10.1124/mol.115.098509] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 03/25/2015] [Indexed: 01/03/2023] Open
|