ArticlePDF Available

A Transient Upregulation of Glutamine Synthetase in the Dentate Gyrus Is Involved in Epileptogenesis Induced by Amygdala Kindling in the Rat

PLOS
PLOS ONE
Authors:

Abstract and Figures

Reduction of glutamine synthetase (GS) function is closely related to established epilepsy, but little is known regarding its role in epileptogenesis. The present study aimed to elucidate the functional changes of GS in the brain and its involvement in epileptogenesis using the amygdala kindling model of epilepsy induced by daily electrical stimulation of basolateral amygdala in rats. Both expression and activity of GS in the ipsilateral dentate gyrus (DG) were upregulated when kindled seizures progressed to stage 4. A single dose of L-methionine sulfoximine (MSO, in 2 µl), a selective GS inhibitor, was administered into the ipsilateral DG on the third day following the first stage 3 seizure (just before GS was upregulated). It was found that low doses of MSO (5 or 10 µg) significantly and dose-dependently reduced the severity of and susceptibility to evoked seizures, whereas MSO at a high dose (20 µg) aggravated kindled seizures. In animals that seizure acquisition had been successfully suppressed with 10 µg MSO, GS upregulation reoccurred when seizures re-progressed to stage 4 and re-administration of 10 µg MSO consistently reduced the seizures. GLN at a dose of 1.5 µg abolished the alleviative effect of 10 µg MSO and deleterious effect of 20 µg MSO on kindled seizures. Moreover, appropriate artificial microRNA interference (1 and 1.5×10(6) TU/2 µl) of GS expression in the ipsilateral DG also inhibited seizure progression. In addition, a transient increase of GS expression and activity in the cortex was also observed during epileptogenesis evoked by pentylenetetrazole kindling. These results strongly suggest that a transient and region-specific upregulation of GS function occurs when epilepsy develops into a certain stage and eventually promotes the process of epileptogenesis. Inhibition of GS to an adequate degree and at an appropriate timing may be a potential therapeutic approach to interrupting epileptogenesis.
Content may be subject to copyright.
A Transient Upregulation of Glutamine Synthetase in the
Dentate Gyrus Is Involved in Epileptogenesis Induced by
Amygdala Kindling in the Rat
Hong-Liu Sun
1,3.
, Shi-Hong Zhang
1.
, Kai Zhong
1
, Zheng-Hao Xu
1
, Bo Feng
1
, Jie Yu
1
, Qi Fang
1
,
Shuang Wang
2
, Deng-Chang Wu
1
, Jian-Min Zhang
2
, Zhong Chen
1,2
*
1Department of Pharmacology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, College of
Pharmaceutical Sciences, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China, 2Epilepsy Center, Second Affiliated Hospital, School of Medicine, Zhejiang
University, Hangzhou, Zhejiang, China, 3Department of Pharmacology, Binzhou Medical University, Yantai, China
Abstract
Reduction of glutamine synthetase (GS) function is closely related to established epilepsy, but little is known regarding its
role in epileptogenesis. The present study aimed to elucidate the functional changes of GS in the brain and its involvement
in epileptogenesis using the amygdala kindling model of epilepsy induced by daily electrical stimulation of basolateral
amygdala in rats. Both expression and activity of GS in the ipsilateral dentate gyrus (DG) were upregulated when kindled
seizures progressed to stage 4. A single dose of L-methionine sulfoximine (MSO, in 2 ml), a selective GS inhibitor, was
administered into the ipsilateral DG on the third day following the first stage 3 seizure (just before GS was upregulated). It
was found that low doses of MSO (5 or 10 mg) significantly and dose-dependently reduced the severity of and susceptibility
to evoked seizures, whereas MSO at a high dose (20 mg) aggravated kindled seizures. In animals that seizure acquisition had
been successfully suppressed with 10 mg MSO, GS upregulation reoccurred when seizures re-progressed to stage 4 and re-
administration of 10 mg MSO consistently reduced the seizures. GLN at a dose of 1.5 mg abolished the alleviative effect of
10 mg MSO and deleterious effect of 20 mg MSO on kindled seizures. Moreover, appropriate artificial microRNA interference
(1 and 1.5610
6
TU/2 ml) of GS expression in the ipsilateral DG also inhibited seizure progression. In addition, a transient
increase of GS expression and activity in the cortex was also observed during epileptogenesis evoked by pentylenetetrazole
kindling. These results strongly suggest that a transient and region-specific upregulation of GS function occurs when
epilepsy develops into a certain stage and eventually promotes the process of epileptogenesis. Inhibition of GS to an
adequate degree and at an appropriate timing may be a potential therapeutic approach to interrupting epileptogenesis.
Citation: Sun H-L, Zhang S-H, Zhong K, Xu Z-H, Feng B, et al. (2013) A Transient Upregulation of Glutamine Synthetase in the Dentate Gyrus Is Involved in
Epileptogenesis Induced by Amygdala Kindling in the Rat. PLoS ONE 8(6): e66885. doi:10.1371/journal.pone.0066885
Editor: Thierry Ame
´de
´e, Centre national de la recherche scientifique, University of Bordeaux, France
Received February 3, 2013; Accepted May 13, 2013; Published June 18, 2013
Copyright: ß2013 Sun et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted
use, distribution, and reproduction in any medium, provided the original author and source are credited.
Funding: This project was supported by grants from the National Natural Science Foundation of China (81030061, 81273506, 81221003, 81173042, 81202516,
81273492). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
Competing Interests: The authors have declared that no competing interests exist.
* E-mail: chenzhong@zju.edu.cn
.These authors contributed equally to this work.
Introduction
Up to one third of epilepsy patients continue to experience
seizures or unacceptable medication-related side effects. Among
patients with temporal lobe epilepsy, more than half develop drug-
resistant seizures [1]. Moreover, most of the available drugs only
inhibit ictogenesis (seizure occurrence), but not epileptogenesis (the
process of developing epilepsy). Intensive studies on the process of
epileptogenesis are of great importance to the development of
therapeutic approaches.
Excessive levels of cerebral glutamate are considered a crucial
factor for epilepsy [2,3]. Glutamate or glutamate analogues
administered to the hippocampus can elicit seizures, whereas
glutamate antagonists block them [4,5]. Moreover, hyperexcit-
ability of dentate granule cells from humans with mesial temporal
lobe epilepsy (MTLE) is found to be glutamate-dependent [3].
Normally, most of the extracellular glutamate is taken up by high-
affinity excitatory amino-acid transporters on adjacent astrocytes
[6,7] where glutamate is rapidly converted to glutamine (GLN)
under the catalysis of glutamine synthetase (GS). GLN is then
transferred back to neurons and converted to glutamate before
repackaging into synaptic vesicles for release [8]. In this cycle,
which is named glutamate-glutamine cycling [9], GS controls the
rate and holds a key position in glutamate homeostasis. GS
deficiency may result in glutamate accumulation in astrocytes and
extracellular space, which may result in neuronal hyperexcitability
[10–12].
Eid et al. [13] reported that both the expression and enzyme
activity of GS are around 40% lower in MTLE hippocampi than
in non-MTLE hippocampi. Similarly, in the chronic phase of
epileptic condition after pilocarpine-induced status epilepticus, the
expression of GS is down-regulated in newly generated astrocytes
[14]. GS activity also shows a significant and region-specific
reduction after pentylenetetrazole (PTZ)-induced repetitive epi-
leptic seizures [15]. So, it has been proposed that the reduction in
GS expression or enzyme activity might be the main reason for
PLOS ONE | www.plosone.org 1 June 2013 | Volume 8 | Issue 6 | e66885
increased extracellular glutamate concentrations in epileptic
animals and patients [16]. On the other hand, Hammer et al.
[17] have recently reported that GS expression in hippocampal
formation increases in the latent phase, while approaches control
levels in the chronic phase in the kainate model of epilepsy.
However, it remains unknown regarding the significance of such
an increase in GS expression. It is likely that function of GS may
change dynamically in epileptogenesis and play different roles
from that in established epilepsy.
Therefore, in the present study we first investigated the dynamic
changes of GS during seizure acquisition using the rat amygdala
kindling model induced by daily electrical stimulation of
basolateral amygdala. We then manipulated the levels of GS in
the DG area using pharmacological and artificial microRNA
interference techniques in an attempt to elucidate its roles in
epileptogenesis induced by amygdala kindling. It is very interesting
to find that a transient upregulation of GS function occurs when
epilepsy develops into a specific stage and eventually promotes the
process of epileptogenesis.
Materials and Methods
Ethics Statement
All experiments were in accordance with the ethical guidelines
approved by the Zhejiang University Animal Experimentation
Committee (Zju2009-02-05-002) and were in complete compli-
ance with the National Institutes of Health Guide for the Care and
Use of Laboratory Animals (NIH Publications No. 80-23, revised
1996). All surgery was performed under chloral hydrate anesthesia
(400 mg/kg, i.p.), and all efforts were made to minimize suffering.
Animals
Animals used in this study were male Sprague-Dawley rats
(280–300 g, Grade II, Certificate No. SCXK2003-0001; provided
by the Experimental Animal Center, Zhejiang Academy of
Medical Science, Hangzhou, China), maintained in individual
cages with a 12-h light-dark cycle (lights on from 8:00 to 20:00).
Water and food were given ad libitum. Experiments were carried
out between 10:00 and 17:00.
Electrodes Implantation and Amygdala Kindling
Under anesthesia, rats were mounted in a stereotaxic apparatus
(Stoelting, USA). Electrodes were implanted into the right
basolateral amygdala (AP: –2.4 mm, L: –4.8 mm, V: –8.8 mm).
The electrodes were made of twisted stainless steel Teflon-coated
wires (diameter 0.2 mm; A.M. Systems, USA) insulated except for
0.5 mm at the tip. The tip separation was 0.7 0.8 mm. An
electrode fixed to the frontal bone served as a reference. The
electrodes were connected to a miniature receptacle. A stainless
steel guide cannula (Reward, China) was implanted into the right
DG (AP: –5.04 mm, L: –3 mm, V: –3.5 mm). In some animals,
another recording electrode binding on the cannula was implanted
together into the same place as the cannula to monitor the EEG in
this area. The receptacle and cannula were embedded in the skull
with dental cement. Animals were allowed to recover from surgery
for 10 days [18–21].
Kindling stimulation of the amygdala consisted of monophasic
square-wave pulses (60 Hz; 1 ms pulse duration; 1 sec train
duration) delivered by a constant current stimulator (Nihon
Kohden, Japan). Electroencephalograms (EEGs) of the amygdala
were recorded with a digital amplifier (NuAmps, Neuroscan
System, USA). Afterdischarge threshold (ADT) was determined
as previously reported [22–25]. In brief, the stimulus intensity
was initiated at 50 mA and increased in increments of 20 mAuntil
at least 5 sec of afterdischarge (AD) was observed in the EEG,
and this current intensity was defined as the ADT. All animals
were subjected to kindling stimulation with the same current
intensity as their own ADT once daily until they were fully
kindled, i.e., the animal exhibited three consecutive stage 5
seizures.
PTZ Kindled Seizures
Rats received an intraperitoneal injection of saline or PTZ
(40 mg/kg, Sigma) every other day as we previously described
[26]. Then the animals were placed in a plexiglas arena
(50 cm630 cm630 cm) and their behaviors were observed for
90 min.
Classification of Seizure Severity
For western blottingblott ng and in PTZ kindling animals?
Seizure severity during kindling was classified according to the
modification of Racine [27]: (1) facial movement; (2) head
nodding; (3) unilateral forelimb clonus; (4) bilateral forelimb
clonus (BFC) and rearing; and (5) BFC and rearing and falling.
Stages 1 to 3 were considered as focal seizures, while stages 4 and 5
were generalized seizures. AD duration and generalized seizure
duration, which was defined as the duration of BFC, were also
recorded.
Drug Intervention
Drugs, all in 2 ml, were infused into the right DG in 10 min with
a disposable dental needle (30 g, Nipro Medical Industries Ltd,
Japan) of which the tip was 0.2 mm below the guide cannula. The
needle was left in place for 5 min before slowly retracted. After the
kindling stimulation, L-methionine sulfoximine (MSO, 5, 10 or
20 mg, Sigma) or saline was injected in a single dose on the day
when the animal showed the first stage 2 seizure or on the third
day following the first stage 3 seizure. GLN (1.5 mg, Sigma) or
saline was given 20 min after MSO treatment and was re-
administered once daily for next 2 consecutive days. Behavioral
seizures and EEG in the amygdala were recorded by video-EEG
monitoring for 24 h after MSO administration.
Lentiviral vectors containing pcDNA
TM
6.2-GW/EmGFPmiR
targeting GS were constructed and purified by Invitrogen
(Shanghai, China) using the following sequences of oligonucleo-
tides: 59-tgctgATCAATGGCCTCCTCAATG CAGTTTTGG-
CCACTGACTGA CTGCATTGAAGGCCATTGAT-39and59-
cctgATCAATGGCCTTCAATGCAGTCAGTCAGTGGCCA-
AAACTGCATTGAGGAGGCCATTGATC -39. Negative vec-
tors were produced using the following sequences of oligonucle-
otides: 59- tgctgAA ATGTACTGCGCGTGGAGACGTTTTG
GCCACTGA CTGACGTCTCCACGCAGTACATTT-39and
59- cctgAAATGTACT GCGTGGAGACGTCAGTCAGTGG-
CCA AAACGTCTCCACGCGCAGTACATTT c-39.Onthe
third day following the first stage 3 seizure, increasing doses of
interference vectors (0.5, 1, 1.5, 2.5610
6
TU diluted to 2 mlin
saline), negative vectors (control) or saline, were injected into the
right DG in the same way as MSO.
ADT was determined just before the injection and again on the
third day after drug injection. At the end of the experiments,
placements of cannulas and electrodes were histologically verified.
Only animals with electrodes and cannulas correctly implanted in
both the basolateral amygdala and the DG were included in the
statistical analysis.
Glutamine Synthetase in Epileptogenesis
PLOS ONE | www.plosone.org 2 June 2013 | Volume 8 | Issue 6 | e66885
Immunohistochemistry
On the second day of each seizure stage, rats were deeply
anesthetized with chloral hydrate and perfused intracardially with
phosphate buffered saline followed by 4% paraformaldehyde
(pH 7.4). The brains were removed and post-fixed in the same
fixative for 4 h at 4uC, then cryoprotected by infiltration with 30%
sucrose overnight. Coronal slices throughout the entire hippo-
campus were cut at 12 mm on a cryostat (CM3050s, Leica,
Germany) and adhered onto gelatin-coated slides. For double
immunofluorescent staining for glial fibrillary acidic protein
(GFAP) and GS, the sections were first incubated with 3% bovine
serum albumin in PBS for 30 min at room temperature, then were
incubated in mixture of rabbit anti-GFAP IgG (Chemicon, USA,
diluted 1:200) and mouse anti-GS IgG (Chemicon, USA, diluted
1:200) in PBS containing 0.3% Triton X-100 overnight at room
temperature. After washing three times for 10 min with PBS,
sections were incubated sequentially in a mixture of FITC-
(Chemicon, USA, diluted 1:200) and Cy3- (Beyotime, China,
diluted 1:200) conjugated secondary antisera for 1 h at room
temperature. Then, after washing three times for 10 min with
PBS, the sections were covered with glass coverslips and observed
under a fluorescence microscope (Olympus, Japan). Images were
captured under identical exposure conditions to guarantee the
clarity and comparability among images. The fluorescence
intensity of different brain regions is also comparable since the
influence of capture conditions on brightness was restricted to the
whole image. Fluorescence intensity was quantified using ImageJ
1.37 software (National Institutes of Health).
Western Blot Analysis and Enzyme Activity Assay
On the second day of each stage in amygdala kindling and PTZ
kindling, animals were decapitated and the brains were removed
without delay, then microdissected into DG, CA3, CA1 and cortex
[28]. Tissues were frozen in liquid nitrogen for further western blot
analysis and enzyme activity assay.
For western blotting, frozen microdissected subregions were
sonicated on ice in homogenisation buffer (1% SDS, 10 mM
sodium phosphate buffer, pH 7.4, 150 mM NaCl, 1 mM
phenylmethylsulphonyl fluoride, 10 mM ethylenediamine tetra-
acetic acid). Protein concentrations were determined with a
bicinchoninic acid (BCA) protein assay kit and measured on a
microplate reader at 570 nm (Biotek, USA). Protein homoge-
nates were mixed with sample loading buffer (62.5 mM Tris-
HCl, 10% glycerol, 2% SDS, 5% 2- mercaptoethanol, 0.025%
bromophenol blue). Protein samples were separated by 12%
SDS-polyacrylamide gels and then electrotransferred onto a
nitrocellulose membrane. After blocking with 5% fat-free milk,
the membranes were incubated with mouse monoclonal
antibody against GS (1:800, Chemicon) or glyceraldehyde-3-
phosphate dehydrogenase (GAPDH, 1:5,000, Kangchen) at 4uC
overnight. After repeated washing, the membranes were reacted
with IRDye 700 anti-mouse molecular probe (Odyssey; LI-
COR) for 2 h. Images were acquired with the Odyssey infrared
imaging system and analyzed by the software program as
specified in the Odyssey software manual. Results were
expressed as GS/GAPDH ratio, and then normalized to the
values measured in control groups.
GS activity was measured by a GS activity assay kit ( Jiancheng,
Nanjing, China; Category No. A047) according to the c-glutamyl
transferase reaction as described previously [15]. Briefly, after
determining the protein concentration as described above, 10 ml
tissue homogenates or standard solution (20 mmol/mL) or distilled
water (control) were incubated with 152 ml reaction mixture at
37uC for 15 min. Reaction was terminated by adding 40 mlofa
stop-solution. Insoluble material was removed by centrifugation at
3,500 g for 10 min at 4uC. After measuring the absorbance (optic
density, OD), 100 ml supernatant was removed to the microplate
and the OD value representing the amount of reaction product, c-
glutamyl hydroxamate was obtained. The factual OD of each well
for further calculation was the difference between the raw OD and
the respective blank OD. Enzymatic activity expressed as units per
milligram of sample protein (U/mg) was calculated based on the
following formula: GS activity (U/mg) = [(OD of the test sample
OD of the control)/(OD of the standard solution OD of the
control)] 6concentration in standard solution (20 mmol/ml) 64/
protein content (mg/ml) [29].
Statistical Analysis
Values are expressed as mean 6SEM. Statistical analysis was
carried out by SPSS 13.0 for Windows. Analysis of group
progression of seizure stage, duration of AD, and generalized
seizures during kindling was performed by two-way analysis of
variance (ANOVA) for repeated measures. Comparison of the
cumulative numbers of stimulationsneededineachseizurestage
and to reach stage 5 during kindling was done with the
nonparametric Mann-Whitney U test. One-way ANOVA was
used for comparison of other indices. For all analyses, the tests
were two-sided and a P,0.05 was considered significant.
Results
Transient Upregulation of GS in the Ipsilateral DG area
during Amygdala Kindling
Immunohistochemical studies showed that GS was co-localized
with GFAP, a specific marker of astrocytes, throughout all
subfields in the brain. The intensity of GS immunoreactivity did
not change noticeably during the stages of focal seizures (stages 1
to 3; Fig. 1A–F and Q). However, in seizure stage 4, GS expression
evidently increased in the ipsilateral DG area (P,0.001; Fig. 1G–I
and Q) without detectable astroglial proliferation as determined by
counting GFAP-positive cells (data not shown). GS expression in
the DG area declined to the control level when the animal was
fully kindled (Fig. 1J–L and Q). Western blotting experiments
confirmed that the amount of GS in the ipsilateral DG area was
about 70% higher than that of control animals (P,0.01; Fig. 2A
and C). No significant change of GS expression was found in
hippocampal CA3 and CA1 subregions (Fig. 1M–P, S and T; Fig.
2B and C) and the cortex (data not shown) at each seizure stage. In
addition, GS activity was 37% higher (P,0.05) in the ipsilateral
DG homogenates from rats in seizure stage 4 (75.5665.67 U/mg)
than in those from controls (58.8863.33 U/mg) (Fig. 2D). No
significant difference in GS activity was found in the contralateral
DG or other subregions, or at other seizure stages (Fig. 2E, F and
G).
To study whether the increase of GS in the DG at stage 4 is
specific for the amygdala kindling model of epilepsy, we assessed
the GS expression and activity in the brain of rats subjected to
PTZ-kindling model of epilepsy. A marked increase in GS
immunoreactivity and activity in the cortex was also detected at
seizure stage 4 (P,0.001; Fig. 3). No significant differences in GS
immunoreactivity and activity were found in other stages or in
other brain regions.
Inhibition of GS in the Ipsilateral DG by MSO Reduced
Evoked Seizures
To determine the role of GS upregulation in kindling
acquisition, MSO (5, 10, and 20 mg) were infused into the
ipsilateral DG on the third day following the first stage 3 seizure
Glutamine Synthetase in Epileptogenesis
PLOS ONE | www.plosone.org 3 June 2013 | Volume 8 | Issue 6 | e66885
Figure 1. GS overexpression in the ipsilateral DG area in the rat amygdala kindling model. Immunoreactivity of GFAP (red) and GS
(green) in the ipsilateral DG (A–L, bar = 50 mm) and the CA3 region (M–P, bar = 20 mm) during seizure acquisition induced by amygdala kindling
(control group, A–C; stages 1–2, not shown; stage 3, D–F; stage 4, G–I; fully kindled, J–L; n = 6/stage). The mean intensity of GS immunoreactivity in
the ipsilateral DG region was significantly increased in stage 4 compared with controls (Q), while no change was observed in GFAP expression (R). GS
expression did not change in CA1 (S) and CA3 (T) regions. Data are shown as mean 6SEM. ***P,0.001 compared with controls.
doi:10.1371/journal.pone.0066885.g001
Glutamine Synthetase in Epileptogenesis
PLOS ONE | www.plosone.org 4 June 2013 | Volume 8 | Issue 6 | e66885
(just before the seizures progressed to stage 4) in order to inhibit
the function of GS enzyme. MSO 5 mg and 10 mg did not evoke
epileptiform discharges in EEG nor visible behavioral seizures,
whereas MSO 20 mg induced epileptiform EEG in all treated rats
and three out of nine presented spontaneous seizures. The
progression of behavioral seizure stages in animals receiving
MSO 5 mg and 10 mg was slower than that in the control group
receiving saline (P,0.05 and P,0.001; Fig. 4A). When animals in
Figure 2. Upregulation of GS function in the ipsilateral DG in the rat amygdala kindling model. Measurements of GS expression in the
ipsilateral DG (A) and CA3 (B) by western blotting and enzyme activity in bilateral DG (D and E), ipsilateral CA3(F) and CA1(G) in the progression of
amygdala kindling (control group, n = 6/stage; stages 1–4 and fully kindled, n = 5–7/stage). Data are shown as mean 6SEM. * P,0.05 and **P,0.01
compared with controls.
doi:10.1371/journal.pone.0066885.g002
Glutamine Synthetase in Epileptogenesis
PLOS ONE | www.plosone.org 5 June 2013 | Volume 8 | Issue 6 | e66885
the control group were all fully kindled, those treated with MSO
5mg and 10 mg were still in stages 4.060.6 and 2.360.7,
respectively (Fig. 4A). Meanwhile, MSO 5 mg and 10 mg shortened
AD durations during kindling (P,0.05 and 0.001, respectively;
Fig. 4B) and the later dose also reduced the duration of generalized
seizures (P,0.05; Fig. 4C)
Further analysis revealed that 5 mg and 10 mg MSO signifi-
cantly increased the number of stimulations needed to reach stage
5 and prolonged the time that animals stayed in stages 0–3
(P,0.01 and 0.001, Fig. 4D and E). Moreover, these treatments
not only prevented the decline of ADT along with seizure
progression observed in the control group, but even significantly
elevated ADT (P,0.01 and 0.001, respectively; Fig. 4F). An
inhibition of seizure severity with MSO 10 mg was also observed
when comparing the average AD duration and generalized seizure
duration of three consecutive stage 5 seizures when the animal was
fully kindled (P,0.05 and 0.01, respectively; Fig. 4G and H). In
contrast, MSO at a high dose (20 mg) remarkably accelerated
kindling progression and aggravated evoked seizures either during
kindling or when the animal was fully kindled (P,0.001, 0.01 and
0.05, respectively; Fig. 4C–H). Representative EEGs recorded pre-
and post-kindling stimulation from the right amygdala on the third
day after drug administration and EEGs at seizure stage 5 are
shown in Fig. 4I and J. Interestingly, the increase in immuno-
staining intensity of GS in the ipsilateral DG reoccurred when the
kindling acquisition re-progressed to seizure stage 4 (P,0.01; Fig.
5A). Re-administration of MSO 10 mg on the third day after
seizures re-progressed to stage 3 consistently inhibited the
progression of kindled seizures (Fig. 5B).
To verify whether the effects of MSO on kindled seizures were
due to GS inhibition, GLN, the product of GS, was administered
following MSO treatment. We found that GLN not only reversed
the deleterious effect of MSO 20 mg, but also abolished the
ameliorative effect of MSO 10 mg(P,0.001; Fig. 6).
However, if administered on the first day after seizures
progressed to stage 2, MSO dose-dependently accelerated seizure
progression (P,0.01; Fig. 7A), as well as prolonged AD and
generalized seizure duration (P,0.01 and 0.05 respectively; Fig.
7B and C). The number of stimulations required to reach stage 5
and the cumulative stimulations in stages 0–3 were also
significantly reduced (P,0.05 and 0.01 respectively; Fig. 7D
and E). Moreover, MSO 10 mg aggravated the decline of ADT
along with seizure acquisition (Fig. 7F). Representative EEGs
recorded pre- and post-kindling stimulation from the right
amygdala on the third day after drug administration are shown
in Fig. 7G.
microRNA Interference of GS in the Ipsilateral DG
Reduced Evoked Seizures
To further verify the role of GS upregulation in the ipsilateral
DG area during kindling, effects of artificial microRNA interfer-
ence lentiviral vectors were evaluated. Negative vectors (control
group) did not affect GS expression and kindled seizures compared
with saline. However, artificial microRNA interference vectors
titer-dependently reduced GS immunoreactivity around the
injected point (Fig. 8). The progression of behavioral seizures in
animals receiving 1 and 1.5610
6
TU/2 ml was markedly slower
than that in the control group (P,0.05 and P,0.001; Fig. 9A).
The AD duration and generalized seizure duration were also
significantly shorted (P,0.05, P,0.001 and P,0.001; respective-
ly; Fig. 9B and C). Moreover, the number of stimulations needed
to reach stage 5 and cumulative stimulations in stage 0–3 were
significantly higher (P,0.05, P,0.001 and P,0.01, P,0.001,
respectively; Fig. 9D and E). ADT in these two groups also
increased (Fig. 9F). In contrast, in the group treated with 2.5610
6
genome copies, the generalized seizure duration was significantly
prolonged (P,0.05; Fig. 9C), and the number of stimulations
needed to reach stage 5 was significantly less than those in other
Figure 3. Upregulation of GS function in the cortex in the rat PTZ-kindling model. GS immunoreactivity (A–E, bar = 20 mm) and activity (F)
in the cortex during PTZ kindling. GS upregulation was detected in seizure stage 4 (C, E) compared with the saline group (A). No change was found in
other stages (B, D, E) and in other subregions (DG, CA3+CA1; F; n = 5/group). Data are shown as mean 6SEM. *P,0.05 and ***P,0.001 compared
with the saline group.
doi:10.1371/journal.pone.0066885.g003
Glutamine Synthetase in Epileptogenesis
PLOS ONE | www.plosone.org 6 June 2013 | Volume 8 | Issue 6 | e66885
groups (P,0.001 and 0.05, respectively; Fig. 9D). No effect was
found with the dose of 0.5 610
6
TU/2 ml (Fig. 9).
Discussion
In the present study, we provided the first evidence that a
transient upregulation of GS function occurred in the ipsilateral
Figure 4. Interruption of GS upregulation by MSO and kindled seizures. MSO (5, 10, 20 mg) was injected into the ipsilateral DG on the third
day following the first stage 3 seizure. (A) behavioral stage progression of seizures, (B) AD duration, (C) generalized seizure duration, (D) numbersof
stimulations required to reach full kindled from MSO administration, (E) numbers of stimulations required to retain in stage 0–3 and stage 4 in
kindling acquisition, (F) difference of ADT between just before drug injection and on the third day after injection, (G) averaged AD duration and (H)
averaged generalized seizure duration of three consecutive stage 5 seizures (n = 9/group). I and J show electrographic examples of afterdischarge
recorded from the ipsilateral amygdala in four groups on the third day after drug injection and after the rats were fully kindled. ES indicates electrical
kindling stimulation. Data are shown as mean 6SEM. *P,0.05, **P,0.01, *** P,0.001 compared with the saline group;
#
P,0.05,
##
P,0.01,
###
P,0.001 compared with each other.
doi:10.1371/journal.pone.0066885.g004
Glutamine Synthetase in Epileptogenesis
PLOS ONE | www.plosone.org 7 June 2013 | Volume 8 | Issue 6 | e66885
DG area in a specific stage during the development of electrical
amygdala kindling. Interrupting GS function with MSO or
microRNA interference significantly inhibited kindling acquisition.
These results indicate that GS is very likely an important
contributor to epileptogenesis.
It has been shown that continuous inhibition of GS activity by
MSO, a specific and irreversible inhibitor of GS, induces
recurrent seizures and neuropathologicalfeaturesinratsthat
are similar to MTLE in humans [11,13,16], and haploinsuffi-
ciency of GS increases susceptibility to experimental febrile
seizures [12]. We also found in the present study that MSO
acutely infused into the DG area of naı
¨ve rats resulted in
seizures that were reversed by glutamine (GLN) replenishment
(data not shown) and that MSO dose-dependently accelerated
kindling progression if administered on the day when the animal
showed the first stage 2 seizure (GS is at normal level at that
time). These results support previous findings that both acute
and chronic impairment of GS function lead to the occurrence
of seizures. On the other hand, the present study revealed that
both expression and activity of GS transiently increased
specifically in the ipsilateral DG area in the early stage of
generalized kindled seizures, and upregulation of GS expression
and activity in the cortex was also detected in stage 4 seizures
induced by PTZ kindling. Hammer et al. [17] reported that GS
expression in the hippocampus is upregulated in the latent
phase, but reduced to control levels in the chronic phase after
kainate-induced status epilepticus, although the significance of
this increase remains unknown. All these data inspire us to
speculate that it is likely that the function of GS changes
dynamically and compartmentally along with the process of
epileptogenesis. We also noticed that the increase in GS activity
was not comparable with that in expression (37% vs 70%).
Previous studies have demonstrated that under epileptic
conditions, GS is nitrated, which leads to decrease in activity
Figure 5. Reoccurrence of GS upregulation. The increase in GS immunoreactivity in the ipsilateral DG reoccurred when kindled seizures in
animals treated with 10 mg MSO re-progressed to stage 4 (A, n = 6/stage) and re-administration of MSO 10 mg injected into the ipsilateral DG on the
third day after seizures re-progressed to stage 3 consistently inhibited seizure progression (B, n = 9/group). Data are shown as mean 6S.E.M. **
P,0.01 compared with the saline group.
doi:10.1371/journal.pone.0066885.g005
Figure 6. Reversion of MSO effects by GLN. MSO (10 mg and 20 mg) was injected into the ipsilateral DG on the third day following the first stage
3 seizure. GLN (1.5 mg) was given 20 min after MSO treatment and was re-administered once daily for next 2 consecutive days. (A) behavioral stage
progression of kindled seizures, (B) AD duration. Data are shown as mean 6SEM. *** P,0.001 compared with the control group.
doi:10.1371/journal.pone.0066885.g006
Glutamine Synthetase in Epileptogenesis
PLOS ONE | www.plosone.org 8 June 2013 | Volume 8 | Issue 6 | e66885
without changes in content [15,30,31]. Therefore, nitration of
GS may explain at least in part the less increase in activity than
in expression.
To assess the role of increased GS function in epileptogenesis,
we infused MSO or artificial microRNA into the ipsilateral DG on
the third day following the first stage 3 seizure (just before the
upregulation of GS in stage 4) to inhibit GS function. Consistent
with previous reports, a high dose of MSO (20 mg) or artificial
microRNA (2.5610
6
TU) that inhibited GS activity excessively
not only accelerated kindling acquisition, but also increased the
susceptibility to and the severity of evoked seizures, providing
further supportive evidence that excessive suppression of GS is
stimulative for epilepsy development. Interestingly, however,
lower doses of MSO or artificial microRNA that inhibited GS to
slight to moderate extent reduced kindling seizures. Since GLN
replenishment reversed the deleterious and ameliorative effects
provided by high and low doses of MSO, respectively, we
concluded that such effects were associated with GS inhibition.
Moreover, when the seizure stage in animals previously treated
with 10 mg MSO re-progressed to stage 4, the up-regulation of
GS expression in the ipsilateral DG reoccurred and the
inhibitory effect of 10 mg MSO on seizure acquisition was
replicated. These data strongly suggest that an upregulation of
GS function may be an indispensable process and serve as a
stimulative factor for the development of amygdala kindling. The
seemingly discrepant facts of GS deficiency in patients with
temporal lobe epilepsy and GS upregulation in certain stage of
amygdala kindling may not indicate that the kindling model is
not representative for the situation in human temporal lobe
epilepsy, but may suggest that a proper functional level of GS is
crucial for the maintenance of normal neuronal activity. GS
dysfunction manifested as either upregulation or impairment
that occurs in different stages or types of epilepsy are associated
with epileptogenesis.
Since GS is responsible for glutamate catabolism, the levels of its
function may be largely dependent on glutamate levels in
extracellular spaces and in astrocytes. Considering that the
extracellular level of glutamate increases along with the develop-
Figure 7. MSO accelerated kindled seizures. MSO (5, 10, and 20 mgin2ml) was injected into the ipsilateral DG on the first day after kindled
seizures progressed to stage 2. (A) behavioral stage progression of seizures, (B) AD duration, (C) generalized seizure duration, (D) numbers of
stimulations required to reach stage 5 from MSO administration, (E) numbers of stimulations required to retain in stage 0–3 and stage 4 in kindling
acquisition, (F) difference of ADT between just before drug administration and on the third day after administration (saline group, n = 6; MSO groups,
n = 9/group). G shows the electrographic examples of afterdischarge recorded from the ipsilateral amygdala in four groups on the third day after
drug administration. ES represents electrical kindling stimulation. Data are shown as mean 6S.E.M. *P,0.05, ** P,0.01 and *** P,0.001 compared
with the saline group. ### P,0.001, as compared with each other.
doi:10.1371/journal.pone.0066885.g007
Glutamine Synthetase in Epileptogenesis
PLOS ONE | www.plosone.org 9 June 2013 | Volume 8 | Issue 6 | e66885
ment of kindled seizures [32], it seems reasonable to speculate that
slight increase of extracellular glutamate in partial seizures (stage
1–3) is not sufficient to evoke GS upregulation in astrocytes. When
seizures were generalized (stage 4) and the extracellular glutamate
levels increase dramatically, GS function is responsively upregu-
lated in order to maintain glutamate homeostasis. As a result, the
rate of glutamate-glutamine conversion is finally accelerated.
However, this acceleration may in turn promote the synthesis and
availability of glutamate and hence contribute to seizure develop-
ment. Actually, it has been demonstrated that an adequate GLN
supply is crucial for epileptiform activity [33–35]. Since c-
aminobutyric acid (GABA), the main inhibitory neurotransmitter
in the central nervous system, is synthesized from glutamate, it is
possible that the acceleration of glutamate synthesis may be
accompanied with the increase in GABA levels. However, kindling
process produces an increased turnover of glutamate relative to
GABA [36] and neuronal hyperactivity is more depend on
glutamate-glutamine cycling [34,35]. Indeed, we measured the
concentration of glutamate and GABA in the ipsilateral DG by
HPLC when kindled seizures progressed to stage 4 and found a
marked increase in the ratio of glutamate and GABA (Fig. S1).
Therefore, the promotive effect of GS upregulation on kindling
acquisition may be explained at least in part by these mechanisms.
This is the first study reporting the role of GS upregulation in
epileptogenesis. Our results suggest that modifying GS function or
interfering the glutamate-glutamine cycle appropriately to main-
tain the stabilization of extracellular glutamate concentration may
be a potential therapeutic intervention for epileptogenesis.
Moreover, our findings also raise the issue that for patients with
high risk of developing epilepsy, GLN intake probably should be
monitored with caution.
Interestingly, the GS upregulation occurred only in the
ipsilateral DG area in the amygdala-kindling model, and only
in the cortex in the PTZ-kindling model. Such an epilepsy type-
related and region-specific GS upregulation indicates that
different brain areas are differentially involved epileptogenesis
induce by different types of insult. Our findings point that the
DG area is crucial for development of epilepsy induced by
amygdala kindling. Although according to available data, there
are no monosynaptic interconnections between the amygdaloid
complex and the DG, the former has rich projections to other
temporal structures that are closely interconnected with the latter
[37]. For example, the amygdala sends fibers to the entorhinal
cortex from which fibers are sent to the DG via the perforant
path, a known pathway that is closely involved in the
propagation and evolution of epileptiform activity induced by
kindling [38]. Along with the progression of kindling with
repeated stimulation, the same stimulus comes to trigger longer
and more widely propagating events that progressively recruit a
larger network, including the DG [38]. Moreover, the DG area
is a regulated gate for the propagation of epileptiform activity,
and granule cells here are more sensitive to excitatory input
[38,39]. An excessive activation of DG neurons under epileptic
condition serves to initiate and sustain seizure activity in the
hippocampal-parahippocampal loop and converts the DG to a
‘‘promoter’’ or ‘‘amplifier’’ of epileptiform discharges [40,41].
Therefore, it is possible that repeated kindling stimulation of the
amygdala gradually induces hyperactivity in dentate neurons.
And subsequently, elevated extracellular glutamate stimulates
glutamate-glutamine cycling and hence promotes epileptogen-
esis. Further studies are needed to elucidate the exact mecha-
nisms underlying the upregulation of GS function in the DG
area during kindling epileptogenesis.
In summary, in the present study, we found a transient but
pronounced upregulation of GS in the ipsilateral DG area during
seizure acquisition in the amygdala kindling model of epilepsy.
Figure 8. microRNA interference and GS expression. Artificial
microRNA interference vectors (0.5, 1.5, and 2.5610
6
TU/2 ml) was
injected into the ipsilateral DG on the third day following the first stage
3 seizure on immunoreactivity. (A–R) GFAP (red) and GS (green) (bar =
50 mm) detected on the third day after injection (control (negative
vector) group, A–C; saline group, D–F; 0.5610
6
TU/2 ml group, G–I;
1610
6
TU/2 ml group, J–L; 1.5610
6
genome copies group, M–O;
2.5610
6
TU/2 ml group, P–R; n = 6/group). The quantitative analysis
revealed a titer-dependent inhibition of GS immunoreactivity in the
ipsilateral DG region (S), while no change in GFAP expression was
observed (T). Data are shown as mean 6SEM. *** P,0.001 compared
with the control group.
doi:10.1371/journal.pone.0066885.g008
Glutamine Synthetase in Epileptogenesis
PLOS ONE | www.plosone.org 10 June 2013 | Volume 8 | Issue 6 | e66885
This increase appears to promote the kindling progression and the
appearance of generalized seizures. Inhibition of GS to an
adequate degree and at the appropriate timing may be a potential
therapeutic approach to interrupting epileptogenesis.
Supporting Information
Figure S1 Glutamate and GABA measurement. Gluta-
mate (A) and GABA (B) content (mg/ml) was measured 24 hrs after
the first stage 3 or 4 seizure in the right DG (RDG) and other
regions in the right hippocampal formation (RHF). (C) shows the
ratio between Glutamate and GABA. Data are shown as mean 6
SEM. * P,0.05, #P= 0.05, compared with the control group.
(TIF)
Author Contributions
Conceived and designed the experiments: HLS SHZ ZC. Performed the
experiments: HLS KZ ZHX BF JY QF SW DCW. Analyzed the data:
HLS KZ SHZ. Contributed reagents/materials/analysis tools: JMZ.
Wrote the paper: HLS SHZ ZC.
Figure 9. Interruption of GS upregulation by microRNA interference and kindled seizures. rtificial microRNA interference vectors (0.5,
1.5, and 2.5 610
6
TU/2 ml) was injected into the ipsilateral DG on the third day following the first stage 3 seizure. (A) behavioral stage progression of
seizures, (B) AD duration, (C) generalized seizure duration, (D) numbers of stimulations required to reach full kindled from administration, (E) numbers
of stimulations required to retain in stage 0–3 and stage 4 in kindling acquisition, (F) difference of ADT between that just before injection and on the
third day after injection (n = 6–10/group). (G) shows electrographic examples of afterdischarge recorded from the ipsilateral amygdala in six groups
on the third day after injection. ES indicates electrical kindling stimulation. Data are shown as mean 6SEM. *P,0.05, **P,0.01, *** P,0.001
compared with the control group;
#
P,0.05,
###
P,0.001 compared with each other.
doi:10.1371/journal.pone.0066885.g009
Glutamine Synthetase in Epileptogenesis
PLOS ONE | www.plosone.org 11 June 2013 | Volume 8 | Issue 6 | e66885
A
References
1. Leppik IE (1992) Intractable epilepsy in adults. Epilepsy Res Suppl 5: 7–11.
2. During MJ, Spencer DD (1993) Extracellular hippocampal glutamate and
spontaneous seizure in the conscious human brain. Lancet 341: 1607–1610.
3. Williamson A (1994) Electrophysiology of epileptic human neocortic al and
hippocampal neurons maintained in vitro. Clin Neurosci 2: 47–52.
4. Olney JW, Collins RC, Sloviter RS (1986). Excitotoxic mechanisms of epileptic
brain damage. In: Delgado-Escueta AV, Ward AA Jr, Woodbury DM, Porter
RJ, editors. Advances in Neurology: Raven Press. pp. 857–877.
5. Olney JW (1978) Neurotoxicity of excitatory amino acids. In: McGeer EG,
Olney JW, McGeer PL, editors. Kainic Acid as A Tool in Neurobiology: Raven
Press. pp. 37–70.
6. Chaudhry FA, Reimer RJ, Edwards RH (2002) The glutamine commute: take
the N line and transfer to the A. J Cell Biol 157: 349–355.
7. Hamberger AC, Chiang GH, Nyle´n ES, Scheff SW, Cotman CW (1979)
Glutamate as a CNS transmitter: I. Evaluation of glucose and glutamine as
precursors for the synthesis of preferentially released glutamate. Brain Res 168:
513–530.
8. Broman J, Hassel B, Rinvik E, Ottersen OP (2000) Biochemistry and anatomy of
transmitter glutamate. In: Ottersen OP, Storm-Mathisen J, editors. Glutamate:
Elsevier Science. pp. 1–44.
9. van den Berg CJ, Garfinkel D (1971) A stimulation study of brain compartments:
metabolism of glutamate and related substances in mouse brain. J Biochem 123:
211–218.
10. de Lanerolle NC, Lee TS, Spencer DD (2010) Astrocytes and epilepsy.
Neurotherapeutics 7: 424–438.
11. Perez EL, Lauritzen F, Wang Y, Lee TS, Kang D, et al. (2012) Evidence for
astrocytes as a potential source of the glutamate excess in temporal lobe epilepsy.
Neurobiol Dis. 47: 331–337.
12. van Gassen KL, van der Hel WS, Hakvoort TB, Lamers WH, de Graan PN
(2009) Haploinsufficiency of glutamine synthetase increases susceptibility to
experimental febrile seizures. Genes Brain Behav 8: 290–295.
13. Eid T, Thomas MJ, Spencer DD, Runde´n-Pran E, Lai JC, et al. (2004) Loss of
glutamine synthe tase in the human e pileptogenic hippocampus: poss ible
mechanism for raised extracellular glutamate in mesial temporal lobe epilepsy.
Lancet 363: 28–37.
14. Kang TC, Kim DS, Kwak SE, Kim JE, Won MH, et al. (2006) Epileptogenic
roles of astroglial death and regeneration in the dentate gyrus of experimental
temporal lobe epilepsy. Glia 54: 258–271.
15. Bidmon HJ, Go¨rg B, Palomero-Gallagher N, Schleicher A, Ha¨ ussinger D, et al.
(2008) Glutamine synthetase becomes nitrated and its activity is reduced during
repetitive seizure activity in the pentylentetrazole model of epilepsy. Epilepsia
49: 1733–1748.
16. Eid T, Ghosh A, Wang Y, Beckstro¨m H, Zaveri HP, et al. (2008) Recurrent
seizures and brain pathology after inhibition of glutamine synthetase in the
hippocampus in rats. Brain 131: 2061–2070.
17. Hammer J, Alvestad S, Osen KK, Skare Ø, Sonnewald U, et al. (2008)
Expression of glutamine synthetase and glutamate dehydrogenase in the latent
phase and chronic phase in the kainate model of temporal lobe epilepsy. Glia 56:
856–868.
18. Yang LX, Jin CL, Zhuge ZB, Wang S, Wei EQ, et al. (2006) Unilateral low-
frequency stimulation of central piriform cortex delays seizure development
induced by amygdaloid kindling. Neuroscience 138: 1089–1096.
19. Wang S, Wu DC , Ding MP, Li Q, Zhuge ZB, et al. (2008) Low frequency
stimulation of cerebellar fastigial nucleus inhibits amygdaloid kindling
acquisition in Sprague-Dawley rats. Neurobiol Dis 29: 52–58.
20. Xu ZH, Wu DC, Fang Q, Zhong K, Wang S, et al. (2010) Therapeutic time
window of low-frequency stimulation at entorhinal cortex for amygdaloid-
kindling seizures in rats. Epilepsia 51: 1861–1864.
21. Zhong K, Wu DC, Jin MM, Xu ZH, Wang Y, et al. (2012) Wide therapeutic
time-window of low-frequency stimulation at the subiculum for temporal lobe
epilepsy treatment in rats. Neurobiol Dis 48: 20–26.
22. Wu DC, Xu ZH, Wang S, Fang Q, Hu DQ, et al. (2008) Time-dependent effect
of low-frequency stimulation on amygdaloid- kindling seizures in rats. Neurobiol
Dis 31: 74–79.
23. Wu DC, Zhu-Ge ZB, Yu CY, Fang Q, Wang S, et al. (2008) Low-frequency
stimulation of the tuberomammillary nucleus facilitates electrical amygdaloid-
kindling acquisition in Sprague-Dawley rats. Neurobiol Dis 32: 151–156.
24. Zhang SH, Sun HL, Fang Q, Zhong K, Wu DC, et al. (2009) Low-frequency
stimulation of the hippocampal CA3 subfield is anti-epileptogenic and anti-
ictogenic in rat amygdaloid kindling model of epilepsy. Neurosci Lett 455: 51–
55.
25. Akbarpour B, Sayyah M, Babapour V, Mahdian R, Beheshti S, et al. (201 2)
Expression of connexin 30 and connexin 32 in hippocampus of rat during
epileptogenesis in a kindling model of epilepsy. Neurosci Bull 28: 729–736.
26. Zhang LS, Chen Z, Huang YW, Hu WW, Wei EQ, et al. (2003) Effects of
endogenous histamine on seizure development of pentylenetetrazole-induced
kindling in rats. Pharmacology 69: 27–32.
27. Racine RJ (1972) Modification of seizure activity by electrical stimulation: II
Motor seizure. Electroencephalogr Clin Neurophysiol 32: 281–294.
28. Amaral DG, Witter MP (1995) Hippocampal formation. In: Paxinos G, editor.
The Rat Nervous System: Academic Press. pp. 443–493.
29. Hu WW, Fang Q, Xu ZH, Yan HJ, He P, et al. (2012) Chronic H1-
antihistamine treatment increases seizure susceptibility after withdrawal by
impairing glutamine synthetase. CNS Neurosci Ther 18: 683–990.
30. Itoh K, Watanabe M (2009) Paradoxical facilitation of pentylenetetrazole-
induced convulsion susceptibility in mice lacking neuronal nitric oxide synthase.
Neuroscience 159: 735–743.
31. Go¨rg B, Qvartskhava N, Voss P, Grune T, Ha¨ussinger D, et al. (2007)
Reversible inhibition of mammalian glutamine synthetase by tyrosine nitration.
FEBS Lett 581: 84–90.
32. Ueda Y, Tsuru N (1995) Simultaneous monitoring of the seizure-related changes
in extracellular glutamate and gamma-aminobutyric acid concentration in
bilateral hippocampi following development of amygdaloid kindling. Epilepsy
Res 20:213–219.
33. Bryant AS, Li B, Beenhakker MP, Huguenard JR (2009) Maintenance of
thalamic epileptiform activity depends on the astrocytic glutamate-glutamine
cycle. J Neurophysiol 102: 2880–2888.
34. Tani H, Bandrowski AE, Parada I, Wynn M, Huguenard JR, et al. (2007)
Modulation of epileptiform activity by glutamine and system A transport in a
model of post-traumatic epilepsy. Neurobiol Dis 25: 230–238.
35. Bacci A, Sancini G, Verderio C, Armano S, Pravettoni E, et al. (2002) Block of
glutamate-glutamine cycle between astrocytes and neurons inhibits epileptiform
activity in hippocampus. J Neurophysiol 88: 2302–2310.
36. Lothman EW, Bennett JP, Perlin JB (1987) Alterations in neurotransmitter
amino acids in hippocampal kindled seizures. Epilepsy Res 1: 313–320.
37. Pitka¨ nen A, Pikkarainen M, Nurminen N, Ylinen A (2000) Reciprocal
connections between the amygdala and the hippocampal formation, perirhinal
cortex, and postrhinal cortex in rat. A review. Ann N Y Acad Sci 911: 369–391.
38. Hsu D (2007) The dentate gyrus as a filter or gate: a look back and a look ahead.
Prog Brain Res 163: 601–613.
39. Heinemann U, Beck H, Dreier JP, Ficker E, Stabel J, et al. (1992) The dentate
gyrus as a regulated gate for the propagation of epileptiform activity. Epilepsy
Res Suppl 7: 273–280.
40. Lothman EW, Stringer JL, Bertram EH (1992) The dentate gyrus as a control
point for seizures in the hippocampus and beyond. Epilepsy Res Suppl 7: 301–
313.
41. Stringer JL, Lothman EW (1992) Bilateral maximal dentate activation is critical
for the appearance of an afterdischarge in the dentate gyrus. Neuroscience 46:
309–314.
Glutamine Synthetase in Epileptogenesis
PLOS ONE | www.plosone.org 12 June 2013 | Volume 8 | Issue 6 | e66885

Supplementary resource (1)

... As described previously [34,35], 24 h and 3 days after KA administration, we used sodium pentobarbital (50 mg/kg, i.p.) to deeply anesthetize five rats per timepoint from each group. We opened the chest and exposed the heart to rapidly inject 250 mL of saline and then 250 mL of 4% paraformaldehyde fixation solution into the left ventricle. ...
... At the same brightness level, fluorescence intensities were compared between regions. As previously described [34,35], Image J V.1.37 software (National Institutes of Health, Bethesda, MD) was used to analyze the fluorescence intensities. ...
Article
Full-text available
Though succinate accumulation is associated with reactive oxygen species (ROS) production and neuronal injury, which play critical roles in epilepsy, it is unclear whether succinate accumulation contributes to the onset of epilepsy or seizures. We sought to investigate changes in succinate, oxidative stress, and mito-SOX levels, as well as mitophagy and neuronal change, in different status epilepticus (SE) rat models. Our results demonstrate that KA-induced SE was accompanied by increased levels of succinate, oxidative stress, and mito-SOX, as well as mitophagy and neuronal degeneration. The similarly increased levels of succinate, oxidative stress, and mito-SOX were also found in pilocarpine-induced SE. Moreover, the reduction of succinate accumulation by the inhibition of succinate dehydrogenase (SDH), malate/aspartate shuttle (MAS), or purine nucleotide cycle (PNC) served to reduce succinate, oxidative stress, and mito-SOX levels, thereby preventing oxidative stress-related neuronal damage and lessening seizure severity. Interestingly, simulating succinate accumulation with succinic acid dimethyl ester may induce succinate accumulation and increased oxidative stress and mito-SOX levels, as well as behavior and seizures in electroencephalograms similar to those observed in rats exposed to KA. Our results indicate that succinate accumulation may contribute to the increased oxidative stress/mitochondrial ROS levels, neuronal degeneration, and SE induced by KA administration. Furthermore, we found that succinate accumulation was mainly due to the inverse catalysis of SDH from fumarate, which was supplemented by the MAS and PNC pathways. These results reveal new insights into the mechanisms underlying SE and that reducing succinate accumulation may be a clinically useful therapeutic target in SE.
... This may reflect a subacute response to increased seizure activity that is subsequently downregulated with chronic epilepsy. The expression of glutamine synthetase has been reported to be transiently upregulated during early stages of epileptogenesis in in vivo rodent models of epilepsy [64,65]. Increased glutamine synthetase expression may represent a compensatory response of reactive astrocytes to seizure-associated activity to metabolise excess glutamate that accumulates during seizures. ...
Article
Full-text available
Refractory epilepsy is the main neurological manifestation of Alpers’ syndrome, a severe childhood-onset mitochondrial disease caused by bi-allelic pathogenic variants in the mitochondrial DNA (mtDNA) polymerase gamma gene (POLG). The pathophysiological mechanisms underpinning neuronal hyperexcitabilty leading to seizures in Alpers’ syndrome remain unknown. However, pathological changes to reactive astrocytes are hypothesised to exacerbate neural dysfunction and seizure-associated cortical activity in POLG-related disease. Therefore, we sought to phenotypically characterise astrocytic pathology in Alpers’ syndrome. We performed a detailed quantitative investigation of reactive astrocytes in post-mortem neocortical tissues from thirteen patients with Alpers’ syndrome, eight neurologically normal controls and five sudden unexpected death in epilepsy (SUDEP) patients, to control for generalised epilepsy-associated astrocytic pathology. Immunohistochemistry to identify glial fibrillary acidic protein (GFAP)-reactive astrocytes revealed striking reactive astrogliosis localised to the primary visual cortex of Alpers’ syndrome tissues, characterised by abnormal-appearing hypertrophic astrocytes. Phenotypic characterisation of individual GFAP-reactive astrocytes demonstrated decreased abundance of mitochondrial oxidative phosphorylation (OXPHOS) proteins and altered expression of key astrocytic proteins including Kir4.1 (subunit of the inwardly rectifying K⁺ ion channel), AQP4 (astrocytic water channel) and glutamine synthetase (enzyme that metabolises glutamate). These phenotypic astrocytic changes were typically different from the pathology observed in SUDEP tissues, suggesting alternative mechanisms of astrocytic dysfunction between these epilepsies. Crucially, our findings provide further evidence of occipital lobe involvement in Alpers’ syndrome and support the involvement of reactive astrocytes in the pathogenesis of POLG-related disease.
... [92][93][94][95] Immunohistochemical staining for calcium-binding protein S100β and glutamine synthetase (GS) labels most (if not all) protoplasmic astrocytes in the neurogenic niche; antibodies against GS also label a subpopulation of RSA. 76,[96][97][98] Glutamate transporter EAAT-2 is widely used as astrocytic marker, yet its immunoreactivity is also detected in the neurogenic region where RSA reside. [72][73][74] Although aquaporin 4 (AQP4, the water channel protein) is detected in some nestin-positive RSA in the SVZ, it is best known as a marker for mature astrocytes and highly expressed in astrocytic endfeet. ...
Article
Full-text available
Adult neurogenesis is a striking example of neuroplasticity, which enables adaptive network remodelling in response to all forms of environmental stimulation in physiological and pathological contexts. Dysregulation or cessation of adult neurogenesis contributes to neuropathology negatively affecting brain functions and hampering regeneration of the nervous tissue, while targeting adult neurogenesis may provide the basis for potential therapeutic interventions. Neural stem cells in the adult mammalian brain are at the core and at the entry point of adult neurogenesis. By their origin and properties these cells belong to astroglia, and are represented by stem radial astrocytes (RSA) which exhibit multipotent "stemness". In the neurogenic niches RSA interact with other cellular components, including protoplasmic astrocytes, which in turn regulate their neurogenic activity. In pathology, RSA become reactive, which affects their neurogenic capabilities, whereas reactive parenchymal astrocytes up-regulate stem cell hallmarks and are able to generate progeny that remain within astrocyte lineage. What makes RSA special is their multipotency, represented by self-renewing capacity capability to generate other cellular types as progeny. A broad understanding of the cellular features of RSA and parenchymal astrocytes provides an insight into the machinery that promotes/suppresses adult neurogenesis, clarifying principles of network remodelling. In this review we discuss the cellular hallmarks, research tools and models of RSA and astrocytes of the subventricular zone along the lateral ventricle and dentate gyrus of the hippocampus. We also discuss RSA in ageing, which has great impact on the proliferative capacity of RSA, as well as the potential of RSA and astrocytes in therapeutic strategies aimed at cell replacement and regeneration.
... Low MSO depressed glutamatergic transmission and epileptiform discharges in cerebral cortical slices in vitro [20,21]. In vivo, intracerebral administration of MSO reduced amygdala kindling-induced seizures [22]. ...
Article
Full-text available
Initial seizures observed in young rats during the 60 min after administration of pilocarpine (Pilo) were delayed and attenuated by pretreatment with a non-convulsive dose of methionine sulfoximine (MSO). We hypothesized that the effect of MSO results from a) glutamine synthetase block-mediated inhibition of conversion of Glu/Gln precursors to neurotransmitter Glu, and/or from b) altered synaptic Glu release. Pilo was administered 60 min prior to sacrifice, MSO at 75 mg/kg, i.p., 2.5 h earlier. [1,2-13C]acetate and [U-13C]glucose were i.p.-injected either together with Pilo (short period) or 15 min before sacrifice (long period). Their conversion to Glu and Gln in the hippocampus and entorhinal cortex was followed using [13C] gas chromatography-mass spectrometry. Release of in vitro loaded Glu surrogate, [3H]d-Asp from ex vivo brain slices was monitored in continuously collected superfusates. [3H]d-Asp uptake was tested in freshly isolated brain slices. At no time point nor brain region did MSO modify incorporation of [13C] to Glu or Gln in Pilo-treated rats. MSO pretreatment decreased by ~37% high potassium-induced [3H]d-Asp release, but did not affect [3H]d-Asp uptake. The results indicate that MSO at a non-convulsive dose delays the initial Pilo-induced seizures by interfering with synaptic Glu-release but not with neurotransmitter Glu recycling.
... Low MSO depressed glutamatergic transmission and epileptiform discharges in cerebral cortical slices in vitro [21,22]. In vivo, intracerebral administration of MSO reduced amygdala kindling-induced seizures [23]. ...
Preprint
Pretreatment with non-convulsive dose of methionine sulfoximine (MSO) attenuated lithi-um-pilocarpine-induced (Li-Pilo) seizures in young rats [1]. We hypothesized that the effect of MSO results from a) glutamine synthetase block-mediated inhibition of conversion of Glu/Gln precursors to neurotransmitter Glu, and/or from b) altered synaptic Glu release. Pilo was admin-istered 60 min prior to sacrifice, MSO at 75 mg/kg, i.p., 2.5 h earlier. [1,2-13C]acetate and [U-13C]glucose were i.p.-injected either together with Pilo (onset) or 15 min before sacrifice (final phase). Their conversion to Glu and Gln in hippocampus and entorhinal cortex was followed us-ing [13C] gas chromatography-mass spectrometry. Release of in vitro loaded [3H]D-Asp from ex vi-vo brain slices was measured in continuously collected superfusates. Protein and mRNA expres-sion were measured by Western Blot and real-time PCR techniques, respectively. At no time point nor brain region did MSO modify incorporation of [13C] to Glu or Gln in Pilo-treated rats. MSO pretreatment decreased by ~37% high potassium-induced [3H]D-Asp release and reduced by ~50% the synaptic vesicular Glu transporter VGLUT1 protein, but not mRNA content in the hippo-campus. The results indicate that MSO at non-convulsive dose mitigates the initial Pilo-induced seizures by interfering with synaptic Glu-release but not with neurotransmitter Glu recycling.
... For example, there is evidence showing a transient increase in GS expression following kainic acid 77 and kindling-induced seizures. 78 On the other hand, pilocarpine-induced SE in rats results in a gradual decrease in GS levels. 79 Although these results are not consistent, they do indicate that changes in the level of GS enzyme in the brain can be associated with epilepsy. ...
Article
Full-text available
Alzheimer's disease (AD) can increase the risk of epilepsy by up to 10‐fold compared to healthy age‐matched controls. However, the pathological mechanisms that underlie this increased risk are poorly understood. Because disruption in brain glutamate homeostasis has been implicated in both AD and epilepsy, this might play a mechanistic role in the pathogenesis of epilepsy in AD. Prior to the formation of amyloid beta (Aβ) plaques, the brain can undergo pathological changes as a result of increased production of amyloid precursor protein (APP) and Aβ oligomers. Impairments in the glutamate uptake ability of astrocytes due to astrogliosis are hypothesized to be an early event occurring before Aβ plaque formation. Astrogliosis may increase the susceptibility to epileptogenesis of the brain via accumulation of extracellular glutamate and resulting excitotoxicity. Here we hypothesize that Aβ oligomers and proinflammatory cytokines can cause astrogliosis and accumulation of extracellular glutamate, which then contribute to the pathogenesis of epilepsy in AD. In this review article, we consider the evidence supporting a potential role of dysfunction of the glutamate‐glutamine cycle and the astrocyte in the pathogenesis of epilepsy in AD.
... Moreover, xenon can significantly inhibit the uptake and efflux of glutamate (Lavaur et al., 2016), and swiftly terminate the synchronous discharge (Uchida et al., 2012). Over excitation induced by elevated concentration of glutamate is closely associated with epileptic development (Sun et al., 2013) and seizures. Neuronal injury and synchronous discharge are the pathological characteristics of epilepsy (During and Spencer, 1993;Chiu et al., 2016;Kim and Kang, 2018). ...
Article
Full-text available
Previous studies have reported the possible neuroprotective effects of xenon treatment. The purpose of this study was to define the range of effective xenon ratio, most effective xenon ratio, and time-window for intervention in the kainic acid (KA) – induced status epilepticus (SE) rat model. Different ratios of xenon (35% xenon, 21% oxygen, 44% nitrogen, 50% xenon, 21% oxygen, 29% nitrogen, 70% xenon, 21% oxygen, and 9% nitrogen) were used to treat the KA-induced SE. Our results confirmed the anti-seizure role of 50 and 70% xenon mixture, with a stronger effect from the latter. Further, 70% xenon mixture was dispensed at three time points (0 min, 15 min delayed, and 30 min delayed) after KA administration, and the results indicated the anti-seizure effect at all treated time points. The results also established that the neuronal injury in the hippocampus and entorhinal cortex (EC), assessed using Fluoro-Jade B (FJB) staining, were reversed by the xenon inhalation, and within 30 min after KA administration. Our study, therefore, indicates the appropriate effective xenon ratio and time-window for intervention that can depress seizures. The prevention of neuronal injury and further reversal of the loss of effective control of depress network in the hippocampus and EC may be the mechanisms underlying the anti-seizure effect of xenon.
... The excised brains were then microdissected into the hippocampus, piriform cortex (PC), remaining cortex (except the PC), and amygdala and were individually sonicated on ice. Their protein content was quantified as previously described (Sun et al., 2013). Homogenates were mixed with sample loading buffer, separated on 12% SDS-polyacrylamide gels, and electrically transferred onto PVDF membranes. ...
Article
Full-text available
Previous studies suggested that the thrombospondin-1/transforming growth factor-β1 (TSP-1/TGF-β1) pathway might be critical in synaptogenesis during development and that the purinergic P2 receptor family could regulate synaptogenesis by modulating TSP-1 signaling. However, it is unclear whether this pathway plays a role in synaptogenesis during epileptic progression. This study was designed to investigate this question by analyzing the dynamic changes and effects of TSP-1 levels on the density of synaptic markers that are related to epileptic seizure activity. In addition, we evaluated whether P2-type receptors could regulate these effects. We generated a rat seizure model via amygdala kindling and inhibited TSP-1 activity using small interfering RNA (siRNA) interference and pharmacological inhibition. We treated the rats with antagonists of P2 or P2Y receptors, pyridoxalphosphate-6-azophenyl-2’,4’-disulfonic (PPADS) or Reactive Blue 2. Following this, we quantified TSP-1 and TGF-β1 immunoreactivity (IR), the density of synaptic markers, and seizure activity. There were significantly more synapses/excitatory synapses in several brain regions, such as the hippocampus, which were associated with progressing epileptic discharges after kindling. These were associated with increased TSP-1 and TGF-β1-IR. Genetic or pharmacologic inhibition of TSP-1 significantly reduced the density of synaptic/excitatory synaptic markers and inhibited the generalization of focal epilepsy. The administration of PPADS or Reactive Blue 2 attenuated the increase in TSP-1-IR and the increase in the density of synaptic markers that follows kindling and abolished most of the epileptic seizure activity. Altogether, our results indicate that the TSP-1/TGF-β1 pathway and its regulation by P2, particularly P2Y-type receptors, may be a critical promoter of synaptogenesis during the progression of epilepsy. Therefore, components of this pathway may be targets for novel antiepileptic drug development.
Article
The contribution of glutamatergic transmission to generation of initial convulsive seizures (CS) is debated. We tested whether pretreatment with a glutamine synthetase (GS) inhibitor, methionine sulfoximine (MSO), affects the onset and progression of initial CS by cholinergic stimulus in juvenile rats. Male rats (24 days old, Sprague Dawley) sequentially received i.p. injections of lithium-carbonate, MSO, methyl-scopolamine, and pilocarpine (Pilo). Pilo was given 150 min after MSO. Animals were continuously monitored using the Racine scale, EEG/EMG and intrahippocampal glutamate (Glu) biosensors. GS activity as measured in hippocampal homogenates, was not altered by MSO at 150 min, showed initial, varied inhibition at 165 (15 min post-Pilo), and dropped down to 11 % of control at 60 min post-Pilo, whereas GS protein expression remained unaltered throughout. Pilo did neither modulate the effect of MSO on GS activity nor affect GS activity itself, at any time point. MSO reduced from 32% to 4% the number of animals showing CS during the first 12 min post-Pilo, delayed by ∼6 min the appearance of electrographic seizures, and tended to decrease EMG power during ∼15 min post-Pilo. The results indicate that MSO impairs an aspect of glutamatergic transmission involved in the transition from the first cholinergic stimulus to the onset of seizures. A continuous rise of extracellular Glu lasting 60 min was insignificantly affected by MSO, leaving the nature of the Glu pool(s) involved in altered glutamatergic transmission undefined.
Article
Full-text available
Introduction Intracerebral hemorrhage (ICH) is a lethal cerebrovascular disorder with a high mortality and morbidity. The pathophysiological mechanisms underlying ICH‐induced secondary injury remain unclear. Methods To examine one of the gaps in the knowledge about ICH pathological mechanisms, isobaric tag for relative and absolute quantification (iTRAQ)‐based liquid chromatography‐tandem mass spectrometry (LC‐MS/MS) was used in collagenase‐induced ICH rats on the 2nd day. Results A total of 6,456 proteins were identified with a 1% false discovery rate (FDR). Of these proteins, 126 and 75 differentially expressed proteins (DEPs) were substantially increased and decreased, respectively. Based on Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and STRING analyses, the protein changes in cerebral hemorrhage were comprehensively evaluated, and the energy metabolism in ICH was anchored. The core position of the nitrogen metabolism pathway in brain metabolism in ICH was found for the first time. Carbonic anhydrase 1 (Ca1), carbonic anhydrase 2 (Ca2), and glutamine synthetase (Glul) participated in this pathway. We constructed the protein–protein interaction (PPI) networks for the energy metabolism of ICH, including the Atp6v1a‐Atp6v0c‐Atp6v0d1‐Ppa2‐Atp6ap2 network. Conclusions It seems that dysregulation of energy metabolism, especially nitrogen metabolism, may be a major cause in secondary ICH injury. This information provides novel insights into secondary events following ICH.
Article
Full-text available
Objective: Understanding the molecular and cellular mechanisms underlying epileptogenesis yields new insights into potential therapies that may ultimately prevent epilepsy. Gap junctions (GJs) create direct intercellular conduits between adjacent cells and are formed by hexameric protein subunits called connexins (Cxs). Changes in the expression of Cxs affect GJ communication and thereby could modulate the dissemination of electrical discharges. The hippocampus is one of the main regions involved in epileptogenesis and has a wide network of GJs between different cell types where Cx30 is expressed in astrocytes and Cx32 exists in neurons and oligodendrocytes. In the present study, we evaluated the changes of Cx30 and Cx32 expression in rat hippocampus during kindling epileptogenesis. Methods: Rats were stereotaxically implanted with stimulating and recording electrodes in the basolateral amygdala, which was electrically stimulated once daily at afterdischarge threshold. Expression of Cx30 and Cx32, at both the mRNA and protein levels, was measured in the hippocampus at the beginning, in the middle (after acquisition of focal seizures), and at the end (after establishment of generalized seizures) of the kindling process, by real-time PCR and Western blot. Results: Cx30 mRNA expression was upregulated at the beginning of kindling and after acquisition of focal seizures. Then it was downregulated when the animals acquired generalized seizures. Overexpression of Cx30 mRNA at the start of kindling was consistent with the respective initial protein increase. Thereafter, no change was found in protein abundance during kindling. Regarding Cx32, mRNA expression decreased after acquisition of generalized seizures and no other significant change was detected in mRNA and protein abundance during kindling. Conclusion: We speculate that Cx32 GJ communication in the hippocampus does not contribute to kindling epileptogenesis. The Cx30 astrocytic network localized to perivascular regions in the hippocampus is, however, overexpressed at the initiation of kindling to clear excitotoxic molecules from the milieu.
Article
This third edition of the standard reference on the nervous system of the rat is a complete and updated revision of the 1994 second edition. All chapters have been extensively updated, and new chapters added covering early segmentation, growth factors, and glia. The book is now aligned with the data available in the Rat Brain in Stereotaxic Coordinates, making it an excellent companion to this bestselling atlas. Physiological data, functional concepts, and correlates to human anatomy and function round out the new edition. Designed to be used in conjunction with the bestselling Rat Brain in Stereotaxic Coordinates. New to this edition is inclusion of physiological data, functional concepts, and correlates to human anatomy and function in each chapter. Contains new chapters on early segmentation of the central nervous system, growth factors and glia.
Article
The reviewed literature leaves us with an impression of the cellular interchange of Glu and its precursors and metabolites (Fig.. 2) that is somewhat different from the classical idea of the 'glutamine" cycle with its near-stoichiometric exchange of Glu and glutamine between neurons and astrocytes. (1) Glu is synthesized from glutamine, as previously thought, but also from neuronal precursors supplied by neuronal pyruvate carboxylation. (2) Transmitter Glu is mostly taken up into astrocytes for conversion to glutamine, but an unknown fraction is metabolized by the astrocytic TCA cycle, either fully to CO2 and water, or only partially, to malate which is converted to pyruvate and hence lactate. (3) Uptake of Glu into astrocytes stimulates astrocytic uptake of serum glucose and export of lactate to the extracellular fluid. (4) Lactate, whether formed from transmitter Glu or serum glucose, may increase regional cerebral blood flow and act as a main neuronal energy substrate. (5) Glutamine is shunted to neurons where it to a large extent is metabolized to CO2 and water and, possibly to a lesser extent, is converted to transmitter Glu. (6) The uptake processes related to the handling of transmitter Glu and glutamine together with the formation of glutamine cause the expenditure of % of the total energy of the serum glucose taken up by the brain.
Article
To investigate the effect of chronic H1-antihistamine treatment on seizure susceptibility after drug withdrawal in nonepileptic rats and to further study its relation to glutamine synthetase (GS), which is the key enzyme for glutamate metabolism and gamma aminobutyric acid (GABA) synthesis. After drug withdrawal from a 2-week treatment with diphenhydramine or pyrilamine, seizure susceptibility was determined by amygdaloid kindling or pentylenetetrazol model; meanwhile, the GS expression or activity was analyzed. The glutamine, glutamate, and GABA contents were measured by high-performance liquid chromatography. Seizure susceptibility significantly increased in amygdaloid kindling and pentylenetetrazol model 10 days after drug withdrawal from a 2-week treatment with H1-antihistamines. Meanwhile, GS activity and expression in the cortex or hippocampus decreased simultaneously with a marked decline of glutamine and GABA content. Comparable inhibition of GS activity by methionine sulfoximine was also sufficient to increase the susceptibility, while supplementation with glutamine reversed the high susceptibility 10 days after diphenhydramine withdrawal. Moreover, the seizure susceptibility increased 10 days after diphenhydramine withdrawal in wild-type mice but not in histidine decarboxylase knockout mice, which lack histamine. Chronic H1-antihistamine treatment produces long-lasting increase in seizure susceptibility in nonepileptic rodents after drug withdrawal and its mechanism involves impairment of GS through blocking the action of histamine.
Article
Increased extracellular brain glutamate has been implicated in the pathophysiology of human refractory temporal lobe epilepsy (TLE), but the cause of the excessive glutamate is unknown. Prior studies by us and others have shown that the glutamate degrading enzyme glutamine synthetase (GS) is deficient in astrocytes in the epileptogenic hippocampal formation in a subset of patients with TLE. We have postulated that the loss of GS in TLE leads to increased glutamate in astrocytes with elevated concentrations of extracellular glutamate and recurrent seizures as the ultimate end-points. Here we test the hypothesis that the deficiency in GS leads to increased glutamate in astrocytes. Rats were chronically infused with methionine sulfoximine (MSO, n=4) into the hippocampal formation to induce GS deficiency and recurrent seizures. A separate group of rats was infused with 0.9% NaCl (saline) as a control (n=6). At least 10days after the start of infusion, once recurrent seizures were established in the MSO-treated rats, the concentration of glutamate was assessed in CA1 of the hippocampal formation by immunogold electron microscopy. The concentration of glutamate was 47% higher in astrocytes in the MSO-treated vs. saline-treated rats (p=0.02), and the ratio of glutamate in astrocytes relative to axon terminals was increased by 74% in the MSO-treated rats (p=0.003). These data support our hypothesis that a deficiency in GS leads to increased glutamate in astrocytes. We additionally propose that the GS-deficient astrocytes in the hippocampal formation in TLE lead to elevated extracellular brain glutamate either through decreased clearance of extracellular glutamate or excessive release of glutamate into the extracellular space from these cells, or a combination of the two.
Article
Daily electrical stimulations of the amygdala and hippocampus at intensities sufficient to evoke after-discharges (ADs) resulted in the development of motor seizures, which could not initially be evoked by these stimulations. The triggering of ADs was critical for this development, as well as for the development of permanent changes in the characteristics of the AD. The wave form of the AD "spikes" became more complex. The frequency of these spikes and the duration of AD increased. The amplitude of the AD spikes increased in the structure stimulated as well as in secondary structures to which the AD was "projected". This increase in amplitude of "projected" spikes often correlated with the appearance of motor seizures. Other electrographic developments are discussed including the appearance of spontaneous "inter-ictal" spiking in the amygdala. It was found that the development of motor seizures by stimulation of the amygdala resulted in an increased ability of the contralateral amygdala, and the septal area, but not of the hippocampus, to drive motor seizures when stimulated ("transfer"). Motor seizure development in the hippocampus transferred to the contralateral hippocampus. These developments were shown, by means of control subjects, with lesions in the primary focus to involve changes outside the primary focus. The implications of these developments with respect to seizure development are discussed.