You are on page 1of 15

Cell. Mol. Life Sci.

DOI 10.1007/s00018-013-1484-0 Cellular and Molecular Life Sciences


Review

Changes in the expression of the glutamate transporter


EAAT3/EAAC1 in health and disease
Massimiliano G. Bianchi · Donatella Bardelli ·
Martina Chiu · Ovidio Bussolati 

Received: 9 August 2013 / Revised: 17 September 2013 / Accepted: 19 September 2013


© Springer Basel 2013

Abstract Excitatory amino acid transporters (EAATs) pathological conditions, such as hypoxia/ischemia, mul-
are high-affinity Na+-dependent carriers of major impor- tiple sclerosis, schizophrenia, and epilepsy. This review
tance in maintaining glutamate homeostasis in the cen- summarizes these results and provides an overall picture
tral nervous system. EAAT3, the human counterpart of of changes in EAAT3/EAAC1 expression in health and
the rodent excitatory amino acid carrier 1 (EAAC1), is disease.
encoded by the SLC1A1 gene. EAAT3/EAAC1 is ubiqui-
tously expressed in the brain, mostly in neurons but also in Keywords EAAC1 · EAAT3 · Glutamate · Oxidative
other cell types, such as oligodendrocyte precursors. While stress · Glutathione · Retinoids
most of the glutamate released in the synapses is taken up
by the “glial-type” EAATs, EAAT2 (GLT-1 in rodents) and
EAAT1 (GLAST), the functional role of EAAT3/EAAC1 Introduction
is related to the subtle regulation of glutamatergic trans-
mission. Moreover, because it can also transport cysteine, EAAT3, the human counterpart of the EAAC1 rodent trans-
EAAT3/EAAC1 is believed to be important for the syn- porter, is a member of the EAAT family of high-affinity,
thesis of intracellular glutathione and subsequent protec- sodium-dependent glutamate carriers encoded by genes
tion from oxidative stress. In contrast to other EAATs, of the SLC1 family [1–3]. EAAT3/EAAC1 is coded by
EAAT3/EAAC1 is mostly intracellular, and several mecha- SLC1A1/Slc1a1 and was the first sodium-dependent mam-
nisms have been described for the rapid regulation of the malian amino acid transporter to be cloned [4]. When
membrane trafficking of the transporter. Moreover, the studying the activity of EAAT3/EAAC1 in cultured human
carrier interacts with several proteins, and this interaction fibroblasts before its cloning, early researchers demon-
modulates transport activity. Much less is known about strated that the transporter can accept the D isomer of
the slow regulatory mechanisms acting on the expression aspartate but not the D isomer of glutamate [5]. This stere-
of the transporter, although several recent reports have oselective anomaly has been consistently confirmed for all
identified changes in EAAT3/EAAC1 protein level and the transporters for anionic amino acids of the SLC1 family
activity related to modulation of its expression at the gene and is one of the functional hallmarks of EAAT carriers.
level. Moreover, EAAT3/EAAC1 expression is altered in Like other EAAT carriers, EAAT3/EAAC1 uses the
transmembrane gradients of Na+, K+, and H+ electrochem-
ical potentials as the driving force to accumulate glutamate
M. G. Bianchi · D. Bardelli · M. Chiu · O. Bussolati (*) 
from the extracellular space [6, 7]. Glutamate is co-trans-
Unit of General Pathology, Department of Biomedical,
Biotechnological and Translational Sciences (SBiBiT), ported with 3 Na+ and 1 H+, while 1 K+ is extruded in the
University of Parma, Via Volturno 39, 43125 Parma, Italy same cycle, ensuring the electrogenicity of the transport
e-mail: ovidio.bussolati@unipr.it cycle and the formation of a high transmembrane gradient
of the transported substrates.
M. G. Bianchi 
Unit of Occupational Medicine, Department of Clinical The expected molecular weight of the transporter is
and Experimental Medicine, University of Parma, Parma, Italy 64 kDa, but electrophoretic migration yields higher values,

13
M. G. Bianchi et al.

ascribed to a high degree of glycosylation. Moreover, West- in Xenopus oocytes injected with the transporter cDNA,
ern-blot experiments have demonstrated high-molecular- including phosphatidylinositol-3 (PI3)-kinase, the phosph-
weight bands, referred to as trimers of the transporter. In oinositide-dependent kinase PDK1, the serum- and gluco-
fact, trimers are thought to constitute the functional unit of corticoid-inducible kinase SGK1, and the phosphatidylino-
EAAT3/EAAC1 in situ [8, 9]. Although experimental data sitol-3-phosphate-5-kinase PIKfyve [35]. Interestingly, the
on the carrier structure are incomplete, EAATs have 8–10 gene encoding PIKfyve is associated with schizophrenia
transmembrane domains, with glycosylation sites in the and the mutant form expressed in patients seems to exert
extracellular loop between TM3 and TM4 and several puta- a dominant-negative effect on EAAT3 trafficking in Xeno-
tive intracellular consensus phosphorylation sites [10]. pus oocytes [36]. In the same experimental model, mTOR
EAAT3/EAAC1 is expressed not only in the central increases EAAT3/EAAC1 activity [37], which, instead, is
nervous system (CNS) but also in Schwann cells [11]. decreased by the mTOR inhibitor AMPK [38].
Moreover, the carrier is also expressed in the intestine and These effects may be due to a direct interaction between
kidney, where it accounts for the absorption/reabsorption EAAT3/EAAC1 and the kinase or, more often, to changes
of dicarboxylic amino acids, and in many other tissues. in the interaction between the transporter and several intra-
Defective expression of functional EAAT3/EAAC1 results cellular proteins, such as the endoplasmic reticulum pro-
in dicarboxylic aminoaciduria (OMIM222730) [12]. In teins addicsin/glutamate transporter-associated protein
the CNS, of the five EAATs characterized so far, EAAT4 3-18 (GTRAP3-18, [39]) and reticulon (RTN2B, [40]),
[13] and EAAT5 [14] are highly expressed in cerebellar which are believed to play opposite regulatory roles in
Purkinje cells and in retinal photoreceptors and bipolar the membrane-targeting of the transporter [41, 42]. Inter-
cells, respectively. In contrast, EAAT1 and EAAT2 pre- estingly, the gene for GTRAP3-18, which is a negative
dominately have a glial distribution, while EAAT3/EAAC1 modulator of EAAT3/EAAC1 activity, maps to human
is highly expressed in glutamatergic and GABAergic neu- chromosome 3p14, a susceptibility locus for cancer and
rons [15], especially in somata and dendrites (with par- epilepsy [43]. GTRAP3-18 is negatively regulated by ADP-
ticular abundance in the neck of dendritic spines [16]) but ribosylation factor-like 6-interacting protein 1 (Ar16ip1),
not in synaptic terminals of axons or spine heads [16–18]. which causes a decreased interaction between GTRAP3-
Other neuronal populations express EAAT3/EAAC1, such 18 and EAAT3/EAAC1 in neurons [44]. Moreover,
as dopaminergic neurons; these neurons are very sensi- EAAT3/EAAC1 trafficking involves several steps depend-
tive to carrier dysfunction [19, 20] or inhibition [21]. ent on Rab11 [45], SNAP-23 (a SNARE protein) [46], and
EAAT3/EAAC1 is also expressed in cells of glial origin, caveolin-1 [47], although evidence for a direct interac-
such as in subsets of oligodendrocytes [22], in immature tion between the transporter and these proteins is lacking.
oligodendrocytes [23], and in oligodendrocyte progenitor The δ-opioid receptor is a G-protein that also modulates
cells [24]. An isolated report describes EAAT3/EAAC1 EAAT3/EAAC1 activity [48]. EAAT3/EAAC1 also inter-
expression also in microglial cells after traumatic brain acts with the cytoskeletal protein α-adducin [49], which is
injury [25]. Mature astrocytes are generally believed to be particularly abundant in dendritic spines [50], although, in
EAAT3/EAAC1-negative, but it should be noted that astro- this case, the functional implications of the interaction have
glial tumor cells, such as rat C6 glioma cells and several not been defined.
human glioma cell models, express the transporter [26]. At Compared to these multiple sites of regulation at the pro-
variance with other EAAT transporters [27, 28], the expres- tein level, the characterization of mechanisms modulating
sion of EAAT3/EAAC1 is mainly intracellular [22, 29, EAAT3/EAAC1 synthesis at the transcriptional level has
30]; only a minor portion of the carriers are targeted to the been much slower. This review aims to recount the infor-
plasma membrane under control conditions, while most are mation available on this subject as well as on pathologi-
associated with cytoplasmic vesicles [15, 16]. cal situations in which the expression of EAAT3/EAAC1
This peculiar distribution has relevant consequences for is altered. Before examining the regulatory mechanisms,
the regulation of transporter activity. In cells of both nerv- a brief summary on the physiological role of EAAT3 is
ous and non-nervous origin, EAAT3/EAAC1 activity is discussed.
modulated by post-translational mechanisms that rapidly
modify its trafficking and abundance on the plasma mem-
brane. EAAT3/EAAC1 abundance on the plasma mem- EAAT3/EAAC1 functions in physiology and pathology
brane is readily enhanced upon PKC activation [31] or
stimulation with PDGF [32]. Several PKC enzymes may In general, the activity of EAAT transporters is consid-
be involved in these regulatory effects at different levels ered the main mechanism responsible for the termination
[31–34]. In addition, other kinases have been implicated in of glutamatergic transmission. As such, EAAT transport-
the regulation of EAAT3/EAAC1 trafficking and activity ers are believed to be important factors for the prevention

13
Changes in the expression of the glutamate transporter

of excitotoxic damage, i.e., neuronal loss due to excessive of Huntington disease [63]. The mouse knocked-out for
influx of calcium mediated by the overstimulation of glu- EAAT3/EAAC1, which presents a decreased glutathione
tamate receptors of the NMDA and AMPA/kainate classes. content in neurons and early onset brain aging, exhibits
However, the roles of the three most important EAATs, significant biochemical and functional improvements if
EAAT1, EAAT2, and EAAT3/EAAC1, are thought to be treated with the antioxidant N-acetyl-cysteine [64].
diverse [51]. This is well exemplified by experiments with The high expression of EAAT3/EAAC1 in GABAergic
knock-out animals. Whereas the knock-outs for GLAST neurons supports its role in the metabolic fueling of GABA
(the rodent counterpart of EAAT1) and GLT-1 (the rodent synthesis. This role is also supported by the increase in par-
EAAT2) exhibit early signs of severe excitotoxic damage oxysmal activity in animals treated with antisense oligonu-
[52], the knock-out for EAAT3/EAAC1 has an apparently cleotides that hinder transporter expression (see below and
normal development [53]. However, these animals present [65]).
symptoms of precocious neurodegeneration and subtle neu- In addition, EAAT3/EAAC1 also has an impor-
rologic damage, associated with cell loss in the cerebral tant function in the regulation of synaptic transmission.
cortex, increased aggressiveness and impaired self-groom- EAAT3/EAAC1 is involved in long-term potentiation and,
ing [54]. In particular, EAAT3/EAAC1-deficient mice hence, in memory and higher cerebral functions [66–69].
exhibit spatial learning/memory dysfunction, loss of dopa- Acute regulation of EAAT3/EAAC1 membrane expression
minergic neurons in the substantia nigra, and movement and activity by volatile anesthetics [70, 71] and carbamaz-
disorders; these disturbances typically appear at senescence epine [72] may represent examples of pharmacological
[20, 54] but have also been reported in young animals [55]. modulation of these mechanisms.
Moreover, patients affected by dicarboxylic aminoaciduria
also display hints of neurologic damage [12, 56].
The prevalent opinion is that the basic mechanism Regulatory mechanisms at the gene and protein levels
for termination of glutamatergic transmission resides
in EAAT1 and EAAT2 function. Consistent with this The expression of EAAT3/EAAC1 varies during brain
hypothesis, these transporters are endowed with higher development [73, 74], and the transporter is expressed
affinity for substrates than EAAT3/EAAC1 and are local- before EAAT1 and EAAT2 both in vitro [75] and in vivo
ized in glial cell processes close to synaptic clefts, while [73]. Early expression suggests a role for EAAT3/EAAC1
EAAT3/EAAC1 is mostly extra-synaptic [16]. Therefore, in neuroprotection of CNS cells during brain development.
EAAT3/EAAC1 may be important in situations where glu- Moreover, because GABAergic neurons reach maturation
tamate spills out from the synaptic zone or reaches abnor- before excitatory glutamatergic cells in the hippocam-
mally high extracellular concentrations, for example under pus, the early expression of EAAT3/EAAC1 may play an
ischemic conditions (see below). However, EAAT3/EAAC1 important role in developing the glutamatergic neuronal
may play an important role in glutamatergic termination in network [76]. These data are consistent with recent evi-
areas such as the hippocampus, where most synapses are dence showing that the EAATs are differentially regulated
not closely surrounded by astrocytic processes [57]. How- during the differentiation of human astroglial progenitors
ever, EAAT3/EAAC1 is far less abundant than EAAT2 also [77]. In particular, EAAT3/EAAC1 is more expressed than
in these regions [18]. EAAT1 and EAAT2 in A2B5-positive cells and decreases
Although glutamate uptake represents the best-charac- upon differentiation to the astrocytic phenotype. Interest-
terized activity of the carrier, EAAT3/EAAC1 also trans- ingly, early precursors of the oligodendroglial cell lineage
ports cysteine [58, 59] and may provide the majority of the are A2B5 positive [78].
influx of the amino acid in neurons [60]. Through cysteine Notwithstanding these data, studies focused on the
uptake, EAAT3/EAAC1 contributes to the intracellular modulation of the SLC1A1 gene under differentiating stim-
synthesis of glutathione [59, 61], one of the most impor- uli are lacking. In Hs683 human oligodendroglioma cells
tant intracellular antioxidants. EAAT3/EAAC1-mediated valproic acid (VPA) and trichostatin A, two inhibitors of
uptake may be the major contributor of cysteine to glu- histone deacetylases (HDACs), strongly increase SLC1A1
tathione synthesis, because the knock-out mouse for xCT, mRNA as well as EAAT3/EAAC1 protein expression and
the catalytic subunit of the sodium-independent transport transport activity [79]. In the same model, VPA signifi-
system for cystine, is apparently normal and shows no cantly also enhances the expression of PDGFRA, an early
changes in brain glutathione content [62]. Indeed, the mod- marker of oligodendrocyte precursors cells (OPCs), while it
ulation of EAAT3/EAAC1 activity correlates with neuronal does not affect the expression of either TUBBIII or GFAP,
glutathione content [41]. Further, the impaired Rab11- markers of neuronal and glial cells, respectively. VPA is a
dependent trafficking of EAAT3/EAAC1 is associated with well-known differentiating agent, and its effects on the dif-
enhanced sensitivity to oxidative stress in a murine model ferentiation of CNS cells have been repeatedly reported. In

13
M. G. Bianchi et al.

particular, VPA promotes neuronal differentiation of multi- is lethal in the early embryonic state, suggesting an indis-
potent adult neural progenitor cells and influences the tim- pensable role in brain development [92]. In rats and mice,
ing of oligodendrocyte differentiation in the developing rat RFX1 is highly expressed in several regions of the brain,
brain [80, 81]. such as the hippocampus, the cerebellum (Purkinje cells),
In another model of immature CNS cells, the rat glioma and the olfactory bulb, with localization in the nuclei
cell line C6, the differentiating agent all–trans retinoic of neurons and microglial cells but not in the nuclei of
acid (ATRA) markedly increases EAAT3/EAAC1 expres- astrocytes [93]. In the study by Feng and Zuo [93], 24 h
sion at both the mRNA and protein levels. ATRA effect after transfection of C6 glioma cells with human RFX1,
is specific for the Slc1a1 gene because it does not affect EAAT3/EAAC1 expression increased by 60 % and trans-
either GLT-1 (Slc1a2) or GLAST (Slc1a3) protein levels porter activity increased by 40 %. Conversely, knock-
[82]. RAR-specific but not RXR-specific inhibitors prevent down of RFX1 expression by antisense oligonucleotides
EAAT3/EAAC1 induction after ATRA treatment, while the decreased EAAT3/EAAC1 expression in rat cortical neu-
RAR-specific agonist Am80 (tamibarotene, retinobenzoic rons in culture. Moreover, transfection with RFX1 stimu-
acid) mimics the ATRA effect. Conversely, EAAT3/EAAC1 lated the transcriptional activity of the SLC1A1 promoter in
induction is prevented by Rarbeta silencing, demonstrating both rat glioma C6 cells and human neuroblastoma SHSY-
that the ATRA effect on EAAT3/EAAC1 strictly depends 5Y cells. Thus, although SLC1A1 mRNA was not directly
upon the RARβ receptor [83]. A consensus site for RARβ measured in that study, these data suggest that the increase
has been identified in the rat Slc1a1 promoter but not in the in EAAT3/EAAC1 expression was due to the interaction of
human SLC1A1 promoter region [83]. Along with Slc1a1 RFX1 with the promoter [93]. However, the natural activa-
induction, the expression of the oligodendrocytic marker tors of RFX1 are not known, and it is difficult to assess its
Plp1 is also enhanced by ATRA in C6 cells [82, 84], sug- physiological role.
gesting that retinoids may be involved in the activation of Although most of the previous studies on
the oligodendrocytic differentiation pathway. EAAT3/EAAC1 function have focused on its role in the
Soon after the cloning of the transporter, increased regulation of glutamate homeostasis, accumulating evi-
expression of EAAT3/EAAC1 was reported in bovine dence suggests that the carrier plays an essential function in
NBL-1 renal epithelial cells incubated under hypertonic controlling cell redox potential (see above). Astrocytes sup-
conditions. Incubation of these cells in medium supple- port neuronal antioxidant capacity by releasing glutathione,
mented with 200 mM sucrose leads to a marked increase in the most important anti-oxidant agent in the CNS, which
EAAT3/EAAC1 mRNA and protein, as well as to the stim- is cleaved to cysteine in brain extracellular space [94].
ulation of anionic amino acid transport [85]. Although it Free cysteine supply is the limiting step for glutathione
has not yet been demonstrated, it is likely that, as with other synthesis, and neuronal cysteine influx depends upon
amino acid transporters [86], EAAT3/EAAC1 induction EAAT3/EAAC1 activity [95]. The activation of the nuclear
also involves the activation of the osmosensitive transcrip- factor erythroid 2-related factor 2 (Nrf2)-antioxidant
tion factor NFAT5/TonEBP [87]. Incubation in the pres- responsive element (ARE) pathway by oxidative stress pro-
ence of tunicamycin, an inhibitor of protein glycosylation motes not only glutathione release from astrocytes but also
that triggers endoplasmic reticulum (ER) stress, increases the transcriptional regulation of neuronal EAAT3/EAAC1
EAAT3/EAAC1 mRNA and protein [88]. In contrast, both in vitro and in vivo [95]. Moreover, Nrf2-depend-
incubation of NBL-1 cells in the absence of amino acids, ent overexpression of EAAT3/EAAC1 protein is associ-
a condition that promotes the expression of other sodium- ated with an increase in neuronal glutathione content, and
dependent transporters, such as SNAT2 [89], stimulates these effects are abrogated in mice genetically deficient
aspartate transport but not EAAT3/EAAC1 expression [90]. in either Nrf2 or EAAT3/EAAC1. Selective overexpres-
Thus, it seems that in NBL-1 renal epithelial cells Slc1a1 is sion of Nrf2 in brain neurons by lentiviral gene transfer
induced by several (but not all) forms of stress associated is sufficient to up-regulate both neuronal EAAT3/EAAC1
with increased protein unfolding and ER stress. Unfortu- protein and glutathione content. These findings support a
nately, these experiments have not been repeated in CNS- mechanism whereby Nrf2 activation can coordinate astro-
derived cell models. cyte glutathione release with neuronal glutathione syn-
Ma et al. [91] have found that a binding sequence for the thesis through transcriptional up-regulation of neuronal
regulatory factor X1 (RFX1) exists in the promoter region EAAT3/EAAC1 expression. EAAT3/EAAC1-mediated
of SLC1A1, but not in the promoters of SLC1A2 (encod- cysteine uptake has also recently been implicated in the
ing EAAT2) and SLC1A3 (encoding EAAT1). RFX pro- increased neuronal glutathione content of mice treated with
teins are transcription factors that bind to X-boxes of DNA caffeine or uric acid [96], although the underlying mech-
sequences. In the mouse brain, RFX1 is expressed from anism has not yet been elucidated. Interestingly, recent
the embryonic state (E15) to adulthood, and its knock-out contributions demonstrate that fumarates can activate

13
Changes in the expression of the glutamate transporter

Nrf2-dependent responses and thereby ameliorate the dis- Four conditions in which alterations in EAAT3/EAAC1
ease course of experimental autoimmune encephalomyeli- expression have been identified will be discussed: hypoxia/
tis (EAE), a rodent model of multiple sclerosis (MS) [97, ischemia, MS, epilepsy, and schizophrenia.
98]. The protective effect of fumarates is associated with
improved preservation of myelin and neurons. Unfortu- Ischemia/hypoxia
nately, no investigation was performed on possible changes
in EAAT3/EAAC1 induced by fumarates in these models. Due to perturbations of ionic gradients, glutamate uptake
In a report by Molteni et al. [99], EAAT3/EAAC1 was through EAAT transporters is impaired during ischemia
up-regulated in parallel with glutamate NMDA receptor and the rise of extracellular glutamate is believed to con-
subunits after chronic periods of exercise, providing further tribute significantly to tissue damage. The possibility that
support for a functional role of the transporter in the modu- changes in the expression of EAAT transporters also occur
lation of glutamatergic transmission. under ischemic conditions has been investigated for many
Overall, these data suggest that the transcriptional regu- years.
lation of EAAT3/EAAC1 expression has peculiar features The potential protective role of EAAT3/EAAC1 was
because most of the reported mechanisms specifically affect proposed following the cloning of the transporter [4]
the transporter, which, conversely, appears insensitive to and discussed in one of the first reviews concerning glu-
well-characterized inducers of EAAT1 and EAAT2 [100, tamate transporters [111]. The first experimental evi-
101]. Similarly, studies on knock-out models for glutamate dence of ischemia-related expression changes was pro-
receptors demonstrate that EAAT1 and EAAT2 expres- vided by Martin et al. [112]; immunoblotting showed that
sion is lower in mGluR3−/− mice, while EAAT3/EAAC1 EAAT3/EAAC1 was reduced by 55 % in the putamen of
is decreased in mGluR2−/− animals [102]. However, newborn piglets 24 h after an ischemic stress caused by
examples of a parallel regulation of EAAT1, EAAT2, and 30 min of hypoxia (arterial O2 saturation, 30 %) followed
EAAT3/EAAC1 proteins have been also reported [103]. by 7 min of airway occlusion (O2 saturation, 5 %). This
decrease was possibly secondary to neuronal loss, con-
sistent with the Slc1a1 mRNA decrease demonstrated in
Pathological conditions involving changes gerbil CA1 hippocampal neurons 24 h after a transient
in EAAT3/EAAC1 expression ischemia [113] and the marked decrease (42–68 %) in
EAAT3/EAAC1 protein detected in the same model [114].
Alterations in EAAT3/EAAC1 have been associated with The latter study compared the decrease of GLT-1, which
several pathological conditions. In particular, there is a precedes neuronal loss, with that of EAAT3/EAAC1, which
strong association between single-nucleotide polymor- parallels cell death. Their results suggest that decreased
phisms (SNPs) in SLC1A1 and obsessive–compulsive dis- EAAT3/EAAC1 may have a role in GABAergic dysfunc-
order [104–107], but these polymorphisms seem more tion. Decrease of EAAT3/EAAC1 expression as a conse-
correlated with changes in transporter function than with quence of neuronal loss has been more recently shown by
its expression. Dicarboxylic aminoaciduria is a Mendelian Han et al. [115] following microsphere embolism in rats.
disorder (OMIM 222730) where EAAT3/EAAC1 function However, the possibility that adaptive changes in ischemic
is defective due to the expression of a mutant, functionally cells may directly affect transporter expression should not
impaired transporter that is also scarcely sorted to the cell be excluded a priori [38].
membrane [12]. Clear-cut, although contrasting, changes A predominant role for GLT-1 in the protection from
in EAAT3/EAAC1 expression have been described in Alz- ischemic damage was proposed by Rao et al. [116], who
heimer’s disease (AD). Results in mouse models of AD showed that knock-down of this transporter, but not
are conflicting. Schallier et al. [108] reported an increase of EAAT3/EAAC1, exacerbated the neuronal damage
in EAAT3/EAAC1 expression (although overwhelmed induced by transient focal cerebral ischemia in rat brain.
by a concomitant decrease in GLT-1 and GLAST levels) Douen et al. [117] also stressed the importance of GLT-
in AβPP23 mice, while a decrease in the expression of all 1, using as a model rats that were preconditioned 3 days
three glutamate transporters has been reported by Cas- before ischemia with cortical spreading depression (CSD)
sano et al. [109] in triple transgenic (3×Tg) AD mice. In and showed relative protection from ischemic damage.
humans, aberrant intracellular accumulation of a detergent These authors noted that extracellular brain glutamate
insoluble EAAT3/EAAC1 was described in hippocampal after ischemia was indeed lower in the preconditioned ani-
neurons of AD patients, although overall cortical expres- mals and that the change was associated with a decrease
sion of the transporter was comparable to controls, suggest- of EAAT1 and EAAT2, but not EAAT3/EAAC1, expres-
ing a defect in transporter processing rather than synthesis sion in the plasma membrane. The authors suggested that
[110]. a lower expression of EAAT transporters contributes to

13
M. G. Bianchi et al.

less glutamate efflux through the reverse operation, origi- stress (expected under ischemic conditions), and main-
nally described as an alternative functional mode of EAAT tenance of zinc homeostasis. N-acetyl-cysteine restored
transporters under hypoxic conditions [118]. A decrease neuronal glutathione, normalized basal zinc levels, and
in Slc1a1 expression under comparable conditions was attenuated ischemia-induced zinc translocation, superoxide
described by Montori et al. [119]. production, and neuron death.
The first experimental contribution pointing to an adap- Given the putative role of EAAT3/EAAC1 as a protec-
tive and/or protective role of increased EAAT3/EAAC1 tive factor under ischemic conditions, attempts to up-regu-
expression in ischemia came in 2000 from the Matute late its expression have been performed as a means to limit
laboratory [120]. These researchers compared the expres- ischemic damage. Chen et al. [130] used intrathecal granu-
sion of the main EAAT transporters in the rat hippocam- locyte colony-stimulating factor (G–CSF) to improve neu-
pus and cerebral cortex in both early (8 h and 1 day) rological defects in the ischemic spinal cord of Wistar rats.
and late (28 days) phases of recovery from transient Using microdialysis, they found that G–CSF significantly
forebrain ischemia. They found a moderate increase in hampers the increase in spinal concentrations of excitatory
EAAT3/EAAC1 protein early after the insult, followed amino acids induced by ischemia or by exogenous intrath-
by a late reduction in both CA1 pyramidal neurons and in ecal administration of glutamate. These effects are associ-
layer V pyramidal neurons. Moreover, the authors observed ated with the increased expression of GLAST, GLT-1 and
a sustained increase in EAAT3/EAAC1 expression in oli- EAAT3/EAAC1.
godendroglial progenitor cells of subcortical white matter
[120], although, in a later study, the same group reported Multiple sclerosis
that EAAT1 and EAAT2 were also up-regulated in this area
[121]. Transient changes in EAAT3/EAAC1 expression Excitotoxicity and oxidative stress have been implicated
during ischemia and the recovery phase were also reported in MS-related damage that involves not only white mat-
by Hwang [122], who described a fast decrease of trans- ter but also gray matter. However, evidence for a role of
porter immunoreactivity in spinal cord neurons just after the EAAT3/EAAC1 transporter in MS is thus far limited,
ischemia/reperfusion, followed by an increase of expres- although, as detailed in the Introduction, the transporter is
sion (after 3 h of reperfusion) and a further subsequent also expressed in cells of oligodendrocytic lineage and, in
decrease. EAAT3/EAAC1 expression was also slowly particular, in oligodendrocyte precursor cells [23, 24, 131].
increased, at both the mRNA and protein levels, under Ohgoh et al. [132], prompted by the evidence of the
“hypoxia-pure” conditions [123] in rat PC12 cells. In this involvement of AMPA/kainate receptor in experimental
case, the maximum increase was reached after 18 h of incu- autoimmune encephalomyelitis (EAE, the animal model
bation at 1 % O2 and corresponded to an eightfold increase of MS) [133], examined the expression of GLT-1, GLAST,
at protein level and a four-fold increase in transport activ- and EAAT3/EAAC1 in the spinal cord of the EAE Lewis
ity. However, GLT-1 was also up-regulated under the same rat. While EAAT3/EAAC1 was dramatically increased
conditions. at both the protein and mRNA levels, GLT-1 and GLAST
These results are, at least in part, consistent with the evi- were down-regulated. The AMPA/kainate receptor inhibitor
dence obtained by Romera et al. [124] in rat cortical cul- NBQX ameliorated EAE symptoms and suppressed changes
tures exposed to sublethal oxygen-glucose deprivation (an in transporter expression. No attempt was made to identify
in vitro model for ischemic preconditioning), and by Pra- the cell type(s) in which changes in carrier expression occur.
dillo et al. [125] in preconditioned rat brain in vivo. In both This issue was instead addressed by Vallejo-Illarramendi
studies, EAAT3/EAAC1 was up-regulated by increased et al. [134] who found an increase of SLC1A3 and SLC1A2
TNF-α signaling. However, in the same studies, TNF- mRNA in optic nerves from MS patients, while SLC1A1
α-independent EAAT2 up-regulation was also reported. mRNA did not change. The expression increase was con-
A generalized up-regulation of EAAT transporters was firmed at the protein level for both EAAT1 (in oligodendro-
described in rat brain upon ischemic pre-conditioning by cytes) and EAAT2 (in astrocytes), while EAAT3/EAAC1
Bigdeli et al. [126, 127]. expression was not investigated. Increased expression of the
Results from EAAC1 knock-out mice support the pro- rodent counterparts of EAAT1 and EAAT2 (at the mRNA,
tective role of the transporter under ischemic conditions protein and function levels) were also observed in rat optic
[128, 129]. In particular, Won et al. demonstrated that nerves incubated with excitotoxic glutamate concentrations.
EAAC1−/− mice subjected to transient cerebral ischemia More recently, Newcombe et al. [135] studied gluta-
exhibit much more widespread hippocampal neuronal mate receptors, transporters, and enzymes in human MS
death than wild-type mice. These authors suggested that lesions. They found that the AMPA GluR-1 receptor subu-
impaired cysteine uptake would severely hinder intracellu- nit was overexpressed in oligodendrocytes at the borders of
lar glutathione synthesis, cell capability to sustain oxidative MS active lesions, while reactive astrocytes, macrophages,

13
Changes in the expression of the glutamate transporter

and neuronal structures expressed a different receptor rep- (not only EAAT3/EAAC1 but also GLAST and GLT-1)
ertoire. Unexpectedly, EAAT3/EAAC1 was expressed in increased within 24 h by kindling but returned to basal
macrophages and reactive astrocytes, leading the authors to levels 30 days after the last seizure. These findings led
propose that this ectopic expression may have a protective, the authors to suggest that glutamate transporters may
although not completely effective, function. contribute to the occurrence of seizures. Interestingly,
The positive effects of retinoids on EAAT3/EAAC1 easily-kindled rats had levels of EAAT3/EAAC1 and
expression (see above) may be important in light of the GAT-1 (the GABA transporter) that were 30 % lower com-
evidence for a positive effect of retinoids in EAE mice pared to epilepsy-resistant animals, again suggesting that
[136, 137]. Klemann et al. attributed the protective effect EAAT3/EAAC1 level is negatively associated with the con-
of the synthetic retinoid Am80 to immunomodulation. On vulsive threshold [141].
the other hand, Huang et al. [137] attributed the protec- Voutsinos-Porche et al. reported the temporal depend-
tive effect of the natural compound 9-cis-retinoic acid to a ence of epilepsy-associated changes in transporter expres-
RXR-γ-dependent stimulation of oligodendrocytic differ- sion, demonstrating that EAAT3/EAAC1 increased in sev-
entiation. Neither study, however, investigated the expres- eral portions of the hippocampus during the acute period of
sion of EAAT3/EAAC1 or other glutamate transporters. status epilepticus, but returned to control levels in the CA1,
In conclusion, while some evidence suggests a role for 2, and 3 layers during the latent period. The authors con-
EAAT3/EAAC1 in the pathophysiology of MS, a definite cluded that “it is not clear to what extent the overexpression
demonstration is still lacking. of EAAT3/EAAC1 contributes to epileptogenesis and in
which area it may represent a preventive or compensatory
Epilepsy or response to injury” [142].
Using the model of kainate-induced convulsions, Sim-
The role of EAAT3/EAAC1 dysregulation in epilepsy was antov et al. [143] reached apparently divergent conclusions.
proposed in the pivotal paper in which antisense EAAT- Kainate acutely lowered EAAT3/EAAC1 in the stratum
knock-out models were investigated for the first time lacunosum moleculare, and the decrease in protein level was
[52]. While the EAAT1 and EAAT2 knock-outs produced associated with a decrease in mRNA. Opposite effects were
severe neurotoxicity and progressive paralysis, the phe- reported for GLT-1, leading the authors to conclude that dif-
notype of the EAAT3/EAAC1 knock-out was milder but ferential regulation of neuronal and glial EAAT may contrib-
associated with epilepsy. Using the same experimental ute to kainate-induced seizures. In another model of experi-
approach, it was then demonstrated that the repression of mental epilepsy due to prenatal exposure to the teratogen
EAAT3/EAAC1 expression was associated with decreased methylazoxymethanol, heterotopic hippocampal neurons
tonic inhibition and to 50 % loss of hippocampal GABA exhibited striking reductions in EAAT3/EAAC1, as well as in
levels, attributable to the impairment of GABA synthesis GluR1 NMDA receptor subunit expression [144]. In contrast,
from extracellular glutamate [65]. a significant increase in the carrier expression was detected in
These early findings spurred many investigations with hippocampal and motor cortex area neurons of rats with chest
experimental models of epilepsy. Compared to controls, compression-induced audiogenic epilepsy [145].
Slc1a1 mRNA was markedly increased in dentate granule Data from human tissues are contradictory. Proper
cells from rats with pilocarpine-induced epilepsy and in et al. [146] reported an increase in EAAT3/EAAC1 in
human dentate granule cells from patients with temporal individual neurons of the hippocampus of TLE patients,
lobe epilepsy (TLE) [138]. In cortical dysplasia, the trans- although the functional consequences of the reported
porter mRNA was elevated in human dysplastic neurons changes are unclear. Increased EAAT3/EAAC1 expres-
compared with non-dysplastic neurons. These changes sion was also reported by Mathern et al. [147], but only in
were confirmed at the protein level and were interpreted as epileptic patients with hippocampal sclerosis. On the other
an attempt to compensate for the higher-than-normal extra- hand, Rakhade and Loweb [148] measured the differential
cellular glutamate levels caused by recurrent seizures [138]. expression of EAAT1–4 at the mRNA and protein levels in
Pilocarpine-induced status epilepticus has been associated electrically mapped human neocortical tissues and found
with an increase in EAAT3/EAAC1 protein in rat hip- significant reductions of EAAT3/EAAC1 and EAAT4)
pocampus and with a ~15-fold increase in Slc1a1 mRNA in at both the mRNA and protein level, not associated with
rat synaptoneurosomes [139], although the authors attrib- changes in the relative neuronal density. The authors con-
uted the change more to the targeting of the messenger to cluded that “regional reductions in EAAT expression at
dendrites than to an effect on transcription [140]. human neocortical epileptic foci could produce increased
Doi et al. [141] examined the induction of epilepsy local glutamate levels that in turn may contribute to both
(kindling) by pentylenetetrazol (PTZ) in rats. They found hyperexcitability and the spontaneous generation of epilep-
that the expression of hippocampal glutamate transporters tic discharges that characterize human epileptic foci” [148].

13
M. G. Bianchi et al.

As far as genetic forms of epilepsy are concerned, Dutuit transport may also contribute to the glutamatergic dysfunc-
et al. [149] described a 32 % decrease in EAAT3/EAAC1 tion implicated in schizophrenia.
expression in the thalamus of young genetic absence epi- Other reports present evidence for a role of
lepsy rats from Strasbourg (GAERS) compared to con- EAAT3/EAAC1 in schizophrenia. Increased carrier tran-
trols. However, the expression of GLT-1 and GLAST also scripts and proteins were reported in schizophrenic subjects
decreased, and the changes were not observed in adult (dorsolateral prefrontal and anterior cingulated cortex) by
animals. Bauer et al. [154]; these findings were, more recently, con-
Interestingly, in human gangliogliomas, which are firmed by Rao et al. [155] in the frontal cortex at both the
neuronal-glial tumors strongly associated with epilepsy, protein and mRNA levels. Conversely, Lauriat et al. [156]
SLC1A1 is among the genes that are down-regulated com- did not observe differences in EAAT3/EAAC1 expression
pared to normal tissue [150]. This alteration has been in either the dorsolateral prefrontal or the primary visual
attributed to a true transcriptional change rather than to a cortex. However, the authors also highlighted the pos-
compensatory response to high extracellular glutamate due sibility that the results were influenced by antipsychotic
to repeated seizures. In contrast, the high expression of treatment that is often prolonged for many years in these
SLC1A1 mRNA detected in the lesions of patients affected patients [156].
by tuberous sclerosis, another condition exhibiting high sei- On the contrary, decreased SLC1A1 transcript expres-
zure incidence, has been considered compensatory [151]. sion in the striatum was observed in schizophrenia (and in
bipolar disorder) by other authors [157] and confirmed in
Schizophrenia schizophrenic patients in a later study [158].

The possibility that glutamatergic transmission is altered in


schizophrenic patients has been debated for several years. Conclusions and perspectives
Changes in the expression of EAAT1 and EAAT2, but
not EAAT3/EAAC1, have been detected in the thalamus The regulation of EAAT3/EAAC1 expression at the gene
[152] or the superior temporal gyrus and the hippocam- level is far less characterized than mechanisms acting on
pus [153]. While these studies do not support a role for transporter trafficking. Nevertheless, accumulating data
EAAT3/EAAC1 in schizophrenia, the findings suggest that, yield an increasingly complex picture (Fig. 1), indicating
in addition to glutamate receptors, alterations in glutamate that at least three levels of control modulate the expression

Fig. 1  Sites of regulation of EAAT3/EAAC1 expression on the modification of transporter trafficking to and from the plasma mem-
plasma membrane. The sketch depicts the three documented sites of brane. For changes in SLC1A1 transcription, transcription factors,
regulation of EAAT3/EAAC1 expression: changes in SLC1A1 tran- chromatin-modifying enzymes, and extracellular stimuli known to be
scription, modulation of carrier interaction with binding proteins and involved in expression changes are detailed

13
Changes in the expression of the glutamate transporter

Table 1  Selected conditions and stimuli associated with changes in the expression of EAAT3/EAAC1
Condition/stimuli SLC1A1/Slc1a1 EAAT3/ Models References
mRNA EAAC1

Alzheimer dementia ND ↑ Mouse (Abeta PP23) hippocampus [108]


ND ↓ Mouse (3×Tg-Alzheimer’s disease) [109]
hippocampus
Dicarboxylic aminoaciduria ND ↓ Canine kidney cell line [12]
Epilepsy (kainate-induced convulsions) ↓ ↓ Rat hippocampus [143]
Epilepsy (methylazoxymethanol) ↓ ↓ Rat hippocampal neurons [144]
Epilepsy (pilocarpine-induced, temporal lobe ↑ ↑ Human hippocampus [138]
epilepsy, TLE) ↑ ↑ Rat hippocampal neurons [139]
↑ ↑ Rat hippocampal granule cells [140]
↓ ↓ Human neocortical tissues [148]
Exercise ↑ ND Rat hippocampus in vivo [99]
Experimental autoimmune encephalomyelitis ↑ ↑ Rat spinal cord [132]
(EAE)
G–CSF ND ↑ Rat spinal cord [130]
HDAC inhibitors ↑ ↑ Human oligodendroglioma Hs683 cells [79]
(valproic acid, trichostatin A)
Hypertonic stress ↑ ↑ Bovine renal epithelial NBL-1 cells [85, 88]
Hypoxia (pure) ↑ ↑ Rat pheochromocytoma PC-12 cells [123]
Ischemia/hypoxia ND ↓ Piglet striatum [112]
↓ ↓ Gerbil hippocampus [113]
↓ ↓ Gerbil hippocampus [114]
↓ ND Rat hippocampus and cerebral cortex [119]
ND ↓ Rat neurons and oligodendrocytes [120]
ND ↑ Rabbit spinal cord [122]
Ischemic preconditioning ↑ ↑ Rat cortical neurons in culture [124]
(oxygen-glucose deprivation) ↑ ↑ Rat brain in vivo [125]
NrF2 ↑ ↑ Rat brain neurons in situ and in vitro [95]
Retinoids (all-trans retinoic acid and Am80, a ↑ ↑ Rat glioma C6 cells [82, 83]
synthetic retinoid)
RFX1 ND ↑ Rat glioma C6 cells; rat brain neurons in situ [91]
Schizophrenia ↑ ↑ Human cortex [154]
↑ ↑ Human frontal cortex [155]
↓ ND Human striatum [157]
Tunicamycin (ER stress?) ↑ ↑ Bovine renal epithelial NBL-1 cells [88]

ND not determined

of the transporter on the plasma membrane: transcriptional is necessary to clarify the role of EAAT3/EAAC1 in these
changes in SLC1A1 expression (Table 1), changes in protein conditions and, more importantly, to envisage novel thera-
interaction, and trafficking modulation. Overall, this infor- peutic approaches.
mation suggests that EAAT3/EAAC1 can be finely regu-
lated at the gene level; however, detailed information is not Acknowledgments  Research in the authors’ laboratory has been
funded by FISM (Fondazione Italiana Sclerosi Multipla onlus), grant
yet available on regulations acting at post-transcriptional or no. 2010/R/9 to OB.
translational sites. With respect to the well-known exam-
ples of fast trafficking modulation, transcriptional regula-
tions are clearly slower but may be more relevant to explain
the somewhat contradictory changes in EAAT3/EAAC1 References
expression described in situations of pathophysiological
1. Amara SG, Fontana AC (2002) Excitatory amino acid transport-
interest and associated with chronic alterations in nerv- ers: keeping up with glutamate. Neurochem Int 41(5):313–318.
ous functions. Improved knowledge of these mechanisms doi:10.1016/S0197-0186(02)00018-9

13
M. G. Bianchi et al.

2. Kanai Y, Hediger MA (2004) The glutamate/neutral amino acid (EAAT3) glutamate transporters expressed by neurons in the
transporter family SLC1: molecular, physiological and pharma- mammalian CNS. J Neurosci 32(17):6000–6013. doi:10.1523/
cological aspects. Pflugers Arch 447(5):469–479. doi:10.1007/ JNEUROSCI.5347-11.2012
s00424-003-1146-4 19. Plaitakis A, Shashidharan P (2000) Glutamate transport and
3. Kanai Y, Hediger MA (2003) The glutamate and neutral metabolism in dopaminergic neurons of substantia nigra: impli-
amino acid transporter family: physiological and pharma- cations for the pathogenesis of Parkinson’s disease. J Neurol
cological implications. Eur J Pharmacol 479(1–3):237–247. 247(Suppl 2):II25–II35
doi:10.1016/j.ejphar.2003.08.073 20. Berman AE, Chan WY, Brennan AM, Reyes RC, Adler BL,
4. Kanai Y, Hediger MA (1992) Primary structure and functional Suh SW, Kauppinen TM, Edling Y, Swanson RA (2011)
characterization of a high-affinity glutamate transporter. Nature N-acetylcysteine prevents loss of dopaminergic neurons in the
360(6403):467–471. doi:10.1038/360467a0 EAAC1−/− mouse. Ann Neurol 69(3):509–520. doi:10.1002/
5. Gazzola GC, Dall’Asta V, Bussolati O, Makowske M, Chris- ana.22162
tensen HN (1981) A stereoselective anomaly in dicarboxylic 21. Nafia I, Re DB, Masmejean F, Melon C, Kachidian P, Kerke-
amino acid transport. J Biol Chem 256(12):6054–6059 rian-Le Goff L, Nieoullon A, Had-Aissouni L (2008) Preferen-
6. Billups B, Rossi D, Oshima T, Warr O, Takahashi M, Sarantis tial vulnerability of mesencephalic dopamine neurons to glu-
M, Szatkowski M, Attwell D (1998) Physiological and patho- tamate transporter dysfunction. J Neurochem 105(2):484–496.
logical operation of glutamate transporters. Prog Brain Res doi:10.1111/j.1471-4159.2007.05146.x
116:45–57 22. Kugler P, Schmitt A (1999) Glutamate transporter
7. Zerangue N, Kavanaugh MP (1996) Flux coupling in a neu- EAAC1 is expressed in neurons and glial cells in the
ronal glutamate transporter. Nature 383(6601):634–637. rat nervous system. Glia 27(2):129–142. doi:10.1002/
doi:10.1038/383634a0 (SICI)1098-1136(199908)27:2<129:AID-GLIA3>3.0.CO;2-Y
8. Nothmann D, Leinenweber A, Torres-Salazar D, Kovermann 23. Domercq M, Sanchez-Gomez MV, Areso P, Matute C
P, Hotzy J, Gameiro A, Grewer C, Fahlke C (2010) Hetero-oli- (1999) Expression of glutamate transporters in rat optic
gomerization of neuronal glutamate transporters. J Biol Chem nerve oligodendrocytes. Eur J Neurosci 11(7):2226–2236.
286(5):3935–3943. doi:10.1074/jbc.M110.187492 doi:10.1046/j.1460-9568.1999.00639.x
9. Leary GP, Holley DC, Stone EF, Lyda BR, Kalachev LV, 24. Domercq M, Matute C (1999) Expression of gluta-
Kavanaugh MP (2011) The central cavity in trimeric glutamate mate transporters in the adult bovine corpus callosum.
transporters restricts ligand diffusion. Proc Natl Acad Sci USA Brain Res Mol Brain Res 67(2):296–302. doi:10.1016/
108(36):14980–14985. doi:10.1073/pnas.1108785108 S0169-328X(99)00072-8
10. Seal RP, Amara SG (1999) Excitatory amino acid transporters: 25. van Landeghem FK, Stover JF, Bechmann I, Bruck W, Unter-
a family in flux. Annu Rev Pharmacol Toxicol 39:431–456. doi: berg A, Buhrer C, von Deimling A (2001) Early expression of
10.1146/annurev.pharmtox.39.1.431 glutamate transporter proteins in ramified microglia after con-
11. Perego C, Di Cairano ES, Ballabio M, Magnaghi V (2011) Neu- trolled cortical impact injury in the rat. Glia 35(3):167–179.
rosteroid allopregnanolone regulates EAAC1-mediated gluta- doi:10.1002/glia.1082
mate uptake and triggers actin changes in Schwann cells. J Cell 26. Palos TP, Ramachandran B, Boado R, Howard BD (1996)
Physiol 227(4):1740–1751. doi:10.1002/jcp.22898 Rat C6 and human astrocytic tumor cells express a neu-
12. Bailey CG, Ryan RM, Thoeng AD, Ng C, King K, Vanslamb- ronal type of glutamate transporter. Brain Res Mol Brain Res
rouck JM, Auray-Blais C, Vandenberg RJ, Broer S, Rasko JE 37(1–2):297–303
(2011) Loss-of-function mutations in the glutamate transporter 27. Chaudhry FA, Lehre KP, van Lookeren Campagne M,
SLC1A1 cause human dicarboxylic aminoaciduria. J Clin Ottersen OP, Danbolt NC, Storm-Mathisen J (1995) Glu-
Invest 121(1):446–453. doi:10.1172/JCI44474 tamate transporters in glial plasma membranes: highly dif-
13. Fairman WA, Vandenberg RJ, Arriza JL, Kavanaugh MP, Amara ferentiated localizations revealed by quantitative ultras-
SG (1995) An excitatory amino-acid transporter with properties tructural immunocytochemistry. Neuron 15(3):711–720.
of a ligand-gated chloride channel. Nature 375(6532):599–603. doi:10.1016/0896-6273(95)90158-2
doi:10.1038/375599a0 28. Dehnes Y, Chaudhry FA, Ullensvang K, Lehre KP, Storm-
14. Arriza JL, Eliasof S, Kavanaugh MP, Amara SG (1997) Excita- Mathisen J, Danbolt NC (1998) The glutamate transporter
tory amino acid transporter 5, a retinal glutamate transporter EAAT4 in rat cerebellar Purkinje cells: a glutamate-gated chlo-
coupled to a chloride conductance. Proc Natl Acad Sci USA ride channel concentrated near the synapse in parts of the den-
94(8):4155–4160 dritic membrane facing astroglia. J Neurosci 18(10):3606–3619
15. Rothstein JD, Martin L, Levey AI, Dykes-Hoberg M, Jin L, 29. Conti F, DeBiasi S, Minelli A, Rothstein JD, Melone M (1998)
Wu D, Nash N, Kuncl RW (1994) Localization of neuronal EAAC1, a high-affinity glutamate transporter, is localized to
and glial glutamate transporters. Neuron 13(3):713–725. astrocytes and GABAergic neurons besides pyramidal cells in
doi:10.1016/0896-6273(94)90038-817 the rat cerebral cortex. Cereb Cortex 8(2):108–116
16. Coco S, Verderio C, Trotti D, Rothstein JD, Volterra A, Mat- 30. Yang W, Kilberg MS (2002) Biosynthesis, intracellular target-
teoli M (1997) Non-synaptic localization of the glutamate trans- ing, and degradation of the EAAC1 glutamate/aspartate trans-
porter EAAC1 in cultured hippocampal neurons. Eur J Neurosci porter in C6 glioma cells. J Biol Chem 277(41):38350–38357.
9(9):1902–1910 doi:10.1074/jbc.M202052200
17. Shashidharan P, Huntley GW, Murray JM, Buku A, Moran T, 31. Gonzalez MI, Kazanietz MG, Robinson MB (2002) Regula-
Walsh MJ, Morrison JH, Plaitakis A (1997) Immunohisto- tion of the neuronal glutamate transporter excitatory amino acid
chemical localization of the neuron-specific glutamate trans- carrier-1 (EAAC1) by different protein kinase C subtypes. Mol
porter EAAC1 (EAAT3) in rat brain and spinal cord revealed Pharmacol 62(4):901–910
by a novel monoclonal antibody. Brain Res 773(1–2):139–148. 32. Fournier KM, Gonzalez MI, Robinson MB (2004) Rapid traf-
doi:10.1016/S0006-8993(97)00921-9 ficking of the neuronal glutamate transporter, EAAC1: evidence
18. Holmseth S, Dehnes Y, Huang YH, Follin-Arbelet VV, Grutle for distinct trafficking pathways differentially regulated by pro-
NJ, Mylonakou MN, Plachez C, Zhou Y, Furness DN, Bergles tein kinase C and platelet-derived growth factor. J Biol Chem
DE, Lehre KP, Danbolt NC (2012) The density of EAAC1 279(33):34505–34513. doi:10.1074/jbc.M404032200

13
Changes in the expression of the glutamate transporter

33. Gonzalez MI, Bannerman PG, Robinson MB (2003) Phorbol transporter, excitatory amino acid carrier 1. J Biol Chem
myristate acetate-dependent interaction of protein kinase Cal- 282(41):29855–29865. doi:10.1074/jbc.M704738200
pha and the neuronal glutamate transporter EAAC1. J Neurosci 48. Xia P, Pei G, Schwarz W (2006) Regulation of the glu-
23(13):5589–5593 tamate transporter EAAC1 by expression and activa-
34. Huang Y, Feng X, Sando JJ, Zuo Z (2006) Critical role of ser- tion of delta-opioid receptor. Eur J Neurosci 24(1):87–93.
ine 465 in isoflurane-induced increase of cell-surface redistribu- doi:10.1111/j.1460-9568.2006.04897.x
tion and activity of glutamate transporter type 3. J Biol Chem 49. Bianchi MG, Gatti R, Torielli L, Padoani G, Gazzola GC,
281(50):38133–38138. doi:10.1074/jbc.M603885200 Bussolati O (2010) The glutamate transporter excitatory
35. Klaus F, Gehring EM, Zurn A, Laufer J, Lindner R, Strutz- amino acid carrier 1 associates with the actin-binding protein
Seebohm N, Tavare JM, Rothstein JD, Boehmer C, Palmada M, alpha-adducin. Neuroscience 169(2):584–595. doi:10.1016/j.
Gruner I, Lang UE, Seebohm G, Lang F (2009) Regulation of neuroscience.2010.05.029
the Na(+)-coupled glutamate transporter EAAT3 by PIKfyve. 50. Seidel B, Zuschratter W, Wex H, Garner CC, Gundelfinger ED
Neurochem Int 54(5–6):372–377 (1995) Spatial and sub-cellular localization of the membrane
36. Fedorenko O, Tang C, Sopjani M, Foller M, Gehring EM, cytoskeleton-associated protein alpha-adducin in the rat brain.
Strutz-Seebohm N, Ureche ON, Ivanova S, Semke A, Lang F, Brain Res 700(1–2):13–24. doi:10.1016/0006-8993(95)00962-P
Seebohm G, Lang UE (2009) PIP5K2A-dependent regulation 51. Nieoullon A, Canolle B, Masmejean F, Guillet B, Pisano P,
of excitatory amino acid transporter EAAT3. Psychopharmacol- Lortet S (2006) The neuronal excitatory amino acid trans-
ogy 206(3):429–435. doi:10.1007/s00213-009-1621-5 porter EAAC1/EAAT3: does it represent a major actor at the
37. Almilaji A, Pakladok T, Guo A, Munoz C, Foller M, brain excitatory synapse? J Neurochem 98(4):1007–1018.
Lang F (2012) Regulation of the glutamate transporter doi:10.1111/j.1471-4159.2006.03978.x
EAAT3 by mammalian target of rapamycin mTOR. 52. Rothstein JD, Dykes-Hoberg M, Pardo CA, Bristol LA, Jin
Biochem Biophys Res Commun 421(2):159–163. L, Kuncl RW, Kanai Y, Hediger MA, Wang Y, Schielke JP,
doi:10.1016/j.bbrc.2012.03.109 Welty DF (1996) Knockout of glutamate transporters reveals
38. Sopjani M, Alesutan I, Dermaku-Sopjani M, Fraser S, Kemp a major role for astroglial transport in excitotoxicity and
BE, Foller M, Lang F (2010) Down-regulation of Na+-cou- clearance of glutamate. Neuron 16(3):675–686. doi:10.1016/
pled glutamate transporter EAAT3 and EAAT4 by AMP- S0896-6273(00)80086-0
activated protein kinase. J Neurochem 113(6):1426–1435. 53. Peghini P, Janzen J, Stoffel W (1997) Glutamate transporter
doi:10.1111/j.1471-4159.2010.06678.x EAAC-1-deficient mice develop dicarboxylic aminoaciduria
39. Lin CI, Orlov I, Ruggiero AM, Dykes-Hoberg M, Lee A, Jack- and behavioral abnormalities but no neurodegeneration. EMBO
son M, Rothstein JD (2001) Modulation of the neuronal gluta- J 16(13):3822–3832. doi:10.1093/emboj/16.13.3822
mate transporter EAAC1 by the interacting protein GTRAP3- 54. Aoyama K, Suh SW, Hamby AM, Liu J, Chan WY, Chen Y,
18. Nature 410(6824):84–88. doi:10.1038/35065084 Swanson RA (2006) Neuronal glutathione deficiency and age-
40. Liu Y, Vidensky S, Ruggiero AM, Maier S, Sitte HH, Rothstein dependent neurodegeneration in the EAAC1 deficient mouse.
JD (2008) Reticulon RTN2B regulates trafficking and func- Nat Neurosci 9(1):119–126. doi:10.1038/nn1609
tion of neuronal glutamate transporter EAAC1. J Biol Chem 55. Lee S, Park SH, Zuo Z (2012) Effects of isoflurane on learning
283(10):6561–6571. doi:10.1074/jbc.M708096200 and memory functions of wild-type and glutamate transporter
41. Aoyama K, Watabe M, Nakaki T (2011) Modulation of neu- type 3 knockout mice. J Pharm Pharmacol 64(2):302–307.
ronal glutathione synthesis by EAAC1 and its interacting pro- doi:10.1111/j.2042-7158.2011.01404.x
tein GTRAP3-18. Amino Acids 42(1):163–169. doi:10.1007/ 56. Smith CP, Weremowicz S, Kanai Y, Stelzner M, Morton CC,
s00726-011-0861-y Hediger MA (1994) Assignment of the gene coding for the
42. Aoyama K, Wang F, Matsumura N, Kiyonari H, Shioi G, Tan- human high-affinity glutamate transporter EAAC1 to 9p24:
aka K, Kinoshita C, Kikuchi-Utsumi K, Watabe M, Nakaki T potential role in dicarboxylic aminoaciduria and neurodegen-
(2012) Increased neuronal glutathione and neuroprotection in erative disorders. Genomics 20(2):335–336. doi:10.1006/g
GTRAP3-18-deficient mice. Neurobiol Dis 45(3):973–982. eno1994.1183
doi:10.1016/j.nbd.2011.12.016 57. Bergles DE, Diamond JS, Jahr CE (1999) Clearance of glu-
43. Butchbach ME, Lai L, Lin CL (2002) Molecular cloning, tamate inside the synapse and beyond. Curr Opin Neurobiol
gene structure, expression profile and functional characteri- 9(3):293–298. doi:10.1016/S0959-4388(99)80043-9
zation of the mouse glutamate transporter (EAAT3) interact- 58. Zerangue N, Kavanaugh MP (1996) Interaction of L-cysteine
ing protein GTRAP3-18. Gene 292(1–2):81–90. doi:10.1016/ with a human excitatory amino acid transporter. J Physiol
S0378-1119(02)00669-8 493(Pt 2):419–423
44. Akiduki S, Ikemoto MJ (2008) Modulation of the neural gluta- 59. Chen Y, Swanson RA (2003) The glutamate transport-
mate transporter EAAC1 by the addicsin-interacting protein ers EAAT2 and EAAT3 mediate cysteine uptake in cor-
ARL6IP1. J Biol Chem 283(46):31323–31332. doi:10.1074/jbc. tical neuron cultures. J Neurochem 84(6):1332–1339.
M801570200 doi:10.1046/j.1471-4159.2003.01630.x
45. Gonzalez MI, Susarla BT, Fournier KM, Sheldon AL, 60. Shanker G, Allen JW, Mutkus LA, Aschner M (2001)
Robinson MB (2007) Constitutive endocytosis and recy- The uptake of cysteine in cultured primary astrocytes
cling of the neuronal glutamate transporter, excitatory and neurons. Brain Res 902(2):156–163. doi:10.1016/
amino acid carrier 1. J Neurochem 103(5):1917–1931. S0006-8993(01)02342-3
doi:10.1111/j.1471-4159.2007.04881.x 61. Himi T, Ikeda M, Yasuhara T, Nishida M, Morita I (2003)
46. Fournier KM, Robinson MB (2006) A dominant-negative Role of neuronal glutamate transporter in the cysteine uptake
variant of SNAP-23 decreases the cell surface expression and intracellular glutathione levels in cultured cortical neu-
of the neuronal glutamate transporter EAAC1 by slow- rons. J Neural Transm 110(12):1337–1348. doi:10.1007/
ing constitutive delivery. Neurochem Int 48(6–7):596–603. s00702-003-0049-z
doi:10.1016/j.neuint.2005.12.030 62. De Bundel D, Schallier A, Loyens E, Fernando R, Miyashita H,
47. Gonzalez MI, Krizman-Genda E, Robinson MB (2007) Cave- Van Liefferinge J, Vermoesen K, Bannai S, Sato H, Michotte
olin-1 regulates the delivery and endocytosis of the glutamate Y, Smolders I, Massie A (2011) Loss of system x(c)- does not

13
M. G. Bianchi et al.

induce oxidative stress but decreases extracellular glutamate in 77. Maragakis NJ, Dietrich J, Wong V, Xue H, Mayer-Proschel M,
hippocampus and influences spatial working memory and lim- Rao MS, Rothstein JD (2004) Glutamate transporter expression
bic seizure susceptibility. J Neurosci 31(15):5792–5803. doi:10. and function in human glial progenitors. Glia 45(2):133–143.
1523/JNEUROSCI.5465-10.2011 doi:10.1002/glia.10310
63. Li X, Valencia A, Sapp E, Masso N, Alexander J, Reeves P, 78. Abney ER, Williams BP, Raff MC (1983) Tracing the
Kegel KB, Aronin N, Difiglia M (2010) Aberrant Rab11- development of oligodendrocytes from precursor cells
dependent trafficking of the neuronal glutamate transporter using monoclonal antibodies, fluorescence-activated
EAAC1 causes oxidative stress and cell death in Huntington’s cell sorting, and cell culture. Dev Biol 100(1):166–171.
disease. J Neurosci 30(13):4552–4561. doi:10.1523/JNEURO doi:10.1016/0012-1606(83)90207-5
SCI.5865-09.2010 79. Bianchi MG, Franchi-Gazzola R, Reia L, Allegri M, Uggeri J,
64. Cao L, Li L, Zuo Z (2012) N-Acetylcysteine reverses exist- Chiu M, Sala R, Bussolati O (2012) Valproic acid induces the
ing cognitive impairment and increased oxidative stress in glutamate transporter excitatory amino acid transporter-3 in
glutamate transporter type 3 deficient mice. Neuroscience. human oligodendroglioma cells. Neuroscience 227:260–270.
doi:10.1016/j.neuroscience.2012.06.044 doi:10.1016/j.neuroscience.2012.09.055
65. Sepkuty JP, Cohen AS, Eccles C, Rafiq A, Behar K, Ganel R, 80. Hsieh J, Nakashima K, Kuwabara T, Mejia E, Gage FH (2004)
Coulter DA, Rothstein JD (2002) A neuronal glutamate trans- Histone deacetylase inhibition-mediated neuronal differentia-
porter contributes to neurotransmitter GABA synthesis and epi- tion of multipotent adult neural progenitor cells. Proc Natl Acad
lepsy. J Neurosci 22(15):6372–6379 Sci USA 101(47):16659–16664. doi:10.1073/pnas.0407643101
66. Levenson J, Weeber E, Selcher JC, Kategaya LS, Sweatt JD, 81. Shen S, Li J, Casaccia-Bonnefil P (2005) Histone modifications
Eskin A (2002) Long-term potentiation and contextual fear affect timing of oligodendrocyte progenitor differentiation in
conditioning increase neuronal glutamate uptake. Nat Neurosci the developing rat brain. J Cell Biol 169(4):577–589. doi:10.10
5(2):155–161. doi:10.1038/nn791 83/jcb.200412101
67. Pita-Almenar JD, Collado MS, Colbert CM, Eskin A (2006) 82. Bianchi MG, Gazzola GC, Tognazzi L, Bussolati O (2008)
Different mechanisms exist for the plasticity of glutamate C6 glioma cells differentiated by retinoic acid overexpress
reuptake during early long-term potentiation (LTP) and late the glutamate transporter excitatory amino acid carrier 1
LTP. J Neurosci 26(41):10461–10471. doi:10.1523/JNEURO (EAAC1). Neuroscience 151(4):1042–1052. doi:10.1016/j.
SCI.2579-06.2006 neuroscience.2007.11.055
68. Andin J, Hallbeck M, Mohammed AH, Marcusson J (2007) 83. Bianchi MG, Gazzola GC, Cagnin S, Kagechika H, Bus-
Influence of environmental enrichment on steady-state solati O (2009) The ATRA-dependent overexpression
mRNA levels for EAAC1, AMPA1 and NMDA2A recep- of the glutamate transporter EAAC1 requires RARbeta
tor subunits in rat hippocampus. Brain Res 1174:18–27. induction. Biochim Biophys Acta 1788(9):1861–1868.
doi:10.1016/j.brainres.2007.06.101 doi:10.1016/j.bbamem.2009.05.005
69. Scimemi A, Tian H, Diamond JS (2009) Neuronal transport- 84. Zhu W, Kanoh M, Ye P, Laszkiewicz I, Royland JE, Wiggins
ers regulate glutamate clearance, NMDA receptor activa- RC, Konat G (1992) Retinoic acid-regulated expression of pro-
tion, and synaptic plasticity in the hippocampus. J Neurosci teolipid protein and myelin-associated glycoprotein genes in
29(46):14581–14595. doi:10.1523/JNEUROSCI.4845-09.2009 C6 glioma cells. J Neurosci Res 31(4):745–750. doi:10.1002/
70. Do SH, Fang HY, Ham BM, Zuo Z (2002) The effects of lido- jnr.490310418
caine on the activity of glutamate transporter EAAT3: the role 85. Ferrer-Martinez A, Felipe A, Nicholson B, Casado J, Pastor-
of protein kinase C and phosphatidylinositol 3-kinase. Anesth Anglada M, McGivan J (1995) Induction of the high-affinity
Analg 95(5):1263–1268 Na(+)-dependent glutamate transport system XAG- by hyper-
71. Huang Y, Zuo Z (2003) Isoflurane enhances the expres- tonic stress in the renal epithelial cell line NBL-1. Biochem J
sion and activity of glutamate transporter type 3 in 310(Pt 2):689–692
C6 glioma cells. Anesthesiology 99(6):1346–1353. 86. Franchi-Gazzola R, Visigalli R, Dall’Asta V, Sala R, Woo SK,
doi:10.1097/00000542-200312000-00016 Kwon HM, Gazzola GC, Bussolati O (2001) Amino acid deple-
72. Lee G, Huang Y, Washington JM, Briggs NW, Zuo Z (2005) tion activates TonEBP and sodium-coupled inositol transport.
Carbamazepine enhances the activity of glutamate transporter Am J Physiol Cell Physiol 280(6):C1465–C1474
type 3 via phosphatidylinositol 3-kinase. Epilepsy Res 66(1– 87. Miyakawa H, Woo SK, Dahl SC, Handler JS, Kwon HM (1999)
3):145–153. doi:10.1016/j.eplepsyres.2005.08.003 Tonicity-responsive enhancer binding protein, a rel-like protein
73. Furuta A, Rothstein JD, Martin LJ (1997) Glutamate transporter that stimulates transcription in response to hypertonicity. Proc
protein subtypes are expressed differentially during rat CNS Natl Acad Sci USA 96(5):2538–2542
development. J Neurosci 17(21):8363–8375 88. McGivan JD, Nicholson B (1999) Regulation of high-affinity
74. Furuta A, Takashima S, Yokoo H, Rothstein JD, Wada K, Iwaki glutamate transport by amino acid deprivation and hyperos-
T (2005) Expression of glutamate transporter subtypes during motic stress. Am J Physiol 277(4 Pt 2):F498–F500
normal human corticogenesis and type II lissencephaly. Brain 89. Gazzola RF, Sala R, Bussolati O, Visigalli R, Dall’Asta V,
Res Dev Brain Res 155(2):155–164. doi:10.1016/j.devbrain Ganapathy V, Gazzola GC (2001) The adaptive regulation
res.2005.01.005 of amino acid transport system A is associated to changes in
75. Guillet B, Lortet S, Masmejean F, Samuel D, Nieoullon ATA2 expression. FEBS Lett 490(1–2):11–14. doi:10.1016/
A, Pisano P (2002) Developmental expression and activ- S0014-5793(01)02126-3
ity of high affinity glutamate transporters in rat cortical pri- 90. Nicholson B, McGivan JD (1996) Induction of high affinity
mary cultures. Neurochem Int 40(7):661–671. doi:10.1016/ glutamate transport activity by amino acid deprivation in renal
S0197-0186(01)00110-3 epithelial cells does not involve an increase in the amount of
76. Lortet S, Canolle B, Masmejean F, Nieoullon A (2008) Plasma transporter protein. J Biol Chem 271(21):12159–12164
membrane expression of the neuronal glutamate transporter 91. Ma K, Zheng S, Zuo Z (2006) The transcription factor regu-
EAAC1 is regulated by glial factors: evidence for different reg- latory factor X1 increases the expression of neuronal gluta-
ulatory pathways associated with neuronal maturation. Neuro- mate transporter type 3. J Biol Chem 281(30):21250–21255.
chem Int 52(7):1373–1382. doi:10.1016/j.neuint.2008.02.007 doi:10.1074/jbc.M600521200

13
Changes in the expression of the glutamate transporter

92. Feng C, Xu W, Zuo Z (2009) Knockout of the regulatory factor candidate gene SLC1A1/EAAC1 in early-onset obsessive-com-
X1 gene leads to early embryonic lethality. Biochem Biophys pulsive disorder. Arch Gen Psychiatry 63(7):778–785. doi:10.10
Res Commun 386(4):715–717. doi:10.1016/j.bbrc.2009.06.111 01/archpsyc.63.7.778
93. Feng C, Zuo Z (2012) Regulatory factor X1-induced down- 106. Stewart SE, Fagerness JA, Platko J, Smoller JW, Scharf JM, Ill-
regulation of transforming growth factor beta2 transcription mann C, Jenike E, Chabane N, Leboyer M, Delorme R, Jenike
in human neuroblastoma cells. J Biol Chem 287(27):22730– MA, Pauls DL (2007) Association of the SLC1A1 glutamate
22739. doi:10.1074/jbc.M111.338590 transporter gene and obsessive-compulsive disorder. Am J Med
94. Vargas MR, Johnson JA (2009) The Nrf2-ARE cytoprotec- Genet B Neuropsychiatr Genet 144B(8):1027–1033. doi:10.100
tive pathway in astrocytes. Expert Rev Mol Med 11:e17. 2/ajmg.b.30533
doi:10.1017/S1462399409001094 107. Veenstra-VanderWeele J, Xu T, Ruggiero AM, Anderson LR,
95. Escartin C, Won SJ, Malgorn C, Auregan G, Berman AE, Chen Jones ST, Himle JA, Kennedy JL, Richter MA, Hanna GL,
PC, Deglon N, Johnson JA, Suh SW, Swanson RA (2011) Arnold PD (2012) Functional studies and rare variant screening
Nuclear factor erythroid 2-related factor 2 facilitates neuronal of SLC1A1/EAAC1 in males with obsessive-compulsive disor-
glutathione synthesis by upregulating neuronal excitatory amino der. Psychiatr Genet 22(5):256–260. doi:10.1097/YPG.0b013e3
acid transporter 3 expression. J Neurosci 31(20):7392–7401. 28353fb63
doi:10.1523/JNEUROSCI.6577-10.2011 108. Schallier A, Smolders I, Van Dam D, Loyens E, De Deyn PP,
96. Aoyama K, Matsumura N, Watabe M, Wang F, Kikuchi-Utsumi Michotte A, Michotte Y, Massie A (2011) Region- and age-
K, Nakaki T (2011) Caffeine and uric acid mediate glutathione specific changes in glutamate transport in the AbetaPP23 mouse
synthesis for neuroprotection. Neuroscience 181:206–215. model for Alzheimer’s disease. J Alzheimers Dis 24(2):287–
doi:10.1016/j.neuroscience.2011.02.047 300. doi:10.3233/JAD-2011-101005
97. Linker RA, Lee DH, Ryan S, van Dam AM, Conrad R, Bista 109. Cassano T, Serviddio G, Gaetani S, Romano A, Dipasquale
P, Zeng W, Hronowsky X, Buko A, Chollate S, Ellrichmann G, P, Cianci S, Bellanti F, Laconca L, Romano AD, Padalino I,
Bruck W, Dawson K, Goelz S, Wiese S, Scannevin RH, Luka- LaFerla FM, Nicoletti F, Cuomo V, Vendemiale G (2012) Gluta-
shev M, Gold R (2011) Fumaric acid esters exert neuroprotec- matergic alterations and mitochondrial impairment in a murine
tive effects in neuroinflammation via activation of the Nrf2 model of Alzheimer disease. Neurobiol Aging 33(6):1121.e1–
antioxidant pathway. Brain 134(Pt 3):678–692. doi:10.1093/ 1121.e12. doi:10.1016/j.neurobiolaging.2011.09.021
brain/awq386 110. Duerson K, Woltjer RL, Mookherjee P, Leverenz JB, Montine
98. Scannevin RH, Chollate S, Jung MY, Shackett M, Patel H, TJ, Bird TD, Pow DV, Rauen T, Cook DG (2009) Detergent-
Bista P, Zeng W, Ryan S, Yamamoto M, Lukashev M, Rhodes insoluble EAAC1/EAAT3 aberrantly accumulates in hip-
KJ (2012) Fumarates promote cytoprotection of central nerv- pocampal neurons of Alzheimer’s disease patients. Brain Pathol
ous system cells against oxidative stress via the nuclear factor 19(2):267–278. doi:10.1111/j.1750-3639.2008.00186.x
(erythroid-derived 2)-like 2 pathway. J Pharmacol Exp Ther 111. Trotti D, Danbolt NC, Volterra A (1998) Glutamate transport-
341(1):274–284. doi:10.1124/jpet.111.190132 ers are oxidant-vulnerable: a molecular link between oxida-
99. Molteni R, Ying Z, Gomez-Pinilla F (2002) Differential effects tive and excitotoxic neurodegeneration? Trends Pharmacol Sci
of acute and chronic exercise on plasticity-related genes in 19(8):328–334. doi:10.1016/S0165-6147(98)01230-9
the rat hippocampus revealed by microarray. Eur J Neurosci 112. Martin LJ, Brambrink AM, Lehmann C, Portera-Cailliau
16(6):1107–1116. doi:10.1046/j.1460-9568.2002.02158.x C, Koehler R, Rothstein J, Traystman RJ (1997) Hypoxia-
100. Rothstein JD, Patel S, Regan MR, Haenggeli C, Huang YH, ischemia causes abnormalities in glutamate transporters and
Bergles DE, Jin L, Dykes Hoberg M, Vidensky S, Chung DS, death of astroglia and neurons in newborn striatum. Ann Neurol
Toan SV, Bruijn LI, Su ZZ, Gupta P, Fisher PB (2005) Beta- 42(3):335–348. doi:10.1002/ana.410420310
lactam antibiotics offer neuroprotection by increasing glutamate 113. Fujita H, Sato K, Wen TC, Peng Y, Sakanaka M (1999) Differ-
transporter expression. Nature 433(7021):73–77. doi:10.1038/ ential expressions of glycine transporter 1 and three glutamate
nature03180 transporter mRNA in the hippocampus of gerbils with transient
101. Ganel R, Ho T, Maragakis NJ, Jackson M, Steiner JP, Rothstein forebrain ischemia. J Cereb Blood Flow Metab 19(6):604–615.
JD (2006) Selective up-regulation of the glial Na+ -dependent doi:10.1097/00004647-199906000-00003
glutamate transporter GLT1 by a neuroimmunophilin ligand 114. Raghavendra Rao VL, Rao AM, Dogan A, Bowen KK, Hatcher
results in neuroprotection. Neurobiol Dis 21(3):556–567. J, Rothstein JD, Dempsey RJ (2000) Glial glutamate trans-
doi:10.1016/j.nbd.2005.08.014 porter GLT-1 down-regulation precedes delayed neuronal
102. Lyon L, Kew JN, Corti C, Harrison PJ, Burnet PW (2008) death in gerbil hippocampus following transient global cer-
Altered hippocampal expression of glutamate receptors and ebral ischemia. Neurochem Int 36(6):531–537. doi:10.1016/
transporters in GRM2 and GRM3 knockout mice. Synapse S0197-0186(99)00153-9
62(11):842–850. doi:10.1002/syn.20553 115. Han F, Shioda N, Moriguchi S, Qin ZH, Fukunaga K (2008)
103. Lin JA, Tsai RY, Lin YT, Lee MS, Cherng CH, Wong CS, Tzeng Downregulation of glutamate transporters is associated with
JI (2008) Amitriptyline pretreatment preserves the antinocicep- elevation in extracellular glutamate concentration following
tive effect of morphine in pertussis toxin-treated rats by lower- rat microsphere embolism. Neurosci Lett 430(3):275–280.
ing CSF excitatory amino acid concentrations and reversing the doi:10.1016/j.neulet.2007.11.021
downregulation of glutamate transporters. Brain Res 1232:61– 116. Rao VL, Dogan A, Todd KG, Bowen KK, Kim BT, Roth-
69. doi:10.1016/j.brainres.2008.07.016 stein JD, Dempsey RJ (2001) Antisense knockdown of the
104. Veenstra-VanderWeele J, Kim SJ, Gonen D, Hanna GL, Lev- glial glutamate transporter GLT-1, but not the neuronal gluta-
enthal BL, Cook EH Jr (2001) Genomic organization of the mate transporter EAAC1, exacerbates transient focal cerebral
SLC1A1/EAAC1 gene and mutation screening in early-onset ischemia-induced neuronal damage in rat brain. J Neurosci
obsessive-compulsive disorder. Mol Psychiatry 6(2):160–167. 21(6):1876–1883
doi:10.1038/sj.mp.4000806 117. Douen AG, Akiyama K, Hogan MJ, Wang F, Dong L, Chow
105. Dickel DE, Veenstra-VanderWeele J, Cox NJ, Wu X, Fischer DJ, AK, Hakim A (2000) Preconditioning with cortical spreading
Van Etten-Lee M, Himle JA, Leventhal BL, Cook EH Jr, Hanna depression decreases intraischemic cerebral glutamate levels
GL (2006) Association testing of the positional and functional and down-regulates excitatory amino acid transporters EAAT1

13
M. G. Bianchi et al.

and EAAT2 from rat cerebral cortex plasma membranes. J Neu- fluid of rats with cord ischemia: role of glutamate trans-
rochem 75(2):812–818 porters. Neuroscience 165(4):1217–1232. doi:10.1016/j.
118. Szatkowski M, Barbour B, Attwell D (1990) Non-vesicu- neuroscience.2009.11.033
lar release of glutamate from glial cells by reversed elec- 131. DeSilva TM, Kabakov AY, Goldhoff PE, Volpe JJ, Rosenberg
trogenic glutamate uptake. Nature 348(6300):443–446. PA (2009) Regulation of glutamate transport in developing rat
doi:10.1038/348443a0 oligodendrocytes. J Neurosci 29(24):7898–7908. doi:10.1523/J
119. Montori S, Martinez-Villayandre B, Dos-Anjos S, Llorente NEUROSCI.6129-08.2009
IL, Burgin TC, Fernandez-Lopez A (2010) Age-depend- 132. Ohgoh M, Hanada T, Smith T, Hashimoto T, Ueno M, Yamani-
ent modifications in the mRNA levels of the rat excita- shi Y, Watanabe M, Nishizawa Y (2002) Altered expression of
tory amino acid transporters (EAATs) at 48hour reperfu- glutamate transporters in experimental autoimmune encepha-
sion following global ischemia. Brain Res 1358:11–19. lomyelitis. J Neuroimmunol 125(1–2):170–178. doi:10.1016/
doi:10.1016/j.brainres.2010.08.020 S0165-5728(02)00029-2
120. Gottlieb M, Domercq M, Matute C (2000) Altered expres- 133. Pitt D, Werner P, Raine CS (2000) Glutamate excitotoxic-
sion of the glutamate transporter EAAC1 in neurons ity in a model of multiple sclerosis. Nat Med 6(1):67–70.
and immature oligodendrocytes after transient forebrain doi:10.1038/71555
ischemia. J Cereb Blood Flow Metab 20(4):678–687. 134. Vallejo-Illarramendi A, Domercq M, Perez-Cerda F, Ravid
doi:10.1097/00004647-200004000-00005 R, Matute C (2006) Increased expression and function of
121. Arranz AM, Gottlieb M, Perez-Cerda F, Matute C (2009) glutamate transporters in multiple sclerosis. Neurobiol Dis
Increased expression of glutamate transporters in subcortical 21(1):154–164. doi:10.1016/j.nbd.2005.06.017
white matter after transient focal cerebral ischemia. Neurobiol 135. Newcombe J, Uddin A, Dove R, Patel B, Turski L, Nishi-
Dis 37(1):156–165. doi:10.1016/j.nbd.2009.09.019 zawa Y, Smith T (2008) Glutamate receptor expression
122. Hwang I, Lee JC, Yoo KY, Cho JH, Jung JY, Kang TC, Oh YS, in multiple sclerosis lesions. Brain Pathol 18(1):52–61.
Kim WK, Won M (2007) Transient ischemia-induced changes doi:10.1111/j.1750-3639.2007.00101.x
of excitatory amino acid carrier 1 in the ventral horn of the lum- 136. Klemann C, Raveney BJ, Klemann AK, Ozawa T, von Hor-
bar spinal cord in rabbits. Neurol Res 29(3):310–316. doi:10.11 sten S, Shudo K, Oki S, Yamamura T (2009) Synthetic retinoid
79/016164107X159153 AM80 inhibits Th17 cells and ameliorates experimental autoim-
123. Kobayashi S, Millhorn DE (2001) Hypoxia regulates glutamate mune encephalomyelitis. Am J Pathol 174(6):2234–2245. doi:1
metabolism and membrane transport in rat PC12 cells. J Neuro- 0.2353/ajpath.2009.081084
chem 76(6):1935–1948 137. Huang JK, Jarjour AA, Nait Oumesmar B, Kerninon C,

124. Romera C, Hurtado O, Botella SH, Lizasoain I, Cardenas A, Williams A, Krezel W, Kagechika H, Bauer J, Zhao C,
Fernandez-Tome P, Leza JC, Lorenzo P, Moro MA (2004) In Baron-Van Evercooren A, Chambon P, Ffrench-Constant C,
vitro ischemic tolerance involves upregulation of glutamate Franklin RJ (2011) Retinoid X receptor gamma signaling
transport partly mediated by the TACE/ADAM17-tumor necro- accelerates CNS remyelination. Nat Neurosci 14(1):45–53.
sis factor-alpha pathway. J Neurosci 24(6):1350–1357. doi:10.1 doi:10.1038/nn.2702
523/JNEUROSCI.1596-03.2004 138. Crino PB, Jin H, Shumate MD, Robinson MB, Coulter DA,
125. Pradillo JM, Hurtado O, Romera C, Cardenas A, Fernandez- Brooks-Kayal AR (2002) Increased expression of the neuronal
Tome P, Alonso-Escolano D, Lorenzo P, Moro MA, Lizas- glutamate transporter (EAAT3/EAAC1) in hippocampal and
oain I (2006) TNFR1 mediates increased neuronal membrane neocortical epilepsy. Epilepsia 43(3):211–218
EAAT3 expression after in vivo cerebral ischemic precon- 139. Ross JR, Porter BE, Buckley PT, Eberwine JH, Robinson MB
ditioning. Neuroscience 138(4):1171–1178. doi:10.1016/j. (2011) mRNA for the EAAC1 subtype of glutamate trans-
neuroscience.2005.12.010 porter is present in neuronal dendrites in vitro and dramatically
126. Bigdeli MR, Hajizadeh S, Froozandeh M, Heidarianpour increases in vivo after a seizure. Neurochem Int 58(3):366–375.
A, Rasoulian B, Asgari AR, Pourkhalili K, Khoshbaten A doi:10.1016/j.neuint.2010.12.012
(2008) Normobaric hyperoxia induces ischemic tolerance 140. Zhang G, Raol YS, Hsu FC, Brooks-Kayal AR (2004)
and upregulation of glutamate transporters in the rat brain Long-term alterations in glutamate receptor and transporter
and serum TNF-alpha level. Exp Neurol 212(2):298–306. expression following early-life seizures are associated with
doi:10.1016/j.expneurol.2008.03.029 increased seizure susceptibility. J Neurochem 88(1):91–101.
127. Bigdeli MR, Rahnema M, Khoshbaten A (2009) Precondition- doi:10.1046/j.1471-4159.2003.02124.x
ing with sublethal ischemia or intermittent normobaric hyper- 141. Doi T, Ueda Y, Nagatomo K, Willmore LJ (2009) Role of glu-
oxia up-regulates glutamate transporters and tumor necrosis tamate and GABA transporters in development of pentylenetet-
factor-alpha converting enzyme in the rat brain. J Stroke Cer- razol-kindling. Neurochem Res 34(7):1324–1331. doi:10.1007/
ebrovasc Dis 18(5):336–342. doi:10.1016/j.jstrokecerebrovas s11064-009-9912-0
dis.2008.12.005 142. Voutsinos-Porche B, Koning E, Clement Y, Kaplan H, Ferran-
128. Won SJ, Yoo BH, Brennan AM, Shin BS, Kauppinen TM, Ber- don A, Motte J, Nehlig A (2006) EAAC1 glutamate transporter
man AE, Swanson RA, Suh SW (2010) EAAC1 gene deletion expression in the rat lithium-pilocarpine model of temporal lobe
alters zinc homeostasis and exacerbates neuronal injury after epilepsy. J Cereb Blood Flow Metab 26(11):1419–1430. doi:10.
transient cerebral ischemia. J Neurosci 30(46):15409–15418. 1038/sj.jcbfm.9600295
doi:10.1523/JNEUROSCI.2084-10.2010 143. Simantov R, Crispino M, Hoe W, Broutman G, Tocco G, Roth-
129. Li L, Zuo Z (2011) Glutamate transporter type 3 knock- stein JD, Baudry M (1999) Changes in expression of neuronal
out reduces brain tolerance to focal brain ischemia in mice. J and glial glutamate transporters in rat hippocampus follow-
Cereb Blood Flow Metab 31(5):1283–1292. doi:10.1038/jc ing kainate-induced seizure activity. Brain Res Mol Brain Res
bfm.2010.222 65(1):112–123. doi:10.1016/S0169-328X(98)00349-0
130. Chen WF, Sung CS, Jean YH, Su TM, Wang HC, Ho JT, 144. Harrington EP, Moddel G, Najm IM, Baraban SC (2007) Altered
Huang SY, Lin CS, Wen ZH (2010) Suppressive effects of glutamate receptor—transporter expression and spontaneous
intrathecal granulocyte colony-stimulating factor on excessive seizures in rats exposed to methylazoxymethanol in utero. Epi-
release of excitatory amino acids in the spinal cerebrospinal lepsia 48(1):158–168. doi:10.1111/j.1528-1167.2006.00838.x

13
Changes in the expression of the glutamate transporter

145. Lu Z, Zhang W, Zhang N, Jiang J, Luo Q, Qiu Y (2008) The transcripts in the thalamus of subjects with schizophrenia. Am J
expression of glutamate transporters in chest compression- Psychiatry 158(9):1393–1399
induced audiogenic epilepsy: a comparative study. Neurol Res 153. Shan D, Lucas EK, Drummond JB, Haroutunian V, Meador-
30(9):915–919. doi:10.1179/174313208X327964 Woodruff JH, McCullumsmith RE (2013) Abnormal expres-
146. Proper EA, Hoogland G, Kappen SM, Jansen GH, Rensen MG, sion of glutamate transporters in temporal lobe areas in elderly
Schrama LH, van Veelen CW, van Rijen PC, van Nieuwenhui- patients with schizophrenia. Schizophr Res 144(1–3):1–8.
zen O, Gispen WH, de Graan PN (2002) Distribution of glu- doi:10.1016/j.schres.2012.12.019
tamate transporters in the hippocampus of patients with phar- 154. Bauer D, Gupta D, Harotunian V, Meador-Woodruff JH,
maco-resistant temporal lobe epilepsy. Brain 125(Pt 1):32–43 McCullumsmith RE (2008) Abnormal expression of glutamate
147. Mathern GW, Mendoza D, Lozada A, Pretorius JK, Dehnes Y, transporter and transporter interacting molecules in prefrontal
Danbolt NC, Nelson N, Leite JP, Chimelli L, Born DE, Saka- cortex in elderly patients with schizophrenia. Schizophr Res
moto AC, Assirati JA, Fried I, Peacock WJ, Ojemann GA, Adel- 104(1–3):108–120. doi:10.1016/j.schres.2008.06.012
son PD (1999) Hippocampal GABA and glutamate transporter 155. Rao JS, Kellom M, Reese EA, Rapoport SI, Kim HW (2012)
immunoreactivity in patients with temporal lobe epilepsy. Neu- Dysregulated glutamate and dopamine transporters in postmor-
rology 52(3):453–472 tem frontal cortex from bipolar and schizophrenic patients. J
148. Rakhade SN, Loeb JA (2008) Focal reduction of neuronal glu- Affect Disord 136(1–2):63–71. doi:10.1016/j.jad.2011.08.017
tamate transporters in human neocortical epilepsy. Epilepsia 156. Lauriat TL, Dracheva S, Chin B, Schmeidler J, McInnes LA,
49(2):226–236. doi:10.1111/j.1528-1167.2007.01310.x Haroutunian V (2006) Quantitative analysis of glutamate trans-
149. Dutuit M, Touret M, Szymocha R, Nehlig A, Belin MF, Didier- porter mRNA expression in prefrontal and primary visual cortex
Bazes M (2002) Decreased expression of glutamate transport- in normal and schizophrenic brain. Neuroscience 137(3):843–
ers in genetic absence epilepsy rats before seizure occurrence. J 851. doi:10.1016/j.neuroscience.2005.10.003
Neurochem 80(6):1029–1038 157. McCullumsmith RE, Meador-Woodruff JH (2002) Striatal
150. Samadani U, Judkins AR, Akpalu A, Aronica E, Crino PB (2007) excitatory amino acid transporter transcript expression in
Differential cellular gene expression in ganglioglioma. Epilep- schizophrenia, bipolar disorder, and major depressive disor-
sia 48(4):646–653. doi:10.1111/j.1528-1167.2007.00925.x der. Neuropsychopharmacology 26(3):368–375. doi:10.1016/
151. White R, Hua Y, Scheithauer B, Lynch DR, Henske EP, Crino S0893-133X(01)00370-0
PB (2001) Selective alterations in glutamate and GABA recep- 158. Nudmamud-Thanoi S, Piyabhan P, Harte MK, Cahir M, Reyn-
tor subunit mRNA expression in dysplastic neurons and giant olds GP (2007) Deficits of neuronal glutamatergic markers in
cells of cortical tubers. Ann Neurol 49(1):67–78 the caudate nucleus in schizophrenia. J Neural Transm Suppl
152. Smith RE, Haroutunian V, Davis KL, Meador-Woodruff 72:281–285
JH (2001) Expression of excitatory amino acid transporter

13

You might also like