151
|
Abstract
Accumulating data, including those from large genetic association studies, indicate that alterations in glutamatergic synapse structure and function represent a common underlying pathology in many symptomatically distinct cognitive disorders. In this review, we discuss evidence from human genetic studies and data from animal models supporting a role for aberrant glutamatergic synapse function in the etiology of intellectual disability (ID), autism spectrum disorder (ASD), and schizophrenia (SCZ), neurodevelopmental disorders that comprise a significant proportion of human cognitive disease and exact a substantial financial and social burden. The varied manifestations of impaired perceptual processing, executive function, social interaction, communication, and/or intellectual ability in ID, ASD, and SCZ appear to emerge from altered neural microstructure, function, and/or wiring rather than gross changes in neuron number or morphology. Here, we review evidence that these disorders may share a common underlying neuropathy: altered excitatory synapse function. We focus on the most promising candidate genes affecting glutamatergic synapse function, highlighting the likely disease-relevant functional consequences of each. We first present a brief overview of glutamatergic synapses and then explore the genetic and phenotypic evidence for altered glutamate signaling in ID, ASD, and SCZ.
Collapse
Affiliation(s)
- Lenora Volk
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205;
| | | | | | | |
Collapse
|
152
|
Mignogna ML, Giannandrea M, Gurgone A, Fanelli F, Raimondi F, Mapelli L, Bassani S, Fang H, Van Anken E, Alessio M, Passafaro M, Gatti S, Esteban JA, Huganir R, D'Adamo P. The intellectual disability protein RAB39B selectively regulates GluA2 trafficking to determine synaptic AMPAR composition. Nat Commun 2015; 6:6504. [PMID: 25784538 PMCID: PMC4383008 DOI: 10.1038/ncomms7504] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 02/03/2015] [Indexed: 01/31/2023] Open
Abstract
RAB39B is a member of the RAB family of small GTPases that controls intracellular vesicular trafficking in a compartment-specific manner. Mutations in the RAB39B gene cause intellectual disability comorbid with autism spectrum disorder and epilepsy, but the impact of RAB39B loss of function on synaptic activity is largely unexplained. Here we show that protein interacting with C-kinase 1 (PICK1) is a downstream effector of GTP-bound RAB39B and that RAB39B-PICK1 controls trafficking from the endoplasmic reticulum to the Golgi and, hence, surface expression of GluA2, a subunit of alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid receptors (AMPARs). The role of AMPARs in synaptic transmission varies depending on the combination of subunits (GluA1, GluA2 and GluA3) they incorporate. RAB39B downregulation in mouse hippocampal neurons skews AMPAR composition towards non GluA2-containing Ca2+-permeable forms and thereby alters synaptic activity, specifically in hippocampal neurons. We posit that the resulting alteration in synaptic function underlies cognitive dysfunction in RAB39B-related disorders. Mutations in the RAB39B gene, which encodes a protein involved in vesicular trafficking, are associated with intellectual disability, but the impact of RAB39B loss of function on synaptic activity is not known. Here the authors show that RAB39B interacts with PICK1, and that this interaction is critical for the translocation of AMPA receptor subunits into the Golgi.
Collapse
Affiliation(s)
- Maria Lidia Mignogna
- 1] Dulbecco Telethon Institute at IRCCS San Raffaele Scientific Institute, Division of Neuroscience, 20132 Milan, Italy [2] F. Hoffmann-La Roche AG, pRED Pharma Research &Early Development, DTA Neuroscience, CH4070 Basel, Switzerland [3] Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Maila Giannandrea
- 1] Dulbecco Telethon Institute at IRCCS San Raffaele Scientific Institute, Division of Neuroscience, 20132 Milan, Italy [2] F. Hoffmann-La Roche AG, pRED Pharma Research &Early Development, DTA Neuroscience, CH4070 Basel, Switzerland
| | - Antonia Gurgone
- 1] Dulbecco Telethon Institute at IRCCS San Raffaele Scientific Institute, Division of Neuroscience, 20132 Milan, Italy [2] Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Francesca Fanelli
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Francesco Raimondi
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Lisa Mapelli
- CNR Institute of Neuroscience, Department of BIOMETRA, University of Milan, 20129 Milan, Italy
| | - Silvia Bassani
- CNR Institute of Neuroscience, Department of BIOMETRA, University of Milan, 20129 Milan, Italy
| | - Huaqiang Fang
- Department of Neuroscience and Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Eelco Van Anken
- IRCCS San Raffaele Scientific Institute, Division of Genetics and Cell Biology, 20132 Milan, Italy
| | - Massimo Alessio
- IRCCS San Raffaele Scientific Institute, Division of Genetics and Cell Biology, 20132 Milan, Italy
| | - Maria Passafaro
- CNR Institute of Neuroscience, Department of BIOMETRA, University of Milan, 20129 Milan, Italy
| | - Silvia Gatti
- F. Hoffmann-La Roche AG, pRED Pharma Research &Early Development, DTA Neuroscience, CH4070 Basel, Switzerland
| | - José A Esteban
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas/Universidad Autónoma de Madrid, Madrid 28049, Spain
| | - Richard Huganir
- Department of Neuroscience and Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Patrizia D'Adamo
- Dulbecco Telethon Institute at IRCCS San Raffaele Scientific Institute, Division of Neuroscience, 20132 Milan, Italy
| |
Collapse
|
153
|
Wildburger NC, Ali SR, Hsu WCJ, Shavkunov AS, Nenov MN, Lichti CF, LeDuc RD, Mostovenko E, Panova-Elektronova NI, Emmett MR, Nilsson CL, Laezza F. Quantitative proteomics reveals protein-protein interactions with fibroblast growth factor 12 as a component of the voltage-gated sodium channel 1.2 (nav1.2) macromolecular complex in Mammalian brain. Mol Cell Proteomics 2015; 14:1288-300. [PMID: 25724910 PMCID: PMC4424400 DOI: 10.1074/mcp.m114.040055] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Indexed: 12/19/2022] Open
Abstract
Voltage-gated sodium channels (Nav1.1–Nav1.9) are responsible for the initiation and propagation of action potentials in neurons, controlling firing patterns, synaptic transmission and plasticity of the brain circuit. Yet, it is the protein–protein interactions of the macromolecular complex that exert diverse modulatory actions on the channel, dictating its ultimate functional outcome. Despite the fundamental role of Nav channels in the brain, information on its proteome is still lacking. Here we used affinity purification from crude membrane extracts of whole brain followed by quantitative high-resolution mass spectrometry to resolve the identity of Nav1.2 protein interactors. Of the identified putative protein interactors, fibroblast growth factor 12 (FGF12), a member of the nonsecreted intracellular FGF family, exhibited 30-fold enrichment in Nav1.2 purifications compared with other identified proteins. Using confocal microscopy, we visualized native FGF12 in the brain tissue and confirmed that FGF12 forms a complex with Nav1.2 channels at the axonal initial segment, the subcellular specialized domain of neurons required for action potential initiation. Co-immunoprecipitation studies in a heterologous expression system validate Nav1.2 and FGF12 as interactors, whereas patch-clamp electrophysiology reveals that FGF12 acts synergistically with CaMKII, a known kinase regulator of Nav channels, to modulate Nav1.2-encoded currents. In the presence of CaMKII inhibitors we found that FGF12 produces a bidirectional shift in the voltage-dependence of activation (more depolarized) and the steady-state inactivation (more hyperpolarized) of Nav1.2, increasing the channel availability. Although providing the first characterization of the Nav1.2 CNS proteome, we identify FGF12 as a new functionally relevant interactor. Our studies will provide invaluable information to parse out the molecular determinant underlying neuronal excitability and plasticity, and extending the relevance of iFGFs signaling in the normal and diseased brain.
Collapse
Affiliation(s)
- Norelle C Wildburger
- From the ‡Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas, 77555-0617; §Neuroscience Graduate Program, Graduate School of Biomedical Sciences, University of Texas Medical Branch, 301 University Blvd., Galveston, Texas, 77555-0617; ¶UTMB Cancer Center, University of Texas Medical Branch, 301 University Blvd., Galveston, Texas, 77555-1074;
| | - Syed R Ali
- From the ‡Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas, 77555-0617
| | - Wei-Chun J Hsu
- ‖Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, 301 University Blvd., Galveston, Texas, 77555-0617
| | - Alexander S Shavkunov
- From the ‡Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas, 77555-0617; ¶UTMB Cancer Center, University of Texas Medical Branch, 301 University Blvd., Galveston, Texas, 77555-1074
| | - Miroslav N Nenov
- From the ‡Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas, 77555-0617
| | - Cheryl F Lichti
- From the ‡Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas, 77555-0617; ¶UTMB Cancer Center, University of Texas Medical Branch, 301 University Blvd., Galveston, Texas, 77555-1074
| | - Richard D LeDuc
- **National Center for Genome Analysis Support, Indiana University, 107 S Indiana Ave., Bloomington, Indiana, 47408
| | - Ekaterina Mostovenko
- From the ‡Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas, 77555-0617; ¶UTMB Cancer Center, University of Texas Medical Branch, 301 University Blvd., Galveston, Texas, 77555-1074
| | - Neli I Panova-Elektronova
- From the ‡Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas, 77555-0617
| | - Mark R Emmett
- ¶UTMB Cancer Center, University of Texas Medical Branch, 301 University Blvd., Galveston, Texas, 77555-1074; ‖Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, 301 University Blvd., Galveston, Texas, 77555-0617
| | - Carol L Nilsson
- From the ‡Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas, 77555-0617; ¶UTMB Cancer Center, University of Texas Medical Branch, 301 University Blvd., Galveston, Texas, 77555-1074
| | - Fernanda Laezza
- From the ‡Department of Pharmacology and Toxicology, University of Texas Medical Branch, 301 University Blvd, Galveston, Texas, 77555-0617;
| |
Collapse
|
154
|
Abstract
Using a comprehensive proteomic approach, Schwenk et al. (2014), in this issue of Neuron, resolve the differential composition of AMPA receptor complexes in brain regions and through development. This work reveals a specificity in AMPA receptor complex assembly that is dynamic in both space and time.
Collapse
Affiliation(s)
- Mark O Collins
- Department of Biomedical Science and Centre for Membrane Interactions and Dynamics (CMIAD), Firth Court, Western Bank, The University of Sheffield, Sheffield S10 2TN, UK.
| |
Collapse
|
155
|
Abstract
Voltage- and ligand-gated ion channels form the molecular basis of cellular excitability. With >400 members and accounting for ∼1.5% of the human genome, ion channels are some of the most well studied of all proteins in heterologous expression systems. Yet, ion channels often exhibit unexpected properties in vivo because of their interaction with a variety of signaling/scaffolding proteins. Such interactions can influence the function and localization of ion channels, as well as their coupling to intracellular second messengers and pathways, thus increasing the signaling potential of these ion channels in neurons. Moreover, functions have been ascribed to ion channels that are largely independent of their ion-conducting roles. Molecular and functional dissection of the ion channel proteome/interactome has yielded new insights into the composition of ion channel complexes and how their dysregulation leads to human disease.
Collapse
|
156
|
Gratacòs-Batlle E, Yefimenko N, Cascos-García H, Soto D. AMPAR interacting protein CPT1C enhances surface expression of GluA1-containing receptors. Front Cell Neurosci 2015; 8:469. [PMID: 25698923 PMCID: PMC4313699 DOI: 10.3389/fncel.2014.00469] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 12/29/2014] [Indexed: 11/30/2022] Open
Abstract
AMPARs mediate the vast majority of fast excitatory synaptic transmission in the brain and their biophysical and trafficking properties depend on their subunit composition and on several posttranscriptional and posttranslational modifications. Additionally, in the brain AMPARs associate with auxiliary subunits, which modify the properties of the receptors. Despite the abundance of AMPAR partners, recent proteomic studies have revealed even more interacting proteins that could potentially be involved in AMPAR regulation. Amongst these, carnitine palmitoyltransferase 1C (CPT1C) has been demonstrated to form an integral part of native AMPAR complexes in brain tissue extracts. Thus, we aimed to investigate whether CPT1C might be able to modulate AMPAR function. Firstly, we confirmed that CPT1C is an interacting protein of AMPARs in heterologous expression systems. Secondly, CPT1C enhanced whole-cell currents of GluA1 homomeric and GluA1/GluA2 heteromeric receptors. However, CPT1C does not alter the biophysical properties of AMPARs and co-localization experiments revealed that AMPARs and CPT1C are not associated at the plasma membrane despite a strong level of co-localization at the intracellular level. We established that increased surface GluA1 receptor number was responsible for the enhanced AMPAR mediated currents in the presence of CPT1C. Additionally, we revealed that the palmitoylable residue C585 of GluA1 is important in the enhancement of AMPAR trafficking to the cell surface by CPT1C. Nevertheless, despite its potential as a depalmitoylating enzyme, CPT1C does not affect the palmitoylation state of GluA1. To sum up, this work suggests that CPT1C plays a role as a novel regulator of AMPAR surface expression in neurons. Fine modulation of AMPAR membrane trafficking is fundamental in normal synaptic activity and in plasticity processes and CPT1C is therefore a putative candidate to regulate neuronal AMPAR physiology.
Collapse
Affiliation(s)
- Esther Gratacòs-Batlle
- Laboratori de Neurobiologia, Area de Neurobiologia Cellular i Molecular, Institut d'Investigació Biomèdica de Bellvitge-IDIBELL L'Hospitalet de Llobregat, Spain ; Department of Pathology and Experimental Therapeutics, Faculty of Medicine, University of Barcelona L'Hospitalet de Llobregat, Spain
| | - Natalia Yefimenko
- Laboratori de Neurobiologia, Area de Neurobiologia Cellular i Molecular, Institut d'Investigació Biomèdica de Bellvitge-IDIBELL L'Hospitalet de Llobregat, Spain ; Department of Pathology and Experimental Therapeutics, Faculty of Medicine, University of Barcelona L'Hospitalet de Llobregat, Spain
| | - Helena Cascos-García
- Laboratori de Neurobiologia, Area de Neurobiologia Cellular i Molecular, Institut d'Investigació Biomèdica de Bellvitge-IDIBELL L'Hospitalet de Llobregat, Spain ; Department of Pathology and Experimental Therapeutics, Faculty of Medicine, University of Barcelona L'Hospitalet de Llobregat, Spain
| | - David Soto
- Laboratori de Neurobiologia, Area de Neurobiologia Cellular i Molecular, Institut d'Investigació Biomèdica de Bellvitge-IDIBELL L'Hospitalet de Llobregat, Spain ; Department of Pathology and Experimental Therapeutics, Faculty of Medicine, University of Barcelona L'Hospitalet de Llobregat, Spain
| |
Collapse
|
157
|
Subramaniyan S, Hajali V, Scherf T, Sase SJ, Sialana FJ, Gröger M, Bennett KL, Pollak A, Li L, Korz V, Lubec G. Hippocampal receptor complexes paralleling LTP reinforcement in the spatial memory holeboard test in the rat. Behav Brain Res 2015; 283:162-74. [PMID: 25639541 DOI: 10.1016/j.bbr.2015.01.036] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 01/18/2015] [Accepted: 01/21/2015] [Indexed: 12/28/2022]
Abstract
The current study was designed to examine learning-induced transformation of early-LTP into late-LTP. Recording electrodes were implanted into the dentate gyrus of the hippocampus in male rats and early-LTP was induced by weak tetanic stimulation of the medial perforant path. Dorsal right hippocampi were removed, membrane proteins were extracted, separated by blue-native gel electrophoresis with subsequent immunoblotting using brain receptor antibodies. Spatial training resulted into reinforcement of LTP and the reinforced LTP was persistent for 6h. Receptor complex levels containing GluN1 and GluN2A of NMDARs, GluA1 and GluA2 of AMPARs, nAchα7R and the D(1A) dopamine receptor were significantly-elevated in rat hippocampi of animals underwent spatial learning, whilst levels of GluA3 and 5-HT1A receptor containing complexes were significantly reduced. Evidence for complex formation between GluN1 and D(1A) dopamine receptor was provided by antibody shift assay, co-immunoprecipitation and mass spectrometric analysis. Thus our results propose that behavioural stimuli like spatial learning reinforce early LTP into late LTP and this reinforced LTP is accompanied by changes in certain receptor levels in the membrane fraction of the rat hippocampus.
Collapse
Affiliation(s)
| | - Vahid Hajali
- Institute of Biology, Otto von Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Thomas Scherf
- Institute of Biology, Otto von Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Sunetra Jitkar Sase
- Department of Paediatrics, Medizinische Universität Wien, Währinger Gürtel 18A, 1090 Wien
| | - Fernando J Sialana
- Department of Paediatrics, Medizinische Universität Wien, Währinger Gürtel 18A, 1090 Wien
| | - Marion Gröger
- Core facilities, Core Facility Imaging, Medizinische Universität Wien, Anna Spiegel Forschungsgebäude, 1090 Vienna, Austria
| | - Keiryn L Bennett
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Lazarettgasse 14, AKH BT25.3 and 1090 Vienna, Austria
| | - Arnold Pollak
- Department of Paediatrics, Medizinische Universität Wien, Währinger Gürtel 18A, 1090 Wien
| | - Lin Li
- Department of Paediatrics, Medizinische Universität Wien, Währinger Gürtel 18A, 1090 Wien
| | - Volker Korz
- Institute of Biology, Otto von Guericke University Magdeburg, 39120 Magdeburg, Germany.
| | - Gert Lubec
- Department of Paediatrics, Medizinische Universität Wien, Währinger Gürtel 18A, 1090 Wien.
| |
Collapse
|
158
|
Martenson JS, Tomita S. Synaptic localization of neurotransmitter receptors: comparing mechanisms for AMPA and GABAA receptors. Curr Opin Pharmacol 2014; 20:102-8. [PMID: 25529200 DOI: 10.1016/j.coph.2014.11.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 11/26/2014] [Accepted: 11/27/2014] [Indexed: 11/30/2022]
Abstract
Ionotropic neurotransmitter receptors mediate fast synaptic transmission by localizing at postsynapses. Changes in receptor number at synapses induce synaptic plasticity. Thus, mechanisms for the synaptic localization of receptors in basal transmission and synaptic plasticity have been investigated extensively. Recent findings reveal that synaptic localization of tetrameric AMPA receptors in basal transmission requires the PDZ binding of TARP auxiliary subunits, which modulate receptor properties and pharmacology. On the other hand, pentameric GABAA receptors require multiple receptor subunits for their synaptic localization in basal transmission. AMPA receptors seem to utilize distinct mechanisms for basal synaptic localization and synaptic insertion during plasticity. Revealing precise mechanisms for receptor synaptic localization may establish new approaches to control synaptic transmission.
Collapse
Affiliation(s)
- James S Martenson
- Program in Cellular Neuroscience, Neurodegeneration and Repair (CNNR), Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Susumu Tomita
- Program in Cellular Neuroscience, Neurodegeneration and Repair (CNNR), Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
159
|
Transcriptional analysis of a whole-body form of long-term habituation in Aplysia californica. ACTA ACUST UNITED AC 2014; 22:11-23. [PMID: 25512573 PMCID: PMC4274328 DOI: 10.1101/lm.036970.114] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Habituation is the simplest form of learning, but we know little about the transcriptional mechanisms that encode long-term habituation memory. A key obstacle is that habituation is relatively stimulus-specific and is thus encoded in small sets of neurons, providing poor signal/noise ratios for transcriptional analysis. To overcome this obstacle, we have developed a protocol for producing whole-body long-term habituation of the siphon-withdrawal reflex (SWR) of Aplysia californica. Specifically, we constructed a computer-controlled brushing apparatus to apply low-intensity tactile stimulation over the entire dorsal surface of Aplysia at regular intervals. We found that 3 d of training (10 rounds of stimulation/day; each round = 15 min brushing at a 10-sec ISI; 15-min rest between rounds) produces habituation with several characteristics favorable for mechanistic investigation. First, habituation is widespread, with SWR durations reduced whether the reflex is evoked by tactile stimulation to the head, tail, or the siphon. Second, long-term habituation is sensitive to the pattern of training, occurring only when brushing sessions are spaced out over 3 d rather than massed into a single session. Using a custom-designed microarray and quantitative PCR, we show that long-term habituation produces long-term up-regulation of an apparent Aplysia homolog of cornichon, a protein important for glutamate receptor trafficking. Our training paradigm provides a promising starting point for characterizing the transcriptional mechanisms of long-term habituation memory.
Collapse
|
160
|
Chen N, Pandya NJ, Koopmans F, Castelo-Székelv V, van der Schors RC, Smit AB, Li KW. Interaction proteomics reveals brain region-specific AMPA receptor complexes. J Proteome Res 2014; 13:5695-706. [PMID: 25337787 DOI: 10.1021/pr500697b] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Fast excitatory synaptic transmission in the brain is mediated by glutamate acting on postsynaptic AMPA receptors. Recent studies have revealed a substantial number of AMPA receptor auxiliary proteins, which potentially contribute to the regulation of AMPA receptor trafficking, subcellular receptor localization, and receptor gating properties. Here we examined the AMPA receptor interactomes from cortex, hippocampus, and cerebellum by comprehensive interaction proteomics. The study reveals that AMPA receptor auxiliary proteins are engaged in distinct brain region-specific AMPA receptors subcomplexes, which might underlie brain region-specific differential regulation of AMPA receptor properties. Depending on the brain region, an interacting protein can be involved in an AMPA and a non-AMPA receptor complex.
Collapse
Affiliation(s)
- Ning Chen
- Department of Molecular and Cellular Neurobiology, and ‡Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, VU University , De Boelelaan 1085, 1081 HV Amsterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
161
|
Abstract
Cornichon homologs (CNIHs) are AMPA-type glutamate receptor (AMPAR) auxiliary subunits that modulate AMPAR ion channel function and trafficking. Mechanisms underlying this interaction and functional modulation of the receptor complex are currently unclear. Here, using proteins expressed from mouse and rat cDNA, we show that CNIH-3 forms a stable complex with tetrameric AMPARs and contributes to the transmembrane density in single-particle electron microscopy structures. Peptide array-based screening and in vitro mutagenesis identified two clusters of conserved membrane-proximal residues in CNIHs that contribute to AMPAR binding. Because CNIH-1 binds to AMPARs but modulates gating at a significantly lower magnitude compared with CNIH-3, these conserved residues mediate a direct interaction between AMPARs and CNIHs. In addition, residues in the extracellular loop of CNIH-2/3 absent in CNIH-1/4 are critical for both AMPAR interaction and gating modulation. On the AMPAR extracellular domains, the ligand-binding domain and possibly a stretch of linker, connecting the ligand-binding domain to the fourth membrane-spanning segment, is the principal contact point with the CNIH-3 extracellular loop. In contrast, the membrane-distal N-terminal domain is less involved in AMPAR gating modulation by CNIH-3 and AMPAR binding to CNIH-3. Collectively, our results identify conserved residues in the membrane-proximal region of CNIHs that contribute to AMPAR binding and an additional unique segment in the CNIH-2/3 extracellular loop required for both physical interaction and gating modulation of the AMPAR. Consistent with the dissociable properties of binding and gating modulation, we identified a mutant CNIH-3 that preserves AMPAR binding capability but has attenuated activity of gating modulation.
Collapse
|
162
|
Cais O, Herguedas B, Krol K, Cull-Candy SG, Farrant M, Greger IH. Mapping the interaction sites between AMPA receptors and TARPs reveals a role for the receptor N-terminal domain in channel gating. Cell Rep 2014; 9:728-40. [PMID: 25373908 PMCID: PMC4405707 DOI: 10.1016/j.celrep.2014.09.029] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 08/12/2014] [Accepted: 09/19/2014] [Indexed: 11/18/2022] Open
Abstract
AMPA-type glutamate receptors (AMPARs) mediate fast neurotransmission at excitatory synapses. The extent and fidelity of postsynaptic depolarization triggered by AMPAR activation are shaped by AMPAR auxiliary subunits, including the transmembrane AMPAR regulatory proteins (TARPs). TARPs profoundly influence gating, an effect thought to be mediated by an interaction with the AMPAR ion channel and ligand binding domain (LBD). Here, we show that the distal N-terminal domain (NTD) contributes to TARP modulation. Alterations in the NTD-LBD linker result in TARP-dependent and TARP-selective changes in AMPAR gating. Using peptide arrays, we identify a TARP interaction region on the NTD and define the path of TARP contacts along the LBD surface. Moreover, we map key binding sites on the TARP itself and show that mutation of these residues mediates gating modulation. Our data reveal a TARP-dependent allosteric role for the AMPAR NTD and suggest that TARP binding triggers a drastic reorganization of the AMPAR complex. The NTD linker has a TARP-dependent and TARP-specific impact on AMPAR gating Peptide arrays reveal binding of TARPs to both extracellular domains of AMPARs A structural reorganization of AMPARs is triggered by TARP binding
Collapse
Affiliation(s)
- Ondrej Cais
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Beatriz Herguedas
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Karolina Krol
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - Stuart G Cull-Candy
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - Mark Farrant
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - Ingo H Greger
- Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK.
| |
Collapse
|
163
|
Abstract
When native and recombinant kainate receptors (KARs) are compared, there is a mismatch in several of their functional properties. While both generate currents, synaptic responses mediated by KARs have rarely observed in cultured hippocampal neurons. The recent discovery of auxiliary proteins for KARs, such as Netos, offers an explanation for these discrepancies. We found that the GluK5 KAR subunit and the ancillary proteins, Neto1 and Neto2, are not expressed by hippocampal neurons in culture. Therefore, we used this model to directly test whether these proteins are required for the synaptic localization of KARs. Transfection of GluK4, GluK5, Neto1, or Neto2 into hippocampal neurons was associated with the appearance of synaptic KAR-mediated EPSCs. However, GluK4 or GluK5 alone produced synaptic activity in a significant proportion of cells and with reliable event frequency. While neurons expressing GluK4 or GluK5 subunits displayed synaptic responses with rapid kinetics, the expression of Neto proteins conferred these synaptic responses with their characteristic slow onset and decay rates. These data reveal some requirements for KAR targeting to the synapse, indicating a fundamental role of high affinity KAR subunits in this process.
Collapse
Affiliation(s)
| | - M Isabel Aller
- Instituto de Neurociencias (CSIC-UMH), 03550 San Juan de Alicante, Spain
| | - Juan Lerma
- Instituto de Neurociencias (CSIC-UMH), 03550 San Juan de Alicante, Spain
| |
Collapse
|
164
|
Regional Diversity and Developmental Dynamics of the AMPA-Receptor Proteome in the Mammalian Brain. Neuron 2014; 84:41-54. [DOI: 10.1016/j.neuron.2014.08.044] [Citation(s) in RCA: 175] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/19/2014] [Indexed: 12/20/2022]
|
165
|
Howe JR. Modulation of non-NMDA receptor gating by auxiliary subunits. J Physiol 2014; 593:61-72. [PMID: 25556788 DOI: 10.1113/jphysiol.2014.273904] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 08/18/2014] [Indexed: 01/15/2023] Open
Abstract
During the past decade, considerable evidence has accumulated that non-NMDA glutamate receptors (both AMPA and kainate subtypes) are modulated by the association of the core tetrameric receptor with auxiliary proteins that are integral components of native receptor assemblies. This short review focuses on the effect of two types of auxiliary subunits on the biophysical properties and kinetic behaviour of AMPA and kainate receptors at the level of single receptor molecules. Type I transmembrane AMPA receptor proteins increase the number of AMPA receptor openings that result from a single receptor activation as well as the proportion of openings to conductance levels above 30 pS, resulting in larger peak ensemble currents that decay more slowly and bi-exponentially. Co-expression of Neto1 and 2 with pore-forming kainate receptor subunits also increases the duration of bursts and destabilizes desensitized states, resulting in a rapid component of recovery and clusters of bursts that produce a slow component in desensitization decays. The distinct gating seen in the presence of auxiliary subunits reflects slow switching between gating modes with different single-channel kinetics and open probability. At any given time, the relative proportions of receptors in each gating mode determine both the shape and the amplitude of synaptic currents.
Collapse
Affiliation(s)
- James R Howe
- Department of Pharmacology, Yale University School of Medicine, SHM B-251, 333 Cedar Street, New Haven, CT, 06520-8066, USA
| |
Collapse
|
166
|
Haering SC, Tapken D, Pahl S, Hollmann M. Auxiliary subunits: shepherding AMPA receptors to the plasma membrane. MEMBRANES 2014; 4:469-90. [PMID: 25110960 PMCID: PMC4194045 DOI: 10.3390/membranes4030469] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Revised: 07/17/2014] [Accepted: 07/25/2014] [Indexed: 11/24/2022]
Abstract
Ionotropic glutamate receptors (iGluRs) are tetrameric ligand-gated cation channels that mediate excitatory signal transmission in the central nervous system (CNS) of vertebrates. The members of the iGluR subfamily of α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptors (AMPARs) mediate most of the fast excitatory signal transmission, and their abundance in the postsynaptic membrane is a major determinant of the strength of excitatory synapses. Therefore, regulation of AMPAR trafficking to the postsynaptic membrane is an important constituent of mechanisms involved in learning and memory formation, such as long-term potentiation (LTP) and long-term depression (LTD). Auxiliary subunits play a critical role in the facilitation and regulation of AMPAR trafficking and function. The currently identified auxiliary subunits of AMPARs are transmembrane AMPA receptor regulatory proteins (TARPs), suppressor of lurcher (SOL), cornichon homologues (CNIHs), synapse differentiation-induced gene I (SynDIG I), cysteine-knot AMPAR modulating proteins 44 (CKAMP44), and germ cell-specific gene 1-like (GSG1L) protein. In this review we summarize our current knowledge of the modulatory influence exerted by these important but still underappreciated proteins.
Collapse
Affiliation(s)
- Simon C Haering
- Department of Biochemistry I-Receptor Biochemistry, Ruhr University Bochum, Universitätsstraße 150, 44780 Bochum, Germany.
| | - Daniel Tapken
- Department of Biochemistry I-Receptor Biochemistry, Ruhr University Bochum, Universitätsstraße 150, 44780 Bochum, Germany.
| | - Steffen Pahl
- Department of Biochemistry I-Receptor Biochemistry, Ruhr University Bochum, Universitätsstraße 150, 44780 Bochum, Germany.
| | - Michael Hollmann
- Department of Biochemistry I-Receptor Biochemistry, Ruhr University Bochum, Universitätsstraße 150, 44780 Bochum, Germany.
| |
Collapse
|
167
|
Khodosevich K, Jacobi E, Farrow P, Schulmann A, Rusu A, Zhang L, Sprengel R, Monyer H, von Engelhardt J. Coexpressed auxiliary subunits exhibit distinct modulatory profiles on AMPA receptor function. Neuron 2014; 83:601-15. [PMID: 25066086 DOI: 10.1016/j.neuron.2014.07.004] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/11/2014] [Indexed: 01/30/2023]
Abstract
Gating properties and surface trafficking of AMPA receptors (AMPARs) are modulated by auxiliary subunits. Here we studied the function of coexpressed auxiliary subunits belonging to two different classes. We focused on TARP γ-8 and CKAMP44 in dentate gyrus (DG) granule cells, since both subunits are highly expressed in this cell type. TARP γ-8 and CKAMP44 decrease the rate of deactivation but have an opposing influence on receptor desensitization, which accounts for their differential modulation of synaptic short-term plasticity. Furthermore, long-term plasticity (LTP) requires TARP γ-8 but not CKAMP44. The coexpression of both auxiliary subunits is necessary for the efficient targeting of AMPARs to the cell surface of DG granule cells. Finally, electrophysiological and biochemical evidence support the notion that CKAMP44 and TARP γ-8 can be contained in the same AMPAR complex.
Collapse
Affiliation(s)
- Konstantin Khodosevich
- Department of Clinical Neurobiology, University Hospital and DKFZ Heidelberg, 69120 Heidelberg, Germany
| | - Eric Jacobi
- Synaptic Signalling and Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), 53175 Bonn, Germany; Synaptic Signalling and Neurodegeneration, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Paul Farrow
- Synaptic Signalling and Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), 53175 Bonn, Germany; Synaptic Signalling and Neurodegeneration, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Anton Schulmann
- Department of Clinical Neurobiology, University Hospital and DKFZ Heidelberg, 69120 Heidelberg, Germany
| | - Alexandru Rusu
- Department of Clinical Neurobiology, University Hospital and DKFZ Heidelberg, 69120 Heidelberg, Germany
| | - Ling Zhang
- Max Planck Institute for Medical Research, 69120 Heidelberg, Germany
| | - Rolf Sprengel
- Max Planck Institute for Medical Research, 69120 Heidelberg, Germany
| | - Hannah Monyer
- Department of Clinical Neurobiology, University Hospital and DKFZ Heidelberg, 69120 Heidelberg, Germany
| | - Jakob von Engelhardt
- Synaptic Signalling and Neurodegeneration, German Center for Neurodegenerative Diseases (DZNE), 53175 Bonn, Germany; Synaptic Signalling and Neurodegeneration, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| |
Collapse
|
168
|
Green T, Nayeem N. The multifaceted subunit interfaces of ionotropic glutamate receptors. J Physiol 2014; 593:73-81. [PMID: 25556789 DOI: 10.1113/jphysiol.2014.273409] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 05/30/2014] [Indexed: 01/14/2023] Open
Abstract
The past fifteen years has seen a revolution in our understanding of ionotropic glutamate receptor (iGluR) structure, starting with the first view of the ligand binding domain (LBD) published in 1998, and in many ways culminating in the publication of the full-length structure of GluA2 in 2009. These reports have revealed not only the central role played by subunit interfaces in iGluR function, but also myriad binding sites within interfaces for endogenous and exogenous factors. Changes in the conformation of inter-subunit interfaces are central to transmission of ligand gating into pore opening (itself a rearrangement of interfaces), and subsequent closure through desensitization. With the exception of the agonist binding site, which is located entirely within individual subunits, almost all modulatory factors affecting iGluRs appear to bind to sites in subunit interfaces. This review seeks to summarize what we currently understand about the diverse roles interfaces play in iGluR function, and to highlight questions for future research.
Collapse
Affiliation(s)
- Tim Green
- Department of Pharmacology, University of Liverpool, Ashton Street, Liverpool L69 3GE, UK
| | | |
Collapse
|
169
|
Mahadevan V, Pressey JC, Acton BA, Uvarov P, Huang MY, Chevrier J, Puchalski A, Li CM, Ivakine EA, Airaksinen MS, Delpire E, McInnes RR, Woodin MA. Kainate receptors coexist in a functional complex with KCC2 and regulate chloride homeostasis in hippocampal neurons. Cell Rep 2014; 7:1762-70. [PMID: 24910435 PMCID: PMC6340141 DOI: 10.1016/j.celrep.2014.05.022] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Revised: 03/31/2014] [Accepted: 05/09/2014] [Indexed: 12/01/2022] Open
Abstract
KCC2 is the neuron-specific K+-Cl(-) cotransporter required for maintaining low intracellular Cl(-), which is essential for fast inhibitory synaptic transmission in the mature CNS. Despite the requirement of KCC2 for inhibitory synaptic transmission, understanding of the cellular mechanisms that regulate KCC2 expression and function is rudimentary. We examined KCC2 in its native protein complex in vivo to identify key KCC2-interacting partners that regulate KCC2 function. Using blue native-polyacrylamide gel electrophoresis (BN-PAGE), we determined that native KCC2 exists in a macromolecular complex with kainate-type glutamate receptors (KARs). We found that KAR subunits are required for KCC2 oligomerization and surface expression. In accordance with this finding, acute and chronic genetic deletion of KARs decreased KCC2 function and weakened synaptic inhibition in hippocampal neurons. Our results reveal KARs as regulators of KCC2, significantly advancing our growing understanding of the tight interplay between excitation and inhibition.
Collapse
Affiliation(s)
- Vivek Mahadevan
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Jessica C Pressey
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Brooke A Acton
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Pavel Uvarov
- Institute of Biomedicine, Anatomy, University of Helsinki, 00014 Helsinki, Finland
| | - Michelle Y Huang
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Jonah Chevrier
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Andrew Puchalski
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Caiwei M Li
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| | - Evgueni A Ivakine
- Hospital for Sick Children Research Institute, Toronto, ON M5G 1X8, Canada
| | - Matti S Airaksinen
- Institute of Biomedicine, Anatomy, University of Helsinki, 00014 Helsinki, Finland
| | - Eric Delpire
- Department of Anesthesiology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Roderick R McInnes
- Hospital for Sick Children Research Institute, Toronto, ON M5G 1X8, Canada; Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada
| | - Melanie A Woodin
- Department of Cell & Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada.
| |
Collapse
|
170
|
Subramaniyan S, Heo S, Patil S, Li L, Hoger H, Pollak A, Lubec G. A hippocampal nicotinic acetylcholine alpha 7-containing receptor complex is linked to memory retrieval in the multiple-T-maze in C57BL/6j mice. Behav Brain Res 2014; 270:137-45. [PMID: 24837029 DOI: 10.1016/j.bbr.2014.05.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Revised: 05/02/2014] [Accepted: 05/05/2014] [Indexed: 12/13/2022]
Abstract
The link between the cholinergic and serotonergic system in cognitive function is well-documented. There is, however, limited information on spatial memory and this formed the rationale to carry out a study with the aim to show a specific link between nicotinic and serotonergic receptor complexes rather than the corresponding subunits, to spatial memory retrieval in a land maze. A total of 46 mice were used and divided into two groups, trained and untrained (yoked) in the multiple-T-Maze (MTM) and following training during the first four days, probe trials for memory retrieval were performed on days 8, 16 and 30. Six hours following scarification, hippocampi were taken for the analysis of native receptor complex levels using blue-native gels followed by immunoblotting with specific antibodies. 5-HT1A-, 5-HT7-, nAChα4- and nACh-α7-containing receptor complexes were observed and were paralleling memory retrievals and receptor complex levels were shown to be significantly different between trained and yoked animals. Only levels of a nicotinic acetylcholine α7 receptor-containing complex at an apparent molecular weight of approximately 480kDa were shown to be linked to memory retrieval on day 8 but not to retrievals on days 16 and 30 when memory extinction has taken place. Correlation between nAChα4-, 5-HT1A- and 5-HT7-containing receptors and latencies on day 16 may point to a probable link in extinction mechanisms. A series of the abovementioned receptor complexes were correlating among each other probably indicating a serotonergic/cholinergic network paralleling spatial memory formation.
Collapse
Affiliation(s)
- Saraswathi Subramaniyan
- Department of Pediatrics, Medical University of Vienna, Austria Währinger Gürtel 18, A 1090 Vienna, Austria
| | - Seok Heo
- Department of Pediatrics, Medical University of Vienna, Austria Währinger Gürtel 18, A 1090 Vienna, Austria
| | - Sudarshan Patil
- Department of Pediatrics, Medical University of Vienna, Austria Währinger Gürtel 18, A 1090 Vienna, Austria
| | - Lin Li
- Department of Pediatrics, Medical University of Vienna, Austria Währinger Gürtel 18, A 1090 Vienna, Austria
| | - Harald Hoger
- Abteilung für Labortierkunde und - genetik, Medical University of Vienna, Brauhausgasse 34, A 2325 Himberg, Austria
| | - Arnold Pollak
- Department of Pediatrics, Medical University of Vienna, Austria Währinger Gürtel 18, A 1090 Vienna, Austria
| | - Gert Lubec
- Department of Pediatrics, Medical University of Vienna, Austria Währinger Gürtel 18, A 1090 Vienna, Austria.
| |
Collapse
|
171
|
Boudkkazi S, Brechet A, Schwenk J, Fakler B. Cornichon2 Dictates the Time Course of Excitatory Transmission at Individual Hippocampal Synapses. Neuron 2014; 82:848-58. [DOI: 10.1016/j.neuron.2014.03.031] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2014] [Indexed: 11/16/2022]
|
172
|
Rubio MD, Drummond JB, Meador-Woodruff JH. Glutamate receptor abnormalities in schizophrenia: implications for innovative treatments. Biomol Ther (Seoul) 2014; 20:1-18. [PMID: 24116269 PMCID: PMC3792192 DOI: 10.4062/biomolther.2012.20.1.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2011] [Accepted: 11/25/2011] [Indexed: 01/18/2023] Open
Abstract
Schizophrenia is a devastating psychiatric illness that afflicts 1% of the population worldwide, resulting in substantial impact to patients, their families, and health care delivery systems. For many years, schizophrenia has been felt to be associated with dysregulated dopaminergic neurotransmission as a key feature of the pathophysiology of the illness. Although numerous studies point to dopaminergic abnormalities in schizophrenia, dopamine dysfunction cannot completely account for all of the symptoms seen in schizophrenia, and dopamine-based treatments are often inadequate and can be associated with serious side effects. More recently, converging lines of evidence have suggested that there are abnormalities of glutamate transmission in schizophrenia. Glutamatergic neurotransmission involves numerous molecules that facilitate glutamate release, receptor activation, glutamate reuptake, and other synaptic activities. Evidence for glutamatergic abnormalities in schizophrenia primarily has implicated the NMDA and AMPA subtypes of the glutamate receptor. The expression of these receptors and other molecules associated with glutamate neurotransmission has been systematically studied in the brain in schizophrenia. These studies have generally revealed region- and molecule-specific changes in glutamate receptor transcript and protein expression in this illness. Given that glutamatergic neurotransmission has been implicated in the pathophysiology of schizophrenia, recent drug development efforts have targeted the glutamate system. Much effort to date has focused on modulation of the NMDA receptor, although more recently other glutamate receptors and transporters have been the targets of drug development. These efforts have been promising thus far, and ongoing efforts to develop additional drugs that modulate glutamatergic neurotransmission are underway that may hold the potential for novel classes of more effective treatments for this serious psychiatric illness.
Collapse
Affiliation(s)
- Maria D Rubio
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, Birmingham, AL 35294-0021, USA
| | | | | |
Collapse
|
173
|
Sauvageau E, Rochdi MD, Oueslati M, Hamdan FF, Percherancier Y, Simpson JC, Pepperkok R, Bouvier M. CNIH4 interacts with newly synthesized GPCR and controls their export from the endoplasmic reticulum. Traffic 2014; 15:383-400. [PMID: 24405750 DOI: 10.1111/tra.12148] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Revised: 12/24/2013] [Accepted: 01/09/2013] [Indexed: 01/02/2023]
Abstract
The molecular mechanisms regulating G protein-coupled receptors (GPCRs) trafficking from their site of synthesis in the endoplasmic reticulum (ER) to their site of function (the cell surface) remain poorly characterized. Using a bioluminescence resonance energy transfer-based proteomic screen, we identified a novel GPCR-interacting protein; the human cornichon homologue 4 (CNIH4). This previously uncharacterized protein is localized in the early secretory pathway where it interacts with members of the 3 family of GPCRs. Both overexpression and knockdown expression of CNIH4 caused the intracellular retention of GPCRs, indicating that this ER-resident protein plays an important role in GPCR export. Overexpression of CNIH4 at low levels rescued the maturation and cell surface expression of an intracellularly retained mutant form of the β2-adrenergic receptor, further demonstrating a positive role of CNIH4 in GPCR trafficking. Taken with the co-immunoprecipitation of CNIH4 with Sec23 and Sec24, components of the COPII coat complex responsible for ER export, these data suggest that CNIH4 acts as a cargo-sorting receptor, recruiting GPCRs into COPII vesicles.
Collapse
Affiliation(s)
- Etienne Sauvageau
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada; Department of Biochemistry, Université de Montréal, Montréal, Canada
| | | | | | | | | | | | | | | |
Collapse
|
174
|
Karataeva AR, Klaassen RV, Ströder J, Ruiperez-Alonso M, Hjorth JJJ, van Nierop P, Spijker S, Mansvelder HD, Smit AB. C-terminal interactors of the AMPA receptor auxiliary subunit Shisa9. PLoS One 2014; 9:e87360. [PMID: 24498314 PMCID: PMC3911953 DOI: 10.1371/journal.pone.0087360] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 12/23/2013] [Indexed: 01/01/2023] Open
Abstract
Shisa9 (initially named CKAMP44) has been identified as auxiliary subunit of the AMPA-type glutamate receptors and was shown to modulate its physiological properties. Shisa9 is a type-I transmembrane protein and contains a C-terminal PDZ domain that potentially interacts with cytosolic proteins. In this study, we performed a yeast two-hybrid screening that yielded eight PDZ domain-containing interactors of Shisa9, which were independently validated. The identified interactors are known scaffolding proteins residing in the neuronal postsynaptic density. To test whether C-terminal scaffolding interactions of Shisa9 affect synaptic AMPA receptor function in the hippocampus, we disrupted these interactions using a Shisa9 C-terminal mimetic peptide. In the absence of scaffolding interactions of Shisa9, glutamatergic AMPA receptor-mediated synaptic currents in the lateral perforant path of the mouse hippocampus had a faster decay time, and paired-pulse facilitation was reduced. Furthermore, disruption of the PDZ interactions between Shisa9 and its binding partners affected hippocampal network activity. Taken together, our data identifies novel interaction partners of Shisa9, and shows that the C-terminal interactions of Shisa9 through its PDZ domain interaction motif are important for AMPA receptor synaptic and network functions.
Collapse
Affiliation(s)
- Anna R. Karataeva
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit University, Amsterdam, The Netherlands
| | - Remco V. Klaassen
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit University, Amsterdam, The Netherlands
| | - Jasper Ströder
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit University, Amsterdam, The Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit University, Amsterdam, The Netherlands
| | - Marta Ruiperez-Alonso
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit University, Amsterdam, The Netherlands
| | - Johannes J. J. Hjorth
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit University, Amsterdam, The Netherlands
| | - Pim van Nierop
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit University, Amsterdam, The Netherlands
| | - Sabine Spijker
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit University, Amsterdam, The Netherlands
| | - Huibert D. Mansvelder
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit University, Amsterdam, The Netherlands
| | - August B. Smit
- Department of Molecular and Cellular Neurobiology, Center for Neurogenomics and Cognitive Research, Neuroscience Campus Amsterdam, Vrije Universiteit University, Amsterdam, The Netherlands
- * E-mail:
| |
Collapse
|
175
|
Samways DSK. Applications for mass spectrometry in the study of ion channel structure and function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 806:237-61. [PMID: 24952185 DOI: 10.1007/978-3-319-06068-2_10] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Ion channels are intrinsic membrane proteins that form gated ion-permeable pores across biological membranes. Depending on the type, ion channels exhibit sensitivities to a diverse range of stimuli including changes in membrane potential, binding by diffusible ligands, changes in temperature and direct mechanical force. The purpose of these proteins is to facilitate the passive diffusion of ions down their respective electrochemical gradients into and out of the cell, and between intracellular compartments. In doing so, ion channels can affect transmembrane potentials and regulate the intracellular homeostasis of the important second messenger, Ca(2+). The ion channels of the plasma membrane are of particular clinical interest due to their regulation of cell excitability and cytosolic Ca(2+) levels, and the fact that they are most amenable to manipulation by exogenously applied drugs and toxins. A critical step in improving the pharmacopeia of chemicals available that influence the activity of ion channels is understanding how their three-dimensional structure imparts function. Here, progress has been slow relative to that for soluble protein structures in large part due to the limitations of applying conventional structure determination methods, such as X-ray crystallography, nuclear magnetic resonance imaging, and mass spectrometry, to membrane proteins. Although still an underutilized technique in the assessment of membrane protein structure, recent advances have pushed mass spectrometry to the fore as an important complementary approach to studying the structure and function of ion channels. In addition to revealing the subtle conformational changes in ion channel structure that accompany gating and permeation, mass spectrometry is already being used effectively for identifying tissue-specific posttranslational modifications and mRNA splice variants. Furthermore, the use of mass spectrometry for high-throughput proteomics analysis, which has proven so successful for soluble proteins, is already providing valuable insight into the functional interactions of ion channels within the context of the macromolecular-signaling complexes that they inhabit in vivo. In this chapter, the potential for mass spectrometry as a complementary approach to the study of ion channel structure and function will be reviewed with examples of its application.
Collapse
Affiliation(s)
- Damien S K Samways
- Department of Biology, Clarkson University, 8 Clarkson Avenue, Potsdam, NY, 13699, USA,
| |
Collapse
|
176
|
Abstract
The study of synaptic plasticity and specifically LTP and LTD is one of the most active areas of research in neuroscience. In the last 25 years we have come a long way in our understanding of the mechanisms underlying synaptic plasticity. In 1988, AMPA and NMDA receptors were not even molecularly identified and we only had a simple model of the minimal requirements for the induction of plasticity. It is now clear that the modulation of the AMPA receptor function and membrane trafficking is critical for many forms of synaptic plasticity and a large number of proteins have been identified that regulate this complex process. Here we review the progress over the last two and a half decades and discuss the future challenges in the field.
Collapse
|
177
|
Isacoff EY, Jan LY, Minor DL. Conduits of life's spark: a perspective on ion channel research since the birth of neuron. Neuron 2013; 80:658-74. [PMID: 24183018 DOI: 10.1016/j.neuron.2013.10.040] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Heartbeats, muscle twitches, and lightning-fast thoughts are all manifestations of bioelectricity and rely on the activity of a class of membrane proteins known as ion channels. The basic function of an ion channel can be distilled into, "The hole opens. Ions go through. The hole closes." Studies of the fundamental mechanisms by which this process happens and the consequences of such activity in the setting of excitable cells remains the central focus of much of the field. One might wonder after so many years of detailed poking at such a seemingly simple process, is there anything left to learn?
Collapse
Affiliation(s)
- Ehud Y Isacoff
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA; Physical Biosciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | | | | |
Collapse
|
178
|
Au PB, Argiropoulos B, Parboosingh JS, Micheil Innes A. Refinement of the critical region of 1q41q42 microdeletion syndrome identifiesFBXO28as a candidate causative gene for intellectual disability and seizures. Am J Med Genet A 2013; 164A:441-8. [DOI: 10.1002/ajmg.a.36320] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 10/06/2013] [Indexed: 12/21/2022]
Affiliation(s)
- P.Y. Billie Au
- Department of Medical Genetics; University of Calgary; Calgary Alberta Canada
| | - Bob Argiropoulos
- Department of Medical Genetics; University of Calgary; Calgary Alberta Canada
- Alberta Children's Hospital Research Institute for Child and Maternal Health; University of Calgary; Calgary Alberta Canada
| | - Jillian S. Parboosingh
- Department of Medical Genetics; University of Calgary; Calgary Alberta Canada
- Alberta Children's Hospital Research Institute for Child and Maternal Health; University of Calgary; Calgary Alberta Canada
| | - A. Micheil Innes
- Department of Medical Genetics; University of Calgary; Calgary Alberta Canada
- Alberta Children's Hospital Research Institute for Child and Maternal Health; University of Calgary; Calgary Alberta Canada
| |
Collapse
|
179
|
Bassani S, Folci A, Zapata J, Passafaro M. AMPAR trafficking in synapse maturation and plasticity. Cell Mol Life Sci 2013; 70:4411-30. [PMID: 23475111 PMCID: PMC11113961 DOI: 10.1007/s00018-013-1309-1] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2012] [Revised: 02/15/2013] [Accepted: 02/18/2013] [Indexed: 12/15/2022]
Abstract
Glutamate ionotropic alpha-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid (AMPA) receptors (AMPARs) mediate most fast excitatory synaptic transmission in the central nervous system. The content and composition of AMPARs in postsynaptic membranes (which determine synaptic strength) are dependent on the regulated trafficking of AMPAR subunits in and out of the membranes. AMPAR trafficking is a key mechanism that drives nascent synapse development, and is the main determinant of both Hebbian and homeostatic plasticity in mature synapses. Hebbian plasticity seems to be the biological substrate of at least some forms of learning and memory; while homeostatic plasticity (also known as synaptic scaling) keeps neuronal circuits stable by maintaining changes within a physiological range. In this review, we examine recent findings that provide further understanding of the role of AMPAR trafficking in synapse maturation, Hebbian plasticity, and homeostatic plasticity.
Collapse
Affiliation(s)
- Silvia Bassani
- CNR Institute of Neuroscience, Department of Medical Pharmacology, University of Milan, Milan, Italy
| | - Alessandra Folci
- CNR Institute of Neuroscience, Department of Medical Pharmacology, University of Milan, Milan, Italy
| | - Jonathan Zapata
- CNR Institute of Neuroscience, Department of Medical Pharmacology, University of Milan, Milan, Italy
| | - Maria Passafaro
- CNR Institute of Neuroscience, Department of Medical Pharmacology, University of Milan, Milan, Italy
- Dulbecco Telethon Institute, Rome, Italy
| |
Collapse
|
180
|
Liu M, Lewis LD, Shi R, Brown EN, Xu W. Differential requirement for NMDAR activity in SAP97β-mediated regulation of the number and strength of glutamatergic AMPAR-containing synapses. J Neurophysiol 2013; 111:648-58. [PMID: 24225540 DOI: 10.1152/jn.00262.2013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
PSD-95-like, disc-large (DLG) family membrane-associated guanylate kinase proteins (PSD/DLG-MAGUKs) are essential for regulating synaptic AMPA receptor (AMPAR) function and activity-dependent trafficking of AMPARs. Using a molecular replacement strategy to replace endogenous PSD-95 with SAP97β, we show that the prototypic β-isoform of the PSD-MAGUKs, SAP97β, has distinct NMDA receptor (NMDAR)-dependent roles in regulating basic properties of AMPAR-containing synapses. SAP97β enhances the number of AMPAR-containing synapses in an NMDAR-dependent manner, whereas its effect on the size of unitary synaptic response is not fully dependent on NMDAR activity. These effects contrast with those of PSD-95α, which increases both the number of AMPAR-containing synapses and the size of unitary synaptic responses, with or without NMDAR activity. Our results suggest that SAP97β regulates synaptic AMPAR content by increasing surface expression of GluA1-containing AMPARs, whereas PSD-95α enhances synaptic AMPAR content presumably by increasing the synaptic scaffold capacity for synaptic AMPARs. Our approach delineates discrete effects of different PSD-MAGUKs on principal properties of glutamatergic synaptic transmission. Our results suggest that the molecular diversity of PSD-MAGUKs can provide rich molecular substrates for differential regulation of glutamatergic synapses in the brain.
Collapse
Affiliation(s)
- Mingna Liu
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | | | | | | | | |
Collapse
|
181
|
Schmidt-Salzmann C, Li L, Bischofberger J. Functional properties of extrasynaptic AMPA and NMDA receptors during postnatal hippocampal neurogenesis. J Physiol 2013; 592:125-40. [PMID: 24218546 DOI: 10.1113/jphysiol.2013.267203] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
In the mammalian hippocampus, new granule cells are continuously generated throughout life. Although it is well known that they rapidly form several thousand new glutamatergic synapses, the underlying mechanisms are not well understood. As extrasynaptic NMDA receptors are believed to support the generation of new spines, we have studied the functional properties of extrasynaptic ionotropic glutamate receptors in newborn granule cells in juvenile rats during and after synaptic integration. Using the fast application of glutamate to outside-out membrane patches, we show that all immature granule cells express functional AMPA and NMDA receptors. The density of AMPA receptors was small in cells starting to receive excitatory synaptic input (∼30 pS μm(-2)) but substantially increased during synaptic integration to finally reach ∼120 pS μm(-2) in fully mature cells. Interestingly, AMPA receptors showed a biphasic change in desensitization time constant which was slowest during synaptic integration and substantially faster before and afterwards. This was paralleled by a change in the non-desensitizing current component which was maximal during synaptic integration and about 50% smaller afterwards. Surprisingly, the NMDA receptor kinetics and density in young cells was already comparable to mature cells (∼10 pS μm(-2)), leading to an enhanced NMDA/AMPA receptor density ratio. Similar to somatic outside-out patches, iontophoretic application of glutamate onto dendrites also revealed an enhanced dendritic NMDA/AMPA ratio in young cells. These data indicate that prolonged AMPA receptor currents in newly generated young granule cells might support the effective activation of extrasynaptic NMDA receptors and therefore constitute a competitive advantage over mature cells for new synapse formation.
Collapse
Affiliation(s)
- Charlotte Schmidt-Salzmann
- J. Bischofberger: Department of Biomedicine, Physiological Institute, University of Basel, Pestalozzistr. 20, CH-4056 Basel, Switzerland.
| | | | | |
Collapse
|
182
|
Abstract
The extensive dendritic arbor of a pyramidal cell introduces considerable complexity to the integration of synaptic potentials. Propagation of dendritic potentials is largely passive, in contrast to regenerative axonal potentials that are maintained by voltage-gated sodium channels, leading to a declination in amplitude as dendritic potentials travel toward the soma in a manner that disproportionally affects distal synaptic inputs. To counteract this amplitude filtering, Schaffer collateral synapses onto CA1 pyramidal cells contain a varying number of AMPA receptors (AMPARs) per synapse that increases with distance from the soma, a phenomenon known as distance-dependent scaling. Here, we undertake an investigation into the molecular mechanisms of distance-dependent scaling. Using dendritic recordings from rat pyramidal neurons, we confirm the basic scaling phenomenon and find that it is expressed and can be manipulated cell autonomously. Finally, we show that it depends on the presence of both a reserve pool of AMPARs and the AMPAR subunit GluA2.
Collapse
|
183
|
Brockie PJ, Jensen M, Mellem JE, Jensen E, Yamasaki T, Wang R, Maxfield D, Thacker C, Hoerndli F, Dunn PJ, Tomita S, Madsen DM, Maricq AV. Cornichons control ER export of AMPA receptors to regulate synaptic excitability. Neuron 2013; 80:129-42. [PMID: 24094107 DOI: 10.1016/j.neuron.2013.07.028] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2013] [Indexed: 12/16/2022]
Abstract
The strength of synaptic communication at central synapses depends on the number of ionotropic glutamate receptors, particularly the class gated by the agonist AMPA (AMPARs). Cornichon proteins, evolutionarily conserved endoplasmic reticulum cargo adaptors, modify the properties of vertebrate AMPARs when coexpressed in heterologous cells. However, the contribution of cornichons to behavior and in vivo nervous system function has yet to be determined. Here, we take a genetic approach to these questions by studying CNI-1--the sole cornichon homolog in C. elegans. cni-1 mutants hyperreverse, a phenotype associated with increased glutamatergic synaptic transmission. Consistent with this behavior, we find larger glutamate-gated currents in cni-1 mutants with a corresponding increase in AMPAR number. Furthermore, we observe opposite phenotypes in transgenic worms that overexpress CNI-1 or vertebrate homologs. In reconstitution studies, we provide support for an evolutionarily conserved role for cornichons in regulating the export of vertebrate and invertebrate AMPARs.
Collapse
Affiliation(s)
- Penelope J Brockie
- Department of Biology, University of Utah, Salt Lake City, UT 84112-0840, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
184
|
Henley JM, Wilkinson KA. AMPA receptor trafficking and the mechanisms underlying synaptic plasticity and cognitive aging. DIALOGUES IN CLINICAL NEUROSCIENCE 2013. [PMID: 23576886 PMCID: PMC3622464 DOI: 10.31887/dcns.2013.15.1/jhenley] [Citation(s) in RCA: 161] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Even in healthy individuals there is an inexorable agerelated decline in cognitive function. This is due, in large part, to reduced synaptic plasticity caused by changes in the molecular composition of the postsynaptic membrane. AMPA receptors (AMPARs) are glutamate-gated cation channels that mediate the overwhelming majority of fast excitatory transmission in the brain. Changes in AMPAR number and/or function are a core feature of synaptic plasticity and age-related cognitive decline, AMPARs are highly dynamic proteins that are subject to highly controlled trafficking, recycling, and/or degradation and replacement. This active regulation of AMPAR synthesis, targeting, synaptic dwell time, and degradation is fundamentally important for memory formation and storage. Further, aberrant AMPAR trafficking and consequent detrimental changes in synapses are strongly implicated in many brain diseases, which represent a vast social and economic burden. The purpose of this article is to provide an overview of the molecular and cellular AMPA receptor trafficking events that control synaptic responsiveness and plasticity, and highlight what is known currently known about how these processes change with age and disease.
Collapse
Affiliation(s)
- Jeremy M Henley
- School of Biochemistry, MRC Centre for Synaptic Plasticity, University of Bristol, Bristol, UK.
| | | |
Collapse
|
185
|
Ontogeny repeats the phylogenetic recruitment of the cargo exporter cornichon into AMPA receptor signaling complexes. Mol Cell Neurosci 2013; 56:10-7. [DOI: 10.1016/j.mcn.2013.02.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Accepted: 02/01/2013] [Indexed: 12/26/2022] Open
|
186
|
Studniarczyk D, Coombs I, Cull-Candy SG, Farrant M. TARP γ-7 selectively enhances synaptic expression of calcium-permeable AMPARs. Nat Neurosci 2013; 16:1266-74. [PMID: 23872597 PMCID: PMC3858651 DOI: 10.1038/nn.3473] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 06/13/2013] [Indexed: 12/14/2022]
Abstract
Regulation of calcium-permeable AMPA receptors (CP-AMPARs) is crucial in normal synaptic function and neurological disease states. Although transmembrane AMPAR regulatory proteins (TARPs) such as stargazin (γ-2) modulate the properties of calcium-impermeable AMPARs (CI-AMPARs) and promote their synaptic targeting, the TARP-specific rules governing CP-AMPAR synaptic trafficking remain unclear. We used RNA interference to manipulate AMPAR-subunit and TARP expression in γ-2-lacking stargazer cerebellar granule cells--the classic model of TARP deficiency. We found that TARP γ-7 selectively enhanced the synaptic expression of CP-AMPARs and suppressed CI-AMPARs, identifying a pivotal role of γ-7 in regulating the prevalence of CP-AMPARs. In the absence of associated TARPs, both CP-AMPARs and CI-AMPARs were able to localize to synapses and mediate transmission, although their properties were altered. Our results also establish that TARPed synaptic receptors in granule cells require both γ-2 and γ-7 and reveal an unexpected basis for the loss of AMPAR-mediated transmission in stargazer mice.
Collapse
Affiliation(s)
- Dorota Studniarczyk
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | | | | | | |
Collapse
|
187
|
Abstract
Large conductance Ca(2+)- and voltage-activated potassium channels (BKCa) shape neuronal excitability and signal transduction. This reflects the integrated influences of transmembrane voltage and intracellular calcium concentration ([Ca(2+)]i) that gate the channels. This dual gating has been mainly studied as voltage-triggered gating modulated by defined steady-state [Ca(2+)]i, a paradigm that does not approximate native conditions. Here we use submillisecond changes of [Ca(2+)]i to investigate the time course of the Ca(2+)-triggered gating of BKCa channels expressed in Chinese hamster ovary cells at distinct membrane potentials in the physiological range. The results show that Ca(2+) can effectively gate BKCa channels and that Ca(2+) gating is largely different from voltage-driven gating. Most prominently, Ca(2+) gating displays a pronounced delay in the millisecond range between Ca(2+) application and channel opening (pre-onset delay) and exhibits slower kinetics across the entire [Ca(2+)]i-voltage plane. Both characteristics are selectively altered by co-assembled BKβ4 or an epilepsy-causing mutation that either slows deactivation or speeds activation and reduces the pre-onset delay, respectively. Similarly, co-assembly of the BKCa channels with voltage-activated Ca(2+) (Cav) channels, mirroring the native configuration, decreased the pre-onset delay to submillisecond values. In BKCa-Cav complexes, the time course of the hyperpolarizing K(+)-current response is dictated by the Ca(2+) gating of the BKCa channels. Consistent with Cav-mediated Ca(2+) influx, gating was fastest at hyperpolarized potentials, but decreased with depolarization of the membrane potential. Our results demonstrate that under experimental paradigms meant to approximate the physiological conditions BKCa channels primarily operate as ligand-activated channels gated by intracellular Ca(2+) and that Ca(2+) gating is tuned for fast responses in neuronal BKCa-Cav complexes.
Collapse
|
188
|
Lovero KL, Blankenship SM, Shi Y, Nicoll RA. SynDIG1 promotes excitatory synaptogenesis independent of AMPA receptor trafficking and biophysical regulation. PLoS One 2013; 8:e66171. [PMID: 23785483 PMCID: PMC3681968 DOI: 10.1371/journal.pone.0066171] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 05/03/2013] [Indexed: 11/23/2022] Open
Abstract
AMPA receptors–mediators of fast, excitatory transmission and synaptic plasticity in the brain–achieve great functional diversity through interaction with different auxiliary subunits, which alter both the trafficking and biophysical properties of these receptors. In the past several years an abundance of new AMPA receptor auxiliary subunits have been identified, adding astounding variety to the proteins known to directly bind and modulate AMPA receptors. SynDIG1 was recently identified as a novel AMPA receptor interacting protein that directly binds to the AMPA receptor subunit GluA2 in heterologous cells. Functionally, SynDIG1 was found to regulate the strength and density of AMPA receptor containing synapses in hippocampal neurons, though the way in which SynDIG1 exerts these effects remains unknown. Here, we aimed to determine if SynDIG1 acts as a traditional auxiliary subunit, directly regulating the function and localization of AMPA receptors in the rat hippocampus. We find that, unlike any of the previously characterized AMPA receptor auxiliary subunits, SynDIG1 expression does not impact AMPA receptor gating, pharmacology, or surface trafficking. Rather, we show that SynDIG1 regulates the number of functional excitatory synapses, altering both AMPA and NMDA receptor mediated transmission. Our findings suggest that SynDIG1 is not a typical auxiliary subunit to AMPA receptors, but instead is a protein critical to excitatory synaptogenesis.
Collapse
Affiliation(s)
- Kathryn L Lovero
- Departments of Cellular and Molecular Pharmacology and Physiology, University of California San Francisco, San Francisco, California, United States of America
| | | | | | | |
Collapse
|
189
|
Drummond JB, Tucholski J, Haroutunian V, Meador-Woodruff JH. Transmembrane AMPA receptor regulatory protein (TARP) dysregulation in anterior cingulate cortex in schizophrenia. Schizophr Res 2013; 147:32-38. [PMID: 23566497 PMCID: PMC3650109 DOI: 10.1016/j.schres.2013.03.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Revised: 03/07/2013] [Accepted: 03/11/2013] [Indexed: 02/09/2023]
Abstract
The glutamate hypothesis of schizophrenia proposes that abnormal glutamatergic neurotransmission occurs in this illness, and a major contribution may involve dysregulation of the AMPA subtype of ionotropic glutamate receptor (AMPAR). Transmembrane AMPAR regulatory proteins (TARPs) form direct associations with AMPARs to modulate the trafficking and biophysical functions of these receptors, and their dysregulation may alter the localization and activity of AMPARs, thus having a potential role in the pathophysiology of schizophrenia. We performed comparative quantitative real-time PCR and Western blot analysis to measure transcript (schizophrenia, N=25; comparison subjects, N=25) and protein (schizophrenia, N=36; comparison subjects, N=33) expression of TARPs (γ subunits 1-8) in the anterior cingulate cortex (ACC) in schizophrenia and a comparison group. TARP expression was also measured in frontal cortex of rats chronically treated with haloperidol decanoate (28.5mg/kg every three weeks for nine months) to determine the effect of antipsychotic treatment on the expression of these molecules. We found decreased transcript expression of TARP γ-8 in schizophrenia. At the protein level, γ-3 and γ-5 were increased, while γ-4, γ-7 and γ-8 were decreased in schizophrenia. No changes in any of the molecules were noted in the frontal cortex of haloperidol-treated rats. TARPs are abnormally expressed at transcript and protein levels in ACC in schizophrenia, and these changes are likely due to the illness and not to the antipsychotic treatment. Alterations in the expression of TARPs may contribute to the pathophysiology of schizophrenia, and represent a potential mechanism of glutamatergic dysregulation in this illness.
Collapse
Affiliation(s)
- Jana B. Drummond
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, CIRC 589A, 1719 6th Ave South, Birmingham, AI 25294 USA
,Corresponding author. . Tel.: 205.996.6164; fax: 205.975.4879
| | - Janusz Tucholski
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, CIRC 589A, 1719 6th Ave South, Birmingham, AI 25294 USA
| | - Vahram Haroutunian
- Department of Psychiatry, Mount Sinai School of Medicine, James J Peters Veteran Adminis Room 4F-20 130 West Kingsbridge Road Bronx, NY 10468 USA
| | - James H. Meador-Woodruff
- Department of Psychiatry and Behavioral Neurobiology, University of Alabama at Birmingham, CIRC 589A, 1719 6th Ave South, Birmingham, AI 25294 USA
| |
Collapse
|
190
|
Oku S, Takahashi N, Fukata Y, Fukata M. In silico screening for palmitoyl substrates reveals a role for DHHC1/3/10 (zDHHC1/3/11)-mediated neurochondrin palmitoylation in its targeting to Rab5-positive endosomes. J Biol Chem 2013; 288:19816-29. [PMID: 23687301 DOI: 10.1074/jbc.m112.431676] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Protein palmitoylation, a common post-translational lipid modification, plays an important role in protein trafficking and functions. Recently developed palmitoyl-proteomic methods identified many novel substrates. However, the whole picture of palmitoyl substrates has not been clarified. Here, we performed global in silico screening using the CSS-Palm 2.0 program, free software for prediction of palmitoylation sites, and selected 17 candidates as novel palmitoyl substrates. Of the 17 candidates, 10 proteins, including 6 synaptic proteins (Syd-1, transmembrane AMPA receptor regulatory protein (TARP) γ-2, TARP γ-8, cornichon-2, Ca(2+)/calmodulin-dependent protein kinase IIα, and neurochondrin (Ncdn)/norbin), one focal adhesion protein (zyxin), two ion channels (TRPM8 and TRPC1), and one G-protein-coupled receptor (orexin 2 receptor), were palmitoylated. Using the DHHC palmitoylating enzyme library, we found that all tested substrates were palmitoylated by the Golgi-localized DHHC3/7 subfamily. Ncdn, a regulator for neurite outgrowth and synaptic plasticity, was robustly palmitoylated by the DHHC1/10 (zDHHC1/11; z1/11) subfamily, whose substrate has not yet been reported. As predicted by CSS-Palm 2.0, Cys-3 and Cys-4 are the palmitoylation sites for Ncdn. Ncdn was specifically localized in somato-dendritic regions, not in the axon of rat cultured neurons. Stimulated emission depletion microscopy revealed that Ncdn was localized to Rab5-positive early endosomes in a palmitoylation-dependent manner, where DHHC1/10 (z1/11) were also distributed. Knockdown of DHHC1, -3, or -10 (z11) resulted in the loss of Ncdn from Rab5-positive endosomes. Thus, through in silico screening, we demonstrate that Ncdn and the DHHC1/10 (z1/11) and DHHC3/7 subfamilies are novel palmitoyl substrate-enzyme pairs and that Ncdn palmitoylation plays an essential role in its specific endosomal targeting.
Collapse
Affiliation(s)
- Shinichiro Oku
- Division of Membrane Physiology, Department of Cell Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan
| | | | | | | |
Collapse
|
191
|
Herring BE, Shi Y, Suh YH, Zheng CY, Blankenship SM, Roche KW, Nicoll RA. Cornichon proteins determine the subunit composition of synaptic AMPA receptors. Neuron 2013; 77:1083-96. [PMID: 23522044 DOI: 10.1016/j.neuron.2013.01.017] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2013] [Indexed: 10/27/2022]
Abstract
Cornichon-2 and cornichon-3 (CNIH-2/-3) are AMPA receptor (AMPAR) binding proteins that promote receptor trafficking and markedly slow AMPAR deactivation in heterologous cells, but their role in neurons is unclear. Using CNIH-2 and CNIH-3 conditional knockout mice, we find a profound reduction of AMPAR synaptic transmission in the hippocampus. This deficit is due to the selective loss of surface GluA1-containing AMPARs (GluA1A2 heteromers), leaving a small residual pool of synaptic GluA2A3 heteromers. The kinetics of AMPARs in neurons lacking CNIH-2/-3 are faster than those in WT neurons due to the fast kinetics of GluA2A3 heteromers. The remarkably selective effect of CNIHs on the GluA1 subunit is probably mediated by TARP γ-8, which prevents a functional association of CNIHs with non-GluA1 subunits. These results point to a sophisticated interplay between CNIHs and γ-8 that dictates subunit-specific AMPAR trafficking and the strength and kinetics of synaptic AMPAR-mediated transmission.
Collapse
Affiliation(s)
- Bruce E Herring
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | | | | | | | | | | |
Collapse
|
192
|
Predicting protein-protein interactions in the post synaptic density. Mol Cell Neurosci 2013; 56:128-39. [PMID: 23628905 DOI: 10.1016/j.mcn.2013.04.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Revised: 04/09/2013] [Accepted: 04/19/2013] [Indexed: 12/27/2022] Open
Abstract
The post synaptic density (PSD) is a specialization of the cytoskeleton at the synaptic junction, composed of hundreds of different proteins. Characterizing the protein components of the PSD and their interactions can help elucidate the mechanism of long-term changes in synaptic plasticity, which underlie learning and memory. Unfortunately, our knowledge of the proteome and interactome of the PSD is still partial and noisy. In this study we describe a computational framework to improve the reconstruction of the PSD network. The approach is based on learning the characteristics of PSD protein interactions from a set of trusted interactions, expanding this set with data collected from large scale repositories, and then predicting novel interaction with proteins that are suspected to reside in the PSD. Using this method we obtained thirty predicted interactions, with more than half of which having supporting evidence in the literature. We discuss in details two of these new interactions, Lrrtm1 with PSD-95 and Src with Capg. The first may take part in a mechanism underlying glutamatergic dysfunction in schizophrenia. The second suggests an alternative mechanism to regulate dendritic spines maturation.
Collapse
|
193
|
Wen S, Schroeter A, Klöcker N. Synaptic plasticity in hepatic encephalopathy - a molecular perspective. Arch Biochem Biophys 2013; 536:183-8. [PMID: 23624147 DOI: 10.1016/j.abb.2013.04.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 04/13/2013] [Accepted: 04/15/2013] [Indexed: 01/04/2023]
Abstract
Hepatic encephalopathy (HE)(1) is a common neuropsychiatric complication of both acute and chronic liver disease. Clinical symptoms may include motor disturbances and cognitive dysfunction. Available animal models of HE mimic the deficits in cognitive performance including the impaired ability to learn and memorize information. This review explores the question how HE might affect cognitive functions at molecular levels. Both acute and chronic models of HE constrain the plasticity of glutamatergic neurotransmission. Thus, long-lasting activity-dependent changes in synaptic efficiency, known as long-term potentiation (LTP) and long-term depression (LTD) are significantly impeded. We discuss molecules and signal transduction pathways of LTP and LTD that are targeted by experimental HE, with a focus on ionotropic glutamate receptors of the AMPA-subtype. Finally, a novel strategy of functional proteomic analysis is presented, which, if applied differentially, may provide molecular insight into disease-related dysfunction of membrane protein complexes, i.e. disturbed ionotropic glutamate receptor signaling in HE.
Collapse
Affiliation(s)
- Shuping Wen
- Institute of Neural and Sensory Physiology, Medical Faculty, University of Düsseldorf D-40225 Düsseldorf, Germany
| | | | | |
Collapse
|
194
|
Serum proteomics in biomedical research: a systematic review. Appl Biochem Biotechnol 2013; 170:774-86. [PMID: 23609910 DOI: 10.1007/s12010-013-0238-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2012] [Accepted: 04/11/2013] [Indexed: 12/22/2022]
Abstract
Proteins that are important indicators of physiological or pathological states may contribute to the early diagnosis of disease, which may provide a basis for identifying the underlying mechanism of disease development. Serum, contains an abundance of proteins, offers an easy and inexpensive approach for disease detection and possesses a high potential to revolutionize the diagnostics. These differentially expressed proteins in serum have become an important role to monitoring the state for disease. Availability of emerging proteomic techniques gives optimism that serum can eventually be placed as a biomedium for clinical diagnostics. Advancements have benefited biomarker research to the point where serum is now recognized as an excellent diagnostic medium for the detection of disease. Comprehensive proteome of human serum fluid with high accuracy and availability has the potential to open new doors for disease biomarker discovery and for disease diagnostics, providing insights useful for future study. Thus, this review presents an overview of the value of serum as a credible diagnostic tool, and we aim to summarize the proteomic technologies currently used for global analysis of serum proteins and to elaborate on the application of serum proteomics to the discovery of disease biomarkers, and discuss some of the critical challenges and perspectives for this emerging field.
Collapse
|
195
|
Upregulation of cornichon transcripts in the dorsolateral prefrontal cortex in schizophrenia. Neuroreport 2013; 23:1031-4. [PMID: 23103966 DOI: 10.1097/wnr.0b013e32835ad229] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Schizophrenia has been proposed to be associated with abnormal glutamatergic neurotransmission. The AMPA subtype of glutamate receptors (AMPARs) mediates fast excitatory synaptic transmission in the brain, and their trafficking and function is regulated in part by AMPAR auxiliary proteins including the cornichons (CNIH) and transmembrane AMPAR-regulatory proteins. Abnormal regulation of AMPARs through altered expression of these auxiliary proteins could induce changes in glutamatergic neurotransmission and thus the pathophysiology of schizophrenia. In this study, transcript expression of cornichon homologs 1-4 was measured in the dorsolateral prefrontal cortex from schizophrenia (N=25) and comparison (N=25) patient groups by comparative quantitative real-time PCR. Significant upregulation of CNIH-1, CNIH-2, and CNIH-3 mRNA expression was found in schizophrenia, with no change in CNIH-4 expression. To determine the effect of antipsychotic treatment on the expression of these genes, cornichon mRNA expression was assayed in the frontal cortex of rats treated chronically with haloperidol decanoate and no changes in any of the cornichon transcripts were found. Abnormal expression of the CNIH family of genes is consistent with cornichon-mediated AMPAR trafficking abnormalities in schizophrenia, and suggests a new mechanism contributing toward the pathophysiology of this illness.
Collapse
|
196
|
Abstract
L-glutamate is the primary neurotransmitter at excitatory synapses in the vertebrate CNS and at arthropod neuromuscular junctions (NMJs). However, the molecular mechanisms that trigger the recruitment of glutamate receptors at the onset of synaptogenesis and promote their stabilization at postsynaptic densities remain poorly understood. We have reported the discovery of a novel, evolutionary conserved molecule, Neto, essential for clustering of ionotropic glutamate receptors (iGluRs) at Drosophila NMJ. Neto is the first auxiliary subunit described in Drosophila and is the only non-channel subunit absolutely required for functional iGluRs. Here we review the role of Drosophila Neto in synapse assembly, its similarities with other Neto proteins and a new perspective on how glutamatergic synapses are physically assembled and stabilized.
Collapse
Affiliation(s)
- Young-Jun Kim
- 1Program in Cellular Regulation and Metabolism; NICHD; NIH; Bethesda, MD USA
| | | |
Collapse
|
197
|
Kopach O, Voitenko N. Extrasynaptic AMPA receptors in the dorsal horn: Evidence and functional significance. Brain Res Bull 2013. [DOI: 10.1016/j.brainresbull.2012.11.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
198
|
AMPA receptor/TARP stoichiometry visualized by single-molecule subunit counting. Proc Natl Acad Sci U S A 2013; 110:5163-8. [PMID: 23479622 DOI: 10.1073/pnas.1218765110] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Members of the transmembrane AMPA receptor-regulatory protein (TARP) family modulate AMPA receptor (AMPA-R) trafficking and function. AMPA-Rs consist of four pore-forming subunits. Previous studies show that TARPs are an integral part of the AMPA-R complex, acting as accessory subunits for mature receptors in vivo. The TARP/AMPA-R stoichiometry was previously measured indirectly and found to be variable and dependent on TARP expression level, with at most four TARPs associated with each AMPA-R complex. Here, we use a single-molecule technique in live cells that selectively images proteins located in the plasma membrane to directly count the number of TARPs associated with each AMPA-R complex. Although individual GFP-tagged TARP subunits are observed as freely diffusing fluorescent spots on the surface of Xenopus laevis oocytes when expressed alone, coexpression with AMPA-R-mCherry immobilizes the stargazin-GFP spots at sites of AMPA-R-mCherry, consistent with complex formation. We determined the number of TARP molecules associated with each AMPA-R by counting bleaching steps for three different TARP family members: γ-2, γ-3, and γ-4. We confirm that the TARP/AMPA-R stoichiometry depends on TARP expression level and discover that the maximum number of TARPs per AMPA-R complex falls into two categories: up to four γ-2 or γ-3 subunits, but rarely above two for γ-4 subunit. This unexpected AMPA-R/TARP stoichiometry difference has important implications for the assembly and function of TARP/AMPA-R complexes.
Collapse
|
199
|
Sumioka A. Auxiliary subunits provide new insights into regulation of AMPA receptor trafficking. J Biochem 2013; 153:331-7. [PMID: 23426437 DOI: 10.1093/jb/mvt015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Glutamate is a major excitatory neurotransmitter in the vertebrate brain. Among the ionotropic glutamate receptors, α-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate (AMPA) glutamate receptors are the major receptors mediating excitatory fast synaptic transmission. AMPA receptors are also responsible for modifying synaptic strength through the regulation of their numbers at synapses. Their high regulatability, therefore, could contribute to the mechanisms of synaptic plasticity. The mechanisms regulating AMPA receptor trafficking have evoked great interest through the decades. Recent studies show that in the brain, AMPA receptors make complexes with transmembrane AMPA regulatory proteins (TARPs), which serve as auxiliary subunits. TARPs are required for AMPA receptor function and trafficking. After the initial discovery of TARPs, several other AMPA receptor auxiliary subunits were identified: CNIH-2, CNIH-3, CKAMP44, SynDIG1, SOL-1, SOL-2 and GSG-1L. This review discusses progress in identifying the role of auxiliary subunits in AMPA receptor trafficking.
Collapse
Affiliation(s)
- Akio Sumioka
- Department of Aging Neurobiology, National Center for Geriatrics and Gerontology, Morioka-cho Gengo 35, Oobu city, Aichi 474-8511, Japan.
| |
Collapse
|
200
|
Casillas-Espinosa PM, Powell KL, O'Brien TJ. Regulators of synaptic transmission: roles in the pathogenesis and treatment of epilepsy. Epilepsia 2013; 53 Suppl 9:41-58. [PMID: 23216578 DOI: 10.1111/epi.12034] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Synaptic transmission is the communication between a presynaptic and a postsynaptic neuron, and the subsequent processing of the signal. These processes are complex and highly regulated, reflecting their importance in normal brain functioning and homeostasis. Sustaining synaptic transmission depends on the continuing cycle of synaptic vesicle formation, release, and endocytosis, which requires proteins such as dynamin, syndapin, synapsin, and synaptic vesicle protein 2A. Synaptic transmission is regulated by diverse mechanisms, including presynaptic modulators of synaptic vesicle formation and release, postsynaptic receptors and signaling, and modulators of neurotransmission. Neurotransmitters released presynaptically can bind to their postsynaptic receptors, the inhibitory γ-aminobutyric acid (GABA)ergic receptors or the excitatory glutamate receptors. Once released, glutamate activates a variety of postsynaptic receptors including α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA), N-methyl-D-aspartate (NMDA), kainate, and metabotropic receptors. The activation of the receptors triggers downstream signaling cascades generating a vast array of effects, which can be modulated by a numerous auxiliary regulatory subunits. Moreover, different neuropeptides such as neuropeptide Y, brain-derived neurotrophic factor (BDNF), somatostatin, ghrelin, and galanin, act as regulators of diverse synaptic functions and along with the classic neurotransmitters. Abnormalities in the regulation of synaptic transmission play a critical role in the pathogenesis of numerous brain diseases, including epilepsy. This review focuses on the different mechanisms involved in the regulation of synaptic transmission, which may play a role in the pathogenesis of epilepsy: the presynaptic modulators of synaptic vesicle formation and release, postsynaptic receptors, and modulators of neurotransmission, including the mechanism by which drugs can modulate the frequency and severity of epileptic seizures.
Collapse
Affiliation(s)
- Pablo M Casillas-Espinosa
- The Departments of Medicine and Neurology, The Royal Melbourne Hospital, The Melbourne Brain Centre, The University of Melbourne, Parkville, Victoria, Australia
| | | | | |
Collapse
|