ArticlePDF Available

Extrasynaptic glutamate release through cystine/glutamate antiporter contributes to ischemic damage

Authors:

Abstract and Figures

During brain ischemia, an excessive release of glutamate triggers neuronal death through the overactivation of NMDA receptors (NMDARs); however, the underlying pathways that alter glutamate homeostasis and whether synaptic or extrasynaptic sites are responsible for excess glutamate remain controversial. Here, we monitored ischemia-gated currents in pyramidal cortical neurons in brain slices from rodents in response to oxygen and glucose deprivation (OGD) as a real-time glutamate sensor to identify the source of glutamate release and determined the extent of neuronal damage. Blockade of excitatory amino acid transporters or vesicular glutamate release did not inhibit ischemia-gated currents or neuronal damage after OGD. In contrast, pharmacological inhibition of the cystine/glutamate antiporter dramatically attenuated ischemia-gated currents and cell death after OGD. Compared with control animals, mice lacking a functional cystine/glutamate antiporter exhibited reduced anoxic depolarization and neuronal death in response to OGD. Furthermore, glutamate released by the cystine/glutamate antiporter activated extrasynaptic, but not synaptic, NMDARs, and blockade of extrasynaptic NMDARs reduced ischemia-gated currents and cell damage after OGD. Finally, PET imaging showed increased cystine/glutamate antiporter function in ischemic rats. Altogether, these data suggest that cystine/glutamate antiporter function is increased in ischemia, contributing to elevated extracellular glutamate concentration, overactivation of extrasynaptic NMDARs, and ischemic neuronal death.
Inhibition of the cystine/glutamate antiporter reduces the OGD-induced current, DC depolarization, and cell death. ( A ) Voltage-clamp recording of OGD-activated currents in the absence or presence of system xc – inhibitors CPG (250 μ M; n = 12) and SAS (250 μ M; n = 10). Both treatments significantly inhibited the OGD-activated current ( n = 25). The mGluR 1 inhibitor AIDA (500 μ M; n = 6) had no effect on anoxic current amplitude or on latency. ( B ) Histograms show the average OGD-induced current amplitude (pA ± SEM) and latency (minutes ± SEM) for each condition. ** P < 0.01 and * P < 0.05 versus OGD. ( C ) AD was recorded as a negative DC field voltage shift in acute cortical slices subjected to OGD in the absence or presence of the ionotropic glutamate receptor antagonists AP5 and CNQX or of the cystine/glutamate antiporter inhibitor CPG. Histogram shows the average DC potential shift (mV ± SEM) for each condition. ** P < 0.01 and * P < 0.05 versus OGD. ( D ) TBOA applied concomitantly with CPG did not enhance the CPG-induced decrease of the OGD current amplitude, but reduced the latency to AD ( n = 6). ( E ) Cell death measured by LDH release in cultures subjected to 45 minutes of OGD applying system xc – inhibitors CPG (250 μ M; n = 5) and SAS (250 μ M; n = 5), NMDAR antagonist MK-801 (50 μ M, n = 5), and mGluR 1 antagonist AIDA (500 μ M; n = 3). LDH was measured after 24 hours of reoxygenation, and data are expressed as the mean ± SEM. *P < 0.05 and **P < 0.01 versus OGD. ( F ) Representative fields demonstrate propidium iodide labeling of organotypic slices treated as in E . Scale bar: 1 mm.
… 
Content may be subject to copyright.
The Journal of Clinical Investigation ReseaRch aRticle
3645
jci.org Volume 124 Number 8 August 2014
Introduction
Brain ischemia is the fourth cause of death and the leading cause
of long-term disability in industrialized countries . Relatively
short periods of blood ow interruption in the brain can pro-
duce irreversible neuronal damage . Energy failure and ox-
ygen deprivation that occur in ischemic episodes induce a loss
of membrane potential in neurons and glia, a process known
as anoxic depolarization AD, which spreads across suscepti-
ble brain tissue as a self-propagating wave-like depolarization
,  and can be initiated by factors that release K and gluta-
mate . Recordings from neurons in hippocampal and cerebel-
lar slices have shown that this AD is associated with a large gluta-
mate-evoked inward current, which can be blocked by a cocktail
of agents blocking ionotropic glutamate receptors , . In par-
ticular, activation of NMDA receptors NMDARs plays a crucial
role in neuronal cell death .
NMDAR-mediated signaling can be either benecial or
deleterious, and this dichotomous behavior has been proposed
to be related to its localization within or outside the synapse
, . Activation of NMDARs in synapses provides plasticity and
cell survival signals, whereas extrasynaptic NMDARs trigger
neurodegeneration refs. ; but see also refs. , . These
opposing signals are transduced to and discriminated by the nu-
cleus on the basis of the dierential phosphorylation state of the
Jacob protein messenger . However, most of these data have
been obtained in vitro, and the role of synaptic or extrasynaptic
NMDARs in ischemic neuronal damage has not been studied in
a more intact preparation.
The functional dichotomy of NMDAR signaling would also
depend on the location of the glutamate source. A major source
of extrasynaptic glutamate is the cystine/glutamate antiporter
, also known as system xc, a solute carrier identied as the
main source of nonsynaptic glutamate in the brain .
This transport system is a membrane-bound, Cl-dependent,
Na-independent antiporter that mediates the cellular uptake of
cystine in exchange for glutamate at a : ratio . Struc-
turally, it is a heterodimer composed of a heavy-chain subunit,
Fhc, and a light-chain–specic subunit, xCT . System
xc is an important source of cystine, which is intracellularly
converted to cysteine, the rate-limiting substrate in glutathione
synthesis . The high rate of oxygen consumption in the brain
renders this antiporter vital to antioxidant defense , and its
expression is rapidly upregulated in vitro under conditions of
oxidative stress , . Nonetheless, the obligate exchange of
During brain ischemia, an excessive release of glutamate triggers neuronal death through the overactivation of NMDA
receptors (NMDARs); however, the underlying pathways that alter glutamate homeostasis and whether synaptic or
extrasynaptic sites are responsible for excess glutamate remain controversial. Here, we monitored ischemia-gated currents
in pyramidal cortical neurons in brain slices from rodents in response to oxygen and glucose deprivation (OGD) as a real-time
glutamate sensor to identify the source of glutamate release and determined the extent of neuronal damage. Blockade of
excitatory amino acid transporters or vesicular glutamate release did not inhibit ischemia-gated currents or neuronal damage
after OGD. In contrast, pharmacological inhibition of the cystine/glutamate antiporter dramatically attenuated ischemia-gated
currents and cell death after OGD. Compared with control animals, mice lacking a functional cystine/glutamate antiporter
exhibited reduced anoxic depolarization and neuronal death in response to OGD. Furthermore, glutamate released by the
cystine/glutamate antiporter activated extrasynaptic, but not synaptic, NMDARs, and blockade of extrasynaptic NMDARs
reduced ischemia-gated currents and cell damage after OGD. Finally, PET imaging showed increased cystine/glutamate
antiporter function in ischemic rats. Altogether, these data suggest that cystine/glutamate antiporter function is increased
in ischemia, contributing to elevated extracellular glutamate concentration, overactivation of extrasynaptic NMDARs, and
ischemic neuronal death.
Extrasynaptic glutamate release through
cystine/glutamate antiporter contributes
to ischemic damage
Federico N. Soria,1 Alberto Pérez-Samartín,1 Abraham Martin,2 Kiran Babu Gona,3 Jordi Llop,3 Boguslaw Szczupak,2
Juan Carlos Chara,1 Carlos Matute,1 and María Domercq1
1Centro de Investigaciones Biomédicas en Red (CIBERNED), Achucarro Basque Center for Neuroscience and Departamento de Neurociencias, Universidad del País Vasco (UPV/EHU), Leioa, Spain.
2Molecular Imaging Unit and 3Radiochemistry Department, CIC biomaGUNE, San Sebastián, Spain.
Related Commentary: p. 3279
Conflict of interest: The authors have declared that no conflict of interest exists.
Submitted: July 9, 2013; Accepted: May 21, 2014.
Reference information: J Clin Invest. 2014;124(8):3645–3655. doi:10.1172/JCI71886.
Downloaded August 22, 2014 from The Journal of Clinical Investigation. doi:10.1172/JCI71886.
The Journal of Clinical Investigation
ReseaRch aRticle
3646 jci.org Volume 124 Number 8 August 2014
accumulation. Although these results do not rule out that OGD
partially reduces glutamate uptake or that a minor population of
EAATs could indeed be working in reverse-uptake mode, they
strongly suggest that another source of glutamate contributes to
the alteration of glutamate homeostasis in ischemia.
We also tested the eect of EAAT blockade in cell death using
organotypic cortical slices . TBOA exacerbated cell death af-
ter OGD and reoxygenation, as assessed by lactic dehydrogenase
LDH release assays Figure D, and see also in Supplemental
Figure  the eect of TBOA in normoxia; supplemental material
available online with this article; doi:./JCIDS. Paral-
lel staining with propidium iodide revealed cell death in the cortex,
which was proportional to the damage quantied by LDH release
Figure E. TBOA-mediated exacerbation in excitotoxic damage in
hypoxia/ischemia has also been observed in cultured neurons 
and organotypic cortical slices , conrming the primary physi-
ological role of EAATs in the clearance of extracellular glutamate.
Inhibition of exocytosis has no eect on ischemia-gated currents
or cell death. An increase in vesicular synaptic glutamate release
and in miniature excitatory postsynaptic currents mEPSCs oc-
curs at early phases of ischemia and before AD , , . We
therefore tested whether vesicular release was contributing to
the increase in AD currents in acute slices by using balomycin
A Baf A;  μM, a vacuolar type H-ATPase inhibitor, to block
neurotransmitter loading into synaptic vesicles. Baf A practically
abolished the mEPSCs Supplemental Figure , thus indicating
an eective inhibition of spontaneous vesicular glutamate re-
lease. In contrast, we found that neither the latency nor the ampli-
tude of the anoxic current was changed when Baf A was present
at the onset of OGD Figure , A and B. Accordingly, neuronal
damage was not signicantly dierent in Baf A–treated  μM
or tetanus toxin–treated TeTN-treated;  μg/ml slices compared
with OGD alone Figure , C and D. These results suggest that
vesicular release after OGD does not signicantly contribute to
AD currents and toxicity and are consistent with an extrasynaptic
source of glutamate in ischemia.
Blockade of the cystine/glutamate exchanger reduces ischemia-
gated currents and neuronal damage after OGD. The cystine/
glutamate antiporter reportedly regulates extracellular gluta-
mate concentration in physiological ,  and pathological
conditions , , . We next analyzed whether ischemia-
induced glutamate release might be caused as a result of cystine
exchange by system xc. To analyze the possible contribution
of the cystine/glutamate antiporter to glutamate release dur-
ing OGD, we applied S-carboxyphenylglycine CPG;  μM
Figure A, a nonsubstrate inhibitor of the antiporter refs. ,
, and see also Supplemental Figure . We found that although
CPG did not change latency to AD, it signicantly reduced its
amplitude Figure , A and B. We further assessed the eect of
CPG on extracellular direct-current DC eld potentials 
and showed that CPG signicantly reduced the ΔVo amplitude
after OGD Figure C. Because CPG is also a competitive in-
hibitor of group I metabotropic glutamate receptors mGluR
, we also tested sulfasalazine SAS;  μM, another non-
substrate inhibitor of the cystine/glutamate antiporter , 
with no anity for glutamate receptors, and obtained a similar
attenuation of the AD current Figure , A and B. In contrast,
glutamate, which is released into the extracellular space, could
be deleterious to neuronal cells and other tissues that are sus-
ceptible to excitotoxic damage. Accordingly, the cystine/gluta-
mate antiporter is implicated in glutamate-associated disorders
such as glioma-derived epileptic seizures , oxidative gluta-
mate toxicity , and excitotoxic oligodendroglial death .
Although ischemia is a disorder that occurs in an environ-
ment of oxidative stress and lack of nutrients, which are induc-
ers of the cystine/glutamate antiporter , its contribution to
glutamate homeostasis alteration and neuronal cell death after
ischemia has not been explored before. Here, we analyze the
contribution of dierent mechanisms of glutamate release, in-
cluding the cystine/glutamate antiporter, and the role of synap-
tic versus extrasynaptic NMDARs in ischemia-gated currents
and neuronal damage.
Results
Inhibition of glutamate transporters shortens the latency to AD and
exacerbates neuronal damage in OGD. Na-dependent excitatory
amino acid transporters EAATs could contribute to alter gluta-
mate homeostasis during ischemic insults by  mechanisms: a
decreased glutamate uptake, b reverse transport, and c hete-
roexchange. Severe chemical ischemic conditions decrease net
glutamate uptake within  to  minutes , and at later stages,
ischemia promotes reverse glutamate transport , . We there-
fore tested the eect of a broad-spectrum nonsubstrate antagonist
of EAATs, DL-threo-β-benzyloxyaspartic acid TBOA;  μM on
OGD-gated currents in pyramidal cells from acute cortical slices,
and its eect on neuronal cell death in organotypic cortical slices.
OGD activated a large inward current within  minutes Figure
, A and B, which corresponds to AD. This current was mainly
mediated by glutamate receptors, since a cocktail of ionotropic
glutamate receptor antagonists  μM AP plus  μM CNQX
applied concomitantly with OGD greatly reduced OGD-gated
currents Figure , A and B. Reduction of AD was not complete,
suggesting that other channels, such as the acid-sensitive anion
channel , the PX receptor , , and pannexin  , also
contribute to the OGD current.
Application of TBOA during OGD did not decrease the am-
plitude of the OGD-activated current Figure , A and B. Fur-
thermore, TBOA reduced the latency to onset of AD in  min-
utes Figure B, as previously observed by others . This
implies that in this paradigm, most transporters are still active
and work in direct uptake mode at the beginning of ischemia,
since its chronic blockade during OGD results in increased con-
centrations of glutamate and reduced latencies to AD. Indeed,
we found that OGD-activated currents in the presence of TBOA
were similarly blocked by glutamate receptor antagonists Fig-
ure , A and B. EAATs reverse as a result of alterations in the
transmembrane gradients once AD has been initiated . Thus,
to ensure that we were blocking EAATs when they are mostly in
reverse-uptake mode, we applied TBOA at the peak of the AD.
Blockade of EAATs after the onset of AD still potentiated the
amplitude of AD current Figure C, suggesting that, despite
the strongly reduced transmembrane sodium and potassium
concentration gradients, EAAT still takes up glutamate during
AD, and its blockade further enhances extracellular glutamate
Downloaded August 22, 2014 from The Journal of Clinical Investigation. doi:10.1172/JCI71886.
The Journal of Clinical Investigation ReseaRch aRticle
3647
jci.org Volume 124 Number 8 August 2014
Next, we investigated whether
pharmacological inhibition of the
cystine/glutamate antiporter re-
duced ischemic damage in cerebral
cortex organotypic slices. Inhi-
bition of system xc with CPG
and SAS  μM signicantly
reduced neuronal damage after
OGD Figure , E and F in a way
similar to the neuroprotection ob-
served with NMDAR antagonist
MK  μM; Figure , E and
F. In contrast, no inhibition of
OGD-induced neuronal damage
was detected with the mGluR an-
tagonist AIDA  μM; Figure ,
E and F. To further conrm the
role played by the cystine/gluta-
mate antiporter, we performed
additional experiments in mutant
mice lacking a functional system
xcSlcasut. These mice 
have a large spontaneous deletion
from intron  to the adjacent sut
region of the Slca gene, leading
to a truncated and nonfunctional
xCT protein. As observed with
the system xc inhibitors, sut mice
showed a signicant reduction in
anoxic currents Figure , A and
B, DC extracellular potential
shifts Figure C, and neuronal
damage after OGD Figure , D
and E. Altogether, these data sug-
gest that the cystine/glutamate
antiporter is a relevant source
of glutamate release during is-
chemia and that its inhibition is
neuroprotective against ischemic
neuronal damage.
Cystine/glutamate antiporter
activates extrasynaptic NMDARs
in cortical neurons. Although the
cystine/glutamate antiporter has been proposed as an important
source of nonsynaptic glutamate , , , there is no direct
link between glutamate release by this transport system and the
activation of extrasynaptic glutamate receptors. Previous data
showed that glutamate release by the cystine/glutamate anti-
porter evokes glutamate receptor–mediated inward currents in
Purkinje cells upon acute exposure to cystine . Hence, we next
analyzed whether glutamate release induced by acute cystine ap-
plication could activate synaptic and/or extrasynaptic NMDARs.
Cortical neurons were clamped at – mV to remove Mg block
from NMDARs, and cystine  mM was applied concomitantly
with glycine  μM. Cystine evoked an inward current in most
cells, with a mean value    pA, while glycine alone only in-
duced small inward currents Figure . Cystine-evoked currents
AIDA  μM, a selective inhibitor of mGluR, did not aect
the OGD-activated current Figure , A and B, excluding any
role of these receptors in the eect observed with CPG. Interest-
ingly, we did not observe an increase in the amplitude of OGD
currents in TBOA-treated slices in the presence of CPG com-
pare Figure , A and B with Figure , B and D, which suggests
that blockade of glutamate uptake by EAATs does not increase
glutamate extracellular levels when the source of glutamate,
cystine/glutamate antiporter, is inhibited. These data are in ac-
cordance with data in normoxic conditions  and reinforce
the important role played by the cystine/glutamate antiporter in
glutamate homeostasis during ischemia. It should be stressed,
however, that the latency to AD was still reduced when TBOA
was present along with CPG Figure B and see also Figure B.
Figure 1. Blockade of EAATs during OGD contributes to extracellular glutamate accumulation in acute slices
and organotypic cultures. (A) Voltage-clamp recording of cortical neurons at 30 mV in acute slices demon-
strates activation of a large current after OGD (n = 32). Ionotropic glutamate receptor antagonists AP5
(100 μM) plus CNQX (30 μM) significantly inhibited the OGD-activated current (n = 9). Nonsubstrate
broad-spectrum EAAT inhibitor DL-TBOA (100 μM) did not significantly change (but tended to increase) the
amplitude of the OGD-induced current and shortened the onset of AD in cortical neurons (n = 12), an eect
abolished when applied concomitantly with AP5 plus CNQX (n = 7). (B) Histograms showing the average am-
plitude (pA ± SEM) and latency (minutes ± SEM) of the OGD-induced current for each condition. Latency was
significantly reduced in the presence of TBOA. *P < 0.05 versus OGD. (C) TBOA increased the OGD-activated
current when applied after the onset of AD (n = 11). (D) Inhibition of EAATs exacerbated OGD-induced cell death
in organotypic slices, as demonstrated by increased LDH release in cultures subjected to 45 minutes of OGD and
24 hours of reoxygenation in the presence or absence of TBOA (100 μM). Data are expressed as the mean ± SEM
(n = 3–5). **P < 0.01 and *P < 0.05 versus OGD. (E) Representative fields demonstrate propidium iodide labeling
in cortical layers of slices treated as in D. Scale bar: 1 mm.
Downloaded August 22, 2014 from The Journal of Clinical Investigation. doi:10.1172/JCI71886.
The Journal of Clinical Investigation
ReseaRch aRticle
3648 jci.org Volume 124 Number 8 August 2014
trasynaptic receptors without interfering with synaptic activity ,
, also reduced the anoxic current Figure , A and C. None of
the antagonists, independently of their subunit specicity, altered
the latency to the anoxic current Figure C. These results suggest
that NRB-containing receptors play a role in AD in ischemia.
Finally, we checked the contribution of synaptic and extra-
synaptic NMDARs to neuronal damage after OGD in organotypic
cortical cultures. The NRA antagonists NVPAAM , 
and TCN  did not reduce cell death signicantly Figure
, D and E. It should be noted, however, that high concentra-
tions of glycine in culture medium during reperfusion may reduce
TCN eectiveness . In contrast, memantine  μM and
RO  μM produced a massive reduction in OGD-induced
cell death Figure , D and E. Neuroprotection with memantine
was similar to that obtained with the NMDAR antagonist MK
 μM; Figure , D and E. Altogether, these data indicate that
activation of extrasynaptic NMDARs contributes substantially to
excitotoxic death during OGD in organotypic cortical cultures and
suggest that these receptors are likely activated by extrasynaptic
tonic release of glutamate from the cystine/glutamate antiporter.
xCT expression and function are increased after ischemia in vitro
and in vivo. The excessive release of glutamate that causes neu-
ronal death during brain ischemia takes place in an environment of
oxidative stress , a condition that favors an increase in the ex-
pression and function of the cystine/glutamate antiporter , .
Therefore, we next analyzed the expression levels of the cata-
lytic subunit of the cystine/glutamate antiporter xCT in cortical
neuron cultures exposed to chemical ischemia in vitro, ischemic
conditions that induced massive cell death .  . and
reproduced metabolic failure in the ischemic core. Real-time
quantitative PCR qPCR analysis demonstrated that xCT mRNA
was signicantly more abundant  in neurons after  hour
of chemical ischemia Figure A. To examine xCT levels by im-
munoblotting, total protein was extracted  hours after chemical
ischemia to allow the translational machinery to produce xCT.
were completely abolished in the presence of the NMDA antago-
nist AP  μM; Figure . To exclude a direct action of cystine
plus glycine,  μM on NMDARs, we applied cystine in the pres-
ence of the cystine/glutamate antiporter blocker CPG  μM.
Cystine-evoked currents were signicantly inhibited by CPG
Figure , suggesting that cystine evokes glutamate release and
indirectly activates NMDARs.
According to their localization in the cell membrane, NMDARs
dier in their subunit composition. Thus, synaptic NMDARs are
enriched in NRA subunits, while NRB subunits are located
mainly in extrasynaptic NMDARs , . This feature allows
for pharmacological blockade of either subtype by using subun-
it-specic antagonists. RO  μM, an NRB antagonist
, almost totally blocked cystine-evoked inward currents Fig-
ure . In contrast, NVPAAM  nM, which preferentially
inhibits NRA-containing receptors , did not change the am-
plitude of cystine-evoked inward currents Figure . These data
suggest that glutamate release in exchange with cystine activates
preferentially extrasynaptic NMDARs.
Antagonists of extrasynaptic, but not of synaptic, NMDARs at-
tenuate ischemia-gated currents and cell death after OGD. If the
cystine/glutamate exchanger regulates extrasynaptic glutamate
levels as previously demonstrated Figure , then glutamate re-
leased during OGD would favor the activation of extrasynaptic
NMDARs over synaptic ones. Hence, we analyzed the contri-
bution of synaptic and extrasynaptic NMDARs to ischemic in-
ward currents and neuronal damage. We observed a signicant
reduction in OGD-activated currents in the presence of ifen-
prodil  μM; Figure , A and C, a known NRB antagonist .
RO  μM, a more potent antagonist of NRB , pro-
duced a -fold reduction in the amplitude of the OGD-activated
current Figure , A and C. In contrast, the NRA antagonists
NVPAAM  nM and TCN  μM ,  did not
change the amplitude of OGD-activated currents Figure , B and
C. Finally, memantine, which at  μM preferentially blocks ex-
Figure 2. Inhibition of vesicular glutamate release does not
change anoxic current or cell death in OGD. (A) OGD-activated
currents (recorded at 30 mV) in the absence or presence of
vesicular fusion inhibitor Baf A1 (μM). (B) Histograms show the
average amplitude (pA ± SEM) and latency (minutes ± SEM) of
the OGD-induced current for each condition. Baf did not change
the amplitude or latency of the OGD-activated current (n = 7). (C)
Cell death measured by LDH release in cultures subjected to 45
minutes of OGD and 24 hours of reperfusion in the presence or
absence of the exocytosis inhibitors Baf (1 μM) and TeNT (1 μg/ml).
Data are expressed as the mean ± SEM (n = 3–5). *P < 0.001 versus
OGD. (D) Representative fields showing propidium iodide labeling
of organotypic slices treated as in C. Scale bar: 1 mm.
Downloaded August 22, 2014 from The Journal of Clinical Investigation. doi:10.1172/JCI71886.
The Journal of Clinical Investigation ReseaRch aRticle
3649
jci.org Volume 124 Number 8 August 2014
of Cl. Specicity was further
assessed using the system xc
competitive inhibitor aminoad-
ipic acid AAA;  μM, which
signicantly blocked C
L-cystine uptake. In accordance
with Western blot data, cystine
uptake by the antiporter was in-
creased in neurons subjected to
chemical ischemia Figure C.
Altogether, these data reveal
an increase in neuronal xCT
expression and function after in
vitro ischemia.
Next we analyzed the func-
tion of cystine/glutamate anti-
porter short times  minutes
and  hours after inducing
transient focal ischemia via
middle cerebral artery occlu-
sion MCAO in rats. In order to
measure xCT function in vivo,
we took advantage of the re-
cently developed radioligand
S---Ffluoropropyl-
L-glutamate FFSPG to im-
age xCT transporter activity
by PET . As previously de-
scribed , the brain showed
low FFSPG uptake. However,
we observed tracer accumu-
lation as a result of higher ac-
tivity of the cystine/glutamate
antiporter in the ipsilateral
hemisphere immediately after
initiation of reperfusion PET
imaging could not be performed
during MCAO, and this accu-
mulation peaked at the striatum
at  hours Figure , D and E.
This is in agreement with the fact
that MCAO induces early striatal
infarction  hours but delayed
necrosis in the cortex  hours
. Altogether, these data indi-
cate that the cystine/glutamate
antiporter increases its expression and activity following ischemia
and further support the relevance of this antiporter in glutamate
homeostasis alteration in stroke.
Discussion
While glutamate-induced excitotoxicity is widely accepted as a
critical event that leads to neuronal death during ischemia, several
other questions have yet to be answered. For instance, the mecha-
nisms behind the alteration in glutamate homeostasis and the bal-
ance between the release and clearance of glutamate in ischemia
are not yet completely understood, and the location of the gluta-
Western blot analysis revealed a -fold increase in xCT protein in
neurons exposed to chemical ischemia Figure B. This increase
in xCT levels at  hours of reoxygenation is consistent with the
fact that reactive oxygen species are generated not only during
ischemia, but also during the reoxygenation period as a result of
the activation of NADPH oxidase . To conrm these nd-
ings, we next examined the cystine transport activity of system
xc by measuring C L-cystine uptake in neurons exposed to
 hour of chemical ischemia plus  hours of reoxygenation. Spe-
cic Cl-dependent cystine uptake by system xc was dened
as the dierential cystine uptake in the presence and absence
Figure 3. Inhibition of the cystine/glutamate antiporter reduces the OGD-induced current, DC depolarization,
and cell death. (A) Voltage-clamp recording of OGD-activated currents in the absence or presence of system xc
inhibitors CPG (250 μM; n = 12) and SAS (250 μM; n = 10). Both treatments significantly inhibited the OGD-acti-
vated current (n = 25). The mGluR1 inhibitor AIDA (500 μM; n = 6) had no eect on anoxic current amplitude or
on latency. (B) Histograms show the average OGD-induced current amplitude (pA ± SEM) and latency (minutes ±
SEM) for each condition. **P < 0.01 and *P < 0.05 versus OGD. (C) AD was recorded as a negative DC field voltage
shift in acute cortical slices subjected to OGD in the absence or presence of the ionotropic glutamate receptor
antagonists AP5 and CNQX or of the cystine/glutamate antiporter inhibitor CPG. Histogram shows the average DC
potential shift (mV ± SEM) for each condition. **P < 0.01 and *P < 0.05 versus OGD. (D) TBOA applied concomi-
tantly with CPG did not enhance the CPG-induced decrease of the OGD current amplitude, but reduced the latency
to AD (n = 6). (E) Cell death measured by LDH release in cultures subjected to 45 minutes of OGD applying system
xc inhibitors CPG (250 μM; n = 5) and SAS (250 μM; n = 5), NMDAR antagonist MK-801 (50 μM, n = 5), and mGluR1
antagonist AIDA (500 μM; n = 3). LDH was measured after 24 hours of reoxygenation, and data are expressed as
the mean ± SEM. *P < 0.05 and **P < 0.01 versus OGD. (F) Representative fields demonstrate propidium iodide
labeling of organotypic slices treated as in E. Scale bar: 1 mm.
Downloaded August 22, 2014 from The Journal of Clinical Investigation. doi:10.1172/JCI71886.
The Journal of Clinical Investigation
ReseaRch aRticle
3650 jci.org Volume 124 Number 8 August 2014
mate receptors that trigger the death signal in ischemia is still con-
troversial. The data reported here provide evidence that glutamate
release by the cystine/glutamate antiporter contributes to ische-
mia-induced anoxic currents and neuronal damage. Electrophys-
iology data suggest that glutamate release by system xc activates
mainly extrasynaptic NMDARs. Accordingly, blocking extrasyn-
aptic, but not synaptic, NMDARs reduces ischemia-induced cur-
rents and cell damage. Finally, in vivo PET imaging indicated that
system xc function was robustly increased during ischemia and
reperfusion, suggesting a further contribution to glutamate re-
lease and cell death. To our knowledge, this is the rst report of
the contribution of the cystine/glutamate antiporter to glutamate
homeostasis alteration and subsequent cell death in ischemia.
Activation of the cell death program in neurons appears to be
linked to the overstimulation of extrasynaptic NMDARs , .
Our data suggest that extrasynaptic NRB-containing receptors
are the main contributors to anoxic currents and neuronal damage
in OGD. Accordingly, previous data showed that cerebral ischemia
recruits death-associated protein kinase  DAPK to the NRB
complex at extrasynaptic sites, and this interaction functions as a
central mediator of stroke damage . In contrast, preblocking
synaptic NMDARs with MK has been reported to protect cul-
tured hippocampal neurons against hypoxic damage . The role
of synaptic and extrasynaptic receptors in neuronal cell death is
currently a matter of intense debate . It is known that NRB
subunits segregate outside the synapses , , whereas NRA
subunits are conned to the postsynaptic membrane . The dif-
ferent availability of endogenous coagonists D-serine and glycine,
which show activation selectivity for NRA and NRB, respec-
tively, also contributes to segregate NMDARs at specic locations
. However, it is possible that such segregation is an oversimpli-
cation , and additional tools to specically block each recep-
tor will be necessary in the future.
The role played by synaptic and extrasynaptic NMDARs in
neuronal cell death in neurological diseases would depend on
the source of glutamate. Glutamate levels outside the synaptic
cleft are maintained by nonvesicular release through system xc
, and glutamate released by the cystine/glutamate anti-
porter is sucient to activate glutamate receptors , , .
We observed that glutamate release by system xc preferentially
activated extrasynaptic NRB receptors. Moreover, nonsubstrate
inhibitors of system xc signicantly attenuated the anoxic cur-
rents and ischemic damage, results conrmed in mutant sut mice,
in which xCT is nonfunctional P  .. This strongly suggests
a role for the cystine/glutamate antiporter as a source of gluta-
mate in ischemia. We thus propose that an important source of
glutamate in this pathology is extrasynaptic, a nding further sup-
ported by our observations that vesicular inhibitor Baf A did not
aect ischemic currents or neuronal damage.
It should be noted that latency to AD was still reduced when
CPG was applied concomitantly with TBOA therefore, glutamate
clearance was inhibited, suggesting that another source of gluta-
mate contributes to altering glutamate homeostasis. Severe ische-
mic conditions may induce the reversion of glutamate transporters
. However, glial glutamate transporters do not readily reverse
and remain functional during ischemia , . In accordance,
Figure 4. AD and OGD-induced cell death are reduced in Slc7a11sut mice, which lack a functional cystine/glutamate antiporter. (A) Voltage-clamp record-
ing of OGD-activated currents in mouse acute slices. The anoxic current amplitude was reduced in the xCT-deficient (sut/sut; n = 13) mice when compared
with that in wild-type mice (+/+; n = 14). However, the latency to AD was unchanged. (B) Histograms show the average amplitude (pA ± SEM) and latency
(minutes ± SEM) of the OGD-induced current for each genotype. *P < 0.05. (C) Simultaneous extracellular DC field potential recording in acute slices from
wild-type and sut/sut mice subjected to OGD. Histogram shows the average DC field potential (mV ± SEM) for each genotype. *P < 0.05. (D) Cell death
measured by LDH release in organotypic cultures from wild-type and sut/sut mice exposed to 45 minutes of OGD and 24 hours of reoxygenation. Data are
expressed as the mean ± SEM (n = 3). (E) Representative fields demonstrate propidium iodide labeling of organotypic slices treated as in D. Scale bar: 1 mm.
Downloaded August 22, 2014 from The Journal of Clinical Investigation. doi:10.1172/JCI71886.
The Journal of Clinical Investigation ReseaRch aRticle
3651
jci.org Volume 124 Number 8 August 2014
we found that blocking glutamate transporters shortened latency
to AD, amplied the anoxic current at the peak of AD, and, conse-
quently, increased neuronal damage see also refs. , . These
data do not exclude the possibility that a small proportion of gluta-
mate transporters i.e., neuronal EAAT function in reverse mode,
though inhibition of EAATs mostly in astrocytes leads to glutamate
accumulation. It is worth noting that the latency to AD was signi-
cantly increased in Eaac–/– , but not in Eaat–/–, mice .
Another possible source of glutamate is vesicular release. An in-
crease in mEPSCs has been described previously at early phases of
ischemia, before AD , , . The increase in mEPSC frequency
was Ca independent and caused by ischemia-induced actin la-
ment depolymerization . However, the increased spontaneous
vesicular transmitter release caused by energy depletion only leads
to extracellular glutamate accumulation when glial glutamate uptake
is blocked . Accordingly, we did not detect any change in anoxic
currents or damage in the presence of Baf A or TeTN. In conclusion,
this work does not challenge the current notion that exocytosis or
EAATs are key contributors to the release of ischemic glutamate, but
suggests that other sources of neurotransmitters and in particular
the cystine/glutamate antiporter are contributing to the process.
Several reports of glutamate release in pathological condi-
tions by this antiporter have been published recently, and a link
between glutamate release by system xc and some CNS diseases
has been established. Thus, xCT silencing diminished glutamate
secretion from gliomas and alleviated neurodegeneration  or
epileptic seizures . Similarly, inhibition of cystine/glutamate
antiporter–mediated glutamate release secondary to either mi-
croglial activation  or Parkinson-inducing toxin -hydroxydo-
pamine  prevented glial and neuronal damage. On the other
hand, expression of the catalytic subunit xCT is augmented in
processes that involve oxidative stress such as inammation ,
viral infection , and tumor proliferation . We also detected
an increase in xCT expression and function in primary cortical
neurons. Furthermore, we have used for the rst time a tracer spe-
cic for the system xc in brain in a live animal to monitor cystine/
glutamate antiporter activity during ischemia and reperfusion. We
found that ischemia induced a rapid increase in system xc activity
in vivo. This augmented function after ischemia would contribute
to further alter glutamate homeostasis during reperfusion.
A better understanding of the alterations of glutamate homeo-
stasis in ischemia is a key challenge for the eld. Protecting neu-
rons from ischemic excitotoxic damage by inhibiting ionotropic
glutamate receptors has been a useful strategy that has rendered
promising results both in vitro and in vivo. However, clinical trials
did not provide satisfactory results, because the use of glutamate
receptor antagonists has many secondary eects that are detri-
mental to patients. Therefore, blocking the source of glutamate
rather than the site of action may be a relevant therapeutic inter-
vention for the prevention of ischemic brain injury.
Methods
Reagents and chemicals. Calcein-AM and all cell culture supplies
were purchased from Invitrogen. The cytotoxicity assay for the
quantication of LDH release was acquired from Roche. L-cystine,
NVPAAM, propidium iodide, SAS, iodoacetate, and TeNT were
obtained from Sigma-Aldrich. Baf A, bicuculine, DAP, CNQX, ifen-
prodil, MK, memantine, RO , CPG, DLTBOA, AIDA, and
TCN were purchased from Tocris.
Mice and genotyping. Mutant Slcasut mice lacking the cystine/
glutamate antiporter  and wild-type CH/HeSnJ mice were pur-
chased from The Jackson Laboratory. We used littermates for all
experiments. For PCR genotyping,  sets of primers were designed:
Rv-E -CAAGGCCTCAAGCCCCTG, which matches to exon
 of the Slca gene only in / mice this region is deleted in sut
mice, and Rv-E -ATTTGACCACAATCTTTGAGACCA,
which hybridizes an alternative exon  E located within ampli-
cation range only in sut mice. The forward primer used in both
cases was Fw-E -TGAAACATGGAAACCGAAATCAC, which
hybridizes to exon  of Slca.
Electrophysiolog. Cortical slices -μm from P to P
Sprague-Dawley rats except for NMDA antagonist experiments, in
which P to P rats were used and from P to P CH/HeSnJ mice
were prepared in articial cerebrospinal uid aCSF; pH . that con-
tained  mM NaCl,  mM NaHCO,  mM NaHPO, . mM KCl,
. mM CaCl,  mM MgCl, and  mM D-glucose constantly bubbled
with  O and  CO. Slices were allowed to recover for at least
 hour and were then transferred to a °C chamber with continuous
ow  ml/minute with aCSF plus bicuculline  μM to block GABA
receptors and cystine  μM. Pyramidal cells of layer V of the cortex
Figure 5. Cystine-evoked (1 mM) inward currents
in pyramidal cortical neurons (n = 21). Currents
were blocked in the presence of CPG (50 μM; n = 5),
the NMDAR antagonist AP5 (50 μM; n = 5), and the
NR2B antagonist RO-256981 (1 μM; n = 6), but not in
the presence of the NR2A antagonist NVP-AAM077
(250 nM; n = 6). *P < 0.05; **P < 0.01.
Downloaded August 22, 2014 from The Journal of Clinical Investigation. doi:10.1172/JCI71886.
The Journal of Clinical Investigation
ReseaRch aRticle
3652 jci.org Volume 124 Number 8 August 2014
Millicell CM culture inserts Millipore and maintained in  neu-
robasal-B,  inactivated horse serum,  HBSS all media from
Invitrogen, . mM glucose,  mM L-glutamine, and X antibiotic-an-
timycotic  U/ml penicillin,  μg/ml streptomycin, and . μg/ml
amphotericin B; Life Technologies at °C and  CO. Slices were
used after  to  days in vitro.
OGD  minutes was achieved by incubating slices in an anaer-
obic chamber, in which O was replaced with N and external glucose
 mM with sucrose in an extracellular solution containing  mM
NaCl, . mM KCl, . mM CaCl,  mM NaHCO, . mM MgCl,
. mM NaHPO, and  μM L-cystine pH .. Organotypic corti-
cal slices were washed with this solution minus glucose for  min-
utes prior to OGD to deplete the remaining glucose from extracellular
spaces. After  minutes of OGD, extracellular solution was replaced
with medium and O supply restored. Antagonists were present during
the preincubation  minutes, OGD  minutes, and reoxygenation
 hours periods. Cell death was determined  hours after OGD by
measuring the release of LDH into the medium, using a colorimetric
assay Cytotoxicity Detection Kit; Roche Diagnostics according to the
manufacturer’s instructions. To evaluate the damaged region within
the slice, cultures were labeled with propidium iodide, the staining for
which correlates with LDH release in models of excitotoxicity  and
hypoxia . Briey, after collecting media for the LDH assay, slices
were incubated in propidium iodide–containing medium  μM for
 hour at °C. Slices were washed  times with culture medium and
photographed using a Nikon AZ uorescence microscope.
were identied visually using infrared dierential interference contrast
DIC microscopy Leica DM LFSA. Voltage-clamp recording pipettes
  M Ω were lled with a solution containing  mM CsCl,  mM NaCl,
. mM CaCl,  mM BAPTA,  mM HEPES,  mM Mg-ATP, and .
mM Na-GTP pH . . Access resistance and holding current were mon-
itored throughout the experiment. To simulate ischemia, glucose was
replaced with  mM sucrose, and  O/ CO was replaced with 
N/ CO. Cells were held at – mV to facilitate the sensing of ische-
mia-evoked currents through NMDARs. All antagonists were applied
concomitantly with ischemia stimulation. For cystine pus, a microp-
erfusion manifold World Precision Instruments containing cystine
 mM plus glycine  μM diluted with bathing solution was connected
to an air-pressured system and placed into layer V of the cortex at a dis-
tance of  μm from the cell soma. Simultaneously with voltage-clamp
recording, AD was recorded as negative extracellular DC potential shift
ΔVo induced by OGD. The DC potential is an extracellular recording
that is considered to provide an index of the polarization of cells sur-
rounding the tip of the glass electrode .. MΩ lled with  M NaCl
see ref. . For mEPSC recordings, refer to Supplemental Methods.
Organotypic slice cultures. Cultures were prepared from coronal
cerebral sections -μm of brains from -day-old Sprague-Dawley
rat pups using the method described by Plenz and Kitai  with minor
modications . Cortex was sliced using a McIllwain tissue chopper
Mickle Laboratory Engineering Co.. Slices containing cortex and
striatum but not hippocampus were selected under a microscope and
dissected to eliminate the corpus callosum. Slices were plated onto
Figure 6. Blockade of extrasynaptic NMDARs attenuates OGD-induced currents and cell death. (A and B) Representative voltage-clamped recordings of
cortical neurons at 30 mV in acute slices. OGD-activated currents were attenuated by the NR2B antagonists ifenprodil (3 μM; n = 9), memantine (10 μM;
n = 11), and RO-256981 (1 μM; n = 7). NR2A antagonists TCN-201 (10 μM; n = 6) and NVP-AAM077 (250 nM; n = 11) failed to reduce OGD-gated currents. (C)
Histograms showing the average amplitude (pA ± SEM) and latency (minutes ± SEM) of OGD-induced currents for each condition. *P < 0.05 versus OGD.
(D) Cell death measured by LDH release in organotypic cultures subjected to 45 minutes of OGD plus 24 hours of reoxygenation in the presence of the NR2B
antagonists memantine (10 μM; n = 4) and RO-256981 (1 μM; n = 3) or of the NR2A antagonists TCN-201 (10 μM; n = 3) and NVP-AAM077 (250 nM; n = 4).
MK801 (50 μM; n = 5) was also used to corroborate NMDA-mediated excitotoxicity. Data are expressed as the mean ± SEM. **P < 0.01 and ***P < 0.001
versus OGD. (E) Representative fields demonstrate propidium iodide labeling of organotypic slices treated as in D. Scale bar: 1 mm.
Downloaded August 22, 2014 from The Journal of Clinical Investigation. doi:10.1172/JCI71886.
The Journal of Clinical Investigation ReseaRch aRticle
3653
jci.org Volume 124 Number 8 August 2014
 groups were analyzed using an unpaired, -tailed Student’s t test.
Comparisons among multiple groups were analyzed by -way ANOVA
followed by Bonferroni’s multiple comparison tests for post hoc analy-
sis. Statistical signicance was considered at P  ..
Study approval. Organotypic cultures, primary cultures, and elec-
trophysiology protocols using rats and wild-type and Slcasut mice
were approved by the Comité de Ética y Bienestar Animal Animal Eth-
ics and Welfare Committee of the UPV/EHU. MCAO and PET studies
were approved by the animal ethics committee of CIC biomaGUNE
and by local authorities and were conducted in accordance with the
directives of the European Union on animal ethics and welfare.
Acknowledgments
The authors would like to thank to H. Gómez, S. Marcos, and S.
Martín for technical assistance with cultures and electrophysiol-
ogy studies; V. Gómez-Vallejo, M. González, and A. Leukona for
technical support with radiosynthesis; and A. Cano, A. Arrieta,
and M. Errasti for technical assistance in the PET studies. This
work was supported by the Instituto de Salud Carlos III fellow-
ship to F.N. Soria; the Fundación Koplowitz; the Spanish Ministry
of Education and Science SAF; the Basque Govern-
ment; the UPV/EHU; and the CIBERNED.
Address correspondence to: Carlos Matute or Maria
Domercq, Dpto. Neurociencias, Universidad del País Vasco,
E Leioa, Spain. Phone: .; E-mail:
carlos.matute@ehu.es C. Matute. Phone: .;
E-mail: maria.domercq@ehu.es M. Domercq.
Primary cortical neuron cultures. Primary neurons were obtained
from the cortical lobes of E Sprague-Dawley rat embryos . Cells
were resuspended in B-supplemented neurobasal medium Life
Technologies plus  FBS and then seeded onto poly-l-ornithine–
coated glass coverslips at a density of . ×  cells/cm. The medium
was replaced with serum-free, B-supplemented neurobasal medium
 hours later. Cultures were essentially free of astrocytes and micro-
glia and were maintained at °C and  CO. Cultures were used
after  days in vitro. See Supplemental Methods for xCT expression
and function assays after in vitro ischemia and glutamine synthetase
assay using primary cortical neurons.
Transient focal ischemia. Transient focal ischemia was produced
by a -hour intraluminal occlusion of the MCA followed by reper-
fusion in adult -week-old male Sprague-Dawley rats obtained
from Janvier  g body weight; n , as described elsewhere
. Briey, rats were anesthetized with  isourane for  to
 minutes in  O, and a .-cm length of  monolament
nylon suture was introduced into the right external carotid artery up
to the level where the MCA branches out, and animals were sutured
and placed in their cages with ad libitum access to food and water.
After  hours, the animals were reanesthetized, and the lament
was removed to allow reperfusion and PET image acquisition. Ani-
mals were studied at  minutes n  and at  hours n  following
the ischemic episode. See Supplemental Methods for details about
the PET assay and radiosynthesis.
Statistics. Data are presented as the mean  SEM unless other-
wise indicated from at least  independent experiments, in which all
conditions were assayed at least in triplicate. Comparisons between
Figure 7. Cystine/glutamate antiporter expression and function are increased in cortical neurons in vitro after chemical ischemia and in vivo after transient
focal ischemia. (A) xCT mRNA levels in neurons exposed to chemical ischemia (1 hour), as measured by qPCR. Data are expressed as the mean ± SEM (n = 4).
(B) xCT protein levels after chemical ischemia (1 hour) plus a 4-hour reoxygenation in cortical neurons. Data were normalized to actin and are expressed as
the mean ± SEM (n = 4). Control and OGD lanes were run on the same gel but were noncontiguous. Full, uncut gels are shown in the Supplemental Material.
(C) Cl-dependent [14C] L-cystine uptake by neurons after 1 hour of chemical ischemia plus 4 hours of reoxygenation. Note that cystine/glutamate antagonist
AAA (250 μM) almost completely inhibited uptake. Data were normalized to protein concentration and are expressed as the mean ± SEM (n = 3). (D) 18[F]FSPG
uptake study by PET imaging in control rats and after MCAO (n = 5 per group). Representative images of control rats and images at 5 minutes and at 5 hours
of reperfusion (RPF) following MCAO. (E) Histogram shows 18[F]FSPG signal expressed as the percentage of injected dose per gram (% ID/g) and normalized
to the contralateral hemisphere in the total infarct area or in the volumes of interest (VOIs) defined in the striatum and cortex at 5 minutes or at 5 hours after
reperfusion. Data are expressed as the mean ± SEM (n = 5). *P < 0.05, **P < 0.01, and ***P < 0.001 versus control.
Downloaded August 22, 2014 from The Journal of Clinical Investigation. doi:10.1172/JCI71886.
The Journal of Clinical Investigation
ReseaRch aRticle
3654 jci.org Volume 124 Number 8 August 2014
. Burke JF, Lisabeth LD, Brown DL, Reeves MJ,
Morgenstern LB. Determining stroke’s rank as a
cause of death using multicause mortality data.
Stroke. ;: .
. Lipton P. Ischemic cell death in brain neurons.
Physiol Re v. ;:.
. Dirnagl U, Iadecola C, Moskowitz MA. Pathobi-
ology of ischaemic stroke: an integrated view.
Trends Neurosci. ;:.
. Somjen GG. Mechanisms of spreading depres-
sion and hypoxic spreading depression-like depo-
larization. Physiol Rev. ; :.
. Rossi DJ, Oshima T, Attwell D. Glutamate release
in severe brain ischaemia is mainly by reversed
uptake. Nature. ;:.
. Hamann M, Rossi DJ, Mohr C, Andrade AL,
Attwell D. The electrical response of cerebellar
Purkinje neurons to simulated ischaemia. Brain.
;pt :.
. Tymianski M. Emerging mechanisms of dis-
rupted cellular signaling in brain ischemia.
Nat Neurosci. ;:.
. Hardingham GE, Bading H. Synaptic versus
extrasynaptic NMDA receptor signalling: impli-
cations for neurodegenerative disorders. Nat Rev
Neurosci. ;:.
. Hardingham GE, Fukunaga Y, Bading H. Extra-
synaptic NMDARs oppose synaptic NMDARs
by triggering CREB shut-off and cell death path-
ways. Nat Neurosci. ;:.
. Zhang SJ, et al. A signaling cascade of nuclear cal-
cium-CREBATF activated by synaptic NMDA
receptors deines a gene repression module that
protects against extrasynaptic NMDA recep-
tor-induced neuronal cell death and ischemic
brain damage. J Neurosci. ;:.
. Zhang SJ, et al. Decoding NMDA receptor
signaling: identiication of genomic programs
specifying neuronal survival and death. Neuron.
;:.
. Tu W, et al. DAPK interaction with NMDA
receptor NRB subunits mediates brain damage
in stroke. Cell. ;: .
. Karpova A, et al. Encoding and transducing the
synaptic or extrasynaptic origin of NMDA receptor
signals to the nucleus. Cell. ;:.
. Stanika RI, et al. Coupling diverse routes of
calcium entry to mitochondrial dysfunction and
glutamate excitotoxicity. Proc Natl Acad Sci U S A.
;:.
. Papouin T, et al. Synaptic and extrasynaptic
NMDA receptors are gated by different endoge-
nous coagonists. Cell. ;: .
. Wroge CM, Hogins J, Eisenman L, Mennerick S.
Synaptic NMDA receptors mediate hypoxic excito-
toxic death. J Neurosci. ;:.
. Baker DA, Xi ZX, Shen H, Swanson CJ, Kali-
vas PW. The origin and neuronal function
of in vivo nonsynaptic glutamate. J Neurosci.
;:.
. Baker DA, Shen H, Kalivas PW. Cystine/gluta-
mate exchange serves as the source for extra-
cellular glutamate: modiications by repeated
cocaine administration. Amino Acids. ;
:.
. Massie A, et al. Dopaminergic neurons of system
xc-deicient mice are highly protected against
-hydroxydopamine-induced toxicity. FASEB J.
;:.
. De Bundel D, et al. Loss of system xc- does not
induce oxidative stress but decreases extracel-
lular glutamate in hippocampus and influences
spatial working memory and limbic seizure sus-
ceptibility. J Neurosci. ;: .
. Bannai S, Kitamura E. Transport interac-
tion of L-cystine and L-glutamate in human
diploid ibroblasts in culture. J Biol Chem.
;: .
 . Lo M, Wang YZ, Gout PW. The xc- cystine/
glutamate antiporter: a potential target for ther-
apy of cancer and other diseases. J Cell Physi ol.
;:.
. Conrad M, Sato H. The oxidative stress-induc-
ible cystine/glutamate antiporter, system x c
–: cystine supplier and beyond. Amino Acids.
;:.
. Gasol E, Jiménez-Vidal M, Chillarón J, Zorzano
A, Palacín M. Membrane topology of system
xc- light subunit reveals a re-entrant loop with
substrate-restricted accessibility. J Biol Chem.
;:.
. Dringen R. Metabolism and functions of glutathi-
one in brain. Prog Neurobiol. ;
:.
. McBean GJ. Cerebral cystine uptake: a tale
of two transporters. Trends Pharmacol Sci.
;:.
 . Sasaki H, et al. Electrophile response element-
mediated induction of the cystine/glutamate
exchange transporter gene expression.
J Biol Chem. ; :.
 . Mysona B, Dun Y, Duplantier J, Ganapathy V,
Smith SB. Effects of hyperglycemia and oxidative
stress on the glutamate transporters GLAST and
system xc- in mouse retinal Müller glial cells.
Cell Tissue Res. ;:.
. Buckingham SC, et al. Glutamate release by pri-
mary brain tumors induces epileptic activity.
Nat Med. ;:.
. Albrecht P, et al. Mechanisms of oxidative gluta-
mate toxicity: the glutamate/cystine antiporter
system xc- as a neuroprotective drug target. CNS
Neurol Disord Drug Targets. ;: .
. Domercq M, et al. System xc- and glutamate
transporter inhibition mediates microglial
toxicity to oligodendrocytes. J Immunol.
;:.
. Lewerenz J, Maher P, Methner A. Regulation of
xCT expression and system x c - function in
neuronal cells. Amino Acids. ;:.
. Jabaudon D, Scanziani M, Gähwiler BH, Gerber
U. Acute decrease in net glutamate uptake during
energy deprivation. Proc Natl Acad Sci U S A.
;:.
 . Gao J, et al. Coupling between NMDA recep-
tor and acid-sensing ion channel contrib-
utes to ischemic neuronal death. Neuron.
;:.
. Domercq M, et al. PX receptors mediate
ischemic damage to oligodendrocytes. Glia.
;:.
. Arbeloa J, Pérez-Samartín A, Gottlieb M, Matute
C. PX receptor blockade prevents ATP excito-
toxicity in neurons and reduces brain damage
after ischemia. Neurobiol Dis. ;
:.
 . Thompson RJ, Zhou N, MacVicar BA. Ischemia
opens neuronal gap junction hemichannels.
Science. ; :.
. Gebhardt C, Körner R, Heinemann U. Delayed
anoxic depolarizations in hippocampal neurons of
mice lacking the excitatory amino acid carrier .
J Cereb Blood Flow Metab. ;: .
. Lossi L, Alasia S, Salio C, Merighi A. Cell death
and proliferation in acute slices and organotypic
cultures of mammalian CNS. Prog Neurobiol.
;:.
. Fujimoto S, Katsuki H, Kume T, Kaneko S,
Akaike A. Mechanisms of oxygen glucose
deprivation-induced glutamate release from
cerebrocortical slice cultures. Neurosci Res.
;: .
. Andrade AL, Rossi DJ. Simulated ischaemia
induces Ca-independent glutamatergic vesicle
release through actin ilament depolymeriza-
tion in area CA of the hippocampus. J Physiol .
;pt :.
. Fleidervish IA, Gebhardt C, Astman N, Gutnick
MJ, Heinemann U. Enhanced spontaneous
transmitter release is the earliest consequence
of neocortical hypoxia that can explain the dis-
ruption of normal circuit function. J Neurosci.
;:.
. Baker DA, et al. Contribution of cystine-gluta-
mate antiporters to the psychotomimetic effects
of phencyclidine. Neuropsychopharmacolog.
;:.
. Patel SA, Warren BA, Rhoderick JF, Bridges RJ. Dif-
ferentiation of substrate and non-substrate inhibi-
tors of transport system xc: an obligate exchanger
of L-glutamate and L-cystine. Neuropharmacolog.
;:.
. Bridges RJ, Natale NR, Patel SA. System xc- cys-
tine/glutamate antiporter: an up date on molec-
ular pharmacology and roles within the CNS.
Br J Pharmacol. ;:–.
. Müller M, S omjen GG. Na dependence and the
role of glutamate receptors and Na channels in
ion fluxes during hypoxia of rat hippocampal slices.
J Neurophysiol. ;:.
. Doherty AJ, Collingridge GL, Jane DE. Antago-
nist activity of alpha-substituted
-carboxyphenylglycine analogues at group I
metabotropic glutamate receptors expressed in
CHO cells. Br J Pharmacol. ;:.
 . Melendez R, Vuthiganon J, Kalivas P. Regulation
of extracellular glutamate in the prefrontal cortex:
focus on the cystine glutamate exchanger and
group I metabotropic glutamate receptors.
J Pharmacol. ;:.
. Chintala S, et al. Slca gene controls produc-
tion of pheomelanin pigment and proliferation
of cultured cells. Proc Natl Acad Sci U S A.
;:.
. Warr O, Takahashi M, Attwell D. Modulation of
extracellular glutamate concentration in rat brain
slices by cystine-glutamate exchange. J Physiol.
; :.
. Tovar KR, Westbrook GL. The incorporation of
NMDA receptors with a distinct subunit compo-
sition at nascent hippocampal synapses in vitro.
J Neurosci. ;:.
. Petralia RS, et al. Organization of NMDA recep-
tors at extrasynaptic locations. Neuroscience.
Downloaded August 22, 2014 from The Journal of Clinical Investigation. doi:10.1172/JCI71886.
The Journal of Clinical Investigation ReseaRch aRticle
3655
jci.org Volume 124 Number 8 August 2014
;:.
. Paoletti P, Bellone C, Zhou Q. NMDA receptor
subunit diversity: impact on receptor properties,
synaptic plasticity and disease. Nat Rev Neurosci.
;:.
. Fischer G, et al. Ro , a highly potent
and selective blocker of N-methyl-D-aspar-
tate receptors containing the NRB subunit.
Characterization in vitro. J Pharmacol Exp Ther.
;:.
. Bartlett TE, et al. Differential roles of NRA and
NRB-containing NMDA receptors in LTP and
LTD in the CA region of two-week old rat hippo-
campus. Neuropharmacolog. ;:.
. Williams K. Ifenprodil discriminates subtypes of
the N-methyl-D-aspartate receptor: selectivity
and mechanisms at recombinant heteromeric
receptors. Mol Pharmacol. ;:.
. Edman S, et al. TCN  selectively blocks Glu-
NA-containing NMDARs in a GluN co-agonist
dependent but non-competitive manner.
Neuropharmacolog. ;:.
. Okamoto S, et al. Balance between synaptic ver-
sus extrasynaptic NMDA receptor activity influ-
ences inclusions and neurotoxicity of mutant
huntingtin. Nat Med. ;:.
. Xia P, Chen HV, Zhang D, Lipton SA. Memantine
preferentially blocks extrasynaptic over syn-
aptic NMDA receptor currents in hippocampal
autapses. J Neurosci. ;:.
. Frizelle PA, Chen PE, Wyllie DJA. Equilibrium
constants for R-S---bromo-phenyl-eth-
ylamino-,-dioxo-,,,-tetrahydroqui-
noxalin--yl-methyl-phosphonic acid NVP-
AAM acting at recombinant NR/NRA and
NR/NRB N-methyl-D-aspartate receptors:
implications for studies of synaptic transmission.
Mol Pharmacol. ;:.
. Abramov AY, Scorziello A, Du chen MR. Three
distinct mechanisms generate oxygen free
radicals in neurons and contribute to cell death
during anoxia and reoxygenation. J Neurosci.
;:.
. Baek S, et al. Exploratory clinical trial of
S---Ffluoropropyl-L-glutamate for imag-
ing xC- transporter using positron emission tomog-
raphy in patients with non-small cell lung or breast
cancer. Clin Cancer Res. ;:.
. Koglin N, et al. Speciic PET imaging of
xC- transporter activity using a F-labeled
glutamate derivative reveals a dominant path-
way in tumor metabolism. Clin Cancer Res.
;:.
. Rojas S, et al. Modest MRI signal intensity
changes precede delayed cortical necrosis
after transient focal ischemia in the rat. Stroke.
;:.
 . Groc L, et al. NMDA receptor surface mobility
depends on NRAB subunits. Proc Natl Acad Sci
U S A. ;:.
. Harris AZ, Pettit DL. Extrasynaptic and synap-
tic NMDA receptors form stable and uniform
pools in rat hippocampal slices. J Physiol.
;pt :–.
. Hamann M, Rossi DJ, Marie H, Attwell D. Knock-
ing out the glial glutamate transporter GLT
reduces glutamate uptake but does not affect hip-
pocampal glutamate dynamics in early simulated
ischaemia. Eur J Neurosci. ;:.
. Savaskan NE, et al. Small interfering RNA-medi-
ated xCT silencing in gliomas inhibits neurode-
generation and alleviates brain edema. Nat Med.
;:.
. Pampliega O, et al. Increased expression of cys-
tine/glutamate antiporter in multiple sclerosis.
J Neuroinflammation. ;:.
. Qin Z, et al. Upregulation of xCT by KSHV-en-
coded microRNAs facilitates KSHV dissemina-
tion and persistence in an environment of oxida-
tive stress. PLoS Pathog. ;:e.
. Ogunrinu TA, Sontheimer H. Hypoxia increases
the dependence of glioma cells on glutathione.
J Biol Chem. ;: .
. Farkas E, Pratt R, Sengpiel F, Obrenovitch TP.
Direct, live imaging of cortical spreading depres-
sion and anoxic depolarisation using a fluores-
cent, voltage-sensitive dye. J Cereb Blood Flow
Metab. ; :.
. Plenz D, Kitai ST. Organotypic cortex-stria-
tum-mesencephalon cultures: the nigrostriatal
pathway. Neurosci Lett. ;:.
. Cavaliere F, Dinkel K, Reymann K. The subven-
tricular zone releases factors which can be protec-
tive in oxygen/glucose deprivation-induced cor-
tical damage: an organotypic study. Exp Neurol.
;:.
. Zimmer J, Kristensen BW, Jakobsen B, Noraberg
J. Excitatory amino acid neurotoxicity and
modulation of glutamate receptor expression
in organotypic brain slice cultures. Amino Acids.
;:.
. Graulich J, et al. Acute neuronal injury after
hypoxia is influenced by the reoxygenation mode
in juvenile hippocampal slice cultures. Brain Res
Dev Brain Res. ;:.
. Ruiz A, Matute C, Alberdi E. Endoplasmic reticu-
lum Ca release through ryanodine and IP
receptors contributes to neuronal excitotoxicity.
Cell Calcium. ;:.
. Justicia C, Pérez-Asensio FJ, Burguete MC, Salom
JB, Planas AM. Administration of transforming
growth factor-α reduces infarct volume after
transient focal cerebral ischemia in the rat.
J Cereb Blood Flow Metab. ;:.
Downloaded August 22, 2014 from The Journal of Clinical Investigation. doi:10.1172/JCI71886.
... Many acute and chronic neurodegenerative disorders are characterized by deficits in glutamate uptake systems, which causes extracellular glutamate concentrations to increase to neurotoxic levels. Hypoxic ischemic conditions can further exacerbate deregulation of glutamate homeostasis through enhanced extrasynaptic glutamate release via the cystine/glutamate antiporter, System XC [11] (Figure 1). Thus, while in healthy condition, the action of glutamate is restricted to the synapse, in neurodegenerative diseases, glutamate receptors located outside synaptic contacts are being activated. ...
... Indeed, the cell pathology common to virtually all neurodegenerative conditions is highly reminiscent of the typical pathological triad triggered by the activation of eNMDARs: loss of structural integrity (i.e., the loss of synapses and dendrites), mitochondrial dysfunction (i.e., the breakdown of the mitochondrial membrane potential, metabolic/energy insufficiency, and increased production of reactive oxygen species), and transcriptional deregulation (i.e., CREB shut-off and reduced expression of activity-regulated neurotrophic/neuroprotective genes) [4,5,38]. One reason for the convergence of different pathomechanisms on toxic eNMDAR signaling is that virtually all neurodegenerative conditions are burdened with faulty or deregulated glutamate uptake systems, resulting in the leakage of synaptically released glutamate and a subsequent rise in glutamate levels at extrasynaptic locations [7,11,[117][118][119]. Deregulated glutamate homeostasis is further enhanced via neuroinflammatory responses and a leaky bloodbrain barrier, both of which are often associated with degenerative processes in the brain. ...
... Indeed, the cell pathology common to virtually all neurodegenerative conditions is highly reminiscent of the typical pathological triad triggered by the activation of eNMDARs: loss of structural integrity (i.e., the loss of synapses and dendrites), mitochondrial dysfunction (i.e., the breakdown of the mitochondrial membrane potential, metabolic/energy insufficiency, and increased production of reactive oxygen species), and transcriptional deregulation (i.e., CREB shut-off and reduced expression of activity-regulated neurotrophic/neuroprotective genes) [4,5,38]. One reason for the convergence of different pathomechanisms on toxic eNMDAR signaling is that virtually all neurodegenerative conditions are burdened with faulty or deregulated glutamate uptake systems, resulting in the leakage of synaptically released glutamate and a subsequent rise in glutamate levels at extrasynaptic locations [7,11,[117][118][119]. Deregulated glutamate homeostasis is further enhanced via neuroinflammatory responses and a leaky blood-brain barrier, both of which are often associated with degenerative processes in the brain. ...
Article
Full-text available
With the discovery that the acquisition of toxic features by extrasynaptic NMDA receptors (NMDARs) involves their physical interaction with the non-selective cation channel, TRPM4, it has become possible to develop a new pharmacological principle for neuroprotection, namely the disruption of the NMDAR/TRPM4 death signaling complex. This can be accomplished through the expression of the TwinF domain, a 57-amino-acid-long stretch of TRPM4 that mediates its interaction with NMDARs, but also using small molecule TwinF interface (TI) inhibitors, also known as NMDAR/TRPM4 interaction interface inhibitors. Both TwinF and small molecule TI inhibitors detoxify extrasynaptic NMDARs without interfering with synaptic NMDARs, which serve important physiological functions in the brain. As the toxic signaling of extrasynaptic NMDARs contributes to a wide range of neurodegenerative conditions, TI inhibitors may offer therapeutic options for currently untreatable human neurodegenerative diseases including Amyotrophic Lateral Sclerosis, Alzheimer’s disease, and Huntington’s disease.
... Besides, the researches on other mechanisms mediating ischemic glutamate release have made progress. Recently, extrasynaptic glutamate release through cystine/glutamate antiporter 25 and volume-regulated anion channel SWELL1 26 is reported to promote ischemic damage. These glutamatereleasing approaches are almost stimulated within several minutes to hours, leading to narrow time window for neuroprotective treatment, though they are critical to the irreversible injury ultimately resulting in the infarction. ...
... Anoxic depolarization (AD) currents in brain slices were induced by oxygen and glucose deprivation (OGD) and recorded as reported 25 . Voltage-clamp recording pipettes (4e6 MU) were filled with a solution containing 135 mmol/L CsCl, 4 mmol/L NaCl, 0.7 mmol/L CaCl 2 , 10 mmol/L BAPTA, 10 mmol/L HEPES, 4 mmol/L Mg-ATP, and 0.5 mmol/L Na 2 -GTP (pH 7.3). ...
... The effective infection of cortex neurons and the overexpression of BEST1 were verified 21 days after the virus microinjection ( Fig. 6C and D). Consistent with previous report 25 , OGD induced an obvious inward current in control slices (infected with AAV-CMV-3Flag-T2A-GFP), corresponding to AD (Fig. 6E). The OGD-induced current in BEST1 WT -overexpressed slices was larger ( Fig. 6E and F, F (2,32) Z 9.308, P Z 0.009), but almost abolished by concomitant application of AP-5 and CNQX with OGD ( Fig. 6E and F, F (2,32) Z 9.308, P Z 0.001), indicating glutamate release through overexpressed BEST1. ...
Article
Full-text available
Many efforts have been made to understand excitotoxicity and develop neuroprotectants for the therapy of ischemic stroke. The narrow treatment time window is still to be solved. Given that the ischemic core expanded over days, treatment with an extended time window is anticipated. Bestrophin 1 (BEST1) belongs to a bestrophin family of calcium-activated chloride channels. We revealed an increase in neuronal BEST1 expression and function within the peri-infarct from 8 to 48 h after ischemic stroke in mice. Interfering the protein expression or inhibiting the channel function of BEST1 by genetic manipulation displayed neuroprotective effects and improved motor functional deficits. Using electrophysiological recordings, we demonstrated that extrasynaptic glutamate release through BEST1 channel resulted in delayed excitotoxicity. Finally, we confirmed the therapeutic efficacy of pharmacological inhibition of BEST1 during 6–72 h post-ischemia in rodents. This delayed treatment prevented the expansion of infarct volume and the exacerbation of neurological functions. Our study identifies the glutamate-releasing BEST1 channel as a potential therapeutic target against ischemic stroke with a wide time window.
... The extrasynaptic glutamate level is sensitive to abnormal and pathological conditions. For example, in rodents, stressful stimuli such as body restraint, forced swimming, and hypoxic insults can selectively increase the extrasynaptic glutamate concentration to over 30 µM or higher [143][144][145]. In stroke and brain injuries, the reduced extracellular volume associated with brain edema elevates the extrasynaptic glutamate concentration as a contributing factor in excitatory neuronal damage [146]. ...
... Compatible with the lower concentrations, extrasynaptic glutamate preferably activates the main population of eNMDARs of higher affinity, e.g., GluN2B-containing NMDARs. GluN2B-containing eNMDARs were proposed to be responsible for ischemia-induced excitotoxicity [179,180], and extrasynaptic glutamate is a primary contributor to ischemic and traumatic damage in the brain [143,146,181]. The activation of eNMDARs, perhaps together with the impaired protective function of sNMDARs, contributes to downstream cascades of necrotic and programmed cell death pathways [179,182]. ...
Article
Full-text available
Stroke and late-onset Alzheimer’s disease (AD) are risk factors for each other; the comorbidity of these brain disorders in aging individuals represents a significant challenge in basic research and clinical practice. The similarities and differences between stroke and AD in terms of pathogenesis and pathophysiology, however, have rarely been comparably reviewed. Here, we discuss the research background and recent progresses that are important and informative for the comorbidity of stroke and late-onset AD and related dementia (ADRD). Glutamatergic NMDA receptor (NMDAR) activity and NMDAR-mediated Ca ²⁺ influx are essential for neuronal function and cell survival. An ischemic insult, however, can cause rapid increases in glutamate concentration and excessive activation of NMDARs, leading to swift Ca ²⁺ overload in neuronal cells and acute excitotoxicity within hours and days. On the other hand, mild upregulation of NMDAR activity, commonly seen in AD animal models and patients, is not immediately cytotoxic. Sustained NMDAR hyperactivity and Ca ²⁺ dysregulation lasting from months to years, nevertheless, can be pathogenic for slowly evolving events, i.e. degenerative excitotoxicity, in the development of AD/ADRD. Specifically, Ca ²⁺ influx mediated by extrasynaptic NMDARs (eNMDARs) and a downstream pathway mediated by transient receptor potential cation channel subfamily M member (TRPM) are primarily responsible for excitotoxicity. On the other hand, the NMDAR subunit GluN3A plays a “gatekeeper” role in NMDAR activity and a neuroprotective role against both acute and chronic excitotoxicity. Thus, ischemic stroke and AD share an NMDAR- and Ca ²⁺ -mediated pathogenic mechanism that provides a common receptor target for preventive and possibly disease-modifying therapies. Memantine (MEM) preferentially blocks eNMDARs and was approved by the Federal Drug Administration (FDA) for symptomatic treatment of moderate-to-severe AD with variable efficacy. According to the pathogenic role of eNMDARs, it is conceivable that MEM and other eNMDAR antagonists should be administered much earlier, preferably during the presymptomatic phases of AD/ADRD. This anti-AD treatment could simultaneously serve as a preconditioning strategy against stroke that attacks ≥ 50% of AD patients. Future research on the regulation of NMDARs, enduring control of eNMDARs, Ca ²⁺ homeostasis, and downstream events will provide a promising opportunity to understand and treat the comorbidity of AD/ADRD and stroke.
... Considering its biochemical functions and its widespread distribution in the CNS, system x c − has been studied in several neurological disorders in which dysregulated excitatory transmission contributes to nervous insults and neurodegeneration. In particular, increased xCT expression has been documented in models of Parkinson's disease, amyotrophic lateral sclerosis, multiple sclerosis as well as in stroke or glioblastoma [19][20][21][22][23][24]. A role for system x c − was also documented in the modulation of inflammatory responses, raising further interest for its implication in pathologies combining neuroinflammation and increased neuronal excitation [19]. ...
Article
Full-text available
Background Despite the high prevalence of neuropathic pain, treating this neurological disease remains challenging, given the limited efficacy and numerous side effects associated with current therapies. The complexity in patient management is largely attributed to an incomplete understanding of the underlying pathological mechanisms. Central sensitization, that refers to the adaptation of the central nervous system to persistent inflammation and heightened excitatory transmission within pain pathways, stands as a significant contributor to persistent pain. Considering the role of the cystine/glutamate exchanger (also designated as system xc⁻) in modulating glutamate transmission and in supporting neuroinflammatory responses, we investigated the contribution of this exchanger in the development of neuropathic pain. Methods We examined the implication of system xc⁻ by evaluating changes in the expression/activity of this exchanger in the dorsal spinal cord of mice after unilateral partial sciatic nerve ligation. In this surgical model of neuropathic pain, we also examined the consequence of the genetic suppression of system xc⁻ (using mice lacking the system xc⁻ specific subunit xCT) or its pharmacological manipulation (using the pharmacological inhibitor sulfasalazine) on the pain-associated behavioral responses. Finally, we assessed the glial activation and the inflammatory response in the spinal cord by measuring mRNA and protein levels of GFAP and selected M1 and M2 microglial markers. Results The sciatic nerve lesion was found to upregulate system xc⁻ at the spinal level. The genetic deletion of xCT attenuated both the amplitude and the duration of the pain sensitization after nerve surgery, as evidenced by reduced responses to mechanical and thermal stimuli, and this was accompanied by reduced glial activation. Consistently, pharmacological inhibition of system xc⁻ had an analgesic effect in lesioned mice. Conclusion Together, these observations provide evidence for a role of system xc⁻ in the biochemical processes underlying central sensitization. We propose that the reduced hypersensitivity observed in the transgenic mice lacking xCT or in sulfasalazine-treated mice is mediated by a reduced gliosis in the lumbar spinal cord and/or a shift in microglial M1/M2 polarization towards an anti-inflammatory phenotype in the absence of system xc⁻. These findings suggest that drugs targeting system xc⁻ could contribute to prevent or reduce neuropathic pain.
... PostC ischemic postconditioning, Ru265 ruthenium red 265; sham, control + Ru265 10 µM Page 10 of 14 that MCU plays a key role in the PostC pathway. During I/R injury, an excessive release of glutamate occurs, leading to over-activation of NMDAR (Bonova et al. 2013;Dávalos et al. 1997;Soria et al. 2014). This cascade leads to cell necrosis or apoptosis by excess Ca 2+ influx into the neuron, triggering a range of downstream pro-death signaling events such as calpain activation, generation of reactive oxygen species and damage to mitochondria (Curcio et al. 2016;Kristián and Siesjö 1998;Lau and Tymianski 2010). ...
Article
Full-text available
The phenomenon of ischemic postconditioning (PostC) is known to be neuroprotective against ischemic reperfusion (I/R) injury. One of the key processes in PostC is the opening of the mitochondrial ATP-dependent potassium (mito-KATP) channel and depolarization of the mitochondrial membrane, triggering the release of calcium ions from mitochondria through low-conductance opening of the mitochondrial permeability transition pore. Mitochondrial calcium uniporter (MCU) is known as a highly sensitive transporter for the uptake of Ca²⁺ present on the inner mitochondrial membrane. The MCU has attracted attention as a new target for treatment in diseases, such as neurodegenerative diseases, cancer, and ischemic stroke. We considered that the MCU may be involved in PostC and trigger its mechanisms. This research used the whole-cell patch-clamp technique on hippocampal CA1 pyramidal cells from C57BL mice and measured changes in spontaneous excitatory post-synaptic currents (sEPSCs), intracellular Ca²⁺ concentration, mitochondrial membrane potential, and N-methyl-d-aspartate receptor (NMDAR) currents under inhibition of MCU by ruthenium red 265 (Ru265) in PostC. Inhibition of MCU increased the occurrence of sEPSCs (p = 0.014), NMDAR currents (p < 0.001), intracellular Ca²⁺ concentration (p < 0.001), and dead cells (p < 0.001) significantly after reperfusion, reflecting removal of the neuroprotective effects in PostC. Moreover, mitochondrial depolarization in PostC with Ru265 was weakened, compared to PostC (p = 0.004). These results suggest that MCU affects mitochondrial depolarization in PostC to suppress NMDAR over-activation and prevent elevation of intracellular Ca²⁺ concentrations against I/R injury. Graphical Abstract
... In addition, the dysfunction of the xc-system that leads to glutathione depletion also occurs in ferroptosis [78][79][80]. Oxidative glutamate intoxication, also known as oxidative glutamate intoxication, is a glutamate-induced cell death mediated by blocking of the xc-system [81]. ...
Article
Full-text available
Diabetic cardiomyopathy (DC) is a serious heart disease caused by diabetes. It is unrelated to hypertension and coronary artery disease and can lead to heart insufficiency, heart failure and even death. Currently, the pathogenesis of DC is unclear, and clinical intervention is mainly symptomatic therapy and lacks effective intervention objectives. Iron overdose mediated cell death, also known as ferroptosis, is widely present in the physiological and pathological processes of diabetes and DC. Iron is a key trace element in the human body, regulating the metabolism of glucose and lipids, oxidative stress and inflammation, and other biological processes. Excessive iron accumulation can lead to the imbalance of the antioxidant system in DC and activate and aggravate pathological processes such as excessive autophagy and mitochondrial dysfunction, resulting in a chain reaction and accelerating myocardial and microvascular damage. In-depth understanding of the regulating mechanisms of iron metabolism and ferroptosis in cardiovascular vessels can help improve DC management. Therefore, in this review, we summarize the relationship between ferroptosis and the pathogenesis of DC, as well as potential intervention targets, and discuss and analyze the limitations and future development prospects of these targets.
... Cytosolic Ca 2+ in ux into the mitochondrial matrix via MCU leads to the elevation of mitochondrial Ca 2+ concentration and induces the regulation of various mitochondrial metabolism: mitochondrial respiration, adenosine triphosphate (ATP) production, mitophagy/autophagy and even the death pathway of apoptosis or necrosis [34][35][36]. Meanwhile, it is known that during I/R injury, an excessive release of glutamate occurs and induces over-activation of NMDAR [37][38][39]. This cascade leads to cell necrosis or apoptosis by excessing Ca 2+ in ux into the neurons and triggering subsequently a range of downstream pro-death signaling events such as calpain activation, reactive oxygen species (ROS) generation and mitochondrial damage [40][41][42]. ...
Preprint
Full-text available
Ischemic postconditioning (PostC) phenomenon is known as the neuroprotection against ischemic reperfusion (I/R) injury. One of the key processes in PostC is opening of mitochondrial ATP dependent potassium (mito-K ATP ) channel and depolarization of mitochondrial membrane potential, which triggers the release of calcium ion from mitochondria through the low conductance opening of mitochondrial permeability transition pore (mPTP). Mitochondrial calcium uniporter (MCU) is known as the highly sensitive transporter for uptake of Ca ²⁺ inwardly existed on the inner mitochondrial membrane. Furthermore, it has attracted attention as a new target of treatments in disease such as neurodegenerative disease, cancer, and ischemic stroke. Thus, we considered that MCU may involve in PostC and trigger its mechanism. In this research, we used the whole-cell patch clamp technique to hippocampal CA1 pyramidal cells from C57BL mice and measured changes in spontaneous excitatory post-synaptic currents (sEPSCs), intracellular Ca ²⁺ concentration, mitochondrial membrane potential and N-methyl-D-aspartate receptor (NMDAR) currents under the inhibition of MCU by Ruthenium red 265 (Ru265) in PostC. Inhibition of MCU increased sEPSCs occurrence (p = 0.008), NMDAR currents (p < 0.001), intracellular Ca ²⁺ concentration (p < 0.001) and dead cells (p < 0.001) significantly after reperfusion, indicating the removal of the neuroprotective effects in PostC. Moreover, mitochondrial depolarization in PostC with Ru265 was gradually decreased after reperfusion (p < 0.001). These results suggest that MCU plays an important role in PostC by maintaining mitochondrial depolarization, which suppresses hyperactivation of NMDARs and prevents the elevation of intracellular Ca ²⁺ concentration against I/R injury.
Chapter
Millions of people suffer from acute hypoxic–ischemic brain injuries (HIBIs) such as stroke and neonatal HIBI worldwide each year, resulting in significant mortality and lifelong morbidity. Therapeutic options for HIBI remain limited, but many investigators are actively seeking out novel therapies based on our evolving understanding of the pathophysiology. Ferroptotic cell death has recently been demonstrated to play a significant part in the complex cellular injury resulting after HIBI. This chapter will discuss the current literature evaluating the role of ferroptosis-related reactive oxygen species release and lipid peroxidation in the pathophysiology of HIBI. Based on the current mechanistic understanding, several investigators have begun to develop interventions for HIBI that target the ferroptosis pathways. As such, this chapter will also discuss many of the ferroptosis inhibitors that have shown neuroprotective effects in cell culture and animal models of HIBI.
Preprint
Full-text available
Ischemic postconditioning (PostC) phenomenon is known as the neuroprotection against ischemic reperfusion (I/R) injury. One of the key processes in PostC is opening of mitochondrial ATP dependent potassium (mito-K ATP ) channel and depolarization of mitochondrial membrane potential, which triggers the release of calcium ion from mitochondria through the low conductance opening of mitochondrial permeability transition pore (mPTP). Mitochondrial calcium uniporter (MCU) is known as the highly sensitive transporter for uptake of Ca ²⁺ inwardly existed on the inner mitochondrial membrane. Furthermore, it has attracted attention as a new target of treatments in disease such as neurodegenerative disease, cancer and ischemic stroke. Thus, we considered that MCU may involve in PostC and trigger its mechanism. In this research, we used the whole-cell patch clamp technique to hippocampal CA1 pyramidal cells from C57BL mice and measured changes in spontaneous excitatory post-synaptic currents (sEPSCs), intracellular Ca ²⁺ concentration, mitochondrial membrane potential and N-methyl-D-aspartate receptor (NMDAR) currents under the inhibition of MCU by Ruthenium red 265 (Ru265) in PostC. Inhibition of MCU increased sEPSCs occurrence (p = 0.008), NMDAR currents (p < 0.001), intracellular Ca ²⁺ concentration (p < 0.001) and dead cells (p < 0.001) significantly after reperfusion, indicating the removal of the neuroprotective effects in PostC. Moreover, the mitochondrial depolarization in PostC with Ru265 was weakened, compared to it in PostC (p = 0.03). These results suggest that MCU affects the mitochondrial depolarization in the PostC mechanism to suppress NMDAR over-activation and prevent the elevation of intracellular Ca ²⁺ concentration against I/R injury.
Article
Glutamate dehydrogenase (GDH) is a key enzyme in mammalian glutamate metabolism. It is located at the intersection of multiple metabolic pathways and participates in a variety of cellular activities. GDH activity is strictly regulated by a variety of allosteric compounds. Here, we review the unique distribution and expressions of GDH in the brain nervous system. GDH plays an essential role in the glutamate-glutamine-GABA cycle between astrocytes and neurons. The dysfunction of GDH may induce the occurrence of many neurodegenerative diseases, such as Parkinson's disease, epilepsy, Alzheimer's disease, schizophrenia, and frontotemporal dementia. GDH activators and gene therapy have been found to protect neurons and improve motor disorders in neurodegenerative diseases caused by glutamate metabolism disorders. To date, no medicine has been discovered that specifically targets neurodegenerative diseases, although several potential medicines are used clinically. Targeting GDH to treat neurodegenerative diseases is expected to provide new insights and treatment strategies.
Article
Full-text available
After the onset of an acute episode of arrested circulation to the brain and consequent cerebral hypoxia, EEG changes and modifications of consciousness ensue within seconds. This in part reflects the rapid effect of hypoxia on the neocortex, where oxygen deprivation leads to impaired neuronal excitability and abnormal synaptic transmission. To identify the cellular mechanisms responsible for the earliest changes in neocortical function and to determine their time course, we have used patch-in-slice recording techniques to investigate the effects of acute hypoxia on the synaptic and intrinsic properties of layer 5 neurons. Coronal slices of mouse somatosensory cortex were maintained at 37°C and challenged with episodes of hypoxia (3–4 min of exposure to 95% N 2 , 5% CO 2 ). In recordings with cell-attached patch electrodes, activation of ATP-sensitive potassium channels first became detectable 211 ± 11 sec (range, 185–240 sec; n = 6 patches) after the onset of hypoxia. Similar recording techniques revealed no alterations in the properties of Na ⁺ currents in the first 4 min after the onset of hypoxia. The earliest hypoxia-induced disturbance was a marked increase in the frequency of spontaneous EPSCs and IPSCs, which began within 15–30 sec of the removal of oxygen. This rapid synaptic effect was not sensitive to TTX and was present in Ca ²⁺ -free perfusate, indicating that the hypoxia had a direct influence on the vesicular release mechanisms. The incoherent, massive increase in miniature PSCs would be expected to deplete the readily releasable pool of vesicles in cortical terminals, and to thereby markedly distort the neuronal interactions that underlie normal circuit function.
Article
Full-text available
The extracellular glutamate concentration ([glu](o)) rises during cerebral ischemia, reaching levels capable of inducing delayed neuronal death. The mechanisms underlying this glutamate accumulation remain controversial. We used N-methyl-D-aspartate receptors on CA3 pyramidal neurons as a real-time, on-site, glutamate sensor to identify the source of glutamate release in an in vitro model of ischemia. Using glutamate and L-trans-pyrrolidine-2,4-dicarboxylic acid (tPDC) as substrates and DL-threo-beta-benzyloxyaspartate (TBOA) as an inhibitor of glutamate transporters, we demonstrate that energy deprivation decreases net glutamate uptake within 2-3 min and later promotes reverse glutamate transport. This process accounts for up to 50% of the glutamate accumulation during energy deprivation. Enhanced action potential-independent vesicular release also contributes to the increase in [glu](o), by approximately 50%, but only once glutamate uptake is inhibited. These results indicate that a significant rise in [glu](o) already occurs during the first minutes of energy deprivation and is the consequence of reduced uptake and increased vesicular and nonvesicular release of glutamate.
Article
Full-text available
NMDA receptors (NMDARs) are glutamate-gated ion channels and are crucial for neuronal communication. NMDARs form tetrameric complexes that consist of several homologous subunits. The subunit composition of NMDARs is plastic, resulting in a large number of receptor subtypes. As each receptor subtype has distinct biophysical, pharmacological and signalling properties, there is great interest in determining whether individual subtypes carry out specific functions in the CNS in both normal and pathological conditions. Here, we review the effects of subunit composition on NMDAR properties, synaptic plasticity and cellular mechanisms implicated in neuropsychiatric disorders. Understanding the rules and roles of NMDAR diversity could provide new therapeutic strategies against dysfunctions of glutamatergic transmission.
Article
Full-text available
The activation of N-methyl-D-aspartate-receptors (NMDARs) in synapses provides plasticity and cell survival signals, whereas NMDARs residing in the neuronal membrane outside synapses trigger neurodegeneration. At present, it is unclear how these opposing signals are transduced to and discriminated by the nucleus. In this study, we demonstrate that Jacob is a protein messenger that encodes the origin of synaptic versus extrasynaptic NMDAR signals and delivers them to the nucleus. Exclusively synaptic, but not extrasynaptic, NMDAR activation induces phosphorylation of Jacob at serine-180 by ERK1/2. Long-distance trafficking of Jacob from synaptic, but not extrasynaptic, sites depends on ERK activity, and association with fragments of the intermediate filament α-internexin hinders dephosphorylation of the Jacob/ERK complex during nuclear transit. In the nucleus, the phosphorylation state of Jacob determines whether it induces cell death or promotes cell survival and enhances synaptic plasticity.
Article
Spreading depression (SD) and the related hypoxic SD-like depolarization (HSD) are characterized by rapid and nearly complete depolarization of a sizable population of brain cells with massive redistribution of ions between intracellular and extracellular compartments, that evolves as a regenerative, “all-or-none” type process, and propagates slowly as a wave in brain tissue. This article reviews the characteristics of SD and HSD and the main hypotheses that have been proposed to explain them. Both SD and HSD are composites of concurrent processes. Antagonists of N-methyl-d-aspartate (NMDA) channels or voltage-gated Na ⁺ or certain types of Ca ²⁺ channels can postpone or mitigate SD or HSD, but it takes a combination of drugs blocking all known major inward currents to effectively prevent HSD. Recent computer simulation confirmed that SD can be produced by positive feedback achieved by increase of extracellular K ⁺ concentration that activates persistent inward currents which then activate K ⁺ channels and release more K ⁺ . Any slowly inactivating voltage and/or K ⁺ -dependent inward current could generate SD-like depolarization, but ordinarily, it is brought about by the cooperative action of the persistent Na ⁺ current I Na,P plus NMDA receptor-controlled current. SD is ignited when the sum of persistent inward currents exceeds persistent outward currents so that total membrane current turns inward. The degree of depolarization is not determined by the number of channels available, but by the feedback that governs the SD process. Short bouts of SD and HSD are well tolerated, but prolonged depolarization results in lasting loss of neuron function. Irreversible damage can, however, be avoided if Ca ²⁺ influx into neurons is prevented.
Article
Basal extracellular glutamate sampled in vivo is present in micromolar concentrations in the extracellular space outside the synaptic cleft, and neither the origin nor the function of this glutamate is known. This report reveals that blockade of glutamate release from the cystine-glutamate antiporter produced a significant decrease (60%) in extrasynaptic glutamate levels in the rat striatum, whereas blockade of voltage-dependent Na+ and Ca2+ channels produced relatively minimal changes (0-30%). This indicates that the primary origin of in vivo extrasynaptic glutamate in the striatum arises from nonvesicular glutamate release by the cystine-glutamate antiporter. By measuring [35S]cystine uptake, it was shown that similar to vesicular release, the activity of the cystine-glutamate antiporter is negatively regulated by group II metabotropic glutamate receptors (mGluR2/3) via a cAMP-dependent protein kinase mechanism. Extracellular glutamate derived from the antiporter was shown to regulate extracellular levels of glutamate and dopamine. Infusion of the mGluR2/3 antagonist (RS)-1-amino-5-phosphonoindan-1-carboxylic acid (APICA) increased extracellular glutamate levels, and previous blockade of the antiporter prevented the APICA-induced rise in extracellular glutamate. This suggests that glutamate released from the antiporter is a source of endogenous tone on mGluR2/3. Blockade of the antiporter also produced an increase in extracellular dopamine that was reversed by infusing the mGluR2/3 agonist (2R,4R)-4-aminopyrrolidine-2,4-dicarboxlylate, indicating that antiporter-derived glutamate can modulate dopamine transmission via mGluR2/3 heteroreceptors. These results suggest that nonvesicular release from the cystine-glutamate antiporter is the primary source of in vivo extracellular glutamate and that this glutamate can modulate both glutamate and dopamine transmission.
Article
(4S)-4-(3-[(18)F]fluoropropyl)-l-glutamate (BAY 94-9392, alias [(18)F]FSPG) is a new tracer to image x(C)(-) transporter activity with positron emission tomography (PET). We aimed to explore the tumor detection rate of [(18)F]FSPG in patients relative to 2-[(18)F]fluoro-2-deoxyglucose ([(18)F]FDG). The correlation of [(18)F]FSPG uptake with immunohistochemical expression of x(C)(-) transporter and CD44, which stabilizes the xCT subunit of system x(C)(-), was also analyzed. Patients with non-small cell lung cancer (NSCLC, n = 10) or breast cancer (n = 5) who had a positive [(18)F]FDG uptake were included in this exploratory study. PET images were acquired following injection of approximately 300 MBq [(18)F]FSPG. Immunohistochemistry was done using xCT- and CD44-specific antibody. [(18)F]FSPG PET showed high uptake in the kidney and pancreas with rapid blood clearance. [(18)F]FSPG identified all 10 NSCLC and three of the five breast cancer lesions that were confirmed by pathology. [(18)F]FSPG detected 59 of 67 (88%) [(18)F]FDG lesions in NSCLC, and 30 of 73 (41%) in breast cancer. Seven lesions were additionally detected only on [(18)F]FSPG in NSCLC. The tumor-to-blood pool standardized uptake value (SUV) ratio was not significantly different from that of [(18)F]FDG in NSCLC; however, in breast cancer, it was significantly lower (P < 0.05). The maximum SUV of [(18)F]FSPG correlated significantly with the intensity of immunohistochemical staining of x(C)(-) transporter and CD44 (P < 0.01). [(18)F]FSPG seems to be a promising tracer with a relatively high cancer detection rate in patients with NSCLC. [(18)F]FSPG PET may assess x(C)(-) transporter activity in patients with cancer. Clin Cancer Res; 18(19); 5427-37. ©2012 AACR.
Article
N-methyl-d-aspartate receptors (NMDARs) are located in neuronal cell membranes at synaptic and extrasynaptic locations, where they are believed to mediate distinct physiological and pathological processes. Activation of NMDARs requires glutamate and a coagonist whose nature and impact on NMDAR physiology remain elusive. We report that synaptic and extrasynaptic NMDARs are gated by different endogenous coagonists, d-serine and glycine, respectively. The regionalized availability of the coagonists matches the preferential affinity of synaptic NMDARs for d-serine and extrasynaptic NMDARs for glycine. Furthermore, glycine and d-serine inhibit NMDAR surface trafficking in a subunit-dependent manner, which is likely to influence NMDARs subcellular location. Taking advantage of this coagonist segregation, we demonstrate that long-term potentiation and NMDA-induced neurotoxicity rely on synaptic NMDARs only. Conversely, long-term depression requires both synaptic and extrasynaptic receptors. Our observations provide key insights into the operating mode of NMDARs, emphasizing functional distinctions between synaptic and extrasynaptic NMDARs in brain physiology.
Article
Stroke has fallen from the second to the fourth leading cause of death in the United States without large declines in stroke incidence or case fatality. We explored whether this decline may be attributable to changes in mortality attribution methodology. Multicause mortality files from 2000 to 2008 were used to compare changes in reporting of stroke as underlying cause of death (UCOD) with changes in death certificates reporting any mention (AMCOD) of stroke. In addition, the UCOD/AMCOD ratio was calculated for the 6 leading organ and disease-specific causes of death. If stroke mortality is underestimated by the system of mortality attribution, we hypothesized that we would find: (1) a greater decline in stroke as UCOD than as AMCOD; and (2) a decline in the UCOD/AMCOD ratio compared with other causes of death. Age-adjusted death rates for stroke as UCOD (61 per 100,000 in 2000 versus 41 in 2008) and AMCOD (102 per 100,000 versus 68) both declined by 33%. The ratio of UCOD to AMCOD for stroke did not change over time (0.595 in 2000 versus 0.598 in 2008). Changes in UCOD/AMCOD ratio for the diagnoses that surpassed stroke as UCOD were too small (no change for lung cancer and a slight increase from 0.49 to 0.52 for chronic lower respiratory diseases) to explain stroke's decline as UCOD. Changes in mortality attribution methodology are not likely responsible for stroke's decline as a leading cause of death. The discordant trends in incidence, case fatality, and mortality require further study.