Elsevier

Brain Research

Volume 886, Issues 1–2, 15 December 2000, Pages 190-207
Brain Research

Interactive report
The AMPAR subunit GluR2: still front and center-stage1

https://doi.org/10.1016/S0006-8993(00)02951-6Get rights and content

Abstract

Abnormal influx of Ca2+ through AMPA-type glutamate receptors (AMPARs) is thought to contribute to the neuronal death associated with a number of brain disorders. AMPARs exist as both Ca2+-impermeable and Ca2+-permeable channels. AMPARs are encoded by four genes designated GluR1 (GluR-A) through GluR4 (GluR-D). The presence of the GluR2 subunit renders heteromeric AMPA receptor assemblies Ca2+-impermeable. Molecular diversity of AMPARs under physiological and pathological conditions is generated by differential spatio-temporal patterns of GluR expression, by alternative RNA splicing and editing and by targeting and trafficking of receptor subunits at dendritic spines. The GluR2 gene is under transcriptional control by the RE1 element specific transcription factor, a gene silencing factor which renders it neuron-specific. GluR2 transcripts are edited by ADAR2 (double-stranded RNA-specific editase 1). AMPAR targeting and trafficking to spines are regulated by synaptic activity and are critical to synaptic plasticity. Recent studies involving animal models of transient forebrain ischemia and epilepsy show that GluR2 mRNA and GluR2 subunit expression are downregulated in vulnerable neurons prior to cell death. Ca2+ imaging and electrical recording from individual pyramidal neurons in hippocampal slices reveal changes in AMPAR functional properties after ischemia. In slices from post-ischemia animals, CA1 neurons with robust action potentials exhibit greatly enhanced AMPA-elicited rises in intracellular Ca2+. Excitatory postsynaptic currents in post-ischemic CA1 exhibit an enhanced Ca2+-dependent component that appears to be mediated by Ca2+-permeable AMPARs. These studies provide evidence for Ca2+ influx through AMPARs in neurons destined to die. To examine whether acute GluR2 downregulation, even in the absence of a neurological insult, can induce neuronal death, we performed knockdown experiments in rats and gerbils with antisense oligonucleotides targeted to GluR2 mRNA. GluR2 antisense oligonucleotide induced neuronal cell death of pyramidal neurons and enhanced pathogenicity of brief ischemic episodes. These observations provide evidence for Ca2+ influx through AMPARs in neurons destined to die and implicate Ca2+-permeable AMPARs in the pathogenesis of ischemia-induced neuronal death.

Introduction

Glutamate receptors mediate excitatory neurotransmission and play a critical role in synaptogenesis and formation of neuronal circuitry, as well as in synaptic plasticity including long-term potentiation and long-term depression. Excessive activation of glutamate receptors is thought to contribute to the neurodegeneration following a wide range of neurological insults including ischemia, trauma, hypoglycemia and epileptic seizures. Chronic neurodegenerative disorders such as Alzheimer’s disease, Huntington’s chorea, AIDS encephalopathy, and amyotrophic lateral sclerosis may also involve glutamate-induced neuronal cell death (for reviews, see [28], [73]).

Considerable interest has focused on the molecular mechanisms underlying glutamate receptor-mediated neuronal death. Glutamate induces neuronal death by eliciting a rise in intracellular free Ca2+, which activates a number of proteases, phospholipases and endoncleases, by generation of free radicals that destroy cellular membranes by lipid peroxidations (for reviews, see [27], [29], [122], [135]) and by induction of apoptosis [29]. Possible mechanisms by which glutamate could elicit a rise in intracellular Ca2+ include: (1) activation of Ca2+-permeable AMPA (α-amino-3-hydroxy-5-methyl-4-isoazole-proprionic acid)-type glutamate receptors (AMPARs) [146], [148]; (2) activation of group 1 metabotropic glutamate receptors (mGluRs), which are positively linked to inositol phosphates; (3) activation of voltage-sensitive Ca2+ channels; and/or (4) de-activation of extrusion and/or sequestration systems [99].

Until recently, AMPARs were thought to be Ca2+-impermeable. It is now well established that the presence of the GluR2 subunit in heteromeric AMPAR assemblies governs the permeability of AMPARs to Ca2+ and Zn2+. In the adult mammalian central nervous system under physiological conditions, the vast majority of cells and tissues express GluR2-containing, Ca2+-impermeable AMPARs. Thus, a change in the level of GluR2 expression would be expected to have significant physiological consequences. The relative expression of GluR2 subunit mRNA and protein in neurons is not static but is regulated in a cell-specific manner during development [108] and may be remodeled after seizures [38], [113], [114] or ischemic insult [38], [44], [111], [114] and by administration of anti-psychotics [35] drugs of abuse [36], [102] or corticosteroids [93]. Ca2+-permeable AMPARs are implicated in synaptogenesis and formation of neuronal circuitry, particularly at times and in cells in which NMDAR expression is low. Ca2+ influx via Ca2+-permeable AMPARs is thought to play a critical role in growth cone movement and experience-dependent pruning of synaptic connections during early development [81].

This article reviews recent studies that address transcriptional and translation regulation and targeting and trafficking of the AMPAR subunit GluR2 under physiological and pathological conditions, with a particular emphasis on transcriptional regulation of GluR2 in ischemia and status epilepticus.

Section snippets

The AMPAR gene family

AMPARs mediate fast excitatory synaptic transmission in the vertebrate central nervous system. AMPARs are assemblies of four subunits, GluR1-4 (or GluRA-D), encoded by separate genes which are differentially expressed throughout the CNS (for review, see [53], [130]). AMPARs assembled from combinations of GluR1, GluR3 and/or GluR4 subunits (lacking the GluR2 subunit) are permeable to Ca2+ and Zn2+ and have doubly rectifying current–voltage relations due to voltage-dependent block by

GluR2 receptor targeting and trafficking

Trafficking and targeting of specific mRNAs and polyribosomes to dendritic spines are thought to play an important role in the modification of synaptic strength during synaptogenesis and in synaptic plasticity (for review, see [7]). New protein synthesis is essential to long-lasting modifications of synaptic strength [4] and the maintenance phase of LTP [80]. Moreover, targeting of mRNAs to dendrites and dendritic spines can be regulated by synaptic transmission and plasticity [47], [90], [134]

Global ischemia-induced suppression of GluR2 mRNA and subunit expression

Global ischemia during cardiac arrest affects 150,000 Americans each year and in many cases results in delayed onset of neurological deficits [112]. In addition, open heart surgery can cause brain ischemia [14], [119], [140]. Transient, severe global or forebrain ischemia, observed in patients during cardiorespiratory arrest, cardiac surgery or experimentally in animals, induces selective and delayed neuronal death [14], [119], [140]. Pyramidal neurons in the CA1 region of the hippocampus are

Genetic approaches to the role of GluR2 in neuronal cell death

Gene inactivation (knockout) and antisense (knockdown) approaches have proven useful in determining the function of a particular protein under physiological and pathological conditions. Altogether four animal models have been developed using genetic techniques: (1) the Q/R editing deficient mouse (lacking intron 11 of the GluR2 gene); (2) the GluR2 knock-out mouse; (3) the GluR2-flip over expressing mouse; and (4) the gerbil acutely treated with GluR2 antisense oligonucleotides (knockdown).

Concluding remarks

This article reviewed the regulation of AMPAR expression at CA1 synapses, with particular emphasis on RNA transcription, editing and splicing, translational regulation, receptor targeting and trafficking, cell surface expression and anchoring. In addition, this article reviewed evidence that severe neurological insults such as global ischemia and limbic seizures trigger a ‘molecular switch’ that shuts off GluR2 subunit expression in neurons destined to die. In neurons that normally express Ca2+

Acknowledgements

This work was supported by NIH grants no. NS 20752 and NS 31282 (to R.S.Z.). M.V.L.B. is the Sylvia and Robert S. Olnick Professor of Neuroscience.

References (159)

  • D.W. Choi

    Calcium: still center-stage in hypoxic-ischemic neuronal death

    Trends Neurosci.

    (1995)
  • J.A. Chong et al.

    REST: a mammalian silencer protein that restricts sodium channel gene expression to neurons

    Cell

    (1995)
  • S.L. Eastwood et al.

    Decreased synaptophysin in the medial temporal lobe in schizophrenia demonstrated using immunoautoradiography

    Neuroscience

    (1995)
  • S.L. Eastwood et al.

    Decreased expression of mRNAs encoding non-NMDA glutamate receptors GluR1 and GluR2 in medial temporal lobe neurons in schizophrenia

    Brain Res. Mol. Brain Res.

    (1995)
  • L.K. Friedman et al.

    GluR2 hippocampal knockdown reveals developmental regulation of epileptogenicity and neurodegeneration

    Brain Res. Mol. Brain Res.

    (1998)
  • J.R. Geiger et al.

    Relative abundance of subunit mRNAs determines gating and Ca2+ permeability of AMPA receptors in principal neurons and interneurons in rat CNS

    Neuron

    (1995)
  • S. Herlitze et al.

    Argiotoxin detects molecular differences in AMPA receptor channels

    Neuron

    (1993)
  • M. Higuchi et al.

    RNA editing of AMPA receptor subunit GluR-B: a base-paired intron-exon structure determines position and efficiency

    Cell

    (1993)
  • T. Himi et al.

    Differential localization of SCG10 and p19/stathmin messenger RNAs in adult rat brain indicates distinct roles for these growth-associated proteins

    Neuroscience

    (1994)
  • R.M. Hong et al.

    Association of N-ethylmaleimide-sensitive factor with synaptic vesicles

    FEBS Lett.

    (1994)
  • R.J. Jackson et al.

    Translational controls impinging on the 5′-untranslated region and initiation factor proteins

    Curr. Opin. Genet. Dev.

    (1997)
  • Z. Jia et al.

    Enhanced LTP in mice deficient in the AMPA receptor GluR2

    Neuron

    (1996)
  • P. Jonas et al.

    Differences in Ca2+ permeability of AMPA-type glutamate receptor channels in neocortical neurons caused by differential GluR-B subunit expression

    Neuron

    (1994)
  • T. Kirino

    Delayed neuronal death in the gerbil hippocampus following ischemia

    Brain Res.

    (1982)
  • R. Kleiman et al.

    Differential subcellular localization of particular mRNAs in hippocampal neurons in culture

    Neuron

    (1990)
  • M. Kohler et al.

    The organization of the gene for the functionally dominant alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptor subunit GluR-B

    J. Biol. Chem.

    (1994)
  • D. Le et al.

    Enhanced neuronal death from focal ischemia in AMPA-receptor transgenic mice

    Brain Res. Mol. Brain Res.

    (1997)
  • Y. Liu et al.

    Functionally distinct double-stranded RNA-binding domains associated with alternative splice site variants of the interferon-inducible double-stranded RNA-specific adenosine deaminase

    J. Biol. Chem.

    (1997)
  • S. Maas et al.

    Mammalian RNA-dependent deaminases and edited mRNAs

    Curr. Opin. Cell Biol.

    (1997)
  • K.C. Martin et al.

    Synapse-specific, long-term facilitation of aplysia sensory to motor synapses: a function for local protein synthesis in memory storage

    Cell

    (1997)
  • J.W. McDonald et al.

    Physiological and pathophysiological roles of excitatory amino acids during central nervous system development

    Brain Res. Brain Res. Rev.

    (1990)
  • T. Melcher et al.

    RED2, a brain-specific member of the RNA-specific adenosine deaminase family

    J. Biol. Chem.

    (1996)
  • T. Melcher et al.

    Editing of alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptor GluR-B pre-mRNA in vitro reveals site-selective adenosine to inosine conversion

    J. Biol. Chem.

    (1995)
  • B. Meldrum et al.

    Excitatory amino acid neurotoxicity and neurodegenerative disease

    Trends Pharmacol. Sci.

    (1990)
  • L. Mittaz et al.

    Cloning of a human RNA editing deaminase (ADARB1) of glutamate receptors that maps to chromosome 21q22.3

    Genomics

    (1997)
  • H. Monyer et al.

    Developmental and regional expression in the rat brain and functional properties of four NMDA receptors

    Neuron

    (1994)
  • H. Monyer et al.

    Glutamate-operated channels: developmentally early and mature forms arise by alternative splicing

    Neuron

    (1991)
  • A. Nishimune et al.

    NSF binding to GluR2 regulates synaptic transmission

    Neuron

    (1998)
  • M.A. O’Connell

    RNA editing: rewriting receptors

    Curr. Biol.

    (1997)
  • E.M. Aronica et al.

    Aurintricarboxylic acid prevents GLUR2 mRNA down-regulation and delayed neurodegeneration in hippocampal CA1 neurons of gerbil after global ischemia

    Proc. Natl. Acad. Sci. USA

    (1998)
  • O. Bar-Peled et al.

    Cultured motor neurons possess calcium-permeable AMPA/kainate receptors

    Neuroreport

    (1999)
  • G.J. Bassell et al.

    The travels of mRNAs through all cells large and small [see comments]

    FASEB J.

    (1999)
  • R. Benne

    RNA editing. The long and the short of it [news; comment]

    Nature

    (1996)
  • M.V.L. Bennett et al.

    The GluR2 hypothesis: Ca(++)-permeable AMPA receptors in delayed neurodegeneration

    Cold Spring Harb. Symp. Quant. Biol.

    (1996)
  • F. Berton et al.

    Developmental regulation of synaptotagmin I, II, III, and IV mRNAs in the rat CNS

    J. Neurosci.

    (1997)
  • M. Blaschke et al.

    A single amino acid determines the subunit-specific spider toxin block of alpha-amino-3-hydroxy-5-methylisoxazole-4-propionate/kainate receptor channels

    Proc. Natl. Acad. Sci. USA

    (1993)
  • J.R. Brorson et al.

    Ca2+ entry via AMPA/KA receptors and excitotoxicity in cultured cerebellar Purkinje cells

    J. Neurosci.

    (1994)
  • R. Brusa et al.

    Early-onset epilepsy and postnatal lethality associated with an editing-deficient GluR-B allele in mice

    Science

    (1995)
  • W. Brysch et al.

    Regional and temporal expression of sodium channel messenger RNAs in the rat brain during development

    Exp. Brain Res.

    (1991)
  • K.E. Burgin et al.

    In situ hybridization histochemistry of Ca2+/calmodulin-dependent protein kinase in developing rat brain

    J. Neurosci.

    (1990)
  • Cited by (213)

    View all citing articles on Scopus
    1

    Published on the World Wide Web on 30 October 2000.

    View full text