101
|
Larsen BR, Holm R, Vilsen B, MacAulay N. Glutamate transporter activity promotes enhanced Na + /K + -ATPase-mediated extracellular K + management during neuronal activity. J Physiol 2016; 594:6627-6641. [PMID: 27231201 DOI: 10.1113/jp272531] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 05/23/2016] [Indexed: 12/14/2022] Open
Abstract
KEY POINTS Management of glutamate and K+ in brain extracellular space is of critical importance to neuronal function. The astrocytic α2β2 Na+ /K+ -ATPase isoform combination is activated by the K+ transients occurring during neuronal activity. In the present study, we report that glutamate transporter-mediated astrocytic Na+ transients stimulate the Na+ /K+ -ATPase and thus the clearance of extracellular K+ . Specifically, the astrocytic α2β1 Na+ /K+ -ATPase subunit combination displays an apparent Na+ affinity primed to react to physiological changes in intracellular Na+ . Accordingly, we demonstrate a distinct physiological role in K+ management for each of the two astrocytic Na+ /K+ -ATPase β-subunits. ABSTRACT Neuronal activity is associated with transient [K+ ]o increases. The excess K+ is cleared by surrounding astrocytes, partly by the Na+ /K+ -ATPase of which several subunit isoform combinations exist. The astrocytic Na+ /K+ -ATPase α2β2 isoform constellation responds directly to increased [K+ ]o but, in addition, Na+ /K+ -ATPase-mediated K+ clearance could be governed by astrocytic [Na+ ]i . During most neuronal activity, glutamate is released in the synaptic cleft and is re-absorbed by astrocytic Na+ -coupled glutamate transporters, thereby elevating [Na+ ]i . It thus remains unresolved whether the different Na+ /K+ -ATPase isoforms are controlled by [K+ ]o or [Na+ ]i during neuronal activity. Hippocampal slice recordings of stimulus-induced [K+ ]o transients with ion-sensitive microelectrodes revealed reduced Na+ /K+ -ATPase-mediated K+ management upon parallel inhibition of the glutamate transporter. The apparent intracellular Na+ affinity of isoform constellations involving the astrocytic β2 has remained elusive as a result of inherent expression of β1 in most cell systems, as well as technical challenges involved in measuring intracellular affinity in intact cells. We therefore expressed the different astrocytic isoform constellations in Xenopus oocytes and determined their apparent Na+ affinity in intact oocytes and isolated membranes. The Na+ /K+ -ATPase was not fully saturated at basal astrocytic [Na+ ]i , irrespective of isoform constellation, although the β1 subunit conferred lower apparent Na+ affinity to the α1 and α2 isoforms than the β2 isoform. In summary, enhanced astrocytic Na+ /K+ -ATPase-dependent K+ clearance was obtained with parallel glutamate transport activity. The astrocytic Na+ /K+ -ATPase isoform constellation α2β1 appeared to be specifically geared to respond to the [Na+ ]i transients associated with activity-induced glutamate transporter activity.
Collapse
Affiliation(s)
- Brian Roland Larsen
- Department Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Rikke Holm
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Bente Vilsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Nanna MacAulay
- Department Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
102
|
Resolution of High-Frequency Mesoscale Intracortical Maps Using the Genetically Encoded Glutamate Sensor iGluSnFR. J Neurosci 2016; 36:1261-72. [PMID: 26818514 DOI: 10.1523/jneurosci.2744-15.2016] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
UNLABELLED Wide-field-of-view mesoscopic cortical imaging with genetically encoded sensors enables decoding of regional activity and connectivity in anesthetized and behaving mice; however, the kinetics of most genetically encoded sensors can be suboptimal for in vivo characterization of frequency bands higher than 1-3 Hz. Furthermore, existing sensors, in particular those that measure calcium (genetically encoded calcium indicators; GECIs), largely monitor suprathreshold activity. Using a genetically encoded sensor of extracellular glutamate and in vivo mesoscopic imaging, we demonstrate rapid kinetics of virally transduced or transgenically expressed glutamate-sensing fluorescent reporter iGluSnFR. In both awake and anesthetized mice, we imaged an 8 × 8 mm field of view through an intact transparent skull preparation. iGluSnFR revealed cortical representation of sensory stimuli with rapid kinetics that were also reflected in correlation maps of spontaneous cortical activities at frequencies up to the alpha band (8-12 Hz). iGluSnFR resolved temporal features of sensory processing such as an intracortical reverberation during the processing of visual stimuli. The kinetics of iGluSnFR for reporting regional cortical signals were more rapid than those for Emx-GCaMP3 and GCaMP6s and comparable to the temporal responses seen with RH1692 voltage sensitive dye (VSD), with similar signal amplitude. Regional cortical connectivity detected by iGluSnFR in spontaneous brain activity identified functional circuits consistent with maps generated from GCaMP3 mice, GCaMP6s mice, or VSD sensors. Viral and transgenic iGluSnFR tools have potential utility in normal physiology, as well as neurologic and psychiatric pathologies in which abnormalities in glutamatergic signaling are implicated. SIGNIFICANCE STATEMENT We have characterized the usage of virally transduced or transgenically expressed extracellular glutamate sensor iGluSnFR to perform wide-field-of-view mesoscopic imaging of cortex in both anesthetized and awake mice. Probes for neurotransmitter concentration enable monitoring of brain activity and provide a more direct measure of regional functional activity that is less dependent on nonlinearities associated with voltage-gated ion channels. We demonstrate functional maps of extracellular glutamate concentration and that this sensor has rapid kinetics that enable reporting high-frequency signaling. This imaging strategy has utility in normal physiology and pathologies in which altered glutamatergic signaling is observed. Moreover, we provide comparisons between iGluSnFR and genetically encoded calcium indicators and voltage-sensitive dyes.
Collapse
|
103
|
Medelin M, Rancic V, Cellot G, Laishram J, Veeraraghavan P, Rossi C, Muzio L, Sivilotti L, Ballerini L. Altered development in GABA co-release shapes glycinergic synaptic currents in cultured spinal slices of the SOD1(G93A) mouse model of amyotrophic lateral sclerosis. J Physiol 2016; 594:3827-40. [PMID: 27098371 DOI: 10.1113/jp272382] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 04/07/2016] [Indexed: 12/20/2022] Open
Abstract
KEY POINTS Increased environmental risk factors in conjunction with genetic susceptibility have been proposed with respect to the remarkable variations in mortality in amyotrophic lateral sclerosis (ALS). In vitro models allow the investigation of the genetically modified counter-regulator of motoneuron toxicity and may help in addressing ALS therapy. Spinal organotypic slice cultures from a mutant form of human superoxide dismutase 1 (SOD1G93A) mouse model of ALS allow the detection of altered glycinergic inhibition in spinal microcircuits. This altered inhibition improved spinal cord excitability, affecting motor outputs in early SOD1(G93A) pathogenesis. ABSTRACT Amyotrophic lateral sclerosis (ALS) is a fatal, adult-onset neurological disease characterized by a progressive degeneration of motoneurons (MNs). In a previous study, we developed organotypic spinal cultures from an ALS mouse model expressing a mutant form of human superoxide dismutase 1 (SOD1(G93A) ). We reported the presence of a significant synaptic rearrangement expressed by these embryonic cultured networks, which may lead to the altered development of spinal synaptic signalling, which is potentially linked to the adult disease phenotype. Recent studies on the same ALS mouse model reported a selective loss of glycinergic innervation in cultured MNs, suggestive of a contribution of synaptic inhibition to MN dysfunction and degeneration. In the present study, we further exploit organotypic cultures from wild-type and SOD1(G93A) mice to investigate the development of glycine-receptor-mediated synaptic currents recorded from the interneurons of the premotor ventral circuits. We performed single cell electrophysiology, immunocytochemistry and confocal microscopy and suggest that GABA co-release may speed the decay of glycine responses altering both temporal precision and signal integration in SOD1(G93A) developing networks at the postsynaptic site. Our hypothesis is supported by the finding of an increased MN bursting activity in immature SOD1(G93A) spinal cords and by immunofluorescence microscopy detection of a longer persistence of GABA in SOD1(G93A) glycinergic terminals in cultured and ex vivo spinal slices.
Collapse
Affiliation(s)
- Manuela Medelin
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Vladimir Rancic
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Giada Cellot
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Jummi Laishram
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | | | - Chiara Rossi
- Neuroimmunology Unit, Division of Neuroscience, Institute of Experimental Neurology (INSPE), San Raffaele Scientific Institute, Milan, Italy
| | - Luca Muzio
- Neuroimmunology Unit, Division of Neuroscience, Institute of Experimental Neurology (INSPE), San Raffaele Scientific Institute, Milan, Italy
| | - Lucia Sivilotti
- Department of Neuroscience, Physiology and Pharmacology, University College London (UCL), London, UK
| | - Laura Ballerini
- Department of Life Sciences, University of Trieste, Trieste, Italy.,International School for Advanced Studies (SISSA/ISAS), Trieste, Italy
| |
Collapse
|
104
|
Danbolt NC, Furness DN, Zhou Y. Neuronal vs glial glutamate uptake: Resolving the conundrum. Neurochem Int 2016; 98:29-45. [PMID: 27235987 DOI: 10.1016/j.neuint.2016.05.009] [Citation(s) in RCA: 158] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 05/03/2016] [Accepted: 05/17/2016] [Indexed: 12/30/2022]
Abstract
Neither normal brain function nor the pathological processes involved in neurological diseases can be adequately understood without knowledge of the release, uptake and metabolism of glutamate. The reason for this is that glutamate (a) is the most abundant amino acid in the brain, (b) is at the cross-roads between several metabolic pathways, and (c) serves as the major excitatory neurotransmitter. In fact most brain cells express glutamate receptors and are thereby influenced by extracellular glutamate. In agreement, brain cells have powerful uptake systems that constantly remove glutamate from the extracellular fluid and thereby limit receptor activation. It has been clear since the 1970s that both astrocytes and neurons express glutamate transporters. However the relative contribution of neuronal and glial transporters to the total glutamate uptake activity, however, as well as their functional importance, has been hotly debated ever since. The present short review provides (a) an overview of what we know about neuronal glutamate uptake as well as an historical description of how we got there, and (b) a hypothesis reconciling apparently contradicting observations thereby possibly resolving the paradox.
Collapse
Affiliation(s)
- N C Danbolt
- The Neurotransporter Group, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.
| | - D N Furness
- School of Life Sciences, Keele University, Keele, Staffs. ST5 5BG, UK
| | - Y Zhou
- The Neurotransporter Group, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| |
Collapse
|
105
|
Leuenberger M, Ritler A, Simonin A, Hediger MA, Lochner M. Concise Asymmetric Synthesis and Pharmacological Characterization of All Stereoisomers of Glutamate Transporter Inhibitor TFB-TBOA and Synthesis of EAAT Photoaffinity Probes. ACS Chem Neurosci 2016; 7:534-9. [PMID: 26918289 DOI: 10.1021/acschemneuro.5b00311] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Glutamate is the major excitatory neurotransmitter in the mammalian brain. Its rapid clearance after the release into the synaptic cleft is vital in order to avoid toxic effects and is ensured by several transmembrane transport proteins, so-called excitatory amino acid transporters (EAATs). Impairment of glutamate removal has been linked to several neurodegenerative diseases and EAATs have therefore received increased attention as therapeutic targets. O-Benzylated l-threo-β-hydroxyaspartate derivatives have been developed previously as highly potent inhibitors of EAATs with TFB-TBOA ((2S,3S)-2-amino-3-((3-(4-(trifluoromethyl)benzamido)benzyl)oxy)succinic acid) standing out as low-nanomolar inhibitor. We report the stereoselective synthesis of all four stereoisomers of TFB-TBOA in less than a fifth of synthetic steps than the published route. For the first time, the inhibitory activity and isoform selectivity of these TFB-TBOA enantio- and diastereomers were assessed on human glutamate transporters EAAT1-3. Furthermore, we synthesized potent photoaffinity probes based on TFB-TBOA using our novel synthetic strategy.
Collapse
Affiliation(s)
- Michele Leuenberger
- Department
of Chemistry and Biochemistry, University of Bern, 3012 Bern, Switzerland
- Swiss National Centre
of Competence in Research, NCCR TransCure, 3008 Bern, Switzerland
| | - Andreas Ritler
- Department
of Chemistry and Biochemistry, University of Bern, 3012 Bern, Switzerland
| | - Alexandre Simonin
- Institute
of Biochemistry and Molecular Medicine, University of Bern, 3012 Bern, Switzerland
- Swiss National Centre
of Competence in Research, NCCR TransCure, 3008 Bern, Switzerland
| | - Matthias A. Hediger
- Institute
of Biochemistry and Molecular Medicine, University of Bern, 3012 Bern, Switzerland
- Swiss National Centre
of Competence in Research, NCCR TransCure, 3008 Bern, Switzerland
| | - Martin Lochner
- Department
of Chemistry and Biochemistry, University of Bern, 3012 Bern, Switzerland
- Swiss National Centre
of Competence in Research, NCCR TransCure, 3008 Bern, Switzerland
| |
Collapse
|
106
|
MacNamee SE, Liu KE, Gerhard S, Tran CT, Fetter RD, Cardona A, Tolbert LP, Oland LA. Astrocytic glutamate transport regulates a Drosophila CNS synapse that lacks astrocyte ensheathment. J Comp Neurol 2016; 524:1979-98. [PMID: 27073064 PMCID: PMC4861170 DOI: 10.1002/cne.24016] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Revised: 04/08/2016] [Accepted: 04/09/2016] [Indexed: 01/24/2023]
Abstract
Anatomical, molecular, and physiological interactions between astrocytes and neuronal synapses regulate information processing in the brain. The fruit fly Drosophila melanogaster has become a valuable experimental system for genetic manipulation of the nervous system and has enormous potential for elucidating mechanisms that mediate neuron-glia interactions. Here, we show the first electrophysiological recordings from Drosophila astrocytes and characterize their spatial and physiological relationship with particular synapses. Astrocyte intrinsic properties were found to be strongly analogous to those of vertebrate astrocytes, including a passive current-voltage relationship, low membrane resistance, high capacitance, and dye-coupling to local astrocytes. Responses to optogenetic stimulation of glutamatergic premotor neurons were correlated directly with anatomy using serial electron microscopy reconstructions of homologous identified neurons and surrounding astrocytic processes. Robust bidirectional communication was present: neuronal activation triggered astrocytic glutamate transport via excitatory amino acid transporter 1 (Eaat1), and blocking Eaat1 extended glutamatergic interneuron-evoked inhibitory postsynaptic currents in motor neurons. The neuronal synapses were always located within 1 μm of an astrocytic process, but none were ensheathed by those processes. Thus, fly astrocytes can modulate fast synaptic transmission via neurotransmitter transport within these anatomical parameters. J. Comp. Neurol. 524:1979-1998, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Sarah E MacNamee
- Department of Neuroscience, University of Arizona, Tucson, Arizona, 85721
| | - Kendra E Liu
- Department of Neuroscience, University of Arizona, Tucson, Arizona, 85721
| | - Stephan Gerhard
- HHMI Janelia Research Campus, Ashburn, Virginia, 20147.,Institute of Neuroinformatics, University of Zurich and ETH Zurich, CH-8057, Zurich, Switzerland
| | - Cathy T Tran
- Department of Neuroscience, University of Arizona, Tucson, Arizona, 85721
| | | | | | - Leslie P Tolbert
- Department of Neuroscience, University of Arizona, Tucson, Arizona, 85721
| | - Lynne A Oland
- Department of Neuroscience, University of Arizona, Tucson, Arizona, 85721
| |
Collapse
|
107
|
Tanui R, Tao Z, Silverstein N, Kanner B, Grewer C. Electrogenic Steps Associated with Substrate Binding to the Neuronal Glutamate Transporter EAAC1. J Biol Chem 2016; 291:11852-64. [PMID: 27044739 DOI: 10.1074/jbc.m116.722470] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Indexed: 12/13/2022] Open
Abstract
Glutamate transporters actively take up glutamate into the cell, driven by the co-transport of sodium ions down their transmembrane concentration gradient. It was proposed that glutamate binds to its binding site and is subsequently transported across the membrane in the negatively charged form. With the glutamate binding site being located partially within the membrane domain, the possibility has to be considered that glutamate binding is dependent on the transmembrane potential and, thus, is electrogenic. Experiments presented in this report test this possibility. Rapid application of glutamate to the wild-type glutamate transporter subtype EAAC1 (excitatory amino acid carrier 1) through photo-release from caged glutamate generated a transient inward current, as expected for the electrogenic inward movement of co-transported Na(+) In contrast, glutamate application to a transporter with the mutation A334E induced transient outward current, consistent with movement of negatively charged glutamate into its binding site within the dielectric of the membrane. These results are in agreement with electrostatic calculations, predicting a valence for glutamate binding of -0.27. Control experiments further validate and rule out other possible explanations for the transient outward current. Electrogenic glutamate binding can be isolated in the mutant glutamate transporter because reactions, such as glutamate translocation and/or Na(+) binding to the glutamate-bound state, are inhibited by the A334E substitution. Electrogenic glutamate binding has to be considered together with other voltage-dependent partial reactions to cooperatively determine the voltage dependence of steady-state glutamate uptake and glutamate buffering at the synapse.
Collapse
Affiliation(s)
- Rose Tanui
- From the Department of Chemistry Binghamton University, Binghamton, New York 13902 and
| | - Zhen Tao
- From the Department of Chemistry Binghamton University, Binghamton, New York 13902 and
| | - Nechama Silverstein
- the Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada Faculty of Medicine, Hebrew University, Jerusalem 91120, Israel
| | - Baruch Kanner
- the Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada Faculty of Medicine, Hebrew University, Jerusalem 91120, Israel
| | - Christof Grewer
- From the Department of Chemistry Binghamton University, Binghamton, New York 13902 and
| |
Collapse
|
108
|
Shimamoto K. Elucidation of Excitatory Neurotransmission and Membrane Protein Integration Mechanisms. BULLETIN OF THE CHEMICAL SOCIETY OF JAPAN 2016. [DOI: 10.1246/bcsj.20150336] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Keiko Shimamoto
- Bioorganic Research Institute, Suntory Foundation for Life Sciences
| |
Collapse
|
109
|
Yasuno Y, Hamada M, Kawasaki M, Shimamoto K, Shigeri Y, Akizawa T, Konishi M, Ohfune Y, Shinada T. (7S)-Kaitocephalin as a potent NMDA receptor selective ligand. Org Biomol Chem 2016; 14:1206-10. [DOI: 10.1039/c5ob02301g] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
A structure–activity relationship (SAR) study of kaitocephalin, known to be a potent naturally occurring NMDA receptor ligand, was performed.
Collapse
Affiliation(s)
- Yoko Yasuno
- Graduate School of Science
- Osaka City University
- Osaka 558-8585
- Japan
| | - Makoto Hamada
- Graduate School of Science
- Osaka City University
- Osaka 558-8585
- Japan
| | | | - Keiko Shimamoto
- Bioorganic Research Institute
- Suntory Foundation for Life Sciences
- Kyoto 619-0284
- Japan
| | - Yasushi Shigeri
- National Institute of Advanced Industrial Science and Technology
- Osaka 563-8577
- Japan
| | - Toshifumi Akizawa
- Analytical Chemistry
- Pharmaceutical Science
- Setsunan University
- Osaka 573-0101
- Japan
| | - Motomi Konishi
- Analytical Chemistry
- Pharmaceutical Science
- Setsunan University
- Osaka 573-0101
- Japan
| | - Yasufumi Ohfune
- Graduate School of Science
- Osaka City University
- Osaka 558-8585
- Japan
| | - Tetsuro Shinada
- Graduate School of Science
- Osaka City University
- Osaka 558-8585
- Japan
| |
Collapse
|
110
|
Matott MP, Ruyle BC, Hasser EM, Kline DD. Excitatory amino acid transporters tonically restrain nTS synaptic and neuronal activity to modulate cardiorespiratory function. J Neurophysiol 2015; 115:1691-702. [PMID: 26719090 DOI: 10.1152/jn.01054.2015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 12/24/2015] [Indexed: 11/22/2022] Open
Abstract
The nucleus tractus solitarii (nTS) is the initial central termination site for visceral afferents and is important for modulation and integration of multiple reflexes including cardiorespiratory reflexes. Glutamate is the primary excitatory neurotransmitter in the nTS and is removed from the extracellular milieu by excitatory amino acid transporters (EAATs). The goal of this study was to elucidate the role of EAATs in the nTS on basal synaptic and neuronal function and cardiorespiratory regulation. The majority of glutamate clearance in the central nervous system is believed to be mediated by astrocytic EAAT 1 and 2. We confirmed the presence of EAAT 1 and 2 within the nTS and their colocalization with astrocytic markers. EAAT blockade withdl-threo-β-benzyloxyaspartic acid (TBOA) produced a concentration-related depolarization, increased spontaneous excitatory postsynaptic current (EPSC) frequency, and enhanced action potential discharge in nTS neurons. Solitary tract-evoked EPSCs were significantly reduced by EAAT blockade. Microinjection of TBOA into the nTS of anesthetized rats induced apneic, sympathoinhibitory, depressor, and bradycardic responses. These effects mimicked the response to microinjection of exogenous glutamate, and glutamate responses were enhanced by EAAT blockade. Together these data indicate that EAATs tonically restrain nTS excitability to modulate cardiorespiratory function.
Collapse
Affiliation(s)
- Michael P Matott
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Brian C Ruyle
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - Eileen M Hasser
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri; and Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| | - David D Kline
- Department of Biomedical Sciences, University of Missouri, Columbia, Missouri, Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri
| |
Collapse
|
111
|
Stein LR, Zorumski CF, Imai SI, Izumi Y. Nampt is required for long-term depression and the function of GluN2B subunit-containing NMDA receptors. Brain Res Bull 2015; 119:41-51. [PMID: 26481044 DOI: 10.1016/j.brainresbull.2015.10.005] [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: 05/10/2015] [Revised: 09/13/2015] [Accepted: 10/12/2015] [Indexed: 01/01/2023]
Abstract
Nicotinamide adenine dinucleotide (NAD(+)) is an essential coenzyme/cosubstrate for many biological processes in cellular metabolism. The rate-limiting step in the major pathway of mammalian NAD(+) biosynthesis is mediated by nicotinamide phosphoribosyltransferase (Nampt). Previously, we showed that mice lacking Nampt in forebrain excitatory neurons (CamKIIαNampt(-/-) mice) exhibited hyperactivity, impaired learning and memory, and reduced anxiety-like behaviors. However, it remained unclear if these functional effects were accompanied by synaptic changes. Here, we show that CamKIIαNampt(-/-) mice have impaired induction of long-term depression (LTD) in the Schaffer collateral pathway, but normal induction of long-term potentiation (LTP), at postnatal day 30. Pharmacological assessments demonstrated that CamKIIαNampt(-/-) mice also display dysfunction of synaptic GluN2B (NR2B)-containing N-methyl-d-aspartate receptors (NMDARs) prior to changes in NMDAR subunit expression. These results support a novel, important role for Nampt-mediated NAD(+) biosynthesis in LTD and in the function of GluN2B-containing NMDARs.
Collapse
Affiliation(s)
- Liana Roberts Stein
- Department of Developmental Biology, Washington University School of Medicine, Campus Box 8103, 660 South Euclid Avenue, St. Louis, MO 63110, USA; Department of Psychiatry, The Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, Campus Box 8134, 660 South Euclid Avenue, St. Louis, MO 63110, USA.
| | - Charles F Zorumski
- Department of Psychiatry, The Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, Campus Box 8134, 660 South Euclid Avenue, St. Louis, MO 63110, USA; Department of Anatomy and Neurobiology, Washington University School of Medicine, Campus Box 8134, 660 South Euclid Avenue, St. Louis, MO 63110, USA.
| | - Shin-Ichiro Imai
- Department of Developmental Biology, Washington University School of Medicine, Campus Box 8103, 660 South Euclid Avenue, St. Louis, MO 63110, USA.
| | - Yukitoshi Izumi
- Department of Psychiatry, The Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, Campus Box 8134, 660 South Euclid Avenue, St. Louis, MO 63110, USA.
| |
Collapse
|
112
|
Huang L, Zhao S, Lu W, Guan S, Zhu Y, Wang JH. Acidosis-Induced Dysfunction of Cortical GABAergic Neurons through Astrocyte-Related Excitotoxicity. PLoS One 2015; 10:e0140324. [PMID: 26474076 PMCID: PMC4608795 DOI: 10.1371/journal.pone.0140324] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 09/24/2015] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Acidosis impairs cognitions and behaviors presumably by acidification-induced changes in neuronal metabolism. Cortical GABAergic neurons are vulnerable to pathological factors and their injury leads to brain dysfunction. How acidosis induces GABAergic neuron injury remains elusive. As the glia cells and neurons interact each other, we intend to examine the role of the astrocytes in acidosis-induced GABAergic neuron injury. RESULTS Experiments were done at GABAergic cells and astrocytes in mouse cortical slices. To identify astrocytic involvement in acidosis-induced impairment, we induced the acidification in single GABAergic neuron by infusing proton intracellularly or in both neurons and astrocytes by using proton extracellularly. Compared the effects of intracellular acidification and extracellular acidification on GABAergic neurons, we found that their active intrinsic properties and synaptic outputs appeared more severely impaired in extracellular acidosis than intracellular acidosis. Meanwhile, extracellular acidosis deteriorated glutamate transporter currents on the astrocytes and upregulated excitatory synaptic transmission on the GABAergic neurons. Moreover, the antagonists of glutamate NMDA-/AMPA-receptors partially reverse extracellular acidosis-induced injury in the GABAergic neurons. CONCLUSION Our studies suggest that acidosis leads to the dysfunction of cortical GABAergic neurons by astrocyte-mediated excitotoxicity, in addition to their metabolic changes as indicated previously.
Collapse
Affiliation(s)
- Li Huang
- Department of Pathophysiology, Bengbu Medical College, Bengbu Anhui, China 233000
| | - Shidi Zhao
- Department of Pathophysiology, Bengbu Medical College, Bengbu Anhui, China 233000
| | - Wei Lu
- Collaborative Innovation Center for Neurodegenerative Disorders in Shandong, Qingdao University, Medical College, 38 Dengzhou, Shandong China 266021
| | - Sudong Guan
- Department of Pathophysiology, Bengbu Medical College, Bengbu Anhui, China 233000
| | - Yan Zhu
- Department of Pathophysiology, Bengbu Medical College, Bengbu Anhui, China 233000
| | - Jin-Hui Wang
- Institute of Biophysics, Chinese Academy of Sciences, Beijing China 100101
| |
Collapse
|
113
|
Takahashi K, Foster JB, Lin CLG. Glutamate transporter EAAT2: regulation, function, and potential as a therapeutic target for neurological and psychiatric disease. Cell Mol Life Sci 2015; 72:3489-506. [PMID: 26033496 PMCID: PMC11113985 DOI: 10.1007/s00018-015-1937-8] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 05/22/2015] [Accepted: 05/26/2015] [Indexed: 12/12/2022]
Abstract
Glutamate is the predominant excitatory neurotransmitter in the central nervous system. Excitatory amino acid transporter 2 (EAAT2) is primarily responsible for clearance of extracellular glutamate to prevent neuronal excitotoxicity and hyperexcitability. EAAT2 plays a critical role in regulation of synaptic activity and plasticity. In addition, EAAT2 has been implicated in the pathogenesis of many central nervous system disorders. In this review, we summarize current understanding of EAAT2, including structure, pharmacology, physiology, and functions, as well as disease relevancy, such as in stroke, Parkinson's disease, epilepsy, amyotrophic lateral sclerosis, Alzheimer's disease, major depressive disorder, and addiction. A large number of studies have demonstrated that up-regulation of EAAT2 protein provides significant beneficial effects in many disease models suggesting EAAT2 activation is a promising therapeutic approach. Several EAAT2 activators have been identified. Further understanding of EAAT2 regulatory mechanisms could improve development of drug-like compounds that spatiotemporally regulate EAAT2.
Collapse
Affiliation(s)
- Kou Takahashi
- Department of Neuroscience, The Ohio State University, 333 West 10th Avenue, Columbus, OH 43210 USA
| | - Joshua B. Foster
- Department of Neuroscience, The Ohio State University, 333 West 10th Avenue, Columbus, OH 43210 USA
| | - Chien-Liang Glenn Lin
- Department of Neuroscience, The Ohio State University, 333 West 10th Avenue, Columbus, OH 43210 USA
| |
Collapse
|
114
|
The Split Personality of Glutamate Transporters: A Chloride Channel and a Transporter. Neurochem Res 2015; 41:593-9. [DOI: 10.1007/s11064-015-1699-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 07/21/2015] [Accepted: 08/13/2015] [Indexed: 02/07/2023]
|
115
|
Fontana ACK. Current approaches to enhance glutamate transporter function and expression. J Neurochem 2015; 134:982-1007. [DOI: 10.1111/jnc.13200] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Revised: 05/19/2015] [Accepted: 05/20/2015] [Indexed: 12/12/2022]
Affiliation(s)
- Andréia C. K. Fontana
- Department of Pharmacology and Physiology; Drexel University College of Medicine; Philadelphia Pennsylvania USA
| |
Collapse
|
116
|
López-Valenzuela CL, Morales-Villagrán A, Medina-Ceja L. A novel method for simultaneous glutamate and extracellular activity measurement in brain slices with high temporal resolution. Talanta 2015; 144:1231-8. [PMID: 26452952 DOI: 10.1016/j.talanta.2015.08.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 08/02/2015] [Accepted: 08/05/2015] [Indexed: 11/30/2022]
Abstract
Measurement of neurotransmitters during normal or altered function in cerebral slices could be an important tool to better understand the relationship between biochemical changes and electrophysiological activity. Some attempts of this analysis have been made; however, the current techniques do not have the appropriate time resolution to establish this relationship. The use of electrochemical biosensors has allowed for good time resolution, but problems related to the reduction of signal noise and biofouling of the electrode surface could be an important issue. In this work, we propose a new alternative to simultaneously measure glutamate and electrical activity with a high temporal resolution in brain slices. This approach is based on the use of enzymatic reactors that generate a fluorescent derivative from glutamate that can be measured at high temporal resolution. The results presented here show a reliable measurement of this neurotransmitter in brain slices obtained from intact animals under the effect of a glutamate transporter blocker DL-threo-beta-benzyloxyaspartate as well as the potassium channel blocker 4-aminopyridine. Differences in the levels of glutamate and high frequency and amplitude discharges as an effect of drug administration were found in brain slices obtained from epileptic rats (p<0.05). In conclusion, this method could be used to measure neurotransmitter concentration online at a near physiological temporal resolution, which can then be correlated to the electrical activity that is simultaneously recorded.
Collapse
Affiliation(s)
- C L López-Valenzuela
- Laboratory of Neurophysiology and Neurochemistry, Department of Cellular and Molecular Biology, CUCBA, University of Guadalajara, Jalisco, Mexico
| | - A Morales-Villagrán
- Laboratory of Neurophysiology and Neurochemistry, Department of Cellular and Molecular Biology, CUCBA, University of Guadalajara, Jalisco, Mexico.
| | - L Medina-Ceja
- Laboratory of Neurophysiology and Neurochemistry, Department of Cellular and Molecular Biology, CUCBA, University of Guadalajara, Jalisco, Mexico
| |
Collapse
|
117
|
de Villiers J, de Villiers M, Geertsema EM, Raj H, Poelarends GJ. Chemoenzymatic Synthesis of ortho-, meta-, and para-Substituted Derivatives of l- threo-3-Benzyloxyaspartate, An Important Glutamate Transporter Blocker. ChemCatChem 2015; 7:1931-1934. [PMID: 26251674 PMCID: PMC4517298 DOI: 10.1002/cctc.201500318] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Indexed: 01/13/2023]
Abstract
A simple, three-step chemoenzymatic synthesis of l-threo-3-benzyloxyaspartate (l-TBOA), as well as l-TBOA derivatives with F, CF3, and CH3 substituents at the aromatic ring, starting from dimethyl acetylenedicarboxylate was investigated. These chiral amino acids, which are extremely difficult to prepare by chemical synthesis, form an important class of inhibitors of excitatory amino acid transporters involved in the regulation of glutamatergic neurotransmission. In addition, a new chemical procedure for the synthesis of racemic mixtures of TBOA and its derivatives was explored. These chemically prepared racemates are valuable reference compounds in chiral-phase HPLC to establish the enantiopurities of the corresponding chemoenzymatically prepared amino acids.
Collapse
Affiliation(s)
- Jandré de Villiers
- Department of Pharmaceutical Biology, Groningen Research Institute of Pharmacy University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen (The Netherlands )
| | - Marianne de Villiers
- Department of Pharmaceutical Biology, Groningen Research Institute of Pharmacy University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen (The Netherlands )
| | - Edzard M Geertsema
- Department of Pharmaceutical Biology, Groningen Research Institute of Pharmacy University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen (The Netherlands )
| | - Hans Raj
- Department of Pharmaceutical Biology, Groningen Research Institute of Pharmacy University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen (The Netherlands ) ; Current address: Chr-Hansen A/S, Boge Alle 10-12 2970 Horsholm (Denmark)
| | - Gerrit J Poelarends
- Department of Pharmaceutical Biology, Groningen Research Institute of Pharmacy University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen (The Netherlands )
| |
Collapse
|
118
|
Ferreira NR, Lourenço C, Barbosa RM, Laranjinha J. Coupling of ascorbate and nitric oxide dynamics in vivo in the rat hippocampus upon glutamatergic neuronal stimulation: A novel functional interplay. Brain Res Bull 2015; 114:13-9. [DOI: 10.1016/j.brainresbull.2015.03.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 02/09/2015] [Accepted: 03/07/2015] [Indexed: 01/01/2023]
|
119
|
Hanson E, Armbruster M, Cantu D, Andresen L, Taylor A, Danbolt NC, Dulla CG. Astrocytic glutamate uptake is slow and does not limit neuronal NMDA receptor activation in the neonatal neocortex. Glia 2015; 63:1784-96. [PMID: 25914127 DOI: 10.1002/glia.22844] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 03/30/2015] [Accepted: 04/07/2015] [Indexed: 01/20/2023]
Abstract
Glutamate uptake by astrocytes controls the time course of glutamate in the extracellular space and affects neurotransmission, synaptogenesis, and circuit development. Astrocytic glutamate uptake has been shown to undergo post-natal maturation in the hippocampus, but has been largely unexplored in other brain regions. Notably, glutamate uptake has never been examined in the developing neocortex. In these studies, we investigated the development of astrocytic glutamate transport, intrinsic membrane properties, and control of neuronal NMDA receptor activation in the developing neocortex. Using astrocytic and neuronal electrophysiology, immunofluorescence, and Western blot analysis we show that: (1) glutamate uptake in the neonatal neocortex is slow relative to neonatal hippocampus; (2) astrocytes in the neonatal neocortex undergo a significant maturation of intrinsic membrane properties; (3) slow glutamate uptake is accompanied by lower expression of both GLT-1 and GLAST; (4) glutamate uptake is less dependent on GLT-1 in neonatal neocortex than in neonatal hippocampus; and (5) the slow glutamate uptake we report in the neonatal neocortex corresponds to minimal astrocytic control of neuronal NMDA receptor activation. Taken together, our results clearly show fundamental differences between astrocytic maturation in the developing neocortex and hippocampus, and corresponding changes in how astrocytes control glutamate signaling.
Collapse
Affiliation(s)
- Elizabeth Hanson
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts.,Neuroscience Program, Tufts Sackler School of Biomedical Sciences, Boston, Massachusetts
| | - Moritz Armbruster
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts
| | - David Cantu
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts
| | - Lauren Andresen
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts.,Neuroscience Program, Tufts Sackler School of Biomedical Sciences, Boston, Massachusetts
| | - Amaro Taylor
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts
| | - Niels Christian Danbolt
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Chris G Dulla
- Department of Neuroscience, Tufts University School of Medicine, Boston, Massachusetts.,Neuroscience Program, Tufts Sackler School of Biomedical Sciences, Boston, Massachusetts
| |
Collapse
|
120
|
Gottipati MK, Bekyarova E, Haddon RC, Parpura V. Chemically functionalized single-walled carbon nanotubes enhance the glutamate uptake characteristics of mouse cortical astrocytes. Amino Acids 2015; 47:1379-88. [PMID: 25837300 DOI: 10.1007/s00726-015-1970-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Accepted: 03/19/2015] [Indexed: 11/28/2022]
Abstract
Using a radioactive glutamate uptake assay and immunolabeling, we report that single-walled carbon nanotubes, chemically functionalized with polyethylene glycol (SWCNT-PEG), delivered as a colloidal solute, cause an increase in the uptake of extracellular glutamate by astrocytes and an increase in the immunoreactivity of the glutamate transporter GLAST on their cell surface, which is likely a consequence of an increase in the immunoreactivity of glial fibrillary acidic protein. Additional corollary is that astrocytes exposed to SWCNT-PEG became larger and stellate, morphological characteristics of maturation and heightened activity of these glial cells. These results imply that SWCNT-PEG could potentially be used as a viable candidate for neural prosthesis applications, perhaps to alleviate the death toll of neurons due to glutamate excitotoxicity, a pathological process observed in brain and spinal cord injuries.
Collapse
Affiliation(s)
- Manoj K Gottipati
- Department of Neurobiology and Department of Biomedical Engineering, University of Alabama at Birmingham, 1719 6th Ave S, CIRC 429, Birmingham, AL, 35294, USA
| | | | | | | |
Collapse
|
121
|
Hinzman JM, DiNapoli VA, Mahoney EJ, Gerhardt GA, Hartings JA. Spreading depolarizations mediate excitotoxicity in the development of acute cortical lesions. Exp Neurol 2015; 267:243-53. [PMID: 25819105 DOI: 10.1016/j.expneurol.2015.03.014] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 03/09/2015] [Accepted: 03/17/2015] [Indexed: 11/19/2022]
Abstract
Spreading depolarizations (SD) are mass depolarizations of neurons and astrocytes that occur spontaneously in acute brain injury and mediate time-dependent lesion growth. Glutamate excitotoxicity has also been extensively studied as a mechanism of neuronal injury, although its relevance to in vivo pathology remains unclear. Here we hypothesized that excitotoxicity in acute lesion development occurs only as a consequence of SD. Using glutamate-sensitive microelectrodes, we found that SD induced by KCl in normal rat cortex elicits increases in extracellular glutamate (11.6±1.3μM) that are synchronous with the onset, sustainment, and resolution of the extracellular direct-current shift of SD. Inhibition of glutamate uptake with d,l-threo-β-benzyloxyaspartate (TBOA, 0.5 and 1mM) significantly prolonged the duration of the direct-current shift (148% and 426%, respectively) and the glutamate increase (167% and 374%, respectively) in a dose-dependent manner (P<0.05). These prolonged events produced significant cortical lesions as indicated by Fluoro-Jade staining (P<0.05), while no lesions were observed after SD in control conditions or after cortical injection of 1mM glutamate (extracellular increase: 243±50.8μM) or 0.5mM TBOA (glutamate increase: 8.5±1.6μM) without SD. We then used an embolic focal ischemia model to determine whether glutamate elevations occur independent of SD in the natural evolution of a cortical lesion. In both the ischemic core and penumbra, glutamate increased only in synchrony with anoxic terminal SD (6.1±1.1μM) and transient SDs (11.8±2.4μM), and not otherwise. Delayed terminal SDs were also observed in two animals at 98 and 150min after ischemic onset and induced similar glutamate elevations. Durations of SDs and glutamate increases were significantly correlated in both normal and ischemic animals (P<0.05). These data suggest that pathologically prolonged SDs are a required mechanism of acute cortical lesion development and that glutamate elevations and the mass electrochemical changes of SD and are merely different facets of the same pathophysiologic process.
Collapse
Affiliation(s)
- Jason M Hinzman
- Department of Neurosurgery, University of Cincinnati (UC) College of Medicine and Neurotrauma Center at UC Neuroscience Institute, Cincinnati, OH, USA.
| | - Vince A DiNapoli
- Department of Neurosurgery, University of Cincinnati (UC) College of Medicine and Neurotrauma Center at UC Neuroscience Institute, Cincinnati, OH, USA; Mayfield Clinic, Cincinnati, OH, USA
| | - Eric J Mahoney
- Department of Neurosurgery, University of Cincinnati (UC) College of Medicine and Neurotrauma Center at UC Neuroscience Institute, Cincinnati, OH, USA
| | - Greg A Gerhardt
- Department of Anatomy and Neurobiology, University of Kentucky Chandler Medical Center, Morris K. Udall Parkinson's Disease Research Center of Excellence, Center for Microelectrode Technology, Spinal Cord and Brain Injury Research Center, Lexington, KY, USA
| | - Jed A Hartings
- Department of Neurosurgery, University of Cincinnati (UC) College of Medicine and Neurotrauma Center at UC Neuroscience Institute, Cincinnati, OH, USA; Mayfield Clinic, Cincinnati, OH, USA
| |
Collapse
|
122
|
Yang KY, Mun JH, Park KD, Kim MJ, Ju JS, Kim ST, Bae YC, Ahn DK. Blockade of spinal glutamate recycling produces paradoxical antinociception in rats with orofacial inflammatory pain. Prog Neuropsychopharmacol Biol Psychiatry 2015; 57:100-9. [PMID: 25445477 DOI: 10.1016/j.pnpbp.2014.10.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2014] [Revised: 10/06/2014] [Accepted: 10/20/2014] [Indexed: 01/06/2023]
Abstract
In our current study, we investigated the role of spinal glutamate recycling in the development of orofacial inflammatory pain. DL-threo-β-benzyloxyaspartate (TBOA) or methionine sulfoximine (MSO) was administered intracisternally to block spinal glutamate transporter and glutamine synthetase activity in astroglia. Intracisternal administration of high dose TBOA (10 μg) produced thermal hyperalgesia in naïve rats but significantly attenuated the thermal hyperalgesia in rats that had been pretreated with interleukin (IL)-1β or Complete Freund's Adjuvant (CFA). In contrast, intracisternal injection of MSO produced anti-hyperalgesic effects against thermal stimuli in CFA-treated rats only. To confirm the paradoxical antinociceptive effects of TBOA and MSO, we examined changes in c-Fos expression in the medullary dorsal horn produced by thermal stimulation in naïve, IL-1β-, or CFA-treated rats, after intracisternal injections of TBOA and MSO. Intracisternal administration of TBOA significantly increased c-Fos immunoreactivity in naïve rats. In contrast, intracisternal administration of TBOA significantly decreased the up-regulation of c-Fos immunoreactivity in the medullary dorsal horn of IL-1β- and CFA-treated rats. However, intracisternal injection of MSO blocked the up-regulation of c-Fos immunoreactivity in CFA-treated rats only. We also investigated the effects of botulinum toxin type A (BoNT-A) on TBOA-induced paradoxical antinociception in CFA-treated rats, as BoNT-A inhibits the release of neurotransmitters, including glutamate. BoNT-A treatment reversed behavioral responses produced by intracisternal administration of TBOA in CFA-treated rats. These results suggest that the paradoxical responses produced by blocking glutamate transporters under inflammatory pain conditions are mediated by the modulation of glutamate release from presynaptic terminals. Moreover, blockade of glutamate reuptake could represent a new therapeutic target for the treatment of chronic inflammatory pain conditions.
Collapse
Affiliation(s)
- Kui Y Yang
- Department of Oral Physiology, School of Dentistry, Kyungpook National University, Daegu, South Korea.
| | - Jun H Mun
- Department of Oral Physiology, School of Dentistry, Kyungpook National University, Daegu, South Korea.
| | - Ki D Park
- Department of Oral Physiology, School of Dentistry, Kyungpook National University, Daegu, South Korea.
| | - Min J Kim
- Department of Oral Physiology, School of Dentistry, Kyungpook National University, Daegu, South Korea.
| | - Jin S Ju
- Department of Oral Physiology, School of Dentistry, Kyungpook National University, Daegu, South Korea.
| | - Seong T Kim
- Department of Orofacial Pain and Oral Medicine, School of Dentistry, Yonsei University, Seoul, South Korea.
| | - Yong C Bae
- Oral Anatomy, School of Dentistry, Kyungpook National University, Daegu, South Korea.
| | - Dong K Ahn
- Department of Oral Physiology, School of Dentistry, Kyungpook National University, Daegu, South Korea.
| |
Collapse
|
123
|
Structural insights into the substrate stereospecificity of D-threo-3-hydroxyaspartate dehydratase from Delftia sp. HT23: a useful enzyme for the synthesis of optically pure L-threo- and D-erythro-3-hydroxyaspartate. Appl Microbiol Biotechnol 2015; 99:7137-50. [PMID: 25715785 DOI: 10.1007/s00253-015-6479-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 01/28/2015] [Accepted: 02/12/2015] [Indexed: 12/22/2022]
Abstract
D-threo-3-Hydroxyaspartate dehydratase (D-THA DH) is a fold-type III pyridoxal 5'-phosphate-dependent enzyme, isolated from a soil bacterium of Delftia sp. HT23. It catalyzes the dehydration of D-threo-3-hydroxyaspartate (D-THA) and L-erythro-3-hydroxyaspartate (L-EHA). To elucidate the mechanism of substrate stereospecificity, crystal structures of D-THA DH were determined in complex with various ligands, such as an inhibitor (D-erythro-3-hydroxyaspartate (D-EHA)), a substrate (L-EHA), and the reaction intermediate (2-amino maleic acid). The C (β) -OH of L-EHA occupied a position close to the active-site Mg(2+), clearly indicating a possibility of metal-assisted C (β) -OH elimination from the substrate. In contrast, the C (β) -OH of an inhibitor was bound far from the active-site Mg(2+). This suggests that the substrate specificity of D-THA DH is determined by the orientation of the C (β) -OH at the active site, whose spatial arrangement is compatible with the 3R configuration of 3-hydroxyaspartate. We also report an optically pure synthesis of L-threo-3-hydroxyaspartate (L-THA) and D-EHA, promising intermediates for the synthesis of β-benzyloxyaspartate, by using a purified D-THA DH as a biocatalyst for the resolution of racemic DL-threo-3-hydroxyaspartate (DL-THA) and DL-erythro-3-hydroxyaspartate (DL-EHA). Considering 50 % of the theoretical maximum, efficient yields of L-THA (38.9 %) and D-EHA (48.9 %) as isolated crystals were achieved with >99 % enantiomeric excess (e.e.). The results of nuclear magnetic resonance signals verified the chemical purity of the products. We were directly able to isolate analytically pure compounds by the recrystallization of acidified reaction mixtures (pH 2.0) and thus avoiding the use of environmentally harmful organic solvents for the chromatographic purification.
Collapse
|
124
|
Akyuz N, Georgieva ER, Zhou Z, Stolzenberg S, Cuendet MA, Khelashvili G, Altman RB, Terry DS, Freed JH, Weinstein H, Boudker O, Blanchard SC. Transport domain unlocking sets the uptake rate of an aspartate transporter. Nature 2015; 518:68-73. [PMID: 25652997 PMCID: PMC4351760 DOI: 10.1038/nature14158] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 12/15/2014] [Indexed: 12/12/2022]
Abstract
Glutamate transporters terminate neurotransmission by clearing synaptically released glutamate from the extracellular space, allowing repeated rounds of signaling and preventing glutamate-mediated excitotoxicity. Crystallographic studies on an archaeal homologue, GltPh, showed that distinct transport domains translocate substrates into the cytoplasm by moving across the membrane within a central trimerization scaffold. Here, we report direct observations of these 'elevator-like' transport domain motions in the context of reconstituted proteoliposomes and physiological ion gradients using single-molecule fluorescence resonance energy transfer (smFRET) imaging. We show that GltPh bearing two “humanizing” mutations exhibits markedly increased transport domain dynamics, which parallels an increased rate of substrate transport, thereby establishing a direct temporal relationship between transport domain motions and substrate uptake. Crystallographic and computational investigations reveal that these mutations favor structurally “unlocked” states with increased solvent occupancy at the interface between the transport domain and the trimeric scaffold.
Collapse
Affiliation(s)
- Nurunisa Akyuz
- Department of Physiology and Biophysics, Weill Cornell Medical College, Cornell University, 1300 York Avenue, New York, New York 10065, USA
| | - Elka R Georgieva
- 1] National Biomedical Center for Advanced ESR Technology, Cornell University, Ithaca, New York 14853, USA [2] Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, USA
| | - Zhou Zhou
- Department of Physiology and Biophysics, Weill Cornell Medical College, Cornell University, 1300 York Avenue, New York, New York 10065, USA
| | - Sebastian Stolzenberg
- Department of Physiology and Biophysics, Weill Cornell Medical College, Cornell University, 1300 York Avenue, New York, New York 10065, USA
| | - Michel A Cuendet
- 1] Department of Physiology and Biophysics, Weill Cornell Medical College, Cornell University, 1300 York Avenue, New York, New York 10065, USA [2] Swiss Institute of Bioinformatics, Quartier Sorge - Batiment Genopode, 1015 Lausanne, Switzerland
| | - George Khelashvili
- Department of Physiology and Biophysics, Weill Cornell Medical College, Cornell University, 1300 York Avenue, New York, New York 10065, USA
| | - Roger B Altman
- Department of Physiology and Biophysics, Weill Cornell Medical College, Cornell University, 1300 York Avenue, New York, New York 10065, USA
| | - Daniel S Terry
- Department of Physiology and Biophysics, Weill Cornell Medical College, Cornell University, 1300 York Avenue, New York, New York 10065, USA
| | - Jack H Freed
- 1] National Biomedical Center for Advanced ESR Technology, Cornell University, Ithaca, New York 14853, USA [2] Department of Chemistry and Chemical Biology, Cornell University, Ithaca, New York 14853, USA
| | - Harel Weinstein
- 1] Department of Physiology and Biophysics, Weill Cornell Medical College, Cornell University, 1300 York Avenue, New York, New York 10065, USA [2] HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medical College, Cornell University, 1305 York Avenue, New York, New York 10065, USA
| | - Olga Boudker
- Department of Physiology and Biophysics, Weill Cornell Medical College, Cornell University, 1300 York Avenue, New York, New York 10065, USA
| | - Scott C Blanchard
- 1] Department of Physiology and Biophysics, Weill Cornell Medical College, Cornell University, 1300 York Avenue, New York, New York 10065, USA [2] Tri-Institutional Training Program in Chemical Biology, 445 East 69th Street, New York, New York 10065, USA
| |
Collapse
|
125
|
McElvain LE, Faulstich M, Jeanne JM, Moore JD, du Lac S. Implementation of linear sensory signaling via multiple coordinated mechanisms at central vestibular nerve synapses. Neuron 2015; 85:1132-44. [PMID: 25704949 DOI: 10.1016/j.neuron.2015.01.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2014] [Revised: 11/10/2014] [Accepted: 01/16/2015] [Indexed: 12/29/2022]
Abstract
Signal transfer in neural circuits is dynamically modified by the recent history of neuronal activity. Short-term plasticity endows synapses with nonlinear transmission properties, yet synapses in sensory and motor circuits are capable of signaling linearly over a wide range of presynaptic firing rates. How do such synapses achieve rate-invariant transmission despite history-dependent nonlinearities? Here, ultrastructural, biophysical, and computational analyses demonstrate that concerted molecular, anatomical, and physiological refinements are required for central vestibular nerve synapses to linearly transmit rate-coded sensory signals. Vestibular synapses operate in a physiological regime of steady-state depression imposed by tonic firing. Rate-invariant transmission relies on brief presynaptic action potentials that delimit calcium influx, large pools of rapidly mobilized vesicles, multiple low-probability release sites, robust postsynaptic receptor sensitivity, and efficient transmitter clearance. Broadband linear synaptic filtering of head motion signals is thus achieved by coordinately tuned synaptic machinery that maintains physiological operation within inherent cell biological limitations.
Collapse
Affiliation(s)
- Lauren E McElvain
- Neurosciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA; Salk Institute for Biological Studies, La Jolla, CA 92037, USA; Champalimaud Neuroscience Programme, Champalimaud Centre for the Unknown, Av. Brasília, Doca de Pedrouços, Lisbon 1400-038, Portugal.
| | | | - James M Jeanne
- Neurosciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jeffrey D Moore
- Neurosciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Sascha du Lac
- Neurosciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, USA; Salk Institute for Biological Studies, La Jolla, CA 92037, USA; Howard Hughes Medical Institute, La Jolla, CA 92037, USA; Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
126
|
EAAT2 (GLT-1; slc1a2) glutamate transporters reconstituted in liposomes argues against heteroexchange being substantially faster than net uptake. J Neurosci 2015; 34:13472-85. [PMID: 25274824 DOI: 10.1523/jneurosci.2282-14.2014] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The EAAT2 glutamate transporter, accounts for >90% of hippocampal glutamate uptake. Although EAAT2 is predominantly expressed in astrocytes, ∼10% of EAAT2 molecules are found in axon terminals. Despite the lower level of EAAT2 expression in glutamatergic terminals, when hippocampal slices are incubated with low concentration of d-aspartate (an EAAT2 substrate), axon terminals accumulate d-aspartate as quickly as astroglia. This implies an unexplained mismatch between the distribution of EAAT2 protein and of EAAT2-mediated transport activity. One hypothesis is that (1) heteroexchange of internal substrate with external substrate is considerably faster than net uptake and (2) terminals favor heteroexchange because of high levels of internal glutamate. However, it is currently unknown whether heteroexchange and uptake have similar or different rates. To address this issue, we used a reconstituted system to compare the relative rates of the two processes in rat and mice. Net uptake was sensitive to changes in the membrane potential and was stimulated by external permeable anions in agreement with the existence of an uncoupled anion conductance. By using the latter, we also demonstrate that the rate of heteroexchange also depends on the membrane potential. Additionally, our data further suggest the presence of a sodium leak in EAAT2. By incorporating the new findings in our previous model of glutamate uptake by EAAT2, we predict that the voltage sensitivity of exchange is caused by the voltage-dependent third Na(+) binding. Further, both our experiments and simulations suggest that the relative rates of net uptake and heteroexchange are comparable in EAAT2.
Collapse
|
127
|
Milanese M, Bonifacino T, Fedele E, Rebosio C, Cattaneo L, Benfenati F, Usai C, Bonanno G. Exocytosis regulates trafficking of GABA and glycine heterotransporters in spinal cord glutamatergic synapses: a mechanism for the excessive heterotransporter-induced release of glutamate in experimental amyotrophic lateral sclerosis. Neurobiol Dis 2014; 74:314-24. [PMID: 25497732 DOI: 10.1016/j.nbd.2014.12.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 11/25/2014] [Accepted: 12/02/2014] [Indexed: 11/29/2022] Open
Abstract
The impact of synaptic vesicle endo-exocytosis on the trafficking of nerve terminal heterotransporters was studied by monitoring membrane expression and function of the GABA transporter-1 (GAT-1) and of type-1/2 glycine (Gly) transporters (GlyT-1/2) at spinal cord glutamatergic synaptic boutons. Experiments were performed by inducing exocytosis in wild-type (WT) mice, in amphiphysin-I knockout (Amph-I KO) mice, which show impaired endocytosis, or in mice expressing high copy number of mutant human SOD1 with a Gly93Ala substitution (SOD1(G93A)), a model of human amyotrophic lateral sclerosis showing constitutively excessive Glu exocytosis. Exposure of spinal cord synaptosomes from WT mice to a 35mM KCl pulse increased the expression of GAT-1 at glutamatergic synaptosomal membranes and enhanced the GAT-1 heterotransporter-induced [(3)H]d-aspartate ([(3)H]d-Asp) release. Similar results were obtained in the case of GlyT-1/2 heterotransporters. Preventing depolarization-induced exocytosis normalized the excessive GAT-1 and GlyT-1/2 heterotransporter-induced [(3)H]d-Asp release in WT mice. Impaired endocytosis in Amph-I KO mice increased GAT-1 membrane expression and [(3)H]GABA uptake in spinal cord synaptosomes. Also the GAT-1 heterotransporter-evoked release of [(3)H]d-Asp was augmented in Amph-I KO mice. The constitutively excessive Glu exocytosis in SOD1(G93A) mice resulted in augmented GAT-1 expression at glutamatergic synaptosomal membranes and GAT-1 or GlyT-1/2 heterotransporter-mediated [(3)H]d-Asp release. Thus, endo-exocytosis regulates the trafficking of GAT-1 and GlyT-1/2 heterotransporters sited at spinal cord glutamatergic nerve terminals. As a consequence, it can be hypothesized that the excessive GAT-1 and GlyT-1/2 heterotransporter-mediated Glu release, in the spinal cord of SOD1(G93A) mice, is due to the heterotransporter over-expression at the nerve terminal membrane, promoted by the excessive Glu exocytosis.
Collapse
Affiliation(s)
- Marco Milanese
- Department of Pharmacy, Unit of Pharmacology and Toxicology and Center of Excellence for Biomedical Research, University of Genoa, 16148 Genoa, Italy
| | - Tiziana Bonifacino
- Department of Pharmacy, Unit of Pharmacology and Toxicology and Center of Excellence for Biomedical Research, University of Genoa, 16148 Genoa, Italy
| | - Ernesto Fedele
- Department of Pharmacy, Unit of Pharmacology and Toxicology and Center of Excellence for Biomedical Research, University of Genoa, 16148 Genoa, Italy
| | - Claudia Rebosio
- Department of Pharmacy, Unit of Pharmacology and Toxicology and Center of Excellence for Biomedical Research, University of Genoa, 16148 Genoa, Italy
| | - Luca Cattaneo
- Department of Pharmacy, Unit of Pharmacology and Toxicology and Center of Excellence for Biomedical Research, University of Genoa, 16148 Genoa, Italy
| | - Fabio Benfenati
- Department of Neuroscience and Brain Technologies, Fondazione Istituto Italiano di Tecnologia, 16163 Genoa, Italy; Department of Experimental Medicine, University of Genoa, 16132 Genoa, Italy
| | - Cesare Usai
- Institute of Biophysics, National Research Council, 16149 Genoa, Italy
| | - Giambattista Bonanno
- Department of Pharmacy, Unit of Pharmacology and Toxicology and Center of Excellence for Biomedical Research, University of Genoa, 16148 Genoa, Italy.
| |
Collapse
|
128
|
Wioland L, Dupont JL, Doussau F, Gaillard S, Heid F, Isope P, Pauillac S, Popoff MR, Bossu JL, Poulain B. Epsilon toxin from Clostridium perfringens acts on oligodendrocytes without forming pores, and causes demyelination. Cell Microbiol 2014; 17:369-88. [PMID: 25287162 DOI: 10.1111/cmi.12373] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 08/29/2014] [Accepted: 09/29/2014] [Indexed: 12/19/2022]
Abstract
Epsilon toxin (ET) is produced by Clostridium perfringens types B and D and causes severe neurological disorders in animals. ET has been observed binding to white matter, suggesting that it may target oligodendrocytes. In primary cultures containing oligodendrocytes and astrocytes, we found that ET (10(-9) M and 10(-7) M) binds to oligodendrocytes, but not to astrocytes. ET induces an increase in extracellular glutamate, and produces oscillations of intracellular Ca(2+) concentration in oligodendrocytes. These effects occurred without any change in the transmembrane resistance of oligodendrocytes, underlining that ET acts through a pore-independent mechanism. Pharmacological investigations revealed that the Ca(2+) oscillations are caused by the ET-induced rise in extracellular glutamate concentration. Indeed, the blockade of metabotropic glutamate receptors type 1 (mGluR1) prevented ET-induced Ca(2+) signals. Activation of the N-methyl-D-aspartate receptor (NMDA-R) is also involved, but to a lesser extent. Oligodendrocytes are responsible for myelinating neuronal axons. Using organotypic cultures of cerebellar slices, we found that ET induced the demyelination of Purkinje cell axons within 24 h. As this effect was suppressed by antagonizing mGluR1 and NMDA-R, demyelination is therefore caused by the initial ET-induced rise in extracellular glutamate concentration. This study reveals the novel possibility that ET can act on oligodendrocytes, thereby causing demyelination. Moreover, it suggests that for certain cell types such as oligodendrocytes, ET can act without forming pores, namely through the activation of an undefined receptor-mediated pathway.
Collapse
Affiliation(s)
- Laetitia Wioland
- Centre National de la Recherche Scientifique Associé à l'Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives UPR3212, 5 rue Blaise Pascal, Strasbourg, cedex F-67084, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
129
|
Pyruvate kinase and aspartate-glutamate carrier distributions reveal key metabolic links between neurons and glia in retina. Proc Natl Acad Sci U S A 2014; 111:15579-84. [PMID: 25313047 DOI: 10.1073/pnas.1412441111] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Symbiotic relationships between neurons and glia must adapt to structures, functions, and metabolic roles of the tissues they are in. We show here that Müller glia in retinas have specific enzyme deficiencies that can enhance their ability to synthesize Gln. The metabolic cost of these deficiencies is that they impair the Müller cell's ability to metabolize Glc. We show here that the cells can compensate for this deficiency by using metabolites produced by neurons. Müller glia are deficient for pyruvate kinase (PK) and for aspartate/glutamate carrier 1 (AGC1), a key component of the malate-aspartate shuttle. In contrast, photoreceptor neurons express AGC1 and the M2 isoform of pyruvate kinase, which is commonly associated with aerobic glycolysis in tumors, proliferating cells, and some other cell types. Our findings reveal a previously unidentified type of metabolic relationship between neurons and glia. Müller glia compensate for their unique metabolic adaptations by using lactate and aspartate from neurons as surrogates for their missing PK and AGC1.
Collapse
|
130
|
Armbruster M, Hampton D, Yang Y, Dulla CG. Laser-scanning astrocyte mapping reveals increased glutamate-responsive domain size and disrupted maturation of glutamate uptake following neonatal cortical freeze-lesion. Front Cell Neurosci 2014; 8:277. [PMID: 25249939 PMCID: PMC4158796 DOI: 10.3389/fncel.2014.00277] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 08/22/2014] [Indexed: 12/27/2022] Open
Abstract
Astrocytic uptake of glutamate shapes extracellular neurotransmitter dynamics, receptor activation, and synaptogenesis. During development, glutamate transport becomes more robust. How neonatal brain insult affects the functional maturation of glutamate transport remains unanswered. Neonatal brain insult can lead to developmental delays, cognitive losses, and epilepsy; the disruption of glutamate transport is known to cause changes in synaptogenesis, receptor activation, and seizure. Using the neonatal freeze-lesion (FL) model, we have investigated how insult affects the maturation of astrocytic glutamate transport. As lesioning occurs on the day of birth, a time when astrocytes are still functionally immature, this model is ideal for identifying changes in astrocyte maturation following insult. Reactive astrocytosis, astrocyte proliferation, and in vitro hyperexcitability are known to occur in this model. To probe astrocyte glutamate transport with better spatial precision we have developed a novel technique, Laser Scanning Astrocyte Mapping (LSAM), which combines glutamate transport current (TC) recording from astrocytes with laser scanning glutamate photolysis. LSAM allows us to identify the area from which a single astrocyte can transport glutamate and to quantify spatial heterogeneity in the rate of glutamate clearance kinetics within that domain. Using LSAM, we report that cortical astrocytes have an increased glutamate-responsive area following FL and that TCs have faster decay times in distal, as compared to proximal processes. Furthermore, the developmental shift from GLAST- to GLT-1-dominated clearance is disrupted following FL. These findings introduce a novel method to probe astrocyte glutamate uptake and show that neonatal cortical FL disrupts the functional maturation of cortical astrocytes.
Collapse
Affiliation(s)
- Moritz Armbruster
- Department of Neuroscience, Tufts University School of Medicine Boston, MA, USA
| | - David Hampton
- Department of Neuroscience, Tufts University School of Medicine Boston, MA, USA
| | - Yongjie Yang
- Department of Neuroscience, Tufts University School of Medicine Boston, MA, USA
| | - Chris G Dulla
- Department of Neuroscience, Tufts University School of Medicine Boston, MA, USA
| |
Collapse
|
131
|
Martinez-Lozada Z, Waggener CT, Kim K, Zou S, Knapp PE, Hayashi Y, Ortega A, Fuss B. Activation of sodium-dependent glutamate transporters regulates the morphological aspects of oligodendrocyte maturation via signaling through calcium/calmodulin-dependent kinase IIβ's actin-binding/-stabilizing domain. Glia 2014; 62:1543-1558. [PMID: 24866099 PMCID: PMC4107011 DOI: 10.1002/glia.22699] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 05/08/2014] [Accepted: 05/09/2014] [Indexed: 02/06/2023]
Abstract
Signaling via the major excitatory amino acid glutamate has been implicated in the regulation of various aspects of the biology of oligodendrocytes, the myelinating cells of the central nervous system (CNS). In this respect, cells of the oligodendrocyte lineage have been described to express a variety of glutamate-responsive transmembrane proteins including sodium-dependent glutamate transporters. The latter have been well characterized to mediate glutamate clearance from the extracellular space. However, there is increasing evidence that they also mediate glutamate-induced intracellular signaling events. Our data presented here show that the activation of oligodendrocyte expressed sodium-dependent glutamate transporters, in particular GLT-1 and GLAST, promotes the morphological aspects of oligodendrocyte maturation. This effect was found to be associated with a transient increase in intracellular calcium levels and a transient phosphorylation event at the serine (S)(371) site of the calcium sensor calcium/calmodulin-dependent kinase type IIβ (CaMKIIβ). The potential regulatory S(371) site is located within CaMKIIβ's previously defined actin-binding/-stabilizing domain, and phosphorylation events within this domain were identified in our studies as a requirement for sodium-dependent glutamate transporter-mediated promotion of oligodendrocyte maturation. Furthermore, our data provide good evidence for a role of these phosphorylation events in mediating detachment of CaMKIIβ from filamentous (F)-actin, and hence allowing a remodeling of the oligodendrocyte's actin cytoskeleton. Taken together with our recent findings, which demonstrated a crucial role of CaMKIIβ in regulating CNS myelination in vivo, our data strongly suggest that a sodium-dependent glutamate transporter-CaMKIIβ-actin cytoskeleton axis plays an important role in the regulation of oligodendrocyte maturation and CNS myelination.
Collapse
Affiliation(s)
- Zila Martinez-Lozada
- Department of Anatomy and Neurobiology, Virginia Commonwealth University Medical Center, Richmond, Virginia 23298, USA
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, México D.F, México
| | - Christopher T. Waggener
- Department of Anatomy and Neurobiology, Virginia Commonwealth University Medical Center, Richmond, Virginia 23298, USA
| | - Karam Kim
- Brain Science Institute, RIKEN, Wako, Saitama 351-0198, Japan
| | - Shiping Zou
- Department of Anatomy and Neurobiology, Virginia Commonwealth University Medical Center, Richmond, Virginia 23298, USA
| | - Pamela E. Knapp
- Department of Anatomy and Neurobiology, Virginia Commonwealth University Medical Center, Richmond, Virginia 23298, USA
| | - Yasunori Hayashi
- Brain Science Institute, RIKEN, Wako, Saitama 351-0198, Japan
- Saitama University Brain Science Institute, Saitama University, Saitama 338-8570, Japan
| | - Arturo Ortega
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, México D.F, México
| | - Babette Fuss
- Department of Anatomy and Neurobiology, Virginia Commonwealth University Medical Center, Richmond, Virginia 23298, USA
| |
Collapse
|
132
|
Ohi Y, Kimura S, Haji A. Modulation of glutamatergic transmission by metabotropic glutamate receptor activation in second-order neurons of the guinea pig nucleus tractus solitarius. Brain Res 2014; 1581:12-22. [DOI: 10.1016/j.brainres.2014.04.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Revised: 04/10/2014] [Accepted: 04/23/2014] [Indexed: 11/30/2022]
|
133
|
Specialized postsynaptic morphology enhances neurotransmitter dilution and high-frequency signaling at an auditory synapse. J Neurosci 2014; 34:8358-72. [PMID: 24920639 DOI: 10.1523/jneurosci.4493-13.2014] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Sensory processing in the auditory system requires that synapses, neurons, and circuits encode information with particularly high temporal and spectral precision. In the amphibian papillia, sound frequencies up to 1 kHz are encoded along a tonotopic array of hair cells and transmitted to afferent fibers via fast, repetitive synaptic transmission, thereby promoting phase locking between the presynaptic and postsynaptic cells. Here, we have combined serial section electron microscopy, paired electrophysiological recordings, and Monte Carlo diffusion simulations to examine novel mechanisms that facilitate fast synaptic transmission in the inner ear of frogs (Rana catesbeiana and Rana pipiens). Three-dimensional anatomical reconstructions reveal specialized spine-like contacts between individual afferent fibers and hair cells that are surrounded by large, open regions of extracellular space. Morphologically realistic diffusion simulations suggest that these local enlargements in extracellular space speed transmitter clearance and reduce spillover between neighboring synapses, thereby minimizing postsynaptic receptor desensitization and improving sensitivity during prolonged signal transmission. Additionally, evoked EPSCs in afferent fibers are unaffected by glutamate transporter blockade, suggesting that transmitter diffusion and dilution, and not uptake, play a primary role in speeding neurotransmission and ensuring fidelity at these synapses.
Collapse
|
134
|
Turecek J, Yuen GS, Han VZ, Zeng XH, Bayer KU, Welsh JP. NMDA receptor activation strengthens weak electrical coupling in mammalian brain. Neuron 2014; 81:1375-1388. [PMID: 24656255 DOI: 10.1016/j.neuron.2014.01.024] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/23/2013] [Indexed: 11/17/2022]
Abstract
Electrical synapses are formed by gap junctions and permit electrical coupling, which shapes the synchrony of neuronal ensembles. Here, we provide a direct demonstration of receptor-mediated strengthening of electrical coupling in mammalian brain. Electrical coupling in the inferior olive of rats was strengthened by activation of NMDA-type glutamate receptors (NMDARs), which were found at synaptic loci and at extrasynaptic loci 20-100 nm proximal to gap junctions. Electrical coupling was strengthened by pharmacological and synaptic activation of NMDARs, whereas costimulation of ionotropic non-NMDAR glutamate receptors transiently antagonized the effect of NMDAR activation. NMDAR-dependent strengthening (1) occurred despite increased input conductance, (2) induced Ca(2+)-influx microdomains near dendritic spines, (3) required activation of the Ca(2+)/calmodulin-dependent protein-kinase II, (4) was restricted to neurons that were weakly coupled, and (5) thus strengthened coupling, mainly between nonadjacent neurons. This provided a mechanism to expand the synchronization of rhythmic membrane potential oscillations by chemical neurotransmitter input.
Collapse
Affiliation(s)
- Josef Turecek
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Genevieve S Yuen
- Department of Psychiatry, New York Presbyterian Hospital-Weill Cornell Medical College, 525 East 68(th) Street, New York, NY 10065, USA
| | - Victor Z Han
- Center for Integrative Brain Research, Seattle Children's Research Institute, 1900 9(th) Avenue, Seattle, WA 98155, USA
| | - Xiao-Hui Zeng
- Center for Integrative Brain Research, Seattle Children's Research Institute, 1900 9(th) Avenue, Seattle, WA 98155, USA
| | - K Ulrich Bayer
- Department of Pharmacology, University of Colorado-Denver School of Medicine, 12800 E. 19(th) Avenue, Aurora, CO 80045, USA
| | - John P Welsh
- Center for Integrative Brain Research, Seattle Children's Research Institute, 1900 9(th) Avenue, Seattle, WA 98155, USA; Department of Pediatrics, University of Washington, 1959 N.E. Pacific Street, Seattle, WA 98195, USA.
| |
Collapse
|
135
|
Scopelliti AJ, Heinzelmann G, Kuyucak S, Ryan RM, Vandenberg RJ. Na+ interactions with the neutral amino acid transporter ASCT1. J Biol Chem 2014; 289:17468-79. [PMID: 24808181 DOI: 10.1074/jbc.m114.565242] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The alanine, serine, cysteine transporters (ASCTs) belong to the solute carrier family 1A (SLC1A), which also includes the excitatory amino acid transporters (EAATs) and the prokaryotic aspartate transporter GltPh. Acidic amino acid transport by the EAATs is coupled to the co-transport of three Na(+) ions and one proton, and the counter-transport of one K(+) ion. In contrast, neutral amino acid exchange by the ASCTs does not require protons or the counter-transport of K(+) ions and the number of Na(+) ions required is not well established. One property common to SLC1A family members is a substrate-activated anion conductance. We have investigated the number and location of Na(+) ions required by ASCT1 by mutating residues in ASCT1 that correspond to residues in the EAATs and GltPh that are involved in Na(+) binding. Mutations to all three proposed Na(+) sites influence the binding of substrate and/or Na(+), or the rate of substrate exchange. A G422S mutation near the Na2 site reduced Na(+) affinity, without affecting the rate of exchange. D467T and D467A mutations in the Na1 site reduce Na(+) and substrate affinity and also the rate of substrate exchange. T124A and D380A mutations in the Na3 site selectively reduce the affinity for Na(+) and the rate of substrate exchange without affecting substrate affinity. In many of the mutants that reduce the rate of substrate transport the amplitudes of the substrate-activated anion conductances are not substantially affected indicating altered ion dependence for channel activation compared with substrate exchange.
Collapse
Affiliation(s)
- Amanda J Scopelliti
- From the Discipline of Pharmacology, School of Medical Sciences, Bosch Institute, University of Sydney, Sydney, New South Wales 2006 and
| | - Germano Heinzelmann
- the School of Physics, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Serdar Kuyucak
- the School of Physics, The University of Sydney, Sydney, New South Wales 2006, Australia
| | - Renae M Ryan
- From the Discipline of Pharmacology, School of Medical Sciences, Bosch Institute, University of Sydney, Sydney, New South Wales 2006 and
| | - Robert J Vandenberg
- From the Discipline of Pharmacology, School of Medical Sciences, Bosch Institute, University of Sydney, Sydney, New South Wales 2006 and
| |
Collapse
|
136
|
Transient inactivation of the ventral hippocampus in neonatal rats impairs the mesolimbic regulation of prefrontal glutamate release in adulthood. Neuropharmacology 2014; 84:19-30. [PMID: 24747179 DOI: 10.1016/j.neuropharm.2014.04.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 03/24/2014] [Accepted: 04/08/2014] [Indexed: 11/21/2022]
Abstract
Cognitive deficits in schizophrenia (SZ) reflect maturational disruptions within a neural system that includes the ventral hippocampus (VH), nucleus accumbens (NAc), basal forebrain, and prefrontal cortex (PFC). A better understanding of these changes may reveal drug targets for more efficacious cognition enhancers. We have utilized an animal model in which the above distributed system is altered, during a sensitive period of development, by transiently inactivating the VH and its efferent projections. We determined the ability of NAc shell activation to evoke prefrontal glutamate release in adult male Wistar rats that had received saline (Sal) or tetrodotoxin (TTX) as neonates (PD7) or as adolescents (PD32). The nucleus accumbens shell (NAcSh) was activated by NMDA infusions (0.05-0.30 μg/0.5 μL). Basal and evoked glutamate levels were measured amperometrically using a glutamate-sensitive microelectrode. There were no differences in basal glutamate levels among the groups tested (overall 1.41 ± 0.26 uM). However, the dose-related stimulation of prefrontal glutamate levels seen in control rats treated with saline on PD7 (4.31 ± 0.22 μM after 0.15 μg) was markedly attenuated in rats treated with TTX on PD7 (0.45 ± 0.12 μM after 0.15 μg). This effect was age-dependent as infusions of TTX on PD32 did not alter the NMDA-induced increases in glutamate release (4.10 ± 0.37 μM after 0.15 μg). Collectively, these findings reveal that transient inactivation of VH transmission, during a sensitive period of development, leads to a functional mesolimbic-cortical disconnection that produces neurochemical and ultimately cognitive impairments resembling those seen in SZ.
Collapse
|
137
|
Fernández-López B, Valle-Maroto SM, Barreiro-Iglesias A, Rodicio MC. Neuronal release and successful astrocyte uptake of aminoacidergic neurotransmitters after spinal cord injury in lampreys. Glia 2014; 62:1254-69. [PMID: 24733772 DOI: 10.1002/glia.22678] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 03/13/2014] [Accepted: 04/03/2014] [Indexed: 12/27/2022]
Abstract
In contrast to mammals, the spinal cord of lampreys spontaneously recovers from a complete spinal cord injury (SCI). Understanding the differences between lampreys and mammals in their response to SCI could provide valuable information to propose new therapies. Unique properties of the astrocytes of lampreys probably contribute to the success of spinal cord regeneration. The main aim of our study was to investigate, in the sea lamprey, the release of aminoacidergic neurotransmitters and the subsequent astrocyte uptake of these neurotransmitters during the first week following a complete SCI by detecting glutamate, GABA, glycine, Hu and cytokeratin immunoreactivities. This is the first time that aminoacidergic neurotransmitter release from neurons and the subsequent astrocytic response after SCI are analysed by immunocytochemistry in any vertebrate. Spinal injury caused the immediate loss of glutamate, GABA and glycine immunoreactivities in neurons close to the lesion site (except for the cerebrospinal fluid-contacting GABA cells). Only after SCI, astrocytes showed glutamate, GABA and glycine immunoreactivity. Treatment with an inhibitor of glutamate transporters (DL-TBOA) showed that neuronal glutamate was actively transported into astrocytes after SCI. Moreover, after SCI, a massive accumulation of inhibitory neurotransmitters around some reticulospinal axons was observed. Presence of GABA accumulation significantly correlated with a higher survival ability of these neurons. Our data show that, in contrast to mammals, astrocytes of lampreys have a high capacity to actively uptake glutamate after SCI. GABA may play a protective role that could explain the higher regenerative and survival ability of specific descending neurons of lampreys.
Collapse
Affiliation(s)
- Blanca Fernández-López
- Department of Cell Biology and Ecology, CIBUS, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | | | | | | |
Collapse
|
138
|
Wasfy MM, Matsui JI, Miller J, Dowling JE, Perkins BD. myosin 7aa(-/-) mutant zebrafish show mild photoreceptor degeneration and reduced electroretinographic responses. Exp Eye Res 2014; 122:65-76. [PMID: 24698764 DOI: 10.1016/j.exer.2014.03.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 03/18/2014] [Accepted: 03/19/2014] [Indexed: 01/20/2023]
Abstract
Mutations in myosin VIIa (MYO7A) cause Usher Syndrome 1B (USH1B), a disease characterized by the combination of sensorineural hearing loss and visual impairment termed retinitis pigmentosa (RP). Although the shaker-1 mouse model of USH1B exists, only minor defects in the retina have been observed during its lifespan. Previous studies of the zebrafish mariner mutant, which also carries a mutation in myo7aa, revealed balance and hearing defects in the mutants but the retinal phenotype has not been described. We found elevated cell death in the outer nuclear layer (ONL) of myo7aa(-/-) mutants. While myo7aa(-/-) mutants retained visual behaviors in the optokinetic reflex (OKR) assay, electroretinogram (ERG) recordings revealed a significant decrease in both a- and b-wave amplitudes in mutant animals, but not a change in ERG threshold sensitivity. Immunohistochemistry showed mislocalization of rod and blue cone opsins and reduced expression of rod-specific markers in the myo7aa(-/-) ONL, providing further evidence that the photoreceptor degeneration observed represents the initial stages of the RP. Further, constant light exposure resulted in widespread photoreceptor degeneration and the appearance of large holes in the retinal pigment epithelium (RPE). No differences were observed in the retinomotor movements of the photoreceptors or in melanosome migration within the RPE, suggesting that myo7aa(-/-) does not function in these processes in teleosts. These results indicate that the zebrafish myo7aa(-/-) mutant is a useful animal model for the RP seen in humans with USH1B.
Collapse
Affiliation(s)
- Meagan M Wasfy
- Department of Molecular and Cellular Biology, Harvard University, 16 Divinity Avenue, Cambridge, MA 02138, USA
| | - Jonathan I Matsui
- Department of Molecular and Cellular Biology, Harvard University, 16 Divinity Avenue, Cambridge, MA 02138, USA
| | - Jessica Miller
- Department of Molecular and Cellular Biology, Harvard University, 16 Divinity Avenue, Cambridge, MA 02138, USA
| | - John E Dowling
- Department of Molecular and Cellular Biology, Harvard University, 16 Divinity Avenue, Cambridge, MA 02138, USA
| | - Brian D Perkins
- Department of Molecular and Cellular Biology, Harvard University, 16 Divinity Avenue, Cambridge, MA 02138, USA.
| |
Collapse
|
139
|
Smith KL, John CS, Sypek EI, Ongür D, Cohen BM, Barry SM, Bechtholt AJ. Exploring the role of central astrocytic glutamate uptake in ethanol reward in mice. Alcohol Clin Exp Res 2014; 38:1307-14. [PMID: 24655029 DOI: 10.1111/acer.12361] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 12/18/2013] [Indexed: 01/16/2023]
Abstract
BACKGROUND Alcoholism is associated with specific brain abnormalities revealed through postmortem studies, including a reduction in glial cell number and dysregulated glutamatergic neurotransmission. Whether these abnormalities contribute to the etiology of alcoholism, are consequences of alcohol use, or both is still unknown. METHODS We investigated the role of astrocytic glutamate uptake in ethanol (EtOH) binge drinking in mice, using the "drinking in the dark" (DID) paradigm by blocking the astrocytic glutamate transporter (GLT-1) with intracerebroventricular (ICV) administration of dihydrokainic acid (DHK). To determine whether astrocytic glutamate uptake regulates the conditioned rewarding effects of EtOH, we examined the effects of ICV DHK on the acquisition and expression of EtOH-induced conditioned place preference. RESULTS Blocking central astrocytic glutamate uptake selectively attenuated EtOH binge drinking behavior in mice. DHK did not alter the acquisition or expression of preference for EtOH-associated cues, indicating that reduced astrocytic glutamate trafficking may decrease binge-like drinking without altering the conditioned rewarding effects of EtOH. CONCLUSIONS Several alternative conclusions are plausible, however, interpreting these data in the context of the human literature, these findings suggest that the reduction of glia in the alcoholic brain may not be a predisposing factor to developing alcoholism and could be a consequence of EtOH toxicity that decreases excessive EtOH intake.
Collapse
Affiliation(s)
- Karen L Smith
- Department of Psychiatry, Harvard Medical School, McLean Hospital, Belmont, Massachusetts; Laboratory of Addictive Disorders, Department of Pharmacology and Experimental Therapeutics, Boston University School of Medicine, Boston, Massachusetts
| | | | | | | | | | | | | |
Collapse
|
140
|
Pinto A, Tamborini L, Mastronardi F, Ettari R, Safoz Y, Bunch L, Nielsen B, Jensen AA, De Micheli C, Conti P. Synthesis of (3-hydroxy-pyrazolin-5-yl)glycine based ligands interacting with ionotropic glutamate receptors. Eur J Med Chem 2014; 75:151-8. [PMID: 24531228 DOI: 10.1016/j.ejmech.2014.01.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Revised: 11/27/2013] [Accepted: 01/20/2014] [Indexed: 11/18/2022]
Abstract
Following the concept that increasing the molecular complexity may enhance the receptor selectivity, we replaced the 3-hydroxy-isoxazoline ring of model compound tricholomic acid with a 3-hydroxy-pyrazoline ring, which could be variously decorated at the N1 position, inserting groups characterized by different electronic and steric properties. Binding assays on rat brain synaptic membranes showed that, depending on the nature of the substituent, some of the new synthesized ligands interacted with either AMPA or KA receptors, with affinities in the mid-micromolar range.
Collapse
Affiliation(s)
- Andrea Pinto
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Mangiagalli 25, 20133 Milano, Italy.
| | - Lucia Tamborini
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Mangiagalli 25, 20133 Milano, Italy
| | - Federica Mastronardi
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Mangiagalli 25, 20133 Milano, Italy
| | - Roberta Ettari
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Mangiagalli 25, 20133 Milano, Italy
| | - Yeliz Safoz
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Mangiagalli 25, 20133 Milano, Italy; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen OE, Denmark
| | - Lennart Bunch
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen OE, Denmark
| | - Birgitte Nielsen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen OE, Denmark
| | - Anders A Jensen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen OE, Denmark
| | - Carlo De Micheli
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Mangiagalli 25, 20133 Milano, Italy
| | - Paola Conti
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, Via Mangiagalli 25, 20133 Milano, Italy
| |
Collapse
|
141
|
Abstract
Retinal ganglion cells (RGCs) receive glutamatergic input from bipolar cells through NMDA- and AMPA-type glutamate receptors. Both GluA2-containing, Ca(2+)-impermeable AMPA receptors (CI-AMPARs) and GluA2-lacking, Ca(2+)-permeable AMPA receptors (CP-AMPARs) contribute to light-evoked responses in ON RGCs; however, specific roles for each subtype are not well understood. Here, we present evidence that light intensity determines the subtype of AMPAR that is activated during the synaptic response in ON RGCs. Using current voltage analysis of the EPSC we show that light intensities near RGC threshold, intensities that travel through the well described primary rod pathway, evoke synaptic currents that are preferentially mediated by CP-AMPARs. Synaptic responses evoked by spontaneous release of transmitter from bipolar cell terminals also preferentially activate CP-AMPARs. Conversely, higher light intensities, most likely carried by secondary rod pathways, activate CI-AMPARs. The same pattern of CP-AMPAR and CI-AMPAR activation was observed in mice containing only functional rods, suggesting that the recruitment of CI-AMPARs at higher light intensity does not require cone stimulation. When glutamate spillover was induced by blocking transporters with TBOA, both the near threshold and spontaneous EPSCs contained a significant CI-AMPAR component. We propose that CI-AMPARs are activated by "spillover" of synaptic glutamate only during bright illumination, or when glutamate uptake is blocked. Glutamate may spill over to more distant sites at the same synapse, or perhaps as far as neighboring synapses. Together, our data suggest that the spatial organization of AMPARs at ON RGCs synapses allows for selective, intensity-dependent activation of AMPARs with distinct subunit composition.
Collapse
|
142
|
Increased extracellular glutamate in the nucleus accumbens promotes excessive ethanol drinking in ethanol dependent mice. Neuropsychopharmacology 2014; 39:707-17. [PMID: 24067300 PMCID: PMC3895249 DOI: 10.1038/npp.2013.256] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 08/02/2013] [Accepted: 08/12/2013] [Indexed: 12/20/2022]
Abstract
Using a well-established model of ethanol dependence and relapse, this study examined adaptations in glutamatergic transmission in the nucleus accumbens (NAc) and their role in regulating voluntary ethanol drinking. Mice were first trained to drink ethanol in a free-choice, limited access (2 h/day) paradigm. One group (EtOH mice) received repeated weekly cycles of chronic intermittent ethanol (CIE) exposure with intervening weeks of test drinking sessions, whereas the remaining mice (CTL mice) were similarly treated but did not receive CIE treatment. Over repeated cycles of CIE exposure, EtOH mice exhibited significant escalation in drinking (up to ∼3.5 g/kg), whereas drinking remained relatively stable at baseline levels (2-2.5 g/kg) in CTL mice. Using in vivo microdialysis procedures, extracellular glutamate (GLUEX) levels in the NAc were increased approximately twofold in EtOH mice compared with CTL mice, and this difference was observed 7 days after final CIE exposure, indicating that this hyperglutamatergic state persisted beyond acute withdrawal. This finding prompted additional studies examining the effects of pharmacologically manipulating GLUEX in the NAc on ethanol drinking in the CIE model. The non-selective glutamate reuptake antagonist, threo-β-benzyloxyaspartate (TBOA), was bilaterally microinjected into the NAc and found to dose-dependently increase drinking in nondependent (CTL) mice to levels attained by dependent (EtOH) mice. TBOA also further increased drinking in EtOH mice. In contrast, reducing glutamatergic transmission in the NAc via bilateral injections of the metabotropic glutamate receptor-2/3 agonist LY379268 reduced drinking in dependent (EtOH) mice to nondependent (CTL) levels, whereas having a more modest effect in decreasing ethanol consumption in CTL mice. Taken together, these data support an important role of glutamatergic transmission in the NAc in regulating ethanol drinking. Additionally, these results indicate that ethanol dependence produces adaptations that favor elevated glutamate activity in the NAc which, in turn, promote excessive levels of ethanol consumption associated with dependence.
Collapse
|
143
|
Medrano MC, Gerrikagoitia I, Martínez-Millán L, Mendiguren A, Pineda J. Functional and morphological characterization of glutamate transporters in the rat locus coeruleus. Br J Pharmacol 2014; 169:1781-94. [PMID: 23638698 DOI: 10.1111/bph.12235] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 04/19/2013] [Accepted: 04/29/2013] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND AND PURPOSE Excitatory amino acid transporters (EAATs) in the CNS contribute to the clearance of glutamate released during neurotransmission. The aim of this study was to explore the role of EAATs in the regulation of locus coeruleus (LC) neurons by glutamate. EXPERIMENTAL APPROACH We measured the effect of different EAAT subtype inhibitors/enhancers on glutamate- and KCl-induced activation of LC neurons in rat slices. EAAT2-3 expression in the LC was also characterized by immunohistochemistry. KEY RESULTS The EAAT2-5 inhibitor DL-threo-β-benzyloxaspartic acid (100 μM), but not the EAAT2, 4, 5 inhibitor L-trans-pyrrolidine-2,4-dicarboxylic acid (100 μM) or the EAAT2 inhibitor dihydrokainic acid (DHK; 100 μM), enhanced the glutamate- and KCl-induced activation of the firing rate of LC neurons. These effects were blocked by ionotropic, but not metabotrobic, glutamate receptor antagonists. DHK (100 μM) was the only EAAT inhibitor that increased the spontaneous firing rate of LC cells, an effect that was due to inhibition of EAAT2 and subsequent AMPA receptor activation. Chronic treatment with ceftriaxone (200 mg·kg(-1) i.p., once daily, 7 days), an EAAT2 expression enhancer, increased the actions of glutamate and DHK, suggesting a functional impact of EAAT2 up-regulation on the glutamatergic system. Immuhistochemical data revealed the presence of EAAT2 and EAAT3 surrounding noradrenergic neurons and EAAT2 on glial cells in the LC. CONCLUSIONS AND IMPLICATIONS These results remark the importance of EAAT2 and EAAT3 in the regulation of rat LC by glutamate. Neuronal EAAT3 would be responsible for terminating the action of synaptically released glutamate, whereas glial EAAT2 would regulate tonic glutamate concentrations in this nucleus.
Collapse
Affiliation(s)
- M C Medrano
- Department of Pharmacology, Faculty of Medicine and Odontology, University of the Basque Country (UPV/ EHU), Bizkaia, Spain
| | | | | | | | | |
Collapse
|
144
|
Abstract
L-Glutamate is the predominant excitatory neurotransmitter in the mammalian central nervous system and plays important roles in a wide variety of brain functions, but it is also a key player in the pathogenesis of many neurological disorders. The control of glutamate concentrations is critical to the normal functioning of the central nervous system, and in this review we discuss how glutamate transporters regulate glutamate concentrations to maintain dynamic signaling mechanisms between neurons. In 2004, the crystal structure of a prokaryotic homolog of the mammalian glutamate transporter family of proteins was crystallized and its structure determined. This has paved the way for a better understanding of the structural basis for glutamate transporter function. In this review we provide a broad perspective of this field of research, but focus primarily on the more recent studies with a particular emphasis on how our understanding of the structure of glutamate transporters has generated new insights.
Collapse
|
145
|
Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, Peters JA, Harmar AJ. The Concise Guide to PHARMACOLOGY 2013/14: transporters. Br J Pharmacol 2013; 170:1706-96. [PMID: 24528242 PMCID: PMC3892292 DOI: 10.1111/bph.12450] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The Concise Guide to PHARMACOLOGY 2013/14 provides concise overviews of the key properties of over 2000 human drug targets with their pharmacology, plus links to an open access knowledgebase of drug targets and their ligands (www.guidetopharmacology.org), which provides more detailed views of target and ligand properties. The full contents can be found at http://onlinelibrary.wiley.com/doi/10.1111/bph.12444/full. Transporters are one of the seven major pharmacological targets into which the Guide is divided, with the others being G protein-coupled receptors, ligand-gated ion channels, ion channels, catalytic receptors, nuclear hormone receptors and enzymes. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading. A new landscape format has easy to use tables comparing related targets. It is a condensed version of material contemporary to late 2013, which is presented in greater detail and constantly updated on the website www.guidetopharmacology.org, superseding data presented in previous Guides to Receptors and Channels. It is produced in conjunction with NC-IUPHAR and provides the official IUPHAR classification and nomenclature for human drug targets, where appropriate. It consolidates information previously curated and displayed separately in IUPHAR-DB and the Guide to Receptors and Channels, providing a permanent, citable, point-in-time record that will survive database updates.
Collapse
Affiliation(s)
- Stephen PH Alexander
- School of Life Sciences, University of Nottingham Medical SchoolNottingham, NG7 2UH, UK
| | - Helen E Benson
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Elena Faccenda
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Adam J Pawson
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | - Joanna L Sharman
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| | | | - John A Peters
- Neuroscience Division, Medical Education Institute, Ninewells Hospital and Medical School, University of DundeeDundee, DD1 9SY, UK
| | - Anthony J Harmar
- The University/BHF Centre for Cardiovascular Science, University of EdinburghEdinburgh, EH16 4TJ, UK
| |
Collapse
|
146
|
Grewer C, Gameiro A, Rauen T. SLC1 glutamate transporters. Pflugers Arch 2013; 466:3-24. [PMID: 24240778 DOI: 10.1007/s00424-013-1397-7] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Revised: 10/21/2013] [Accepted: 10/22/2013] [Indexed: 12/13/2022]
Abstract
The plasma membrane transporters for the neurotransmitter glutamate belong to the solute carrier 1 family. They are secondary active transporters, taking up glutamate into the cell against a substantial concentration gradient. The driving force for concentrative uptake is provided by the cotransport of Na(+) ions and the countertransport of one K(+) in a step independent of the glutamate translocation step. Due to eletrogenicity of transport, the transmembrane potential can also act as a driving force. Glutamate transporters are expressed in many tissues, but are of particular importance in the brain, where they contribute to the termination of excitatory neurotransmission. Glutamate transporters can also run in reverse, resulting in glutamate release from cells. Due to these important physiological functions, glutamate transporter expression and, therefore, the transport rate, are tightly regulated. This review summarizes recent literature on the functional and biophysical properties, structure-function relationships, regulation, physiological significance, and pharmacology of glutamate transporters. Particular emphasis is on the insight from rapid kinetic and electrophysiological studies, transcriptional regulation of transporter expression, and reverse transport and its importance for pathophysiological glutamate release under ischemic conditions.
Collapse
Affiliation(s)
- Christof Grewer
- Department of Chemistry, Binghamton University, PO Box 6000, Binghamton, 13902-6000, NY, USA,
| | | | | |
Collapse
|
147
|
Melzer N, Hicking G, Bittner S, Bobak N, Göbel K, Herrmann AM, Wiendl H, Meuth SG. Excitotoxic neuronal cell death during an oligodendrocyte-directed CD8+ T cell attack in the CNS gray matter. J Neuroinflammation 2013; 10:121. [PMID: 24093512 PMCID: PMC3853237 DOI: 10.1186/1742-2094-10-121] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2013] [Accepted: 09/24/2013] [Indexed: 12/27/2022] Open
Abstract
Background Neural-antigen reactive cytotoxic CD8+ T cells contribute to neuronal dysfunction and degeneration in a variety of inflammatory CNS disorders. Facing excess numbers of target cells, CNS-invading CD8+ T cells cause neuronal cell death either via confined release of cytotoxic effector molecules towards neurons, or via spillover of cytotoxic effector molecules from 'leaky’ immunological synapses and non-confined release by CD8+ T cells themselves during serial and simultaneous killing of oligodendrocytes or astrocytes. Methods Wild-type and T cell receptor transgenic CD8+ T cells were stimulated in vitro, their activation status was assessed by flow cytometry, and supernatant glutamate levels were determined using an enzymatic assay. Expression regulation of molecules involved in vesicular glutamate release was examined by quantitative real-time PCR, and mechanisms of non-vesicular glutamate release were studied by pharmacological blocking experiments. The impact of CD8+ T cell-mediated glutamate liberation on neuronal viability was studied in acute brain slice preparations. Results Following T cell receptor stimulation, CD8+ T cells acquire the molecular repertoire for vesicular glutamate release: (i) they upregulate expression of glutaminase required to generate glutamate via deamination of glutamine and (ii) they upregulate expression of vesicular proton-ATPase and vesicular glutamate transporters required for filling of vesicles with glutamate. Subsequently, CD8+ T cells release glutamate in a strictly stimulus-dependent manner. Upon repetitive T cell receptor stimulation, CD25high CD8+ T effector cells exhibit higher estimated single cell glutamate release rates than CD25low CD8+ T memory cells. Moreover, glutamate liberation by oligodendrocyte-reactive CD25high CD8+ T effector cells is capable of eliciting collateral excitotoxic cell death of neurons (despite glutamate re-uptake by glia cells and neurons) in intact CNS gray matter. Conclusion Glutamate release may represent a crucial effector pathway of neural-antigen reactive CD8+ T cells, contributing to excitotoxicity in CNS inflammation.
Collapse
Affiliation(s)
- Nico Melzer
- Department of Neurology, University of Münster, Albert-Schweitzer-Campus 1, Münster 48149, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
148
|
Magi S, Arcangeli S, Castaldo P, Nasti AA, Berrino L, Piegari E, Bernardini R, Amoroso S, Lariccia V. Glutamate-induced ATP synthesis: relationship between plasma membrane Na+/Ca2+ exchanger and excitatory amino acid transporters in brain and heart cell models. Mol Pharmacol 2013; 84:603-14. [PMID: 23913256 DOI: 10.1124/mol.113.087775] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
It is known that glutamate (Glu), the major excitatory amino acid in the central nervous system, can be an essential source for cell energy metabolism. Here we investigated the role of the plasma membrane Na(+)/Ca(2+) exchanger (NCX) and the excitatory amino acid transporters (EAATs) in Glu uptake and recycling mechanisms leading to ATP synthesis. We used different cell lines, such as SH-SY5Y neuroblastoma, C6 glioma and H9c2 as neuronal, glial, and cardiac models, respectively. We first observed that Glu increased ATP production in SH-SY5Y and C6 cells. Pharmacological inhibition of either EAAT or NCX counteracted the Glu-induced ATP synthesis. Furthermore, Glu induced a plasma membrane depolarization and an intracellular Ca(2+) increase, and both responses were again abolished by EAAT and NCX blockers. In line with the hypothesis of a mutual interplay between the activities of EAAT and NCX, coimmunoprecipitation studies showed a physical interaction between them. We expanded our studies on EAAT/NCX interplay in the H9c2 cells. H9c2 expresses EAATs but lacks endogenous NCX1 expression. Glu failed to elicit any significant response in terms of ATP synthesis, cell depolarization, and Ca(2+) increase unless a functional NCX1 was introduced in H9c2 cells by stable transfection. Moreover, these responses were counteracted by EAAT and NCX blockers, as observed in SH-SY5Y and C6 cells. Collectively, these data suggest that plasma membrane EAAT and NCX are both involved in Glu-induced ATP synthesis, with NCX playing a pivotal role.
Collapse
Affiliation(s)
- Simona Magi
- Department of Biomedical Sciences and Public Health, School of Medicine, University Politecnica of Marche, Ancona, Italy (S.M., S.Ar., P.C., A.A.N., S.Am., V.L.); Department of Experimental Medicine, Second University of Naples, Naples, Italy (L.B., E.P.); and Department of Clinical and Molecular Biomedicine, School of Medicine, University of Catania, Catania, Italy (R.B.)
| | | | | | | | | | | | | | | | | |
Collapse
|
149
|
Bortz DM, Mikkelsen JD, Bruno JP. Localized infusions of the partial alpha 7 nicotinic receptor agonist SSR180711 evoke rapid and transient increases in prefrontal glutamate release. Neuroscience 2013; 255:55-67. [PMID: 24095692 DOI: 10.1016/j.neuroscience.2013.09.047] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 09/11/2013] [Accepted: 09/20/2013] [Indexed: 10/26/2022]
Abstract
The ability of local infusions of the alpha 7 nicotinic acetycholine receptor (α7 nAChR) partial agonist SSR180711 to evoke glutamate release in prefrontal cortex was determined in awake rats using a microelectrode array. Infusions of SSR180711 produced dose-dependent increases in glutamate levels. The lower dose (1.0μg in 0.4μL) evoked a rapid rise (∼1.0s) in glutamate (1.41±0.30μM above baseline). The higher dose (5.0μg) produced a similarly rapid, yet larger increase (3.51±0.36μM above baseline). After each dose, the glutamate signal was cleared to basal levels within 7-18s. SSR180711-evoked glutamate was mediated by the α7 nAChR as co-infusion of the selective α7 nAChR antagonist α-bungarotoxin (10.0μM)+SSR1808711 (5.0μg) reduced the effect of 5.0μg alone by 87% (2.62 vs. 0.35μM). Finally, the clearance of the SSR180711 (5.0μg)-evoked glutamate was bidirectionally affected by drugs that inhibited (threo-beta-benzyl-oxy-aspartate (TβOA), 100.0μM) or facilitated (ceftriaxalone, 200mg/kg, i.p.) excitatory amino acid transporters. TβOA slowed both the clearance (s) and rate of clearance (μM/s) by 10-fold, particularly at the mid-late stages of the return to baseline. Ceftriaxone reduced the magnitude of the SSR180711-evoked increase by 65%. These results demonstrate that pharmacological stimulation of α7 nAChRs within the prefrontal cortex is sufficient to evoke rapid yet transient increases in glutamate levels. Such increases may underlie the cognition-enhancing effects of the drug in animals; further justifying studies on the use of α7 nAChR-positive modulators in treating cognition-impairing disorders in humans.
Collapse
Affiliation(s)
- D M Bortz
- Department of Psychology, The Ohio State University, Columbus, OH, United States
| | | | | |
Collapse
|
150
|
Reevaluation of the beam and radial hypotheses of parallel fiber action in the cerebellar cortex. J Neurosci 2013; 33:11412-24. [PMID: 23843513 DOI: 10.1523/jneurosci.0711-13.2013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The role of parallel fibers (PFs) in cerebellar physiology remains controversial. Early studies inspired the "beam" hypothesis whereby granule cell (GC) activation results in PF-driven, postsynaptic excitation of beams of Purkinje cells (PCs). However, the "radial" hypothesis postulates that the ascending limb of the GC axon provides the dominant input to PCs and generates patch-like responses. Using optical imaging and single-cell recordings in the mouse cerebellar cortex in vivo, this study reexamines the beam versus radial controversy. Electrical stimulation of mossy fibers (MFs) as well as microinjection of NMDA in the granular layer generates beam-like responses with a centrally located patch-like response. Remarkably, ipsilateral forepaw stimulation evokes a beam-like response in Crus I. Discrete molecular layer lesions demonstrate that PFs contribute to the peripherally generated responses in Crus I. In contrast, vibrissal stimulation induces patch-like activation of Crus II and GABAA antagonists fail to convert this patch-like activity into a beam-like response, implying that molecular layer inhibition does not prevent beam-like responses. However, blocking excitatory amino acid transporters (EAATs) generates beam-like responses in Crus II. These beam-like responses are suppressed by focal inhibition of MF-GC synaptic transmission. Using EAAT4 reporter transgenic mice, we show that peripherally evoked patch-like responses in Crus II are aligned between parasagittal bands of EAAT4. This is the first study to demonstrate beam-like responses in the cerebellar cortex to peripheral, MF, and GC stimulation in vivo. Furthermore, the spatial pattern of the responses depends on extracellular glutamate and its local regulation by EAATs.
Collapse
|