• Tidak ada hasil yang ditemukan

Directory UMM :Data Elmu:jurnal:B:Brain Research:Vol886.Issue1-2.Nov2000:

N/A
N/A
Protected

Academic year: 2017

Membagikan "Directory UMM :Data Elmu:jurnal:B:Brain Research:Vol886.Issue1-2.Nov2000:"

Copied!
18
0
0

Teks penuh

(1)

www.elsevier.com / locate / bres

Interactive report

1

The AMPAR subunit GluR2: still front and center-stage

*

Hidenobu Tanaka, Sonja Y. Grooms, Michael V.L. Bennett, R. Suzanne Zukin

Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA Accepted 26 September 2000

Abstract

21

Abnormal influx of Ca through AMPA-type glutamate receptors (AMPARs) is thought to contribute to the neuronal death associated

21 21

with a number of brain disorders. AMPARs exist as both Ca -impermeable and Ca -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

21

assemblies Ca -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

21

downregulated in vulnerable neurons prior to cell death. Ca 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

21

with robust action potentials exhibit greatly enhanced AMPA-elicited rises in intracellular Ca . Excitatory postsynaptic currents in

21 21

post-ischemic CA1 exhibit an enhanced Ca -dependent component that appears to be mediated by Ca -permeable AMPARs. These

21

studies provide evidence for Ca 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

21

and enhanced pathogenicity of brief ischemic episodes. These observations provide evidence for Ca influx through AMPARs in neurons

21

destined to die and implicate Ca -permeable AMPARs in the pathogenesis of ischemia-induced neuronal death.  2000 Elsevier Science B.V. All rights reserved.

Theme: Neurotransmitters, modulators, transporters, and receptors

Topic: Excitatory amino acid receptors: ligand-gated channels

Keywords: Excitatory amino acid receptor; AMPA receptor; Calcium; Neurotoxicity; Ischemia; Seizure

1. Introduction

mission and play a critical role in synaptogenesis and

formation of neuronal circuitry, as well as in synaptic

Glutamate receptors mediate excitatory neurotrans-

plasticity including long-term potentiation and long-term

depression. Excessive activation of glutamate receptors is

thought to contribute to the neurodegeneration following a

1

Published on the World Wide Web on 30 October 2000.

wide range of neurological insults including ischemia,

Abbreviations: ADAR, Double-stranded RNA adenosine deaminase;

trauma, hypoglycemia and epileptic seizures. Chronic

AMPA, a-amino-3-hydroxy-5-methyl-4-isoxazole-propionic acid;

AM-neurodegenerative disorders such as Alzheimer’s disease,

PARs, AMPA receptors; EAA, excitatory amino acids; EPSCs, excitatory

postsynaptic currents; NBQX, 2,3-dihydroxy-6-nitro-7-sulfamoylbenzo

Huntington’s chorea, AIDS encephalopathy, and

amyot-(f)quinoxiline; PMSF, phenylmethylsulfonyl fluoride; NSF, N-methyl-

rophic lateral sclerosis may also involve glutamate-induced

maleimide sensitive fusion protein; NRSF, neural restrictive silencing

neuronal cell death (for reviews, see [28,73]).

factor; REST, RE1 element specific transcription factor

Considerable interest has focused on the molecular

*Corresponding author. Tel.: 11-718-430-2160; fax: 1

1-718-430-mechanisms underlying glutamate receptor-mediated

neu-8932.

E-mail address: zukin@aecom.yu.edu (R.S. Zukin).

ronal death. Glutamate induces neuronal death by eliciting

(2)

21

a rise in intracellular free Ca

, which activates a number

[42,52] voltage-dependent block by polyamines [13,32]

of proteases, phospholipases and endoncleases, by gene-

and single channel conductance [141] of recombinant

ration of free radicals that destroy cellular membranes by

AMPARs expressed in Xenopus oocytes and mammalian

lipid peroxidations (for reviews, see [27,29,122,135]) and

cells.

by induction of apoptosis [29]. Possible mechanisms by

AMPARs are ligand-gated channels and, by analogy to

21

which glutamate could elicit a rise in intracellular Ca

the nicotinic acetylcholine receptor, are thought to be

21

include: (1) activation of Ca

-permeable AMPA (

a

-

tetrameric or pentameric assemblies arranged around a

amino-3-hydroxy-5-methyl-4-isoazole-proprionic

acid)-

central aqueous pore.

type glutamate receptors (AMPARs) [146,148]; (2) activa-

The current model of GluR subunit topology in the

tion

of

group

1

metabotropic

glutamate

receptors

membrane includes: (1) a large extracellular

amino-termi-(mGluRs), which are positively linked to inositol phos-

nal domain; (2) three transmembrane-spanning domains

21

phates; (3) activation of voltage-sensitive Ca

channels;

(TM1, TM3 and TM4); (3) a fourth amphipathic segment

and / or (4) de-activation of extrusion and / or sequestration

(TM2) that forms a channel-lining reentrant hairpin loop,

1

systems [99].

similar in structure to the pore-forming region of K

21

Until recently, AMPARs were thought to be Ca

-

channels [158]; (4) a binding domain for agonists formed

impermeable. It is now well established that the presence

from segments of the amino-terminal domain and

extracel-of the GluR2 subunit in heteromeric AMPAR assemblies

lular loop [138]; and (5) an intracellular C-terminal

21 21

governs the permeability of AMPARs to Ca

and Zn

.

domain. The dominance of the GluR2 subunit in

determin-21

In the adult mammalian central nervous system under

ing permeability to Ca

and other divalent ions is

physiological conditions, the vast majority of cells and

attributed to the presence of a positively charged arginine

21

tissues express GluR2-containing, Ca

-impermeable AM-

(R) in place of a glutamine (Q) residue within TM2, which

PARs. Thus, a change in the level of GluR2 expression

forms the selectivity filter of AMPARs [21,57]. Although

would be expected to have significant physiological conse-

most

hippocampal

neurons

express

predominantly

quences. The relative expression of GluR2 subunit mRNA

heteromeric AMPARs, they may also express GluR1

and protein in neurons is not static but is regulated in a

homomers [80,157].

cell-specific manner during development [108] and may be

AMPARs are differentially expressed throughout

neu-remodeled after seizures [38,113,114] or ischemic insult

rons of the mammalian central nervous system. Studies

[38,44,111,114] and by administration of anti-psychotics

involving patch-clamp recording combined with RT-PCR

21

[35] drugs of abuse [36,102] or corticosteroids [93]. Ca

-

(reverse transcriptase-polymerase chain reaction)

demon-21

permeable AMPARs are implicated in synaptogenesis and

strate that AMPAR permeability to Ca

varies inversely

formation of neuronal circuitry, particularly at times and in

with abundance of GluR2 mRNA in a wide range of cell

21

cells in which NMDAR expression is low. Ca

influx via

types (Table 1). Excitatory principal neurons such as

21

Ca

-permeable AMPARs is thought to play a critical role

hippocampal [12] and neocortical [60] pyramidal cells and

in growth cone movement and experience-dependent prun-

dentate gyrus granule cells [42] express abundant GluR2

21

ing of synaptic connections during early development [81].

mRNA and exhibit low AMPAR Ca

-permeability. Thus,

This article reviews recent studies that address transcrip-

a change in GluR2 expression would be expected to have

tional and translation regulation and targeting and traffick-

significant physiological consequences.

ing of the AMPAR subunit GluR2 under physiological and

pathological conditions, with a particular emphasis on

2.1. GluR2 RNA editing

transcriptional regulation of GluR2 in ischemia and status

epilepticus.

The functionally critical arginine (R) residue within

TM2 is not encoded by the GluR2 gene, but rather arises

by adenosine-specific RNA editing of the double-stranded

2. The AMPAR gene family

pre-mRNA, which converts an adenosine residue in the

glutamine codon to an inosine [50,136] (Fig. 1). RNA

AMPARs mediate fast excitatory synaptic transmission

editing, the process by which genomically encoded

in-in the vertebrate central nervous system. AMPARs are

formation is enzymatically modified, is an important mode

assemblies of four subunits, GluR1-4 (or GluRA-D),

of receptor regulation [8,78,97]. To date, three

structurally-encoded by separate genes which are differentially ex-

related RNA-editing enzymes with adenosine deaminase

pressed throughout the CNS (for review, see [53,130]).

activity have been identified in mammalian tissues:

AMPARs assembled from combinations of GluR1, GluR3

ADAR1 (dsRNA-specific adenosine deaminase, dsRAD or

and / or GluR4 subunits (lacking the GluR2 subunit) are

DRADA [63,74,98,107]; ADAR2 (dsRNA-specific editase

21 21

permeable to Ca

and Zn

and have doubly rectifying

1, RED-1 [43,69,82,85,97]; and RED2 (dsRNA-specific

current–voltage relations due to voltage-dependent block

editase 2) [82]). Each of the editing enzymes contains two

by intracellular polyamines [21,52,152]. The presence of

to three dsRNA binding domains within the

amino-termi-21

(3)

carboxy-Table 1

a

Studies supporting the GluR2 hypothesis

Condition Cell type Refs

Suppression of GluR2 gene expression

Developing brain various [108,111]

Transient global ischemia CA1 pyramidal cells [3,44,49,109–111,113]

Oxygen–glucose dissociated hippocampal [159]

deprivation neurons

Status epilepticus CA3 pyramidal cells [36–38,45,70,71,111,113,114]

Mutant spastic rats cerebellar Purkinje and [79]

granule cells

Schizophrenia parahippocampal [34]

pyramidal cells

Amyotrophic lateral spinal motor neurons [153]

sclerosis

Alzheimer’s disease entorhinal cortex [144]

GluR2 knockdown-induced neuronal death

Adult rat and gerbil hippocampal pyramidal [100]

neurons

Developing brain hippocampal pyramidal [39]

neurons

21

Expression of Ca -permeable AMPARs

Developing brain retinal ganglion cells [120,139]

Transient global ischemia CA1 pyramidal cells [44,146,147]

Oxygen–glucose deprivation dissociated hippocampal [159]

neurons

Editing-deficient GluR2 mice CA3 pyramidal cells [16]

21

Neurotoxicity mediated by Ca -permeable AMPARs

Primary cultures cerebellar Purkinje cells [15]

Primary cultures neocortical neurons [77,151,156]

primary cultures spinal motor neurons [5]

cell line oligodendroglial lineage

Oxygen–glucose dissociated hippocampal [159]

deprivation neurons

Editing-deficient GluR2 mice CA3 pyramidal cells [16]

a

Modified from [109].

(4)

terminal domain [6]. Whereas ADAR1 and ADAR2 are

at the R / G site and splicing of the flip / flop cassettes of

expressed in most mammalian tissue; RED2 is expressed

GluR1-4 are developmentally regulated. During early

exclusively in the brain [63,82,97].

postnatal life, mammalian neurons express only GluR flip

ADAR2 is an extremely efficient and specific double-

splice forms; expression of GluR flop forms occurs at a

stranded RNA editase. In neurons of mammalian brain,

later stage of development, leading to a reduction in the

ADAR2 edits primary transcripts encoding glutamate

steady-state phase of glutamate-evoked currents [87,88].

receptor subunits at the ‘Q / R’ site, thereby altering the

gating and ionic permeability properties of the transmitter-

2.3. Transcriptional regulation of GluR2

activated channel [83,124,131]. The other main target of

ADAR2 is the serotonin 5HT-2C receptor, a member of

Recent studies indicate the presence of a functional

the large superfamily of G protein-coupled receptors [23].

repressor element 1 (RE1-like silencer) in the proximal

ADAR2 edits the 5HT-2C pre-mRNA at a site near its 5

9

promoter of the GluR2 gene [92] and show that under

end to convert a genomically-encoded isoleucine residue to

physiological conditions, the GluR2 gene is under

tran-valine. Fully edited versions of the serotonin 5HT-2C

scriptional control by REST (RE1 element specific

tran-receptor couple with less efficiency to G proteins.

scription factor, also known as NRSF or XBR), a gene

In neonatal and adult rat brain, virtually 100% of GluR2

silencing factor which renders it highly neuron selective

mRNA is edited at the Q / R site to yield GluR2 subunits

[56,92] (Fig. 2). Recombinant REST represses

transcrip-21

that form Ca

-impermeable AMPARs. The unedited form

tion of the GluR2 gene by recruiting the co-repressors

is detectable only in prenatal life (E14 to P0) but never

Sin3A and histone deacetylase to the RE1 site of the

exceeds 1% of total GluR2 mRNA [21]. In human brain

GluR2 promoter [56].

[96], Q / R editing is somewhat less in substantia nigra

REST, a gene silencing factor which binds the RE1

(72%), striatum (89%) and fetal tissue (96%), suggesting

regulatory element, is thought to serve a critical role in

that editing efficiency may vary among species and / or

differentiation by repression of a subset of neuron-specific

developmental stages.

genes (Fig. 2). Known target genes including NaCh II

In the AMPAR family, desensitization kinetics are

[17,51], calbindin I [1], synaptotagmin IV [10], neuronal

controlled by editing of GluR2 through GluR4 at a site

nicotinic ACh receptor

a

2

[154] and AMPAR subunit

preceding the putative fourth transmembrane region [75].

GluR2 [56,90]. REST, a member of the Kruppel family of

Editing at the ‘R / G’ site is specific for GluR2, -3 and -4,

zinc finger transcription factor proteins, contains two

and is about 80–90% complete in adult rat brain. The

repressor motifs within the N- and C-terminal domains and

degree of RNA editing at the R / G site increases with age

nine zinc finger motifs (contained within the central

through the embryonic and postnatal periods in a subunit-

portion of the protein). Molecular diversity of REST is

and splice variant-specific manner [75].

conferred by alternative RNA splicing which generates at

least six splice variants, many of which are truncated and

2.2. GluR2 RNA splicing

exhibit reduced trans-repressor activity and DNA binding

capacity [104]. The structure of the REST gene and

In addition to RNA editing, further molecular diversity

regulation by alternative RNA splicing are conserved in

of AMPAR subunits is conferred by alternative RNA

human, mouse and rat.

splicing of GluR1-4. AMPAR subunits exist as either of

In early embryogenesis, REST is expressed at high

two distinct isoforms termed ‘flip’ and ‘flop’, which are

levels specifically in non-neural tissues and in

undifferen-generated by alternative splicing of a 114 bp region

tiated neural precursor cells [30,127,129] and is thought to

immediately adjacent to the R / G editing site. Alternative

serve a critical role by repression of a subset of

neuron-splicing introduces one of two functionally critical casset-

specific genes [30,127,128]. The pattern of REST

expres-tes of 38 amino acids (flip or flop) [135] into the

sion in the immature nervous system is inversely correlated

extracellular loop of the GluR subunit. RNA editing at the

with patterns of known target genes. In adult rat brain

R / G site and splicing at the flip / flop site act cooperatively

under physiological conditions, REST is expressed by

to control the desensitization and recovery rates of

neuronal cells, but less abundantly than in non-neuronal

AMPAR

responses

[131].

The

allosteric

modulator

cells. Highest levels of REST expression occur in neurons

cyclothiazide strongly attenuates desensitization in flip but

of the hippocampus, pons / medulla and midbrain. Although

not in flop splice variants of recombinant AMPARs [105].

all splice forms are expressed in neuronal cells, two are

Consistent with their extracellular position, neither the

specific. These splice forms contain short

neuron-21

(5)

Fig. 2. Models of neuron restrictive silencer element (NRSE), RE-1-silencing trancription factor (REST), REST function and GluR2 promoter. (A) DNA sequence of the neuron restrictive silencer element (NRSE) consensus, which serves as a recognition sequence for REST. (B) Schematic representation of the repressor element 1(RE-1)-silencing transcription factor (REST) protein. (C) Function of NRSF / REST in non-neuronal cells and neuronal cells. (D) Schematic showing the location of the silencer in the GluR2 promoter showing silencer element. A,B, reprinted from [61]; C, reprinted from [127]; D, reprinted from [92].

Under pathological conditions, REST expression is

mRNAs encoding them are localized to the spines of

markedly upregulated in hippocampal neurons. Kainic

hippocampal neurons. A number of proteins implicated in

acid-induced status epilepticus induces a dramatic, but

synaptic plasticity and the mRNAs encoding them are

transient upregulation [56] of REST in dentate granule

localized to spines of hippocampal neurons. These include

cells and a pronounced, long-term (24 h or longer)

microtubule-associated protein 2 (MAP2) [40,65], the

a

21

upregulation of REST in hippocampal pyramidal neurons

subunit of Ca

/ calmodulin-dependent protein kinase II

a

following severe limbic seizures [104]. These observations

(CamKII

a

) [19], the activity-related cytokseletal protein

raise the possibility that other neuronal insults as, for

ArC [19], the RNA binding protein, CREB [31], RNA

example, global ischemia, also induce REST expression in

polymerase III transcript BC1 [145], and AMPAR and

neurons destined to die.

NMDAR mRNAs [41,86] (for review, see [7]). Thus,

translational regulation of specific transcripts within

de-2.4. Translational regulation of the GluR2 transcript

ndrites would be expected to provide fine control over the

temporal and spatial extent of gene expression, including

Recent interest in translational control of synaptic

those for glutamate receptor subunits.

(6)

which regulate the initiation and elongation of mature

this mechanism comes from the finding of pools of

transcripts, and (2) translational repressor and enhancer

AMPARs within spines [118], the high concentration of

factors. In addition, translational efficiency is influenced by

NSF in the hippocampus [54,116], its high localization

transcript-specific structural motifs, which serve as recog-

within hippocampal PSDs [155], and its accumulation in

nition sequences for the RNA binding proteins. These

PSDs following transient cerebral ischemia [55].

include: (1) sequences residing in the 5

9

(and 3

9

) UTR; (2)

alternate or additional 5

9

-UTR AUG codons (or their

cognate short open reading frames); (3) consensus se-

4. Regulation of GluR2 expression in

quences for RNA binding proteins; and / or (4) the exact

neurodegenerative disorders

nucleotide context of the initiator AUG [58].

The GluR2 transcript appears to be under negative

4.1. Ischemia

translational control. The presence of the 5

9

-UTR region in

the GluR2 transcript suppresses translation of GluR2 in

4.1.1. Global ischemia-induced suppression of GluR2

Xenopus oocytes and in cell-free reticulocyte lysates [92].

mRNA and subunit expression

The longest GluR2 transcripts identified in vivo, beginning

Global ischemia during cardiac arrest affects 150,000

481 bases downstream from the AUG, exhibit greatly

Americans each year and in many cases results in delayed

reduced translational efficiency (by about 40- to 50-fold)

onset of neurological deficits [112]. In addition, open heart

relative to the GluR2 transcript containing only a seven-

surgery can cause brain ischemia [14,119,140]. Transient,

base leader when assayed in reticulocyte lysates [67,92].

severe global or forebrain ischemia, observed in patients

The region with the greatest suppressor activity (

|

20-fold)

during cardiorespiratory arrest, cardiac surgery or

ex-overlaps all identified transcription initiation sites and

perimentally in animals, induces selective and delayed

includes two of the five upstream AUG codons and a

neuronal death [14,119,140]. Pyramidal neurons in the

40-nucleotide imperfect GU repeat. Although the mecha-

CA1 region of the hippocampus are particularly

vulner-nism for translational suppression by the GluR2 5

9

-UTR

able. Histological evidence of degeneration, exhibiting the

has yet to be identified, the upstream AUGs on their own

hallmarks of apoptosis, is not observed until two to three

exert only minor suppression of translation [91].

days after induction of ischemia in rats [25,64,115,121].

The delayed cell death after ischemia requires an early rise

21 21

in Ca

in CA1 and initial translocation of Zn

from

3. GluR2 receptor targeting and trafficking

presynaptic neurons at CA1 synapses. During the ischemic

21

episode cells depolarize, exhibit a rise in intracellular Ca

Trafficking and targeting of specific mRNAs and poly-

and become inexcitable. Following reperfusion, cells

ap-ribosomes to dendritic spines are thought to play an

pear morphologically normal, exhibit normal intracellular

21

important role in the modification of synaptic strength

Ca

and regain the ability to generate action potentials for

during synaptogenesis and in synaptic plasticity (for

24 to 72 h after the ischemic insult. Ultimately,

intracellu-21 21

review, see [7]). New protein synthesis is essential to

lar Ca

and / or Zn

rises in vulnerable neurons and cell

long-lasting modifications of synaptic strength [4] and the

death ensues.

maintenance phase of LTP [80]. Moreover, targeting of

This delayed neurodegeneration, which may have

clini-mRNAs to dendrites and dendritic spines can be regulated

cal relevance, is thought to be triggered by a transient rise

by synaptic transmission and plasticity [47,90,134,143].

in glutamate release during the ischemic episode [142],

21

Individual spines along a single dendrite may express

followed by late and excessive Ca

influx through

AMPARs which differ in subunit composition and func-

glutamate receptor channels [29,135]. Although NMDARs

21 21

tional properties including conductance, kinetics and Ca

are highly Ca

-permeable, there is now general consensus

permeability [70,123]. Recent studies support an NSF(N-

that antagonists of AMPARs, like the quinoxalinedione

methyl-maleimide

sensitive

fusion

protein)-dependent

NBQX, appear to be much more effective than NMDA

mechanism for insertion of GluR2-containing AMPARs

antagonists in preventing CA1 cell death following severe

and reveal a rapid recycling of functional AMPARs at CA1

global ischemia, even when given as late as 24 h after

synapses under physiological conditions (Fig. 3). Immuno-

ischemia [18,94,133]. These observations indicate that

precipitation experiments demonstrate that GluR2 exists

AMPARs are critical mediators of ischemia-induced

neuro-together

with

NSF

in

a

complex

in

hippocampus

nal death and that neuronal damage is not irreversible for

[103,137]. Yeast two-hybrid studies reveal that GluR2 (and

24 h after the insult.

(7)

Fig. 3. Model of NSF modulation of AMPA receptor function. (A) Regulation of receptor insertion. Interaction of NSF with C terminus of the GluR2 or GluR4 subunit in subsynaptic vesicles (1) may regulate the association or ‘docking’ (2) of the vesicles with the postsynaptic membrane fusion machinery and promote the insertion of AMPA receptors into the synaptic plasma membrane (3). (B) Regulation of receptor function or clustering. Interaction of NSF with the C terminus of the GluR2 or GluR4 subunit in the synaptic plasma membrane (1) regulates the conformation (2) of the GlurR subunits and modulates channel function and / or the association of the receptors with synaptic proteins involved in synaptic targeting. Reprinted from [137].

mRNA downregulation directly precedes neural damage,

lated into changes in subunit abundance. Studies involving

immunofluorescent labeling of brain sections and Western

and does not occur in the CA3 or dentate gyrus regions

blot analysis of microdissected hippocampal subfields

which are resistant to ischemia-induced damage. GluR3

indicate that global ischemia triggers a pronounced and

mRNA levels also decrease, but to a lesser degree (to

cell-specific reduction in GluR2 subunit abundance in CA1

about 50% of control values); GluR1 and NR1 levels

pyramidal neurons [101]. At 72 h after ischemia, virtually

remain constant. The GluR1 / GluR2 ratio is markedly

all CA1 pyramidal neurons exhibited greatly reduced

increased in CA1 at 24 h, but not in CA3 or dentate gyrus.

GluR2 immunolabeling throughout their somata and

de-Because the presence of the edited GluR2 subunit greatly

21

ndritic processes (Fig. 5). GluR2 immunolabeling was

reduces the Ca

permeability of AMPA channels, these

21

unchanged in pyramidal cells of the CA3 and granule cells

findings predict expression of functional of Ca

-perme-of the dentate gyrus, regions resistant to ischemia-induced

able AMPARs in CA1 neurons after global ischemia, but at

damage. In contrast, immunolabeling of the AMPAR

times preceding neuronal death. Moreover, timing of the

GluR1 was unchanged in CA1, CA3 and DG. Western

switch is consistent with a causal role for

AMPAR-me-21

analysis indicated that GluR2 subunit abundance was

diated Ca

influx in ischemia-induced damage. Since

markedly reduced in CA1 at 60 and 72 h after the ischemic

editing of GluR2 mRNA at the Q / R site is unaltered after

insult, times prior to the onset of neuronal death; GluR1

global ischemia [106,125,159], the change in AMPAR

abundance was unchanged in all subfields at all times

functional properties would appear to be a direct

conse-examined. These findings, together with our previous

quence of altered gene expression.

21

observation of enhanced AMPA-elicited Ca

influx in

Because AMPAR subunit expression is known to be

post-ischemic CA1 neurons [44] demonstrate expression of

under translational control, an important question was

21

(8)

Fig. 5. Global ischemia induces downregulation of GluR2, but not GluR1, immunolabeling in CA1 pyramidal neurons. GluR1 and GluR2 immunoreactivity in brain sections from (A, C) a control (sham-operated) and (B, D) an experimental animal 72 h after ischemia. (B) GluR2 immunoreactivity decreased in CA1 pyramidal cells after ischemia, although (D) GluR1 immunoreactivity was unchanged. so: stratum oriens; sp: stratum pyramidale; sr: stratum radiatum. Similar results were observed in experimental and control animals.

21

acetyl spermine [147], channel blockers selective for Ca

-permeable AMPARs [11,48]. These findings are consistent

with a mechanism whereby post-ischemic CA1 neurons

21

generate slow EPSCs mediated by newly expressed Ca

-permeable AMPARs. Many post-ischemic CA1 pyramidal

Fig. 4. Expression of GluR2 mRNA is reduced specifically in CA1 after

neurons are irreversibly depolarized by prolonged low

ischemia. Photomicrographs of autoradiograms of GluR1, GluR2, a NR1

frequency stimulation of the Schaffer collateral /

commisur-mRNA in situ hybridization in coronal sections of gerbil brain at the level

al input [149]. Moreover, post-ischemic neurons fail to

of the dorsal hippocampus from control animals (A, C, E) and from

exhibit long-term potentiation following tetanic stimulation

experimental animals 72 h after ischemia (B, D, F) are shown. GluR2

[64]. Hippocampal neurons in culture subjected to

subleth-mRNA was dramatically reduced in the pyramidal cell layer of the

vulnerable CA1 but not in the pyramdial cell layer of CA3 or in the

al oxygen–glucose deprivation exhibit increased

AMPA-21

granule cell layer of the dentate gyrus, areas that do not undergo

or kainate-induced Ca

accumulation, increased

sensitivi-neurodegeneration. GluR1 and NR1 mRNAs were somewhat reduced in

ty of AMPARs to Joro spider toxin, and increased

vul-CA1 72 h after ischemia. Reprinted from [44].

nerability to AMPAR-mediated excitotoxity, suggesting

21

increased formation of GluR2-lacking, Ca

-permeable

AMPARs [159].

vulnerable neurons. Thus, the present study provides an

To determine whether downregulation of GluR2

expres-21

important link in the causal chain between global ischemia

sion translates into enhanced Ca

influx through

AM-and delayed death of CA1 pyramidal neurons.

PARs into CA1 neurons, we used a combination of

electrophysiological intracellular recording and optical

21

4.1.2. Ischemia-induced changes in glutamate receptor

imaging with the Ca

indicator dye fura-2 [44]. In

function

hippocampal slices from gerbils 48 h after ischemia,

21

Global ischemia induces a number of functional changes

AMPA elicits only a small rise in [Ca

] , which is not

i 21

in the hippocampal CA1 indicative of increased expression

significantly different from Ca

rises in control neurons.

21

of Ca

-permeable AMPARs. AMPAR-mediated excitat-

At 72 h after the ischemic insult individual CA1 neurons

ory postsynaptic currents (EPSCs) at the CA1 / Schaffer

that retain the ability to fire action potentials exhibit a

21

collateral synapse are prolonged after ischemia and exhibit

greatly enhanced AMPA-elicited rise in [Ca

] . Basal

i 21

(9)

pyramidal cells (Fig. 6). These findings indicate that

AMPAR antagonist, 6-cyano-7-nitro-quinoxiline-2,3-dione

21

AMPAR functional responses are altered following global

disodium, or of the Ca

permeable AMPAR channel

21

ischemia and provide direct evidence for Ca

-influx

blocker, 1-naphthyl acetyl spermine, afforded protection

directly through AMPARs in vulnerable CA1 neurons at

against antisense-induced cell death. This finding indicates

21

times after brief global ischemia and preceding obvious

that antisense-induced cell death is mediated by Ca

cell loss.

permeable AMPARs. GluR2 antisense and brief sublethal

To examine whether acute downregulation of the GluR2

global ischemia acted synergistically to cause degeneration

subunit, even in the absence of a neurological insult, can

of pyramidal neurons, consistent with a common

mecha-cause neuronal cell death, we performed GluR2 ‘knoc-

nism of cell death. These findings demonstrate that

dow-kdown’ experiments in rats and gerbils with antisense

nregulation of GluR2 is sufficient to induce delayed death

oligonucleotides targeted to GluR2 mRNA [100].

of specific neuronal populations.

21

Knockdown of receptor subunits has proven valuable in

Recent evidence implicates Zn

entry as a critical

investigation of receptor function in vivo and in vitro.

component of neuronal death after transient global

is-21

GluR2 antisense induced delayed death of pyramidal

chemia in rats [66]. After a brief ischemic insult, Zn

is

neurons in CA1 and CA3. Antisense-induced neurodegene-

translocated from presynaptic terminals and accumulates in

ration was preceded by a reduction in GluR2 mRNA, as

degenerating neurons in CA1 [66]. This accumulation

indicated by in situ hybridization analysis, and in GluR2

precedes the onset of neurodegeneration and is prevented

21

protein, as indicated by Western blot analysis. GluR2

by intraventricular administration of the Zn

chelator

antisense suppressed GluR2 mRNA in the dentate gyrus,

Ca-EDTA just prior to induction of ischemia. Ca-EDTA

but did not cause cell death (Fig. 7). Administration of the

administration 1 h after reperfusion is not neuroprotective,

21

Fig. 6. AMPA-elicited inward current and rise in intracellular free Ca concentration in a CA1 pyramidal neuron after ischemia. (A) Inward current elicited by AMPA (30mM with 10mM cyclothiazide (CTZ)) in a CA1 pyramidal neuron in a hippocampal slice from an animal 72 h after ischemia.

21 1

AMPA and CTZ were bath applied for 30 s (red bar) in saline containing NMDA, Ca and Na channel blockers. Then the AMPA was washed out with

21 1

saline containing the NMDA, Ca and Na channel blockers. After|5 min, CNQX (20mM) was added to the other blockers to cause more rapid recovery. In the control neuron illustrated in B, the AMPA-elicited inward current in the presence of 30mM CTZ was of somewhat lower amplitude but similar in time course. (B) Optical imaging Optical imaging (excitation wavelength, 350 nm) of individual CA1 pyramidal neurons injected with fura-2 in hippocampal slices from a control animal (upper row) and an experimental animal 72 h after ischemia (lower row, same cell as in A). (a) Image taken before bath application of agonist (time indicated in current trace above); (b) image taken at peak inward current after application of AMPA (30mM with

21

10mM CTZ); (c) image taken after recovery to near baseline current. Color represents Ca concentration determined from the ratio of fluorescence

21

obtained at two excitation wavelengths (350 nm and 380 nm); calibration at right. AMPA elicited little change in intracellular free Ca in the control

21

neuron. In contrast, AMPA elicited a rise in Ca in the soma of the postischemic neuron and a smaller increase in its proximal dendrites. Red circles in the

21

(10)
(11)

21

indicating that Zn

entry during or immediately after

ischemia is toxic. Moreover, ischemia induces mRNA

expression of the transporter gene ZNT-1 in CA1 neurons

that are destined to die, presumably in response to the

21 21

Zn

accumulation, since Zn

induces expression of the

gene in cultured neurons [150]. Although one can interpret

the induction as a homeostatic response, it does not prevent

cell death. Since GluR2-lacking AMPARs are permeable to

21 21

Zn

[132], delayed entry of Zn

may contribute to

degeneration. If so, Ca-EDTA treatment delayed by several

days after the ischemic insult would also be

neuroprotec-tive.

21

Finally, evidence in support of a role for Ca

-perme-able AMPARs in ischemia-induced damage comes from

studies involving dissociated primary cultures of cortical

21

neurons [24]. This study reveals that activation of Ca

-permeable AMPARs, in addition to activation of NMDA

channels, leads to the generation of oxygen radicals, a

well-established cause of cell damage.

Together, these studies provide substantial evidence that

suppression of GluR2 expression in CA1 after transient

global ischemia leads to assembly and insertion of

GluR2-lacking AMPARs at CA1 synapses. The change in subunit

composition is consonant with observed changes in

AMPAR functional properties and increased influx of toxic

21 21

Ca

and Zn

[9,109].

4.2. Status epilepticus

Kainic acid-induced status epilepticus leads to delayed,

selective death of pyramidal neurons of the hippocampus.

Status epilepticus in adult rats triggers a pronounced

Fig. 8. Status epilepticus induces downregulation of GluR2, but not

suppression of GluR2 mRNA in vulnerable CA1 and CA3

GluR1 mRNA in hippocampal CA1 and CA3. (A) GluR2 mRNA

pyramidal neurons prior to the onset of neuronal death, as

expression in rat hippocampus after status epilepticus. (Upper) Film

assessed by in situ hybridization [38,113] (Fig. 8). To

autoradiograms of GluR2 mRNA expression in the hippocampus of status epilepticus and control rats. Downregulation of GluR2 mRNA expression

examine whether the observed changes in GluR2 mRNA

is first detected at 12 h after KA injection within CA1 and CA3, but

are translated into changes in subunit expression, we

remains stable in DG. (Lower) Time-course of GluR2 mRNA expression

performed immunolabeling and Western blot analysis [45]

after onset of status epilepticus. Mean densities in hippocampal CA1 and

(Fig. 9). Double immunolabeling revealed individual CA1

CA3 decreased and increased in DG. (B) GluR1 mRNA expression in rat

and CA3 pyramidal neurons expressing abundant GluR1

hippocampus after status epilepticus. (Upper) Film autoradiograms of GluR1 mRNA expression at 6, 12, 16, 20, 24 h after KA-induced status

and greatly reduced GluR2. GluR2 immunolabeling was

epilepticus and in saline controls. GluR1 mRNA expression remains

enhanced in granule cells of the dentate gyrus, a region

stable at all time points assayed, although there is a modest decrease in

resistant to seizure-induced damage. Quantitative Western

CA1 and CA3 at 24 h. (Lower) Mean densities of GluR1 mRNA remain

blot analysis revealed a reduction in GluR2 subunit

unchanged. Values are expressed as a percentage of saline injected

abundance by about 15% in CA1 and by about 30% in

controls. Error bars represent the standard error of the mean. *P,0.05,

Student’s unpaired t test. Reprinted from [45].

CA3 at 20 h, with no change in dentate gyrus. Thus,

GluR2 subunit abundance was regulated in a cell-specific

manner. GluR1 subunit abundance was unchanged in all

subfields at all times examined. These findings indicate

induces status epilepticus, followed by delayed

neurode-that AMPAR subunit composition is remodeled in response

generation of pyramidal neurons in the hippocampal CA3.

21

to neuronal injury and support a role for Ca

-permeable

Status epilepticus triggered suppression of GluR2 mRNA,

21

AMPAR as critical mediators in the neuronal death associ-

consistent with expression of GluR2-lacking, Ca

-perme-ated with status epilepticus.

able AMPARs in neurons destined to die. In pup rats,

(12)

Fig. 9. Status epilepticus downregulates GluR2 subunit expression in CA3. (Upper) Co-localization of GluR1 and GluR2 immunoreactivity in CA3 pyramidal neurons. Saline injected control sections stained for GluR1 (A) and GluR2 (C). GluR2 immunoreactivity decreased in CA3 pyramidal cells 22h after kainate administration (B), although GluR1 immunoreactivity was unchanged (D). Co-localization of GluR1 and GluR2 occurred primarily in stratum pyramidale (E,F). (Lower) Status epilepticus decreases GluR2 but not GluR1 protein levels in CA1 and CA3. Quantitative Western blot analysis of GluR1 and GluR2 protein expression in microdissected hippocampal subfields (see Insert in A) after status epilepticus. (A) GluR2 protein levels decreased in CA3a–b at 16 h after KA injections, and further declined at 20 and 24 h. Similar but smaller changes were seen in CA1, but were not significant until 24 h. No changes in the level of GluR2 protein were observed in DG-CA3c. The plotted ratios are the means of the ratios for individual animals. Error bars represent SEMs. **P,0.001; *P,0.05; Student’s upaired t test. Reprinted from [45].

(13)

subunit expression vary with development: GluR2 dow-

in synaptic plasticity during brain development, and failure

21

nregulation occur only at those ages at which seizure-

to decrease Ca

-permeability of AMPARs at later stages

induced damage is observed. These findings provide

may lead to aberrant development.

21

evidence for Ca

-permeable AMPARs in the causal chain

Why do neurons which express little or no GluR2 (e.g.,

of events between severe limbic seizures and delayed

hippocampal interneurons) and hippocampal pyramidal

neuronal death.

neurons of the GluR2(

2

/

2

) knockout mouse [59] survive?

Viability of the GluR2 knock-out mice in contrast to that

of the GluR2 editing deficient mice could occur for any of

5. Genetic approaches to the role of GluR2 in

a number of reasons. These include (1) compensatory

21

neuronal cell death

increases in Ca

buffering and extrusion (as, for example,

21

enhanced expression of Ca

-binding proteins [68,117]),

Gene inactivation (knockout) and antisense (knoc-

(2) reduced AMPAR currents, due to slowed receptor

kdown) approaches have proven useful in determining the

assembly or reduced expression of GluR1 and GluR3,

function of a particular protein under physiological and

and / or (3) expression of receptors with altered properties,

pathological conditions. Altogether four animal models

such as number, localization and interaction of AMPARs

have been developed using genetic techniques: (1) the

with signaling and / or anchoring proteins and enhanced

Q / R editing deficient mouse (lacking intron 11 of the

desensitization [42]. The viability of these neurons suggest

GluR2 gene); (2) the GluR2 knock-out mouse; (3) the

that acute knockdown of GluR2 in neurons that normally

GluR2-flip over expressing mouse; and (4) the gerbil

express high levels of GluR2 (and relatively rapid changes

21

acutely treated with GluR2 antisense oligonucleotides

in AMPAR Ca

permeability) may be necessary to induce

(knockdown). This section briefly reviews the conse-

neuronal death. It should, however, be noted that the

quences of these genetic manipulations on brain develop-

GluR2 (

2

/

2

) knockout was made on a 129 / SvEMS

3

ment and susceptibility to excitotoxic cell death.

C57BL / 6 hybrid, a strain with high resistance to

gluta-Heterozygous transgenic mice engineered for a Q / R

mate-induced excitotoxicity [42,126]. Moreover, Jia et al.

editing deficient GluR2 allele express AMPARs with

(1996) [59] did not evaluate the susceptibility of the

21

increased Ca

-permeability, particularly in hippocampal

corresponding wild-type mice to excitotoxic cell death.

and neocortical principal neurons [16]. The primary conse-

Modifications other than downregulation of GluR2

quence is the onset of spontaneous and recurrent seizures

expression can increase excitotoxicity. The presence of the

[16]. The mice develop recurrent seizures and die within

GluR2-flip splice-variant subunit in heteromeric AMPAR

the first three weeks of life, with cell loss in the hippocam-

leads to a larger current flow through these channels

pus. In these animals, unedited GluR2 may contribute to

[42,89] Transgenic mice that over-express GluR2-flip

21

the formation of a greater number of Ca

-permeable

show enhanced susceptibility to excitotoxic

glutamate-AMPARs than in the GluR2 knockout mice (see below).

mediated damage after permanent middle cerebral artery

Transgenic mice with targeted disruption of the GluR2

occlusion, and glutamate excitotoxicity is increased

rela-gene (‘GluR2 knock-out mice’) differ considerably from

tive to that of wild-type in neurons cultured from the

GluR2Q / R editing deficient mice; the knock-out mice are

transgenic mice [72]. Excitotoxicity may be caused by

21 21

viable and fertile. The knock-out mice exhibit a nine-fold

increased Ca

influx through voltage-gated Ca

chan-21

increase in kainate-elicited Ca

-influx into individual

nels and through NMDAR channels [26,73,84] following

CA1 pyramidal neurons and increased inward rectification

increased depolarization mediated by AMPARs containing

of kainate-elicited responses and of the AMPAR com-

primarily the flip isoform of GluR2 [42,89].

ponent of EPSCs [59]. The passive membrane properties at

GluR2 knockdown as a technique offers the advantage

the resting potential are unchanged, except that input

of examining the effects of GluR2 suppression in an

resistance is increased, perhaps due to reduced cell size.

animal that has developed under normal conditions.

In-EPSCs are little changed in amplitude and the decay rate is

jection of antisense oligonucleotides directly into the brain

unchanged (unlike the EPSCs in post ischemic gerbil

of gerbils and rats has been used to demonstrate a probable

[146]. The AMPAR-mediated component of the EPSCs is

causal relationship between downregulation of GluR2

reduced relative to the NMDAR-mediated component,

expression and delayed neuronal cell death [100]. This

possibly due to reduction in AMPAR density as a result of

study demonstrates that knockdown of GluR2 by

intraven-inefficient receptor assembly. LTP in GluR2 knock-out

tricular injection of specific antisense oligonucleotides

animals is increased and has a substantial NMDAR-in-

leads to death of CA1 and CA3 neurons. Scrambled

dependent component. These data strongly suggest that

antisense administered under the same conditions is

with-21 21

LTP can be mediated by Ca

influx through Ca

-

out effect. Since the induced neurotoxicity is blocked by

permeable AMPARs (see also [46]). The GluR2 knockout

1-naphthyl acetyl spermine, the cause of death is likely to

21 21

(14)

neuro-nal damage) leads to greater cell death than is observed for

References

antisense administration to control animals.

[1] H. Abe, M. Watanabe, T. Yamakuni, R. Kuwano, Y. Takahashi, H.

Given the prominent role of GluR2 in normal

physi-Kondo, Localization of gene expression of calbindin in the brain of

ology, brain development, and excitotoxicity, an important

adult rats, Neurosci. Lett. 138 (1992) 211–215.

future direction would be to apply spatial and temporal

[2] S. Akbarian, M.A. Smith, E.G. Jones, Editing for an AMPA receptor

restrictions to the regulation of GluR2 expression as, for

subunit RNA in prefrontal cortex and striatum in Alzheimer’s

example, by conditional knockout and / or knockin ap-

disease, Huntington’s disease and schizophrenia, Brain Res. 699

(1995) 297–304.

proaches. Such studies would be expected to aid in our

[3] E.M. Aronica, J.A. Gorter, S. Grooms, J.A. Kessler, M.V. Bennett,

understanding of the role of the GluR2 subunit during

R.S. Zukin, D.M. Rosenbaum, Aurintricarboxylic acid prevents

synaptogenesis and formation of neuronal circuitry and in

GLUR2 mRNA down-regulation and delayed neurodegeneration in

the neurodegeneration associated with stroke and epilepsy.

hippocampal CA1 neurons of gerbil after global ischemia, Proc.

Natl. Acad. Sci. USA 95 (1998) 7115–7120.

[4] C.H. Bailey, P. Montarolo, M. Chen, E.R. Kandel, S. Schacher, Inhibitors of protein and RNA synthesis block structural changes that accompany long-term heterosynaptic plasticity in Aplysia,

6. Concluding remarks

Neuron 9 (1992) 749–758.

[5] O. Bar-Peled, R.J. O’Brien, J.H. Morrison, J.D. Rothstein, Cultured

This article reviewed the regulation of AMPAR expres-

motor neurons possess calcium-permeable AMPA / kainate receptors,

sion at CA1 synapses, with particular emphasis on RNA

Neuroreport 10 (1999) 855–859.

transcription, editing and splicing, translational regulation,

[6] B.L. Bass, RNA editing and hypermutation by adenosine deamina-tion, Trends. Biochem. Sci. 22 (7) (1997) 157–162, Published

receptor targeting and trafficking, cell surface expression

erratum appears in Trends. Biochem. Sci. 22 (7) (1997) 278.

and anchoring. In addition, this article reviewed evidence

[7] G.J. Bassell, Y. Oleynikov, R.H. Singer, The travels of mRNAs

that severe neurological insults such as global ischemia

through all cells large and small [see comments], FASEB J. 13

and limbic seizures trigger a ‘molecular switch’ that shuts

(1999) 447–454.

off GluR2 subunit expression in neurons destined to die. In

[8] R. Benne, RNA editing. The long and the short of it [news;

21 comment], Nature 380 (1996) 391–392.

neurons that normally express Ca

-impermeable

chan-21 [9] M.V.L. Bennett, D.E. Pellegrini-Giampietro, J.A. Gorter, E. Aronica,

nels, Ca

entry via GluR2-lacking AMPARs could

con-J.A. Connor, R.S. Zukin, The GluR2 hypothesis: Ca(11

)-perme-tribute significantly to delayed cell death. In addition to

able AMPA receptors in delayed neurodegeneration, Cold Spring

21

their role in neurological disorders, Ca

-permeable AM-

Harb. Symp. Quant. Biol. 61 (1996) 373–384.

PARs are thought to serve a number of physiological

[10] F. Berton, C. Iborra, J.A. Boudier, M.J. Seagar, B. Marqueze, Developmental regulation of synaptotagmin I, II, III, and IV mRNAs

functions, including strengthening of synaptic transmission

in the rat CNS, J. Neurosci. 17 (1997) 1206–1216.

in certain spinal neurons [46], induction of long-term

[11] M. Blaschke, B.U. Keller, R. Rivosecchi, M. Hollmann, S.

potentiation as observed in the GluR2 knockout mouse

Heinemann, A. Konnerth, A single amino acid determines the

21 1

[59], and possibly activation of Ca

-dependent K

chan-

subunit-specific spider toxin block of

alpha-amino-3-hydroxy-5-nels as well as inactivation of NMDARs [22,60]. Increased

methylisoxazole-4-propionate / kainate receptor channels, Proc. Natl.

21 Acad. Sci. USA 90 (1993) 6528–6532.

Ca

permeability through AMPARs may also be involved

[12] P. Bochet, E. Audinat, B. Lambolez, F. Crepel, J. Rossier, M. Iino,

in the development of kindled seizures [114]. The

underly-K. Tsuzuki, S. Ozawa, Subunit composition at the single-cell level

21

ing mechanism by which AMPAR Ca

permeability

explains functional properties of a glutamate-gated channel, Neuron

increases under physiological or pathological conditions

12 (1994) 383–388.

[13] D. Bowie, M.L. Mayer, Inward rectification of both AMPA and

reported to date involves a suppression of GluR2 gene

kainate subtype glutamate receptors generated by

polyamine-me-expression, rather than a reduction in Q / R editing

diated ion channel block, Neuron 15 (1995) 453–462.

[2,21,62,106]. Reduction in GluR2 expression has also

[14] J. Brillman, Central nervous system complications in coronary artery

been observed in other vulnerable neuronal populations

bypass graft surgery, Neurol. Clin. 11 (1993) 475–495.

21

including cerebellar Purkinje and granule cells in mutant

[15] J.R. Brorson, P.A. Manzolillo, R.J. Miller, Ca entry via AMPA / KA receptors and excitotoxicity in cultured cerebellar Purkinje cells,

spastic rats [79], pyramidal cells of the parahippocampal

J. Neurosci. 14 (1994) 187–197.

gyrus in schizophrenia [33], and spinal motor neurons in

[16] R. Brusa, F. Zimmermann, D.S. Koh, D. Feldmeyer, P. Gass, P.H.

amyotrophic lateral sclerosis [153] (Table 1). These

exam-Seeburg, R. Sprengel, Early-onset epilepsy and postnatal lethality

ples indicate the more general applicability of the GluR2

associated with an editing-deficient GluR-B allele in mice, Science

hypothesis to neuropsychiatric diseases and disorders

270 (1995) 1677–1680.

[17] W. Brysch, O.D. Creutzfeldt, K. Luno, R. Schlingensiepen, K.H.

involving glutamate-induced cell death.

Schlingensiepen, Regional and temporal expression of sodium channel messenger RNAs in the rat brain during development, Exp. Brain Res. 86 (1991) 562–567.

[18] A.M. Buchan, H. Li, S. Cho, W.A. Pulsinelli, Blockade of the

Acknowledgements

AMPA receptor prevents CA1 hippocampal injury following severe but transient forebrain ischemia in adult rats, Neurosci. Lett. 132

This work was supported by NIH grants no. NS 20752

(1991) 255–258.

and NS 31282 (to R.S.Z.). M.V.L.B. is the Sylvia and

[19] K.E. Burgin, M.N. Waxham, S. Rickling, S.A. Westgate, W.C.

(15)

calmodulin-dependent protein kinase in developing rat brain, J. Bennett, S.L. Moshe, R.S. Zukin, Kainate-induced status epilepticus alters glutamate and GABAA receptor gene expression in adult rat Neurosci. 10 (1990) 1788–1798.

hippocampus: an in situ hybridization study, J. Neurosci. 14 (1994) [20] N. Burnashev, H. Monyer, P.H. Seeburg, B. Sakmann, Divalent ion

2697–2707. permeability of AMPA receptor channels is dominated by the edited

[39] L.K. Friedman, J. Veliskova, GluR2 hippocampal knockdown re-form of a single subunit, Neuron 8 (1992) 189–198.

veals developmental regulation of epileptogenicity and neurodegene-[21] N. Burnashev, R. Schoepfer, H. Monyer, J.P. Ruppersberg, W.

ration, Brain Res. Mol. Brain Res. 61 (1998) 224–231. Gunther, P.H. Seeburg, B. Sakmann, Control by asparagine residues

[40] C.C. Garner, R.P. Tucker, A. Matus, Selective localization of of calcium permeability and magnesium blockade in the NMDA

messenger RNA for cytoskeletal protein MAP2 in dendrites, Nature receptor, Science 257 (1992) 1415–1419.

336 (1988) 674–677. [22] N. Burnashev, A. Villarroel, B. Sakmann, Dimensions and ion

[41] A.H. Gazzaley, D.L. Benson, G.W. Huntley, J.H. Morrison, Dif-selectivity of recombinant AMPA and kainate receptor channels and

ferential subcellular regulation of NMDAR1 protein and mRNA in their dependence on Q / R site residues, J. Physiol. (Lond.) 496

dendrites of dentate gyrus granule cells after perforant path transec-(1996) 165–173.

tion, J. Neurosci. 17 (1997) 2006–2017. [23] C.M. Burns, H. Chu, S.M. Rueter, L.K. Hutchinson, H. Canton, E.

[42] J.R. Geiger, T. Melcher, D.S. Koh, B. Sakmann, P.H. Seeburg, P. Sanders-Bush, R.B. Emeson, Regulation of serotonin-2C receptor

Jonas, H. Monyer, Relative abundance of subunit mRNAs de-G-protein coupling by RNA editing, Nature 387 (1997) 303–308,

termines gating and Ca21 permeability of AMPA receptors in See comments.

principal neurons and interneurons in rat CNS, Neuron 15 (1995)

21

[24] S.G. Carriedo, H.Z. Yin, S.L. Sensi, J.H. Weiss, Rapid Ca entry 193–204.

21

through Ca -permeable AMPA / Kainate channels triggers marked [43] A. Gerber, M.A. O’Connell, W. Keller, Two forms of human intracellular Ca21rises and consequent oxygen radical production, double-stranded RNA-specific editase 1 (hRED1) generated by the J. Neurosci. 18 (1998) 7727–7738. insertion of an Alu cassette, RNA 3 (1997) 453–463.

[25] J. Chen, R.L. Zhu, M. Nakayama, K. Kawaguchi, K. Jin, R.A. [44] J.A. Gorter, J.J. Petrozzino, E.M. Aronica, D.M. Rosenbaum, T. Stetler, R.P. Simon, S.H. Graham, Expression of the apoptosis- Opitz, M.V. Bennett, J.A. Connor, R.S. Zukin, Global ischemia effector gene, Bax, is up-regulated in vulnerable hippocampal CA1 induces downregulation of Glur2 mRNA and increases AMPA neurons following global ischemia, J. Neurochem. 67 (1996) 64–71. receptor-mediated Ca21 influx in hippocampal CA1 neurons of [26] D.W. Choi, Calcium-mediated neurotoxicity: relationship to specific gerbil, J. Neurosci. 17 (1997) 6179–6188.

channel types and role in ischemic damage, Trends Neurosci. 11 [45] S.Y. Grooms, T. Opitz, M.V. Bennett, R.S. Zukin, Status epilepticus

(1988) 465–469. decreases glutamate receptor 2 mRNA and protein expression in

[27] D.W. Choi, Cerebral hypoxia: some new approaches and unanswered hippocampal pyramidal cells before neuronal death, Proc. Natl. questions, J. Neurosci. 10 (1990) 2493–2501. Acad. Sci. USA 97 (2000) 3631–3636.

[28] D.W. Choi, Excitotoxic cell death, J. Neurobiol. 23 (1992) 1261– [46] J.G. Gu, C. Albuquerque, C.J. Lee, A.B. MacDermott, Synaptic

21

1276. strengthening through activation of Ca -permeable AMPA

recep-[29] D.W. Choi, Calcium: still center-stage in hypoxic-ischemic neuronal tors, Nature 381 (1996) 793–796.

death, Trends Neurosci. 18 (1995) 58–60. [47] Y. Hayashi, S.H. Shi, J.A. Esteban, A. Piccini, J.C. Poncer, R. [30] J.A. Chong, J. Tapia-Ramirez, S. Kim, J.J. Toledo-Aral, Y. Zheng, Malinow, Driving AMPA receptors into synapses by LTP and M.C. Boutros, Y.M. Altshuller, M.A. Frohman, S.D. Kraner, G. CaMKII: requirement for GluR1 and PDZ domain interaction, Mandel, REST: a mammalian silencer protein that restricts sodium Science 287 (2000) 2262–2267.

channel gene expression to neurons, Cell 80 (1995) 949–957. [48] S. Herlitze, M. Raditsch, J.P. Ruppersberg, W. Jahn, H. Monyer, R. [31] P. Crino, K. Khodakhah, K. Becker, S. Ginsberg, S. Hemby, J. Schoepfer, V. Witzemann, Argiotoxin detects molecular differences

Eberwine, Presence and phosphorylation of transcription factors in in AMPA receptor channels, Neuron 10 (1993) 1131–1140. developing dendrites, Proc. Natl. Acad. Sci. USA 95 (1998) 2313– [49] C. Heurteaux, I. Lauritzen, C. Widmann, M. Lazdunski, Essential

1

2318. role of adenosine, adenosine A1 receptors, and ATP-sensitive K

[32] S.D. Donevan, M.A. Rogawski, Intracellular polyamines mediate channels in cerebral ischemic preconditioning, Proc. Natl. Acad. Sci.

21

inward rectification of Ca -permeable alpha-amino-3-hydroxy-5- USA 92 (1995) 4666–4670.

methyl-4-isoxazolepropionic acid receptors, Proc. Natl. Acad. Sci. [50] M. Higuchi, F.N. Single, M. Kohler, B. Sommer, R. Sprengel, P.H.

USA 92 (1995) 9298–9302. Seeburg, RNA editing of AMPA receptor subunit GluR-B: a

base-[33] S.L. Eastwood, P.J. Harrison, Decreased synaptophysin in the medial paired intron-exon structure determines position and efficiency, Cell temporal lobe in schizophrenia demonstrated using immuno- 75 (1993) 1361–1370.

autoradiography, Neuroscience 69 (1995) 339–343. [51] T. Himi, T. Okazaki, H. Wang, T.H. McNeill, N. Mori, Differential [34] S.L. Eastwood, B. McDonald, P.W. Burnet, J.P. Beckwith, R.W. localization of SCG10 and p19 / stathmin messenger RNAs in adult Kerwin, P.J. Harrison, Decreased expression of mRNAs encoding rat brain indicates distinct roles for these growth-associated proteins, non-NMDA glutamate receptors GluR1 and GluR2 in medial Neuroscience 60 (1994) 907–926.

21

temporal lobe neurons in schizophrenia, Brain Res. Mol. Brain Res. [52] M. Hollmann, M. Hartley, S. Heinemann, Ca permeability of

29 (1995) 211–223. KA-AMPA — gated glutamate receptor channels depends on

[35] L.W. Fitzgerald, A.Y. Deutch, G. Gasic, S.F. Heinemann, E.J. subunit composition, Science 252 (1991) 851–853.

Nestler, Regulation of cortical and subcortical glutamate receptor [53] M. Hollmann, S. Heinemann, Cloned glutamate receptors, Annu. subunit expression by antipsychotic drugs, J. Neurosci. 15 (1995) Rev. Neurosci. 17 (1994) 31–108.

2453–2461. [54] R.M. Hong, H. Mori, T. Fukui, Y. Moriyama, M. Futai, A.

[36] L.W. Fitzgerald, J. Ortiz, A.G. Hamedani, E.J. Nestler, Drugs of Yamamoto, Y. Tashiro, M. Tagaya, Association of

N-abuse and stress increase the expression of GluR1 and NMDAR1 ethylmaleimide-sensitive factor with synaptic vesicles, FEBS Lett. glutamate receptor subunits in the rat ventral tegmental area: 350 (1994) 253–257.

common adaptations among cross-sensitizing agents, J. Neurosci. 16 [55] B.R. Hu, M. Park, M.E. Martone, W.H. Fischer, M.H. Ellisman, J.A.

(1996) 274–282. Zivin, Assembly of proteins to postsynaptic densities after transient

[37] L.K. Friedman, A.R. Koudinov, Unilateral GluR2(B) hippocampal cerebral ischemia, J. Neurosci. 18 (1998) 625–633.

(16)

[57] R.I. Hume, R. Dingledine, S.F. Heinemann, Identification of a site in Kuner, H. Monyer, M. Higuchi, A. Bach, P.H. Seeburg, Control of glutamate receptor subunits that controls calcium permeability, kinetic properties of AMPA receptor channels by nuclear RNA

Science 253 (1991) 1028–1031. editing, Science 266 (1994) 1709–1713.

[58] R.J. Jackson, M. Wickens, Translational controls impinging on the [76] H. Lomeli, J. Mosbacher, T. Melcher, T. Hoger, J.R. Geiger, T. 59-untranslated region and initiation factor proteins, Curr. Opin. Kuner, H. Monyer, M. Higuchi, A. Bach, P.H. Seeburg, Control of

Genet. Dev. 7 (1997) 233–241. kinetic properties of AMPA receptor channels by nuclear RNA

editing, Science 266 (1994) 1709–1713. [59] Z. Jia, N. Agopyan, P. Miu, Z. Xiong, J. Henderson, R. Gerlai, F.A.

Taverna, A. Velumian, J. MacDonald, P. Carlen, W. Abramow- [77] Y.M. Lu, H.Z. Yin, J. Chiang, J.H. Weiss, Ca(21)-permeable

21 Newerly, J. Roder, Enhanced LTP in mice deficient in the AMPA AMPA / kainate and NMDA channels: high rate of Ca influx receptor GluR2, Neuron 17 (1996) 945–956. underlies potent induction of injury, J. Neurosci. 16 (1996) 5457–

5465. [60] P. Jonas, C. Racca, B. Sakmann, P.H. Seeburg, H. Monyer,

21

Differences in Ca permeability of AMPA-type glutamate receptor [78] S. Maas, T. Melcher, P.H. Seeburg, Mammalian RNA-dependent channels in neocortical neurons caused by differential GluR-B deaminases and edited mRNAs, Curr. Opin. Cell Biol. 9 (1997) subunit expression, Neuron 12 (1994) 1281–1289. 343–349.

[61] F.S. Jones, R. Meech, Knockout of REST / NRSF shows that the [79] J.E. Margulies, R.W. Cohen, M.S. Levine, J.B. Watson, Decreased protein is a potent repressor of neuronally expressed genes in GluR2(B) receptor subunit mRNA expression in cerebellar neurons non-neural tissues, Bioessays 21 (1999) 372–376. at risk for degeneration, Dev. Neurosci. 15 (1993) 110–120. [62] W. Kamphuis, F.E. de Leeuw, D.S.F. Lopes, Ischaemia does not [80] K.C. Martin, A. Casadio, H. Zhu, J.C. Rose, M. Chen, C.H. Bailey,

alter the editing status at the Q / R site of glutamate receptor-A, -B, E.R. Kandel, Synapse-specific, long-term facilitation of aplysia -5 and -6 subunit mRNA, Neuroreport 6 (1995) 1133–1136. sensory to motor synapses: a function for local protein synthesis in

memory storage, Cell 91 (1997) 927–938. [63] U. Kim, Y. Wang, T. Sanford, Y. Zeng, K. Nishikura, Molecular

cloning of cDNA for double-stranded RNA adenosine deaminase, a [81] J.W. McDonald, M.V. Johnston, Physiological and pathophysiologi-candidate enzyme for nuclear RNA editing, Proc. Natl. Acad. Sci. cal roles of excitatory amino acids during central nervous system

USA 91 (1994) 11457–11461. development, Brain Res. Brain Res. Rev. 15 (1990) 41–70.

[64] T. Kirino, Delayed neuronal death in the gerbil hippocampus [82] T. Melcher, S. Maas, A. Herb, R. Sprengel, M. Higuchi, P.H. following ischemia, Brain Res. 239 (1982) 57–69. Seeburg, RED2, a brain-specific member of the RNA-specific adenosine deaminase family, J. Biol. Chem. 271 (1996) 31795– [65] R. Kleiman, G. Banker, O. Steward, Differential subcellular

locali-31798. zation of particular mRNAs in hippocampal neurons in culture,

Neuron 5 (1990) 821–830. [83] T. Melcher, S. Maas, M. Higuchi, W. Keller, P.H. Seeburg, Editing [66] J.Y. Koh, S.W. Suh, B.J. Gwag, Y.Y. He, C.Y. Hsu, D.W. Choi, The of alpha-amino-3-hydroxy-5-methylisoxazole-4-propionic acid re-role of zinc in selective neuronal death after transient global cerebral ceptor GluR-B pre-mRNA in vitro reveals site-selective adenosine ischemia, Science 272 (1996) 1013–1016. to inosine conversion, J. Biol. Chem. 270 (1995) 8566–8570. [67] M. Kohler, H.C. Kornau, P.H. Seeburg, The organization of the gene [84] B. Meldrum, J. Garthwaite, Excitatory amino acid neurotoxicity and

for the functionally dominant alpha-amino-3-hydroxy-5-methylisox- neurodegenerative disease, Trends Pharmacol. Sci. 11 (1990) 379– azole-4-propionic acid receptor subunit GluR-B, J. Biol. Chem. 269 387.

(1994) 17367–17370. [85] L. Mittaz, H.S. Scott, C. Rossier, P.H. Seeburg, M. Higuchi, S.E. [68] M. Kondo, R. Sumino, H. Okado, Combinations of AMPA receptor Antonarakis, Cloning of a human RNA editing deaminase subunit expression in individual cortical neurons correlate with (ADARB1) of glutamate receptors that maps to chromosome expression of specific calcium-binding proteins, J. Neurosci. 17 21q22.3, Genomics 41 (1997) 210–217.

(1997) 1570–1581. [86] K. Miyashiro, M. Dichter, J. Eberwine, On the nature and

differen-[69] F. Lai, C.X. Chen, K.C. Carter, K. Nishikura, Editing of glutamate tial distribution of mRNAs in hippocampal neurites: implications for receptor B subunit ion channel RNAs by four alternatively spliced neuronal functioning, Proc. Natl. Acad. Sci. USA 91 (1994) 10800– DRADA2 double-stranded RNA adenosine deaminases, Mol. Cell 10804, See comments.

Biol. 17 (1997) 2413–2424. [87] H. Monyer, N. Burnashev, D.J. Laurie, B. Sakmann, P.H. Seeburg, [70] A.S. Landsend, M. Amiry-Moghaddam, A. Matsubara, L. Developmental and regional expression in the rat brain and func-Bergersen, S. Usami, R.J. Wenthold, O.P. Ottersen, Differential tional properties of four NMDA receptors, Neuron 12 (1994) 529– localization of delta glutamate receptors in the rat cerebellum: 540.

coexpression with AMPA receptors in parallel fiber-spine synapses [88] H. Monyer, P.H. Seeburg, W. Wisden, Glutamate-operated channels: and absence from climbing fiber-spine synapses, J. Neurosci. 17 developmentally early and mature forms arise by alternative

splic-(1997) 834–842. ing, Neuron 6 (1991) 799–810.

[71] W. Lason, J. Turchan, B. Przewlocka, D. Labuz, J. Mika, R. [89] J. Mosbacher, R. Schoepfer, H. Monyer, N. Burnashev, P.H. Przewlocki, Seizure-related changes in the glutamate R2 and R5 Seeburg, J.P. Ruppersberg, A molecular determinant for submil-receptor genes expression in the rat hippocampal formation, J. lisecond desensitization in glutamate receptors, Science 266 (1994)

Neural Transm. 104 (1997) 125–133. 1059–1062.

[72] D. Le, S. Das, Y.F. Wang, T. Yoshizawa, Y.F. Sasaki, M. Takasu, A. [90] I.A. Muslimov, E. Santi, P. Homel, S. Perini, D. Higgins, H. Tiedge, Nemes, M. Mendelsohn, P. Dikkes, S.A. Lipton, N. Nakanishi, RNA transport in dendrites: a cis-acting targeting element is Enhanced neuronal death from focal ischemia in AMPA-receptor contained within neuronal BC1 RNA, J. Neurosci. 17 (1997) 4722– transgenic mice, Brain Res. Mol. Brain Res. 52 (1997) 235–241. 4733.

[73] S.A. Lipton, P.A. Rosenberg, Excitatory amino acids as a final [91] S.J. Myers, R. Dingledine, K. Borges, Genetic regulation of common pathway for neurologic disorders [see comments], N. Engl. glutamate receptor ion channels, Annu. Rev. Pharmacol. Toxicol. 39

J. Med. 330 (1994) 613–622. (1999) 221–241.

[74] Y. Liu, C.X. George, J.B. Patterson, C.E. Samuel, Functionally [92] S.J. Myers, J. Peters, Y. Huang, M.B. Comer, F. Barthel, R. distinct double-stranded RNA-binding domains associated with Dingledine, Transcriptional regulation of the GluR2 gene: neural-alternative splice site variants of the interferon-inducible double- specific expression, multiple promoters, and regulatory elements, J. stranded RNA-specific adenosine deaminase, J. Biol. Chem. 272 Neurosci. 18 (1998) 6723–6739.

(1997) 4419–4428. [93] S.M. Nair, T.R. Werkman, J. Craig, R. Finnell, M. Joels, J.H.

Gambar

Fig. 1. Proposed secondary structure of GluR subunits depicting critical sites conferring functional diversity on AMPA receptors
Fig. 2. Models of neuron restrictive silencer element (NRSE), RE-1-silencing trancription factor (REST), REST function and GluR2 promoter
Fig. 3. Model of NSF modulation of AMPA receptor function. (A) Regulation of receptor insertion
Fig. 5. Global ischemia induces downregulation of GluR2, but notGluR1, immunolabeling in CA1 pyramidal neurons
+5

Referensi

Dokumen terkait

Dalam skripsi yang berjudul “ Implementasi Pendekatan Taktis dalam Pembelajaran Aktivitas Penguasaan Bola pada Permainan Bolabasket ” ini mengungkapkan mengenai

mengendalikan pelaksanaan program dan kegiatan serta penetapan kebijakan teknis pada Dinas yang meliputi Sekretariat, Bidang Perdagangan, Bidang Pasar, Bidang Tertib Niaga,

Faktanya adalah masyarakat Kobe Kulo belum memahami keberadaan taman nasional baik secara fisik, batasan maupun manfaatnya, sedangkan masyarakat Tayawi memiliki persepsi

Penelitian ini bertujuan untuk mengetahui potensi cadangan karbon yang tersimpan dalam biomassa tegakan hutan lindung Long Ketrok, yaitu hutan lindung yang dikelola oleh masyarakat

”Apakah pendekatan taktis dapat meningkatkan hasil penguasaan bola dalam permainan bolabasket pada pembelajaran Penjas di SMK Cipta Skill Bandung?”.

[r]

Karya tulis ilmiah (KTI) merupakan laporan dari kegiatan nyata yang dilakukan para guru di kelasnya dalam upaya meningkatkan mutu pembelajaran (ini tentunya

Kepala Satuan Polisi Pamong Praja PEMERINTAH KOTA