SlideShare a Scribd company logo
Yale University
EliScholar – A Digital Platform for Scholarly Publishing at Yale
Yale Medicine Thesis Digital Library School of Medicine
January 2019
Localized Hippocampal Glutamine Synthetase
Knockout: A Novel Model Of Mesial Temporal
Lobe Epilepsy
Maxwell Gerard Farina
Follow this and additional works at: https://elischolar.library.yale.edu/ymtdl
This Open Access Thesis is brought to you for free and open access by the School of Medicine at EliScholar – A Digital Platform for Scholarly
Publishing at Yale. It has been accepted for inclusion in Yale Medicine Thesis Digital Library by an authorized administrator of EliScholar – A Digital
Platform for Scholarly Publishing at Yale. For more information, please contact elischolar@yale.edu.
Recommended Citation
Farina, Maxwell Gerard, "Localized Hippocampal Glutamine Synthetase Knockout: A Novel Model Of Mesial Temporal Lobe
Epilepsy" (2019). Yale Medicine Thesis Digital Library. 3491.
https://elischolar.library.yale.edu/ymtdl/3491
Localized Hippocampal Glutamine
Synthetase Knockout: a Novel Model of
Mesial Temporal Lobe Epilepsy
a thesis submitted to the
Yale University School of Medicine
in partial fulfillment for the
degree of Doctor of Medicine
Maxwell G. Farina
Advisor: Tore Eid, MD, PhD
Thesis Committee Members: Peter Tattersall, PhD,
Nihal DeLanerolle, DPhil, DSc & Ellen Foxman, MD, PhD
May 2019
Abstract
LOCALIZED HIPPOCAMPAL GLUTAMINE SYNTHETASE KNOCKOUT: A
NOVEL MODEL OF MESIAL TEMPORAL LOBE EPILEPSY.
Maxwell Farina and Tore Eid. Departments of Laboratory Medicine and Neurosurgery,
Yale University, School of Medicine, New Haven, CT.
The purpose of this study was to create and optimize a model of mesial temporal lobe epilepsy through
selective depletion of glutamine synthetase (GS) in the mouse hippocampus. Following validation of the
model, preliminary studies attempted to characterize morphological astrocytic and synaptic changes that re-
sult from GS deficiency. Aim 1 established a novel mouse model of GS knockout in hippocampal astrocytes.
Aim 2 tested whether localized hippocampal knockout of GS causes mice to exhibit an epilepsy-like phe-
notype. Aim 3 characterized the cellular effects of localized GS loss. To generate the knockout, Glul-floxed
C57BL/6J mice were injected with four different adeno-associated viral vectors containing Cre-recombinase
expression cassettes. Mice were also implanted with intracranial depth or screw electrodes and monitored
for spontaneous seizures using 24-hour video-EEG recording for two weeks. To assess for provoked seizure
sensitivity, seizures were induced with pentylenetetrazol (PTZ) prior to perfusion fixation. Brains were per-
fused, sectioned, and immunostained for analysis using standard and STED fluorescence microscopy. Knock-
out of GS, as evidenced by loss of GS immunoreactivity, was found over a greater area in brain regions in-
jected with the AAV5 CMV and AAV8 GFAP serotypes. In addition, within each GS knockout region,
AAV8 GFAP exhibited a significantly greater efficiency of knockdown compared to AAV5 CMV Legacy
and AAV8 CMV (83.1% decreased fluorescence intensity, p=0.0003) and compared to AAV5 CMV (20.2%
decreased fluorescence intensity, p=0.018). AAV8 GFAP exhibited near perfect target specificity (98.7% of
GFP+ cells were astrocytes), while AAV5 CMV Legacy, AAV5 CMV, and AAV8 CMV targeted mostly neu-
rons with varied degrees astrocyte labeling detected (10.0%, 21.3%, and 12.7% astrocytes, respectively. Sixty
percent (3/5) of mice injected with AAV8 GFAP exhibited an epilepsy-like phenotype including sponta-
neous recurrent seizures that were clustered in the morning hours. Twenty-five percent (1/4) of control mice
seized spontaneously over the same period. Additionally, focal GS knockout mice demonstrated significantly
lower time to initial clonic twitch following PTZ administration compared to control mice (mean ± SEM:
41.2 ± 3.2 seconds vs. 65.83 ± 12.9 seconds, respectively; p=0.044). The effect on time to convulsive seizure
was not statistically significant, though there was a trend of knockout animals proceeding to convulsions
in less time (74.2 ± 9.4 seconds vs. 100.0 ± 18.0 seconds, p=0.20). Finally, examination of synaptic mark-
ers revealed decreased expression of PSD-95 surrounding GS- astrocytes compared to GS+ astrocytes, with
sampled relative intensity of 0.57 ± 0.04 (p=0.002). Relative intensity (RI) of synaptophysin and gephyrin
appeared to be unchanged in the sampled areas (synaptophysin RI 0.94 ± 0.15, p=0.87; gephyrin RI 0.94 ±
0.04, p=0.23). In this study, we created a novel model of mesial temporal lobe epilepsy by selectively knock-
ing out GS in the hippocampal astrocytes of mice. Development of this monogenetic knockout model with
effects restricted to the hippocampus and adjacent structures has the potential to more fully elucidate the
impact of GS loss in this treatment-resistant disease. Initial examination of synaptic markers in GS depleted
areas highlights the importance of glutamatergic synaptic transmission in epilepsy pathology.
ii
Acknowledgments
This work was made possible by the knowledge, skills, and support conferred by
countless friends and mentors through the years. In particular, I am thankful to Tore Eid,
who exemplifies selfless mentorship; my father, who taught me the importance of ask-
ing questions; and my mother, who volunteered to help my fourth-grade science class
dissect a shark, and in doing so, instilled in me an unending love for science, its system-
atic search for truth, and the uniquely fundamental way in which it connects us to one
another.
Financial support for the work presented in this thesis was provided in part by the Na-
tional Heart, Lung, and Blood Institute of the National Institutes of Health (Maxwell
Farina, award number T35HL007649) and NIH grant S10-OD020142 (Yale Confocal Mi-
croscopy Core).
iii
Abbreviations
AAV Adeno-associated virus
AMPA α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid
ATP Adenosine triphosphate
CA 1-3 Cornu ammonis subfields 1-3 of the hippocampus
CMV Cytomegalovirus
CNS Central nervous system
DH Dentate hilus of the hippocampus
EAAT Excitatory amino acid transporters
EC Entorhinal cortex
GABA γ-aminobutyric acid
GFAP Glial fibrillary acid protein
GFP Green fluorescent protein
GLUL Glutamine ammonia ligase
Gr Granule cell layer of the dentate gyrus
GS Glutamine synthetase
LV Lateral ventricle
Mol Molecular layer of the dentate gyrus
MSO Methionine sulfoximine
MTLE Mesial temporal lobe epilepsy
NMDA n-methyl-D-aspartate
PTZ Pentylenetetrazol
SUB Subiculum
TCA Tricarboxylic acid
iv
Contents
Introduction 2
Mesial Temporal Lobe Epilepsy . . . . . . . . . . . . . . . . . . . . . . . . . . 2
Glutamine Synthetase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
Chemical Models of Mesial Temporal Lobe Epilepsy . . . . . . . . . . . . . . . 9
Genetic Models of Glutamine Synthetase Depletion . . . . . . . . . . . . . . . 12
Specific Aims 17
Methods 18
Animals and Reagents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
Surgery: Viral Injection and Electrode Implantation . . . . . . . . . . . . . . . . 20
Video-EEG Monitoring and Seizure Precipitation Studies . . . . . . . . . . . . 23
Fixation, Immunofluorescence, and Microscopy . . . . . . . . . . . . . . . . . . 23
Statistical Analysis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24
Results 25
Knockout of Glutamine Synthetase . . . . . . . . . . . . . . . . . . . . . . . . 25
Seizure Findings . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29
Synaptic Changes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32
Discussion 34
References 38
1
Introduction
Epilepsy is a chronic neurological disorder characterized by recurrent, unprovoked
seizures; i.e. sudden and transient episodes of abnormal electrical brain activity that result
in a change in clinical state. The clinical presentation of epilepsy is widely varied, as seizure
activity can take many forms including staring, unresponsiveness, stereotyped movements,
loss of muscle tone, stiffness, and limb-jerking. Likely due in part to its potentially dra-
matic appearance, epilepsy is one of the oldest recognized health conditions, with extensive
descriptions originating on Babylonian cuneiform tablets dating to 4000 BC [1]. Today,
it is estimated that epilepsy affects approximately 2% of the worldwide population [2]. Of
these cases, about one-third are refractory (i.e., inadequately controlled by ≥2 appropriately
selected antiepileptic drugs) [3]; some forms of epilepsy are refractory at much higher rates.
Indeed, there are dozens of “epilepsy syndromes,” each of which is characterized by consis-
tently occurring seizure type, age of onset, electroencephalographic findings, precipitating
factors, genetic markers, clinical course, prognosis, and response to pharmacotherapy.
Mesial Temporal Lobe Epilepsy
Of the epilepsy syndromes, mesial temporal lobe epilepsy (MLTE) is the most common
treatment resistant variant in adults [4]. It is estimated that 70% of MTLE patients are in-
adequately controlled with medication alone [2]. Detailed epidemiological data on MTLE
is scarce, as an incomplete understanding of disease pathophysiology has led to imprecise
INTRODUCTION 3
nomenclature and a lack of consensus on diagnostic criteria [5]. However, a recent meta-
analysis inferred an incidence of 8.9 cases per 100,000 people per year, a prevalence of 1.9
cases per 1000 people, and an estimated patient population of 615,600 individuals in the
United States alone [6]. MTLE most commonly starts before the age of 18 and gradually
increases in intensity and pharmacoresistance over time [7]. Patients often have a past med-
ical history that includes childhood febrile convulsions. While the causes of MTLE remain
to be elucidated, the most widely accepted theory posits that these childhood febrile con-
vulsions or other predisposing injuries act as an early insult that culminates in hippocampal
damage and eventual development of MTLE [5]. Though this theory, initially posited by
Meyer in 1954, serves as a useful framework for understanding MTLE, it is important to
highlight that a large number of MTLE patients have no history of predisposing injury that
precedes onset of the epileptic syndrome.
The clinical presentation of MTLE is varied, as the entity referred to as MTLE is widely
believed to be a heterogeneous collection of different pathophysiologies; however, cer-
tain patterns appear to be more characteristic of MTLE patients. MTLE seizure episodes
often begin with a vegetative aura, often described as “an epigastric or substernal rising
sensation,” [5]. Other common aura symptoms include a sudden sense of fear, delusions,
hallucinations, and olfactory or gustatory sensations [7]. As the complex partial seizure be-
gins, behavioral arrest and staring occur. Next follow automatisms including lip smacking
and chewing, and while motor symptoms are less common in MTLE, dystonic postur-
ing of the contralateral arm does occur and is useful as a lateralizing feature [5]. The com-
plex partial seizure typically continues for 1-2 minutes and can include head deviation, and
clonic-tonic activity, uncommonly culminating in convulsion. While the postictal period
is variable, it is not uncommon for patients to exhibit significant confusion and (in the case
of dominant temporal lobe onset) several minutes of postictal aphasia. Postictal memory
impairment can also occur in MTLE, sometimes rendering the patient amnestic for several
hours despite apparently normal behavior.
INTRODUCTION 4
Epilepsy syndromes are largely classified by EEG patterns. Though neither EEG find-
ings nor clinical presentation are pathognomonic for MTLE, interictal scalp electroen-
cephalogram often demonstrates temporal-lobe spike and sharp waves and focal slowing
[5]. Hippocampal sclerosis – the atrophy and scarring of the hippocampus – often cited as
the hallmark of MTLE, is also difficult to recognize using magnetic resonance imaging, as
these changes are often subtle and bilateral, interfering with the qualitative asymmetry re-
quired to identify the pathology. All of these factors make definitive diagnosis of MTLE
challenging.
Untreated MTLE is often described as a chronic, progressive disorder in which seizures
increase in duration and intensity over time [5]. While MTLE is typically very respon-
sive to pharmacotherapy at the onset of disease, seizures frequently become pharmacoresis-
tant by early adulthood. In addition to often debilitating seizures, patients are commonly
plagued by additional long-term sequelae of MTLE including global cognitive dysfunction,
impaired episodic memory recall, poor working memory performance, executive dysfunc-
tion, impaired task-switching, decreased alertness, and difficulties with language and word
retrieval [8]. MTLE patients also exhibit features of depression, anxiety, and obsessive-
compulsive disorder with greater frequency than the general population [9]. In particular,
depression is a widely known comorbidity of temporal lobe epilepsy, with an incidence
of 30% and prevalence of 50% among MTLE patients [10]. Indeed, suicide is the leading
cause of death in patients with refractory MTLE; one study found that of the deaths of en-
rolled MTLE patients during a 9-year follow up, 50% were suicides [11]. Suicidal ideation
is a major driver of the finding that MTLE patients had a standardized mortality ratio of
4.86.
The pathogenesis of MTLE is poorly understood. Broadly, based on our understand-
ing of neuronal function, it can be inferred that epilepsy is caused by a departure from the
homeostatic balance of excitatory and inhibitory forces in human neuronal networks that
predisposes neurons to inappropriate synchronous excitation [5]. This imbalance can re-
INTRODUCTION 5
sult from a preponderance of excitatory signaling or a paucity of inhibitory signaling, and
in reality, MTLE is likely caused by a complex combination of both of these (and other)
factors. Investigation of excitatory pathways reveals several etiological hypotheses includ-
ing mossy fiber sprouting which forms new recurrent excitatory connections in the den-
tate gyrus [12], increased expression of certain voltage-gated sodium channels [13], and in-
creased expression of the AMPA and NMDA glutamate receptors [14]. Hypotheses regard-
ing compromise of balancing inhibitory signaling include loss of hippocampal interneu-
rons [15], shortened duration of inhibitory GABAA synaptic potentials [16], and lack of
GABAB-mediated use-dependent synaptic depression [17]. Potential extra-neuronal eti-
ologies include impairment of the blood-brain-barrier [18], shifts in hormonal neuromod-
ulators [19], and a host of astrocyte-related changes in neurotransmitter metabolism, ion
redistribution, and direct synaptic interaction [2]. In short, the pathophysiology of MTLE
is staggeringly complex and likely represents a multifactorial system which spans dozens
of cell types, receptors, and molecules. While recent years have brought a vast amount
of progress in the understanding of these various pathways, much work remains. A large
number of MTLE cases are poorly controlled, and there have been no new epilepsy drug
therapies developed in the last decade. Further work on animal models of MTLE and cor-
relation to human disease will be crucial to categorizing the various subtypes of temporal
lobe epilepsy, identifying their most relevant and high-impact target pathways, and design-
ing molecules and interventions that improve the symptomology and long-term survival of
patients who suffer from MTLE.
Glutamine Synthetase
In 1993, During and Spencer reported that extracellular hippocampal glutamate levels were
elevated in MTLE patients not only following complex partial seizures but also preceding
any electroencephalographic or clinical evidence of seizure [20]. Such human evidence,
INTRODUCTION 6
taken with the findings that administration of glutamate analogues in animal models is suf-
ficient to trigger seizures [21], implicates high levels of extracellular glutamate as a pos-
sible causative factor in epilepsy. A subsequent study utilizing magnetic resonance spec-
troscopy found that epileptic hippocampus contains both increased glutamate levels and
decreased glutamine levels [22]. These findings pointed Eid and colleagues towards poten-
tial involvement of glutamine synthetase, an enzyme found in astrocytes that functions to
convert excitatory neurotransmitter glutamate into glutamine. In an effort to understand
the origin of the pathological metabolic disturbances, Eid and colleagues examined tis-
sue levels of glutamine synthetase in the human hippocampus, and in 2004, they reported
that compared to control tissues, glutamine synthetase expression and enzymatic activity
were reduced by 40% and 38%, respectively, in MTLE [23]. The prospect of identifying a
well-characterized enzymatic defect as a contributor of seizure generation has significant
implications for therapeutic target identification, thus glutamine synthetase has become an
important subject of investigation in recent years.
Glutamine synthetase (GS) is an enzyme encoded by the strikingly ubiquitous gluta-
mate ammonia ligase (Glul) gene, which is found in all known living species. The eukary-
otic variant of GS is structured as two adjacent pentameric rings consisting of ten identical
subunits [24], and the enzyme serves as a catalyst of the ATP-dependent synthesis of glu-
tamine from substrates glutamate and ammonia. In mammals, GS expression is limited to
specific tissues: liver, kidney, pancreas, adipose tissue, skin, and the central nervous system
(CNS) [25]. Within the CNS, GS is exclusively expressed in astrocytes [26], emphasizing
the indispensable role that astrocytes play in the maintenance of the homeostatic cellular
and metabolic environment of the CNS.
This astrocytic regulation of extracellular metabolite levels—particularly in proximity to
the synapse—is most relevant to epileptogenesis through the glutamate-glutamine cycle. In
a recent textbook chapter, Eid et. al., succinctly describe the glutamate-glutamine cycle in
the following steps:
INTRODUCTION 7
1. Exocytosis of glutamate from axon terminals into the synaptic cleft.
2. Binding of glutamate to postsynaptic glutamate receptors.
3. Diffusion of glutamate away from the synapse with possible binding to extrasynaptic
receptors and uptake into astrocytes or neurons via high-affinity excitatory amino
acid transporters (EAATs).
4. Conversion of glutamate and ammonia to glutamine in the astroglial cytoplasm via
the enzyme glutamine synthetase.
5. Shuttling of glutamine to neurons via glutamine transporters.
6. Conversion of glutamine to glutamate and ammonia via the enzyme glutaminase.
7. Concentration of glutamate in synaptic vesicles via vesicular glutamate transporters.
[25]
As is evident in Figure 1, one astrocytic function is to remove glutamate – the most
abundant neurotransmitter in the vertebrate nervous system [27] – from the synaptic cleft.
This astrocytic intake of synaptic glutamate serves two purposes: 1) ensuring a precise post-
synaptic glutamate response (and modulation of time course, pattern, and extent of said
response) [28], and 2) prevention of the neuronal death that occurs in the presence of el-
evated glutamate levels for extended periods (a phenomenon referred to as excitotoxicity)
[29]. Once inside the astrocyte, glutamate is degraded by a variety of pathways including
conversion to glutathione and metabolism by the TCA cycle (into lactate, aspartate, or
recycled glutamate), though it appears that the largest fraction ( 50%) is converted to glu-
tamine by GS [30]. Finally, the astrocyte exports glutamine for use by neurons, which can
easily convert glutamine to functional glutamate via glutaminase. Glutamine is a preferable
metabolite for extracellular transport, as it is not associated with the same excitotoxicity
phenomenon as glutamate.
INTRODUCTION 8
Figure 1. The Glutamate-
Glutamine cycle. Image
from Eid, T., et al. (2016).
”The Glutamate-Glutamine
Cycle in Epilepsy.” Adv Neu-
robiol 13: 351-400.
Genetic cases of GS loss in humans are extremely rare, though findings from these pa-
tients support the importance of GS in healthy CNS function. Of the three noted cases
of homozygous Glul mutation, all patients developed severe epileptic encephalopathy, and
two of the thee experienced fatal multi-organ failure and resulting neonatal death [31, 32].
Individuals with Alzheimer’s dementia have also been found to have reduced expression of
GS [33], and abnormalities of tissue GS levels have been associated with many other condi-
tions including amyotrophic lateral sclerosis [34], Huntington’s disease [35], depression [36],
schizophrenia [37], and suicidal ideation [38].
Given the multitude of studies linking GS to human disease as well as the enzyme’s
compelling biochemical relationship to CNS health and functional neurotransmission,
much attention has been placed on further elucidation of the specific role GS has in
epileptogenesis, seizure initiation, and seizure propagation. However, human studies are
often limited in sample size, access to relevant tissues, and, of course, interventional scope.
Thus, animal models of epilepsy have been and remain crucial to the experimental process
of determining the precise relationship between glutamine synthetase and human disease.
INTRODUCTION 9
Chemical Models of Mesial Temporal Lobe Epilepsy
For over 50 years, animal models have provided insight into the mechanisms of epilepsy
and formed the foundation for discovering novel anti-epileptic drugs found in the clinic
today [39]. However, this suite of pharmacotherapies continues to be largely ineffective
in the treatment of refractory epilepsies—particularly MTLE. Some hypothesize that the
lack of advancement in MTLE treatment stems from fundamental differences between fea-
tures of epilepsy animal models in widespread use and the features specific to MTLE [40].
Staying cognizant of this important observation, the following discussion will focus on the
most common models with high relevance to MTLE specifically. In addition, it is impor-
tant to note that there are a vast number of other, non-chemical animal models of epilepsy
which will not be discussed here. While models of induction such as traumatic brain in-
jury, ischemic brain damage, and hyperthermia are undoubtedly useful, their widespread
and often non-specific effects complicate the study of complex, interconnected biochemi-
cal pathways [41].
One of the earliest examples of an MTLE model was first described by Ben-Ari and
Lagowska, who injected kainic acid into the unilateral rat amygdala [42]. Kainic acid is a
cyclic analog of glutamate that produces excitatory post-synaptic potentials upon binding
as an agonist to the ionotropic kainate receptor and as a partial agonist to AMPA receptors
[43]. Kainic acid can be administered locally as an injection to the desired brain region or
via systemic routes including subcutaneous, intraperitoneal, and intravenous [44]. While
systemic administration certainly simplifies the experimental procedure, it also carries sev-
eral disadvantages compared to direct CNS delivery: 1) extrahippocampal neuronal damage
is far more extensive than found in human MTLE, 2) unlike the typically unilateral hip-
pocampal damage seen in MTLE, systemic kainic acid damages both hippocampi, and 3)
animal mortality is high due to the systemic effects of kainic acid [45]. In contrast, intracra-
nial administration of kainic acid largely avoids these shortcomings, and in particular, in-
trahippocampal administration in mice closely resembles human MTLE both in terms of
INTRODUCTION 10
histological findings and seizure patterns. Following unilateral injection of 0.2 µg of kainic
acid into the dorsal hippocampus, mice enter status epilepticus which is typically noncon-
vulsive [46]. Following a two week latency period, mice enter a chronic epileptic phase in
which EEG demonstrates isolated high-voltage sharp waves and hippocampal paroxysmal
discharges which are restricted to the ipsilateral hippocampus [47]. In the study by Riban
et. al., all mice developed spontaneous chronic seizures, and approximately half of the co-
hort experienced secondary generalization. Histopathological findings in this model are
highly consistent with findings in human MTLE including neuronal loss in CA1 and CA3
subfields and dentate hilus as well as reactive gliosis predominantly restricted to the ipsilat-
eral hippocampus [46, 47].
Pilocarpine, a cholinergic muscarinic agonist, was first used as a rodent epileptogenic
shortly after kainic acid [48]. As seen with kainic acid, pilocarpine can be administered
through a variety of systemic and intracerebral routes, however administration via intrahip-
pocampal injection results in less extra-hippocampal damage, lower subject mortality, more
consistent induction of initial status epilepticus, and more prompt recovery from status
epilepticus [49]. In a large study, following unilateral injection of 2.4 µg of pilocarpine
into the hippocampus, 76% of animals experienced status epilepticus, which took a form
similar to the initial status epilepticus in the kainic acid model [50]. After a period of la-
tency averaging two weeks (range 2-30 days), 71% of animals demonstrated spontaneous
recurring seizures. In the pilocarpine model, abnormal EEG discharges typically begin
in the hippocampus and are associated with orofacial automatisms prior to development
of head and forelimb clonus, rearing, and falling [51]. As in human MTLE progression,
seizure frequency increases over time in the pilocarpine model [52]. Upon neuropatho-
logical examination, findings include neuronal loss in CA1, CA3, and dentate hilus, as well
as mossy fiber sprouting in the dentate gyrus [51]. Though extra-hippocampal damage
is more limited in local injection compared to systemic administration, the hippocampal
injection technique does yield some neurodegeneration in the cortex, thalamus, and amyg-
INTRODUCTION 11
dala [49]. The kainic acid and pilocarpine models induce neuronal damage in similar re-
gions, however the timing of such damage is quite different, as pilocarpine induced neural
loss is fast-appearing, whereas neurodegeneration is delayed following administration of
kainic acid [53].
The kainic acid and pilocarpine models of epilepsy have maintained their relevance for
decades, and they remain standards for the investigation of MTLE. However, understand-
ing the exact relevance of GS in epileptogenesis requires more specific methods of epilepsy
induction, particularly when attempting to understand whether GS plays a causative role
in the disease. In an effort to determine the direct effects of GS loss in vivo, in 2008, Eid
and colleagues infused methionine sulfoximine (MSO) into the hippocampi of otherwise
normal rats [54]. MSO, an irreversible glutamine synthetase inhibitor, is first phosphory-
lated by and then irreversibly binds to the enzyme [55]. Ten days after initiation of contin-
uous MSO infusion into the unilateral hippocampus, GS activity was found to be signifi-
cantly reduced compared to saline infused controls (82% reduction in low-dose MSO [.625
µg/hr] and 97% reduction in high-dose MSO [2.5 µg/hr]) [54]. Interestingly, GS activity
in the contralateral hippocampus was significantly reduced in the high-dose animals but
was unchanged in low-dose animals; drug diffusion effects may explain this finding.
Remarkably, 98% of MSO-infused animals were noted to have recurrent behavioral
seizures, whereas none of the saline infused animals seized [54]. This finding is the first
compelling evidence suggesting that GS depletion is sufficient to cause epileptogenesis.
Further, the seizure patterns seen in the MSO-infused animals mimicked patterns seen in
MTLE patients: 1) seizures were found to be temporally clustered, 2) EEG suggested a
mild degree of secondary generalization (i.e., mean 5.3 depth-electrode detected seizures
per day vs. mean 1.0 epidural electrode detected seizures per day), and 3) seizure behaviors
included immobilization, chewing and whisker twitching, forelimb clonus, and rearing and
falling. Upon histological examination of brain sections, MSO-infused animals demon-
strated dose-dependent neuronal loss in the hippocampus and adjacent structures. A subset
INTRODUCTION 12
of animals showed MTLE-like pathology including hippocampal shrinkage, CA1-CA3
neuronal loss, and relative sparing of neurons in the subiculum and granule cell layer of the
dentate gyrus. Interestingly, many animals that received the low dose of MSO exhibited
minimal neuronal loss despite extensive seizure activity, and the authors suggest that such
a result may indicate that GS depletion is more relevant to MTLE development than the
often-discussed mesial temporal sclerosis pattern.
The aforementioned MSO study has highlighted the relationship between GS and
MTLE as a crucial realm of investigation. Indeed, subsequent experiments have remon-
strated the model’s validity [56], addressed new areas of MTLE pathology [57], and as-
sessed the relevance of therapeutic interventions [58]. However, as with almost all chemical
agents, selectivity and off-target effects must be discussed. MSO is known to deplete tissue
glutathione [59], increase astrocyte glycogen [60], and activate neurons independent of its
action on GS [61]. While the 2008 MSO study accounted for glutathione depletion, and
found that there was no discernible effect on tissue glutathione in the low-dose MSO an-
imals [54], the control of other off-target effects of MSO poses a significant challenge. In
order to truly verify and further study the effects of specific absence of GS alone, genetic
approaches to GS depletion must be undertaken.
Genetic Models of Glutamine Synthetase Depletion
Since the first use of the Cre-lox recombination system in mammalian cells in 1988 [62],
this remarkable technology has gained widespread acceptance and validation as a tool for
the selective deletion of genes in certain model organisms. Following the establishment
of the NIH’s Neuroscience Research Cre-driver projects [63], the first model of Cre-lox
mediated GS knockout was reported in 2007 [64]. The researchers found that in a whole-
body (i.e., non-conditional) knockout of GS, the mice were not viable; embryo death oc-
curred when the embryo migrates from the oviduct into the uterus (embryonic day 3.5).
INTRODUCTION 13
This finding was largely unsurprising, as GS plays many crucial functions—both synthetic
and detoxifying—in several tissues throughout the body. In order to improve viability of
the mice and further study the effects of GS knockout, a follow-up experiment from the
same group restricted Cre expression to GFAP+ cells, thus limiting the GS knockout to
CNS astrocytes [65]. These mice with CNS knockout of GS were reported to be the first
viable animal model of genetic deletion of GS, however, even this more limited knockout
model resulted in early death (approximately post-natal day 3) that precluded the study of
potential epileptogenesis.
Very recently, Zhou et. al., reported creation of a novel GS knockout that was further
restricted, yielding viable mice that seize spontaneously [66]. By taking advantage of the
Emx1-Cre mouse line described previously [67], GS knockout was restricted to astrocytes
in the hippocampus and neocortex. These Emx1-Cre GS knockout mice were found to
exhibit recurrent spontaneous seizures, thus establishing the method as a tool for studying
epileptogenesis; indeed, the preliminary study itself shed light on several aspects of MTLE
pathology. Perhaps most notably, the researchers found that following deletion of GS,
changes in brain biochemistry as well as glial and vascular morphology were present for
several weeks prior to the development of recurrent seizures. This temporal dissociation
in a GS-specific epilepsy model provides strong evidence that GS-related epileptogenesis is
unlikely to be mediated by an excitotoxic mechanism, and instead, is related to “a patho-
logical process involving reactive astrocytes and impaired neurovascular coupling,” [66].
The Emx1-Cre GS knockout is certainly the most precise model of GS-mediated
MTLE to date. In addition, the high level of similarity between Emx1-Cre GS knock-
out and human MTLE tissue pathology and phenotype provides strong validation to the
gliopathy hypothesis of epileptogenesis [68]. However, the most notable limitation of the
Emx1-Cre knockout model is the overly-widespread distribution of GS knockout. Tis-
sue specific Cre-recombinase expression is restricted by the viability and availability of
transgenic mice with Cre expression driven by a particular gene of interest (e.g., GFAP or
INTRODUCTION 14
Emx1, as described above). Further spatial specificity of Cre expression is only feasible by
alternate techniques. In the Emx1-Cre GS knockout model, the observed strong deletion
of GS in the neocortex, while expected, is not representative of human MTLE, in which
GS depletion is typically present in the hippocampus and amygdala [23]. In fact, assessment
of human MTLE neocortex has revealed that GS activity appears to be unchanged in this
region [69]. In addition, tissue-specific Cre expression systems do not allow for lateraliza-
tion of the desired knockout, and the bilateral GS loss found in the Emx1-Cre model can-
not be refined to more closely approximate the unilateral pathology often found in MTLE.
In order to overcome these limitations of tissue-specific Cre-driven knockout, our goal
was to generate GS-knockout mice using local delivery of Cre via a direct injection of
Cre-expressing viral vector. The recombination of loxP sites driven by Cre derived from
a vector rather than from crossing with a Cre-expressing mouse line was first described
in 1995, when an adenovirus was used to transfect cells in vitro in an effort to prove that
such extrinsic methods of delivery were capable of inducing recombination of a floxed
gene [70]. Early studies using the technique faced substantial complications due to the
immunogenicity of adenovirus and other vectors, however in 2002, a newly engineered
adeno-associated virus (AAV) was used successfully in vivo to express the reporter gene
green fluorescent protein (GFP) in the mouse brain [71]. AAVs and lentivirus vectors are
unique in that they both have been engineered to remove all viral genes, thus rendering
them minimally immunogenic and virtually nontoxic [72, 73]. While many viral vectors
rely on active cell division in order to incorporate vector DNA into the host genome and
sustain protein production, AAVs and lentiviruses have the ability to transduce both divid-
ing and non-dividing cells, making them ideal candidates for transduction of the CNS, in
which many cell populations are non-dividing. AAVs retain this ability by maintaining the
viral genome episomally within the host cell [74], whereas lentiviruses have the unique ca-
pability of entering the nucleus in order to incorporate the plasmid in the absence of cell
division [75]. One of the most significant points of differentiation between lentiviruses and
INTRODUCTION 15
AAVs is their capsid size, as lentivirus has a typical diameter of 100nm and AAVs have a
diameter of approximately 20nm. Indeed, this substantial size difference has implications
for vector diffusibility through tissue following injection; the smaller AAV demonstrates 3-
to 5-fold greater viral spread than comparable lentivirus injections [76, 77]. The increased
diffusibility of AAVs come at the expense of payload, as more complex or multi-genetic
expression cassettes are not capable of fitting within the smaller AAV genome of approxi-
mately 4.7 kbp [78].
Given the relatively small size of the Cre and reporter GFP expression cassettes, the im-
portance of maximum viral spread from injection site, and the extensive customizability of
AAV packages, we elected to utilize AAVs in this study. However, within the AAV family,
There are numerous catalogued AAV serotypes, each of which has a unique capsid struc-
ture and surface proteins that influence cellular tropism and specificity [79]. In a highly
exhaustive review of transduction characteristics of the six most common AAV serotypes
currently used, Aschauer, Kreuz, and Rumpel present useful head-to-head comparisons in
efficiency of transgene expression, cell-type specificity, impact on microglia, and capabil-
ity for retrograde axonal transport [80]. Among the findings presented was evidence that
serotypes AAV5 and AAV8 were both highly effective in transduction of astrocytes. When
examined specifically, background-scaled reporter fluorescence of hippocampal astrocytes
was approximately 10-fold greater in AAV8 transduced cells compared to AAV5 transduced
cells, though both serotypes performed well compared to most other AAV types. As this
extensive dataset of serotype comparisons was conducted entirely with expression cassettes
driven by CMV promoters, we selected three of our four investigational viruses to utilize
CMV as well. In addition, given research suggesting that AAVs utilizing the intermedi-
ate filament glial fibrillary acidic protein (GFAP) improve selectivity of astrocytes and may
also improve transgene expression [81], we also elected to include an AAV8 serotype with
GFAP promoters. By comparing the knockdown efficiency of what appear to be four of
the most effective astrocytic transducers, we aim to identify the optimal vehicle for knock-
INTRODUCTION 16
down of GS in the mouse hippocampus via a methodology that allows for the spatial and
temporal control required for precise emulation of human MTLE.
Specific Aims
The purpose of this study was to create and optimize a model of mesial temporal lobe
epilepsy through selective depletion of glutamine synthetase (GS) in the mouse hippocam-
pus. In addition, preliminary studies characterized morphological astrocytic and synaptic
changes that result from the GS deficiency.
The goal of Aim 1 was to establish a novel mouse model of glutamine synthetase (GS)
knockout in hippocampal astrocytes. Cohorts of homozygous GS-floxed C57BL/6 mice
were injected with Cre-expressing virus into the bilateral dentate gyri, subiculi, and en-
torhinal cortices. Each cohort was injected with a virus of different serotype and promoter
sequence in order to determine which virus maximally depletes GS at the injection sites.
The goal of Aim 2 was to test whether localized hippocampal knockout of GS causes
mice to exhibit an epilepsy-like phenotype. Knockout and control mice were monitored
for severity and frequency of seizures using 24-hour video EEG recordings, and prior to
sacrifice, seizure proclivity was measured during a pentylenetetrazol injection study. Such
studies evaluated hippocampal GS knockout as a useful model of human epilepsy.
The goal of Aim 3 was to characterize the cellular effects of local GS loss. Given GS’s
role in converting glutamate to glutamine, proteins such as gephyrin (GABA synapse
marker), PSD-95 (glutamate synapse marker), and synaptophysin (global synapse marker)
are hypothesized to display altered tissue distribution. We will examine this using stimu-
lated emission depletion confocal microscopy. Mice created in Aim 1 allow us to identify
downstream effects of GS knockout and better characterize the process of epileptogenesis.
Methods
MGF performed all procedures described below with the exception of the preparation of
GS recombineering vector. Roni Dhaher assisted with seizure precipitation studies. Some
of the surgeries were performed by Mani Sandhu.
Animals and Reagents
All animal care and use procedures were approved by the Institutional Animal Care and
Use Committee of Yale University, and experiments were performed in accordance with
current guidelines. Care was taken to avoid suffering and minimize the number of exper-
imental animals required for the study. Mice were housed on a 12-hour light/dark cycle
in individually ventilated cages at constant temperature (22 ± 0.7 °C) and humidity (56
± 6%) and were fed with Harlan Teklad 2018 (Harlan Laboratories Inc., Indianapolis, IN,
USA) with access to food and water ad libitum. Biopsies from ear or tail were collected for
determining genotype.
In order to create a conditional knockout of GS, male and female C57BL/6J Glulf/f
mice were generated (see Figure 2). GS targeting vector was prepared by recombineer-
ing as described by Lee et. al., [82]. Briefly, approximately 12 kb of Glul genomic frag-
ment containing the entire Glul sequence was retrieved from the bacterial artificial chro-
mosome (BAC) clone, RP24-326N10 (obtained from the BACPAC Resources Center
at the Children’s Hospital Oakland Research Institute, Oakland, CA) by gap repair. The
5’LoxP site was inserted in intron 1 approximately 628 nucleotides 5’ of exon 2, and the
METHODS 19
Figure 2. Conditional knock-
out generation. Selective
deletion of the Glul gene in
the mesial temporal lobe was
achieved by injecting Cre-
expressing virus directly into
the hippocampi of Glul-floxed
mice.
second 3’LoxP sequence together with the Frt-PGKneo-Frt selectable marker was in-
serted in intron 6 approximately 140 nucleotides 5’ of exon 7. This vector containing ap-
proximately 4kb and 2.9kb of 5’-long and 3’-short arms, respectively, was then linearized
by NotI digestion, purified, and then electroporated into ES cells, which were derived
from F1(129sv/C57BL/6J) blastocyst. ES cells were cultured in the presence of G418 and
Gancyclovir after electroporation according to Wurst and Joyner [83], and drug resis-
tant colonies were picked and cultured in 96-well plates. Drug resistant ES clones were
screened by nested long-range PCR using primers specific to genomic sequences outside
the homology arms and LoxP sites to identify targeted ES clones. Targeted clones were
expanded and screened again to confirm their identity prior to the generation of chimeric
animals by aggregation with CD1 morula. Chimeric males were then bred with ROSA26-
Flpe female [84] to remove the PGKneo cassette to generate the final Glul floxed allele.
The resulting floxed F1 mice were then backcrossed with C57BL/6 mice for over 20 gen-
erations.
Site specific recombination of the loxP sites was achieved using a variety of viruses ex-
pressing Cre recombinase and GFP under various promoters in two separate expression
METHODS 20
cassettes (see example in Figure 3). The AAV5 CMV Legacy virus (SignaGen Laboratories,
Rockville, MD), which expressed Cre and GFP under CMV promoters within a capsid
from AAV serotype 5, was readily available from prior experiments conducted in our lab.
To compare viral transfection efficiency, three other virus types were acquired through
custom order from the Gene Therapy Center Vector Core at the University of North Car-
olina (Chapel Hill, NC). These viruses include AAV5 CMV (an additional AAV serotype 5
expressing Cre and GFP under separate CMV promoters), AAV8 CMV (an AAV serotype
8 expressing Cre and GFP under separate CMV promoters), and AAV8 GFAP (an AAV
serotype 8 expressing Cre and GFP under separate GFAP promoters). In addition, a corre-
sponding control virus was ordered for each of the four experimental viruses. Each control
virus was procured from the same source and was of identical construction to its corre-
sponding experimental virus other than lacking the Cre expression cassette. Prior to injec-
tion, all viruses were diluted to the same concentration to account for titer mismatch.
Primary antibodies used in this study included chicken anti-GFP (Aves Labs, Tigard,
OR), rabbit anti-GS (Sigma, St. Louis, MO), mouse anti-synaptophysin (Sigma), mouse
anti-gephyrin (Synaptic Systems, Göttingen, Germany), and mouse anti-PSD-95 (EMD
Millipore, Billerica, MA). Secondary antibodies Alexa Fluor 488 goat anti-chicken IgG,
Alexa Fluor 555 goat anti-rabbit IgG, Alexa Fluor 647 goat anti-mouse IgG, and Pro-
Long Diamond Antifade Mountant were from Life Technologies (Eugene, OR). All other
reagents, unless otherwise noted, were obtained from Sigma Chemical Co. (St. Louis,
MO).
Surgery: Viral Injection and Electrode Implantation
Mice were inducted into anesthesia with 5% Isoflurane (Baxter, Deerfield, IL) in O2 and
placed in a stereotaxic frame (David Kopf Instruments, Tujunga, CA). For the duration of
surgery, Isoflurane was maintained at 1.5-3% and titrated based on respiratory status and re-
METHODS 21
Figure 3. Vector map of the viral plasmid construct, showing AAV5 CMV as an example. Upon
entry into CMV-expressing cells, transcription and translation yields functional Cre and eGFP pro-
tein.
sponse to intermittent leg pinch. Burr holes were made into the skull bilaterally, and virus
was injected into the brain parenchyma using the following validated coordinates using
bregma as reference: Dentate Gyrus- AP -3.2mm, ML ±3.1mm, DV -3.5mm; Subiculum-
AP -3.7mm, ML ±3.5mm, DV -3.44mm; Entorhinal Cortex- AP -4.1mm, ML ±3.8mm,
DV -3.5mm. Virus was delivered stereotaxically using a 5µL 26s gauge syringe (Hamil-
ton, Reno, NV). At each of the six injection sites per animal, 0.5µL of virus was injected
at a rate of 0.05µL per 30 seconds. In order to minimize viral reflux upon needle removal,
following delivery at each injection site, the needle was left to rest for 2 minutes and then
raised out of the parenchyma at a rate not exceeding 0.5mm per minute.
METHODS 22
In the viral comparison study, 16 animals were injected as above, split equally (n=2 each)
between Cre-expressing and control viruses of the serotypes AAV5 CMV (Legacy), AAV5
CMV, AAV8 CMV, and AAV8 GFAP. Concurrent with viral injection, stainless steel
screw electrodes (Plastics One, Roanoke, VA) were positioned in the epidural space over
the parietal cortex bilaterally. A third screw electrode was placed in the epidural space over
the cerebellum as a reference, and a fourth screw electrode was implanted in the occipital
bone as a ground. The female ends of the electrodes were inserted into a pedestal (Plastics
One) which was cemented onto the skull with UV light cured acrylated urethane adhesive
(Loctite 3106 Light Cure Adhesive, Henkel Corp., Rocky Hill, CT) to form a headcap.
Post-operative analgesia was achieved using a single injection of Meloxicam SR at a dose of
4 mg/kg administered prior to removal of Isoflurane.
In the video-EEG study, 16 animals were injected with virus as described above, split
equally between Cre-expressing and control viruses only of serotype AAV8 GFAP (based
on findings of the viral comparison study; see Results section). Four weeks after viral in-
jection surgery, recording electrodes were implanted in various arrangements: one subset
was implanted with bilateral depth electrodes located in each hippocampus (n=6), another
subset was implanted with unilateral screw electrodes over the parietal cortex (n=7), and
a final subset was implanted with a single recording depth electrode in the hippocampus
and a reference depth electrode in the white matter of the cerebellum (n=3). Headcaps
were created as described above. Analgesia was achieved using one 0.05mg/kg pre-op dose
of buprenorphine (Reckitt Benckiser Healthcare, Hull, England), one 0.05mg/kg post-
op dose of buprenorphine, and approximately 1mg/kg/day of Meloxicam in the drinking
water for 48 hours after surgery.
METHODS 23
Video-EEG Monitoring and Seizure Precipitation Studies
Four to six weeks after viral injection surgery, mice were placed individually in custom-
made Plexiglas cages. A 6-channel cable was connected to the animal’s electrode pedestal
on one end and to a commutator (Plastics One) on the other. A second cable connected
the commutator to the digital EEG recording unit (CEEGraph Vision LTM, Natus/Bio-
Logic Systems Corp., San Carlos, CA). Digital cameras with infrared light detection ca-
pability were used to record animal behavior. The digital video signal was encoded and
synchronized to the digital EEG signals. Seizures were identified by visual inspection of the
EEG record. The Racine criteria [85] were modified and used to classify the seizures from
the video records, as follows: Stage 1, immobilization, eye blinking, twitching of the vib-
rissae, and mouth movements; Stage 2, head nodding, often accompanied by severe facial
clonus; Stage 3, forelimb clonus; Stage 4, rearing without falling; Stage 5, rearing, falling,
and generalized convulsions; Stage 6, violent generalized convulsions with repeated jump-
ing.
Ten weeks after viral injection, seizure precipitation studies were conducted by measur-
ing the time to first clonic twitch and time to generalized convulsions following intraperi-
toneal injection of the convulsant agent pentylenetetrazol (Sigma Aldrich, St. Louis, MO)
at a dose of 90mg/kg (diluted to 9mg/mL in normal saline). Following induction of con-
vulsive seizure, animals were fully anesthetized using Isoflurane and sacrificed as described
below.
Fixation, Immunofluorescence, and Microscopy
For staining, mice were fully anesthetized with Isoflurane and perfused transcardially
with saline (to remove intravascular blood) followed by 4% formaldehyde in 0.1M PB,
pH 7.4 for 5 minutes. The brains were rapidly removed from the skull and post-fixed in
the same fixative overnight at 4°C and then sectioned on a Vibratome at 50µm. Sections
METHODS 24
were rinsed (3 x 5 minutes) in TBST (TBS with 0.5% Triton X-100), treated with 1M
ethanolamine in 0.1M NaPi pH 7.4 for 30 minutes, washed in TBST (3 x 5 minutes), in-
cubated for 1 hour in TBST containing 10% newborn calf serum and 3% bovine serum
albumin followed by overnight incubation with primary antibodies. Next, sections were
washed with TBST (3 x 10 minutes), incubated with secondary antibodies (Alexa 488 di-
luted to 1:1000, Alexa 555 and Alexa 647 diluted to 1:500), washed with TBST (3 x 10
minutes), and mounted onto gel-coated slides with ProLong Diamond Antifade Mountant.
Samples were examined using an Olympus BX61 wide-angle fluorescence microscope
with Olympus DP71 camera attachment for image recording. Super-resolution images
were acquired using a Leica TCS SP8 Gated STED 3X at maximal total magnification
of approximately 5000x. Triple-labeling studies were conducted using a custom proto-
col consisting of sequential scans in order of descending STED depletion wavelength, as
follows: Alexa 647 (pulsed white-light laser tuned to 647nm, HyD photodetector tuned
to 680-730nm, 775nm STED depletion beam), Alexa 555 (pulsed white-light laser tuned
to 555nm, HyD photodetector tuned to 550-620nm, 660nm STED depletion beam), and
Alexa 488 (pulsed white-light laser tuned to 488nm, HyD photodetector tuned to 500-
570nm, 592nm STED depletion beam).
Statistical Analysis
Statistical analyses and graph plotting were conducted in Igor Pro 8.02 (WaveMetrics).
Statistical significance was assessed by Student’s t-test. Differences between groups were
judged to be significant when p-values were smaller than 0.05. Error bars indicate the
mean ± standard error of the mean (SEM) except when stated. No samples or animals
were excluded from the analysis. For PTZ seizure precipitation studies, experimenters
were blinded to the viral vector used in each animal when recording time to seizure events.
Results
Knockout of Glutamine Synthetase
To evaluate whether injection of each virus into the mesial temporal lobe resulted in vi-
able transfection of CNS cells, we first stained formaldehyde-fixed brain sections for GFP,
the reporter protein carried by each viral plasmid. GFP fluorescence was visualized in all
animals, confirming that the selected injection coordinates resulted in viral transfection of
the dentate gyrus, subiculum, and entorhinal cortex (see Figure 4a). Despite maintaining
consistent injection sites and quantity of virus injected for all viral serotypes, the degree
of observed transfection area varied substantially between virus types (see Figure 4b), sug-
gesting that different virus types may have differing abilities to diffuse through tissues or to
transfect cells in regions of lower viral concentration (i.e., at the periphery of the injection
field). Transfection area was determined by measuring the number of pixels that emitted
FITC filtered fluorescence (excitation 470-490nm, emission 510nm long-pass) at an in-
tensity above that of background emission and below that of obvious artifact (e.g., edge
effect). Measurements were taken at the section level which appeared to have maximal
affected area based on qualitative comparison and spanned an entire 40x field in order to
accommodate assessment of all three injection sites in one hemisphere. The most striking
finding was that the AAV8 GFAP virus appeared to transfect a substantially larger area than
both AAV5 CMV viruses and the AAV8 CMV virus (approximately 2.5 x 105
pixels vs. 1.2
x 105
, 1.4 x 105
, and 7.4 x 104
pixels, respectively).
RESULTS 26
Figure 4. Injection locations and viral spread. (A) GFP is detected in transfected cells of the hip-
pocampus and adjacent structures in a representative 40x field. EC- entorhinal cortex, DH- dentate
hilus, Gr- granule cell layer of the dentate gyrus, Mol- molecular layer of the dentate gyrus, SUB-
subiculum, LV- lateral ventricle. Scale bar = 0.5mm (B) Viral spread is measured as GFP+ pixels
present in the mesial temporal lobe.
We first confirmed that GS knockout was restricted to areas containing virus-transfected
cells by assessing co-localization of GFP and GS (see Figure 5). Next, area of GS knockout
was quantified for each virus at three magnifications in order to assess the proportion of
knockout in the entire mesial temporal lobe (40x), in the hippocampal formation (100x),
and in the dentate gyrus (200x). Area was determined by measuring the number of pixels
emitting TRITC filtered fluorescence (excitation 520-550nm, emission 580nm long-pass)
at an intensity above that of background emission and below that of distant areas of tis-
sue exhibiting normal expression of GS. As seen in Figure 6, the AAV5 CMV and AAV8
GFAP viruses decreased expression of GS in a substantially greater area than the other two
viruses.
Next, the efficiency of GS knockout was assessed for each virus type. In order to calcu-
late the extent of knockdown, five regions of interest (circular, radius 8 pixels) were placed
in the areas of apparently maximal knockdown visualized in the most qualitatively affected
40x field, and mean TRITC wavelength fluorescence intensity was measured in these re-
RESULTS 27
Figure 5. Representative section of brain transfected with AAV8 GFAP virus. Green represents
virus-transfected cells, while red represents presence of GS. GS knockout is seen only in areas with
transfected cells. Scale bar = 0.2mm
gions (see Figure 7a). The intensity was scaled by a control figure measured as the average
fluorescence intensity of three regions of interest which appeared to exhibit normal GS ex-
pression in the same section. The resulting relative intensity figure is the complement of
knockdown efficiency, expressed as a percentage. As seen in Figure 7b, knockdown ef-
ficiency was not statistically significant for AAV5 CMV Legacy (95% CI -14.5% to 2.4%,
p=0.12) or AAV8 CMV (95% CI -10.7% to 2.9%, p=0.19). Both the AAV5 CMV virus
and the AAV8 GFAP virus knocked down GS significantly (respectively, 95% CIs 47.8%
to 78.1% and 78.8% to 87.5%, p=0.0003 and 0.000007). In addition, the increased knock-
down of AAV8 GFAP over AAV5 CMV was statistically significant (p=0.018).
Finally, each virus’ selectivity for astrocytes was assessed, as cell-type specificity is an im-
portant factor that mitigates confounding cellular effects of off-target Cre and GFP ex-
pression and improves visualization of cells of interest in microscopy studies. Selectivity
RESULTS 28
800
600
400
200
Pixels
(x10
3
)
AAV5 CMV
(Legacy)
AAV5 CMV AAV8 CMV AAV8 GFAP
at 40x
at 100x
at 200x
Figure 6. Area of GS knockout for each virus
at various magnifications. AAV5 CMV and
AAV8 GFAP outperformed the AAV5 CMV
Legacy and AAV8 CMV viruses.
Figure 7. Quantification of GS knockout intensity. (A) Example of ROI selection in a section
transfected with AAV5 CMV. ROIs 1-5 are placed in areas of knockout whereas ROIs 6-8 are used
as control regions to scale intensity measures. Red stained areas are not infected with virus and ex-
press GS normally. Scale bar = 0.5mm (B) Knockdown efficiency following infection with each virus
type (*P<0.05 and **P<0.001).
RESULTS 29
Figure 8. Assessment of astrocyte specificity in transfected cell population. (A) Representative
field from AAV8 GFAP demonstrating astrocyte morphology in nearly all GFP+ cells. Scale bar =
0.1mm (B) Proportion of GFP+ cells demonstrating astrocyte morphology after transfection with
each virus type.
was assessed by manual count of GFP+ cells within a representative 200x field within the
dentate gyrus and classification of cells as “astrocyte” or “other” based on morphology. As
expected, AAV8 GFAP exhibited near perfect target specificity (98.7% of GFP+ cells were
astrocytes). As seen in Figure 8, AAV5 CMV Legacy, AAV5 CMV, and AAV8 CMV tar-
geted mostly neurons with varied degrees astrocyte labeling detected (10.0%, 21.3%, and
12.7% astrocytes, respectively.)
Seizure Findings
To determine if intrahippocampal knockout of GS is sufficient to cause recurrent sponta-
neous seizures, mice injected with Cre-expressing AAV8 GFAP (n=5) and control virus
(n=4) were monitored for two weeks with continuous video and intracranial EEG record-
ing. Epileptiform activity was assessed visually (example in Figure 9). Of the 6 animals im-
planted with bilateral hippocampal depth electrodes, 5 died due to post-operative compli-
cations prior to commencement of EEG recording. Thus, the remaining 10 animals were
implanted either with screw electrodes only (n=7) or a single recording hippocampal depth
RESULTS 30
Figure 9. Example EEG tracing during Racine stage 3 seizure. (A) Seizure progression and termi-
nation is demonstrated in top tracing with normal control trace below for comparison. Scale bar
represents 5 seconds. (B) Highlighted area in (A) is expanded to detail epileptic waveform.
electrode (n=3). Seizures were noted to occur in 60% (3/5) of the animals injected with
Cre-expressing virus and in 25% (1/4) of the animals injected with control virus (see Table
1). Seizures tended to occur in clusters (11/19 seizure events occurred within 2-hours of
another seizure event in the same animal) and in the early morning hours (14/19 seizure
events occurred between midnight at 8:00 AM. Observed behaviors during seizure events
were varied between animals, however, individual animals tended to exhibit similar features
during each event. Common seizure behaviors included facial automatisms, tail extension,
lordosis, rearing, and uncontrolled jumping.
In addition to comparing development of spontaneous seizures, sensitivity to provo-
cation of seizures was also measured. Fifteen mice transfected with one of the four ex-
perimental viruses (n=9) or four control viruses (n=6) were observed to determine time
to first myoclonic twitch and time to convulsive seizure following intraperitoneal injec-
tion of PTZ. As seen in Figure 10, animals with GS knocked out exhibited the first clonic
twitch after an average of 41.2 ± 3.2 seconds (mean ± SEM), which was significantly less
time than control animals, averaging a delay of 65.83 ± 12.9 seconds (p=0.044). The effect
RESULTS 31
Table 1. Day-by-day report of noted spontaneous behavioral seizures in the 9 continuous video-
EEG-recorded mice. Dark grey boxes indicate that recording was stopped due to death of the
animal or deterioration of clinical state requiring euthanasia. Electrode arrangements are as follows:
Depth, Bil. depth recording electrodes in both hippocampi; Depth, Uni.- depth recording electrode
in a single hippocampus and a depth reference electrode in the cerebellum; Screw- screw recording
electrode above the parietal cortex.
on time to convulsive seizure was not statistically significant, though there was a trend of
knockout animals proceeding to convulsions in less time (74.2 ± 9.4 seconds vs. 100.0 ±
18.0 seconds, p=0.20).
120
100
80
60
40
20
0
Delay
from
PTZ
Administration
(s)
*
Clonic Twitch Convulsions
GS Knockout
Control
Figure 10. PTZ-induced precipitation of pro-
voked seizures. GS knockout mice demon-
strate increased susceptibility to provoked
seizures, as illustrated by a shorter time to
clonic twitch compared to mice transfected
with control virus (p<0.05). There was a sim-
ilar trend in time to convulsion, though this
effect was not significant (p=0.20).
RESULTS 32
Synaptic Changes
A preliminary assessment of observed synaptic changes caused by GS loss was also per-
formed by performing fluorescence STED microscopy on brain sections from mice in-
jected with AAV8 GFAP Cre-expressing virus. Sections were triple-labeled with primary
antibodies against GFP, GS, and one of three synaptic markers: synaptophysin, PSD-95, or
gephyrin. First, changes in synaptic marker expression between GS+ and GS- areas were
assessed visually in a 200x field containing the dentate gyrus (Figure 10a). The expected
absence synaptophysin, PSD-95, and gephyrin within the granule cell layer of the dentate
gyrus was noted, and no other gross changes were apparent. Next, more nuanced changes
in synaptic marker expression were assessed by measuring the synaptic fluorescence in-
tensity in selected regions within the visible territories of both GFP-/GS+ (unaffected)
and GFP+/GS- (knockout) astrocytes. Expression of PSD-95 appeared to be decreased in
synapses proximal to GS- astrocytes, with sampled relative intensity of 0.57 ± 0.04 (mean
± SEM, p=0.002, see Figure 10b). Relative expression of synaptophysin and gephyrin
appeared to be unchanged in the sampled areas (synaptophysin relative expression 0.94 ±
0.15, p=0.87; gephyrin relative expression 0.94 ± 0.04, p=0.23).
RESULTS 33
Figure 11. Assessment of synaptic marker changes associated with loss of GS. (A) Example 200x
field illustrating staining pattern of synaptophysin in the dentate gyrus, which is grossly unchanged
in areas with GS knockdown. (B) Quantification of relative intensity of synaptic markers in normal
GS+ astrocytes (arrow in panel A) compared to GFP+/GS- astrocytes (arrowhead in panel A).
PSD-95 expression appears to be reduced in GS- astrocytes (p=0.002), while synaptophysin and
gephyrin expression appear to be unchanged. (C)Comparison of a synaptophysin colocalization
with single astrocytic process of a GS+ astrocyte (left panel) and a GS knockout astrocyte (right
panel) at 5000x magnification. Scale bars = 5µm
Discussion
Development of a monogenetic knockout model with effects restricted to the hip-
pocampus and adjacent structures has the potential to more fully elucidate the impact of
GS loss in this treatment-resistant disease. In this study, we demonstrated the use of a novel
model of MTLE by selectively knocking out GS in the hippocampal astrocytes of mice.
Studies of MTLE in humans and in other experimental models show that loss of GS is
closely associated with MTLE pathology [23, 25, 54], however the exact role of GS in
MTLE and the biochemical pathways through which it may exert its effects remain un-
clear.
In order to first maximize GS knockout, we compared the expression patterns of GS
after Glul-floxed C57BL/6J mice were injected with four different adeno-associated viral
vectors containing Cre-recombinase expression cassettes. Injection volumes and viral titers
were matched between viruses, however knockout of GS was found over a greater area in
the AAV5 CMV and AAV8 GFAP serotypes. In addition, within each GS knockout re-
gion, AAV8 GFAP exhibited a significantly greater efficiency of knockdown compared to
other serotypes and most selectively targeted astrocytes. Thus, AAV8 GFAP was selected
for use in video-EEG and synaptic microscopy studies. When hippocampal GS knockout
mice were assessed with video-EEG over a 12-day period, 60% (3/5) animals exhibited a
phenotype comparable to human MTLE including spontaneous recurrent seizures clustered
in the morning hours. 25% (1/4) of control animals also seized recurrently when moni-
DISCUSSION 35
tored over the same period. Additionally, GS knockout mice appeared more susceptible
to provoked seizures, as delay to clonic twitch following PTZ injection was reduced by 24
seconds (p=0.044). Finally, examination of synaptic markers in the territory of GS+ and
GS- astrocytes revealed a significant decrease in PSD-95, while expression of synaptophysin
and gephyrin appeared unchanged.
Several limitations of the research presented must be discussed in order to most appro-
priately interpret the study results. First, the small sample size prevents definitive conclu-
sions from being drawn in the seizure proclivity studies, particularly given the development
of spontaneous seizures in one of the control animals injected with non-Cre-expressing
virus. It should be noted that the epileptic control animal was the sole surviving animal of
the 6 mice implanted with bilateral depth electrodes, which may suggest that the seizures
resulted from physical trauma to the brain parenchyma during electrode implantation. As-
certaining the cause of this (and any subsequent) control animal seizures will be crucial in
validation of the model and confirmation that seizures in experimental animals are caused
by GS depletion rather than physical trauma associated with viral injection and/or cellu-
lar effects of viral transfection or intracellular GFP. We also encountered challenges with
a high incidence of premature headcap removal (5/9 mice). This further limited data col-
lected for video-EEG, seizure precipitation, and histology studies, and, importantly, po-
tentially exposed experimental animals to undue discomfort. Subsequent iterations of EEG
implantation should attempt to use alternate materials or methods of headcap mounting
in order to reduce the frequency of such events. Small sample size also limits the general-
izability of microscopy data, which in some cases, required analysis of brain sections from
a single animal transfected with certain viral serotypes. Finally, while the virally mediated
localization of GS knockout has the potential to fully mimic the spatial pattern of GS loss
seen in human MTLE, this iteration of the model does not. The experimenters elected to
knockout GS in the dentate gyrus, subiculum, and entorhinal cortex bilaterally as an ini-
tial proof-of-concept, however subsequent iterations will more closely model the typically
DISCUSSION 36
unilateral findings of GS depletion.
The results presented lay the foundation for informative follow-up investigations. First,
bolstering of sample size will clarify the efficacy of virus-mediated GS knockout in elicit-
ing spontaneous seizures and will allow more granular study of epileptogenesis at various
time points following GS loss. The extent of GS knockdown should also be confirmed
using tissue-based methods such as laser capture micro-dissection followed by qPCR. As-
sessing local knockout mice for comorbidities found in human MTLE, such as depression
and anxiety, will also lend further evaluate how well the model mimics human disease.
Additionally, following thorough validation of the local GS knockout model, more ex-
pansive microscopy experiments should be undertaken to fully capitalize on the poten-
tial advantages conferred by super-resolution microscopy techniques such as STED. Mi-
croscopy studies will include the examination of the changes induced by GS-loss on vas-
culature, molecular transporters, microglia, and synapses, including further investigation of
our finding that PSD-95 expression appeared to be reduced in the vicinity of GS knock-
out astrocytes. Given PSD-95’s known binding activity with neural potassium channels and
glutamate receptors AMPA and NMDA [86] as well as the well-characterized BDNF me-
diated downregulation of NMDA receptor expression in the context of excitotoxicity [87],
our findings may suggest that such changes may vary within each cell-to-cell microenvi-
ronment. Additional examination of this observed PSD-95 downregulation and apparent
sparing of synaptophysin (which is nearly ubiquitous in all synapses) and gephyrin (found at
inhibitory synapses) may shed light on the specific importance of excitatory synaptic reg-
ulation in epileptogenesis. Further studies will also produce more information about the
prevalence of seizures in local GS knockout mice, and should there be GS knockout mice
which do not seize (as there were in this study), the unique cellular features present in such
mice may provide useful long-term insights for therapeutic targeting and drug develop-
ment.
The most notable shortcoming of the virus-mediated localized knockout of GS is the
DISCUSSION 37
highly technical and time-consuming nature of subject generation. Even after Glul-floxed
mice have been created and bred to produce homozygous-floxed animals, injection of virus
is a relatively involved process which requires careful diligence and time. The necessity
to use stereotaxic technique, deliver virus extremely gradually, and repeat the procedure
over multiple injection sites results in a multi-hour surgical procedure for each individ-
ual subject which benefits negligibly from batching of procedures. For this reason, local
GS knockout may not be an ideal model for all MTLE investigations. However, the novel
precision of the technique does address recent critiques about indiscriminate selection of
animal models [88]. This approach also improves on the emulative accuracy of models cur-
rently in use in epilepsy basic science and translational research, which has been cited as a
contributor to the recent paucity of novel therapeutics in the field of treatment-resistant
epilepsies such as MTLE [2]. The novel model presented here has the capability to mimic
the spatial and temporal disease pattern of human MTLE with greater accuracy and flex-
ibility than any previously reported GS-depletion model of epilepsy, and with further
validation, virus-mediated knockout of hippocampal GS has the potential to become the
gold-standard model in MTLE research.
References
1. Wilson, J.K. and E.H. Reynolds, Translation and analysis of a cuneiform text form-
ing part of a Babylonian treatise on epilepsy. Medical history, 1990. 34(2): p. 185-
198.
2. Steinhauser, C., M. Grunnet, and G. Carmignoto, Crucial role of astrocytes in tem-
poral lobe epilepsy. Neuroscience, 2016. 323: p. 157-69.
3. Stafstrom, C.E. and L. Carmant, Seizures and Epilepsy: An Overview for Neurosci-
entists. Cold Spring Harbor Perspectives in Medicine, 2015. 5(6).
4. Bernhardt, B., et al., Imaging structural and functional brain networks in temporal
lobe epilepsy. Frontiers in Human Neuroscience, 2013. 7(624).
5. Thom, M. and E.H. Bertram, Chapter 14 - Temporal lobe epilepsy, in Handbook of Clin-
ical Neurology, H. Stefan and W.H. Theodore, Editors. 2012, Elsevier. p. 225-240.
6. Asadi-Pooya, A.A., et al., Prevalence and Incidence of Drug-Resistant Mesial Tem-
poral Lobe Epilepsy in the United States. World Neurosurgery, 2017. 99: p. 662-
666.
7. Brodie, M.S., SC; Kwan, P, Epilepsy. 5 ed. Fast Facts. 2012: Health Press Limited.
146.
8. Allone, C., et al., Neuroimaging and cognitive functions in temporal lobe epilepsy:
A review of the literature. Journal of the Neurological Sciences, 2017. 381: p. 7-15.
9. Hermann, B.P., et al., Comorbid Psychiatric Symptoms in Temporal Lobe Epilepsy:
Association with Chronicity of Epilepsy and Impact on Quality of Life. Epilepsy &
Behavior, 2000. 1(3): p. 184-190.
10. Garcia, C.S., Depression in temporal lobe epilepsy: a review of prevalence, clinical
features, and management considerations. Epilepsy research and treatment, 2012.
2012: p. 1-12.
11. Andrade-Machado, R., et al., Mortality in patients with refractory temporal lobe
epilepsy at a tertiary center in Cuba. Epilepsy & Behavior, 2015. 53: p. 154-160.
12. Sutula, T., Mossy fiber synaptic reorganization in the epileptic human temporal lobe.
Annals of neurology. 26(3): p. 321-330.
REFERENCES 39
13. Aronica, E., et al., Induction of neonatal-sodium channel II and III (α-isoform mR-
NAs in neurons and microglia after status epilepticus in the rat hippocampus. Euro-
pean Journal of Neuroscience, 2001. 13(6): p. 1261-1266.
14. Mathern, G.W., Hippocampal AMPA and NMDA mRNA levels and subunit im-
munoreactivity in human temporal lobe epilepsy patients and a rodent model of
chronic mesial limbic epilepsy. Epilepsy research. 32(1-2): p. 154-171.
15. de Lanerolle, N.C., Hippocampal interneuron loss and plasticity in human temporal
lobe epilepsy. Brain research. 495(2): p. 387-395.
16. Mangan, P.S. and E.H. Bertram Iii, Shortened-duration GABA(A) receptor-
mediated synaptic potentials underlie enhanced CA1 excitability in a chronic model
of temporal lobe epilepsy. Neuroscience, 1997. 80(4): p. 1101-1111.
17. Mangan, P.S. and E.W. Lothman, Profound disturbances of pre- and postsynaptic
GABA(B)-receptor-mediated processes in region CA1 in a chronic model of tempo-
ral lobe epilepsy. Journal of Neurophysiology, 1996. 76(2): p. 1282-1296.
18. Rigau, V., et al., Angiogenesis is associated with blood-brain barrier permeability in
temporal lobe epilepsy. Brain, 2007. 130(7): p. 1942-1956.
19. Mtchedlishvili, Z., E.H. Bertram, and J. Kapur, Diminished allopregnanolone en-
hancement of GABAA receptor currents in a rat model of chronic temporal lobe
epilepsy. Journal of Physiology, 2001. 537(2): p. 453-465.
20. During, M.J., Extracellular hippocampal glutamate and spontaneous seizure in the
conscious human brain. Lancet. 341(8861): p. 1607-1610.
21. Olney, J.W., Excitotoxic mechanisms of epileptic brain damage. Advances in neurol-
ogy, 1986. 44: p. 857-877.
22. Petroff, O.A.C., Glutamate-glutamine cycling in the epileptic human hippocampus.
Epilepsia (Copenhagen). 43(7): p. 703-710.
23. Eid, T., et al., Loss of glutamine synthetase in the human epileptogenic hippocam-
pus: possible mechanism for raised extracellular glutamate in mesial temporal lobe
epilepsy. The Lancet, 2004. 363(9402): p. 28-37.
24. Krajewski, W.W., et al., Crystal structures of mammalian glutamine synthetases illus-
trate substrate-induced conformational changes and provide opportunities for drug
and herbicide design. (1089-8638 (Electronic)).
25. Eid, T., et al., The Glutamate-Glutamine Cycle in Epilepsy. Adv Neurobiol, 2016.
13: p. 351-400.
26. Norenberg Md Fau - Martinez-Hernandez, A. and A. Martinez-Hernandez, Fine
structural localization of glutamine synthetase in astrocytes of rat brain. (0006-8993
(Print)).
REFERENCES 40
27. Meldrum, B.S., Glutamate as a neurotransmitter in the brain: review of physiology
and pathology. (0022-3166 (Print)).
28. Danbolt, N.C., Glutamate uptake. (0301-0082 (Print)).
29. Olney, J.W., Brain lesions, obesity, and other disturbances in mice treated with
monosodium glutamate. (0036-8075 (Print)).
30. McKenna, M.C., The glutamate-glutamine cycle is not stoichiometric: fates of glu-
tamate in brain. Journal of neuroscience research. 85(15): p. 3347-3358.
31. Haberle, J., et al., Congenital glutamine deficiency with glutamine synthetase muta-
tions. (1533-4406 (Electronic)).
32. Haberle, J., et al., Natural course of glutamine synthetase deficiency in a 3 year old
patient. (1096-7206 (Electronic)).
33. Le Prince, G., et al., Glutamine synthetase (GS) expression is reduced in senile de-
mentia of the Alzheimer type. 1995(0364-3190 (Print)).
34. Tumani, H., et al., Glutamine synthetase in cerebrospinal fluid, serum, and brain: a
diagnostic marker for Alzheimer disease? 1999(0003-9942 (Print)).
35. Carter, C.J., Loss of glutamine synthetase activity in the brain in Huntington’s dis-
ease. 1981(0140-6736 (Print)).
36. Rajkowska, G., Astrocyte pathology in major depressive disorder: insights from hu-
man postmortem brain tissue. Current drug targets, 2013. 14(11): p. 1225-1236.
37. Bruneau, E.G., Increased expression of glutaminase and glutamine synthetase
mRNA in the thalamus in schizophrenia. Schizophrenia research, 2005. 75(1): p.
27-34.
38. Kalkman, H.O., Circumstantial evidence for a role of glutamine-synthetase in sui-
cide. Medical hypotheses, 2011. 76(6): p. 905-907.
39. Löscher, W., Critical review of current animal models of seizures and epilepsy used
in the discovery and development of new antiepileptic drugs. Seizure, 2011. 20(5): p.
359-368.
40. Depaulis, A. and S. Hamelin, Animal models for mesiotemporal lobe epilepsy: The
end of a misunderstanding? Revue Neurologique, 2015. 171(3): p. 217-226.
41. Löscher, W., Animal models of epilepsy and epileptic seizures. Antiepileptic Drugs.
Handbook of Experimental Pharmacology, 1999: p. 19-62.
42. Ben-Ari, Y. and J. Lagowska, Epileptogenic action of intra-amygdaloid injection of kainic
acid. Comptes rendus hebdomadaires des seances de l’Academie des sciences. Serie
D: Sciences naturelles, 1978. 287(8): p. 813-816.
REFERENCES 41
43. Fritsch, B., et al., Role of GluK1 Kainate Receptors in Seizures, Epileptic Dis-
charges, and Epileptogenesis. The Journal of Neuroscience, 2014. 34(17): p. 5765.
44. Lévesque, M. and M. Avoli, The kainic acid model of temporal lobe epilepsy. Neu-
roscience & Biobehavioral Reviews, 2013. 37(10, Part 2): p. 2887-2899.
45. Jefferys, J., C. Steinhäuser, and P. Bedner, Chemically-induced TLE models: Topical
application. Journal of Neuroscience Methods, 2016. 260: p. 53-61.
46. Bouilleret, V., et al., Recurrent seizures and hippocampal sclerosis following
intrahippocampal kainate injection in adult mice: Electroencephalography,
histopathology and synaptic reorganization similar to mesial temporal lobe epilepsy.
Neuroscience, 1999. 89(3): p. 717-729.
47. Riban, V., et al., Evolution of hippocampal epileptic activity during the develop-
ment of hippocampal sclerosis in a mouse model of temporal lobe epilepsy. Neuro-
science, 2002. 112(1): p. 101-111.
48. Turski, W.A., et al., Limbic seizures produced by pilocarpine in rats: Behavioural,
electroencephalographic and neuropathological study. Behavioural Brain Research,
1983. 9(3): p. 315-335.
49. Castro, O.W., et al., Comparative neuroanatomical and temporal characterization of
FluoroJade-positive neurodegeneration after status epilepticus induced by systemic
and intrahippocampal pilocarpine in Wistar rats. Brain Research, 2011. 1374: p. 43-
55.
50. Furtado, M.A., et al., Study of spontaneous recurrent seizures and morphological
alterations after status epilepticus induced by intrahippocampal injection of pilo-
carpine. Epilepsy Behavior, 2011. 20(2): p. 257-266.
51. De A. Furtado, M., et al., Behavioral, Morphologic, and Electroencephalo-
graphic Evaluation of Seizures Induced by Intrahippocampal Microinjection of Pi-
locarpine. Epilepsia, 2002. 43(s5): p. 37-39.
52. Arida, R.M., et al., The course of untreated seizures in the pilocarpine model of
epilepsy. Epilepsy Research, 1999. 34(2): p. 99-107.
53. Covolan, L. and L.E.A.M. Mello, Temporal profile of neuronal injury following pi-
locarpine or kainic acid-induced status epilepticus. Epilepsy Research, 2000. 39(2):
p. 133-152.
54. Eid, T., et al., Recurrent seizures and brain pathology after inhibition of glutamine
synthetase in the hippocampus in rats. Brain, 2008. 131(Pt 8): p. 2061-70.
55. Krajewski, W.W., S.L. Jones Ta Fau - Mowbray, and S.L. Mowbray, Structure of
Mycobacterium tuberculosis glutamine synthetase in complex with a transition-state
mimic provides functional insights. Proc Natl Acad Sci, U. S. A., 2005(0027-8424
(Print)).
REFERENCES 42
56. Dhaher, R., et al., Effects of site-specific infusions of methionine sulfoximine on
the temporal progression of seizures in a rat model of mesial temporal lobe epilepsy.
Epilepsy, Res, 2015(1872-6844 (Electronic)).
57. Wang, H., et al., Network evolution in mesial temporal lobe epilepsy revealed by
diffusion tensor imaging. Epilepsia, 2017(1528-1167 (Electronic)).
58. Dhaher, R., et al., 5-aminovaleric acid suppresses the development of severe seizures
in the methionine sulfoximine model of mesial temporal lobe epilepsy. Neurobiol,
Dis, 2014(1095-953X (Electronic)).
59. Shaw, C.A. and J.S. Bains, Synergistic versus antagonistic actions of glutamate and
glutathione: the role of excitotoxicity and oxidative stress in neuronal disease. Cellu-
lar and Molecular Biology, 2002. 48(2): p. 127-136.
60. Bernard-Hélary, K., et al., In vivo and in vitro glycogenic effects of methionine sul-
foximine are different in two inbred strains of mice. Brain Research, 2002. 929(2):
p. 147-155.
61. Kam, K. and R. Nicoll, Excitatory synaptic transmission persists independently of
the glutamate-glutamine cycle. Journal of Neuroscience, 2007. 27(34): p. 9192-
9200.
62. Sauer, B. and N. Henderson, Site-specific DNA recombination in mammalian cells
by the Cre recombinase of bacteriophage P1. Proceedings of the National Academy
of Sciences, 1988. 85(14): p. 5166.
63. NIH blueprint for neuroscience: Cre driver network. Available from:
http://www.neuroscienceblueprint.nih.gov/factSheet/CreDriver.htm.
64. He, Y., et al., Glutamine synthetase is essential in early mouse embryogenesis. De-
velopmental Dynamics, 2007. 236(7): p. 1865-1875.
65. He, Y., et al., Glutamine synthetase deficiency in murine astrocytes results in neona-
tal death. GLIA, 2010. 58(6): p. 741-754.
66. Zhou, Y., et al., Selective deletion of glutamine synthetase in the mouse cerebral
cortex induces glial dysfunction and vascular impairment that precede epilepsy and
neurodegeneration. Neurochem Int, 2018.
67. Gorski, J.A., et al., Cortical Excitatory Neurons and Glia, But Not GABAergic
Neurons, Are Produced in the Emx1-Expressing Lineage. The Journal of Neuro-
science, 2002. 22(15): p. 6309.
68. Coulter, D.A. and T. Eid, Astrocytic regulation of glutamate homeostasis in epilepsy.
Glia, 2012. 60(8): p. 1215-1226.
69. Steffens, M., et al., Unchanged glutamine synthetase activity and increased NMDA
receptor density in epileptic human neocortex: Implications for the pathophysiology
of epilepsy. Neurochemistry International, 2005. 47(6): p. 379-384.
REFERENCES 43
70. Anton, M. and F.L. Graham, Site-specific recombination mediated by an adenovirus
vector expressing the Cre recombinase protein: a molecular switch for control of
gene expression. Journal of Virology, 1995. 69(8): p. 4600.
71. Kaspar, B.K., et al., Adeno-associated virus effectively mediates conditional gene
modification in the brain. Proceedings of the National Academy of Sciences, 2002.
99(4): p. 2320.
72. Blömer, U., et al., Highly efficient and sustained gene transfer in adult neurons with
a lentivirus vector. Journal of Virology, 1997. 71(9): p. 6641.
73. Kaspar, B.K., et al., Targeted Retrograde Gene Delivery for Neuronal Protection.
Molecular Therapy, 2002. 5(1): p. 50-56.
74. Salganik, M., M.L. Hirsch, and R.J. Samulski, Adeno-associated virus as a mammalian
DNA vector. Microbiology spectrum, 2015. 3(4).
75. Sakuma, T., M.A. Barry, and Y. Ikeda, Lentiviral vectors: Basic to translational.
Biochemical Journal, 2012. 443(3): p. 603-618.
76. Cearley, C.N. and J.H. Wolfe, Transduction characteristics of adeno-associated virus
vectors expressing cap serotypes 7, 8, 9, and Rh10 in the mouse brain. Molecular
Therapy, 2006. 13(3): p. 528-537.
77. Linterman, K.S., et al., Lentiviral-mediated gene transfer to the sheep brain: Impli-
cations for gene therapy in batten disease. Human Gene Therapy, 2011. 22(8): p.
1011-1020.
78. Blessing, D. and N. Déglon, Adeno-associated virus and lentivirus vectors: a refined
toolkit for the central nervous system. Current Opinion in Virology, 2016. 21: p.
61-66.
79. Agbandje-McKenna, M. and J. Kleinschmidt, AAV capsid structure and cell interactions,
in Methods in Molecular Biology. 2011. p. 47-92.
80. Aschauer, D.F., S. Kreuz, and S. Rumpel, Analysis of Transduction Efficiency,
Tropism and Axonal Transport of AAV Serotypes 1, 2, 5, 6, 8 and 9 in the Mouse
Brain. PLOS ONE, 2013. 8(9): p. e76310.
81. von Jonquieres, G., et al., Glial Promoter Selectivity following AAV-Delivery to the
Immature Brain. PLOS ONE, 2013. 8(6): p. e65646.
82. Lee, E.C., et al., A highly efficient Escherichia coli-based chromosome engineering
system adapted for recombinogenic targeting and subcloning of BAC DNA. Ge-
nomics, 2001. 73(0888-7543 (Print)).
83. Wurst, W.J., A.L., Production of Targeted Embryonic Stem Cell Clones, in Targeting, Gene,
A Practical Approach, A.L. Joyner, Editor. 1999, Oxford University Press: New York.
REFERENCES 44
84. Soriano, P., The PDGF� receptor is required for neural crest cell development and
for normal patterning of the somites. Development, 1997. 124(14): p. 2691-2700.
85. Racine, R.J., et al., Rates of motor seizure development in rats subjected to electri-
cal brain stimulation: strain and inter-stimulation interval effects. Electroencephalog-
raphy and Clinical Neurophysiology, 1973. 35(5): p. 553-556.
86. Sheng, M. and C. Sala, PDZ Domains and the Organization of Supramolecular
Complexes. Annual Review of Neuroscience, 2001. 24(1): p. 1-29.
87. Brandoli, C., et al., Brain-Derived Neurotrophic Factor and Basic Fibroblast Growth
Factor Downregulate NMDA Receptor Function in Cerebellar Granule Cells. The
Journal of Neuroscience, 1998. 18(19): p. 7953.
88. Denayer, T., T. Stöhr, and M. Van Roy, Animal models in translational medicine:
Validation and prediction. New Horizons in Translational Medicine, 2014. 2(1): p.
5-11.

More Related Content

What's hot

Diaz-Arrastia, Ramon
Diaz-Arrastia, RamonDiaz-Arrastia, Ramon
Diaz-Arrastia, Ramon
National Neurotrauma Symposium
 
Subacute sclerosing panencephalitis (SSPE) In Iraq
Subacute sclerosing panencephalitis (SSPE) In IraqSubacute sclerosing panencephalitis (SSPE) In Iraq
Subacute sclerosing panencephalitis (SSPE) In Iraq
iosrjce
 
A Proposed Mechanism for Autoimmune Mediated Motor Dysfunctions, and a Hypoth...
A Proposed Mechanism for Autoimmune Mediated Motor Dysfunctions, and a Hypoth...A Proposed Mechanism for Autoimmune Mediated Motor Dysfunctions, and a Hypoth...
A Proposed Mechanism for Autoimmune Mediated Motor Dysfunctions, and a Hypoth...
ramdab
 
Poster-Kelly Berger
Poster-Kelly BergerPoster-Kelly Berger
Poster-Kelly Berger
Kelly Berger
 
Genome sequencing uncovers MS phenocopies in primary progressive multiple scl...
Genome sequencing uncovers MS phenocopies in primary progressive multiple scl...Genome sequencing uncovers MS phenocopies in primary progressive multiple scl...
Genome sequencing uncovers MS phenocopies in primary progressive multiple scl...
Sherman Jia
 
Neurology Today
Neurology TodayNeurology Today
Neurology Today
Katerina Akassoglou
 
Diapositivas seminario biomol
Diapositivas seminario biomolDiapositivas seminario biomol
Diapositivas seminario biomol
IsabellaMadrigalAlva
 
The Laboratory Diagnosis Of Tuberous Sclerosis
The Laboratory Diagnosis Of Tuberous SclerosisThe Laboratory Diagnosis Of Tuberous Sclerosis
The Laboratory Diagnosis Of Tuberous Sclerosis
atss
 
Ferretti et al., BBI 2016
Ferretti et al., BBI 2016Ferretti et al., BBI 2016
Ferretti et al., BBI 2016
Maria Teresa Ferretti
 
Bio341.Final.new
Bio341.Final.newBio341.Final.new
Bio341.Final.new
Bradley Christman
 
Molecular biology seminar - mlmh-3
Molecular biology   seminar - mlmh-3Molecular biology   seminar - mlmh-3
Molecular biology seminar - mlmh-3
marialuisamendezhern
 
Human Genetics Paper
Human Genetics PaperHuman Genetics Paper
Human Genetics Paper
Joseph Thompson
 
BDSRA 2015 CLN10 Burrow, Spaeth, Sisk, Hallinan
BDSRA 2015 CLN10 Burrow, Spaeth, Sisk, HallinanBDSRA 2015 CLN10 Burrow, Spaeth, Sisk, Hallinan
BDSRA 2015 CLN10 Burrow, Spaeth, Sisk, Hallinan
Batten Disease Support and Research Association
 
Dr. Francesc Palau - 'Neuropatías periféricas hereditarias'
Dr. Francesc Palau - 'Neuropatías periféricas hereditarias'Dr. Francesc Palau - 'Neuropatías periféricas hereditarias'
Dr. Francesc Palau - 'Neuropatías periféricas hereditarias'
Fundación Ramón Areces
 
Diapos seminario microglía
Diapos seminario microglía Diapos seminario microglía
Diapos seminario microglía
IsabellaMadrigalAlva
 
Marc Dhenain Alzforum Webinar - Dec 7, 2016
Marc Dhenain Alzforum Webinar - Dec 7, 2016Marc Dhenain Alzforum Webinar - Dec 7, 2016
Marc Dhenain Alzforum Webinar - Dec 7, 2016
Alzforum
 
Hypothermia
HypothermiaHypothermia
Hypothermia
Hikmatullah Arif
 
stem cell and Traumatic brain injury
stem cell and Traumatic brain injurystem cell and Traumatic brain injury
stem cell and Traumatic brain injury
aishudiva
 
BDSRA 2015 CLN1 Hofmann
BDSRA 2015 CLN1 HofmannBDSRA 2015 CLN1 Hofmann
Scientifi c Journal of Multiple Sclerosis
Scientifi c Journal of Multiple SclerosisScientifi c Journal of Multiple Sclerosis
Scientifi c Journal of Multiple Sclerosis
SciRes Literature LLC. | Open Access Journals
 

What's hot (20)

Diaz-Arrastia, Ramon
Diaz-Arrastia, RamonDiaz-Arrastia, Ramon
Diaz-Arrastia, Ramon
 
Subacute sclerosing panencephalitis (SSPE) In Iraq
Subacute sclerosing panencephalitis (SSPE) In IraqSubacute sclerosing panencephalitis (SSPE) In Iraq
Subacute sclerosing panencephalitis (SSPE) In Iraq
 
A Proposed Mechanism for Autoimmune Mediated Motor Dysfunctions, and a Hypoth...
A Proposed Mechanism for Autoimmune Mediated Motor Dysfunctions, and a Hypoth...A Proposed Mechanism for Autoimmune Mediated Motor Dysfunctions, and a Hypoth...
A Proposed Mechanism for Autoimmune Mediated Motor Dysfunctions, and a Hypoth...
 
Poster-Kelly Berger
Poster-Kelly BergerPoster-Kelly Berger
Poster-Kelly Berger
 
Genome sequencing uncovers MS phenocopies in primary progressive multiple scl...
Genome sequencing uncovers MS phenocopies in primary progressive multiple scl...Genome sequencing uncovers MS phenocopies in primary progressive multiple scl...
Genome sequencing uncovers MS phenocopies in primary progressive multiple scl...
 
Neurology Today
Neurology TodayNeurology Today
Neurology Today
 
Diapositivas seminario biomol
Diapositivas seminario biomolDiapositivas seminario biomol
Diapositivas seminario biomol
 
The Laboratory Diagnosis Of Tuberous Sclerosis
The Laboratory Diagnosis Of Tuberous SclerosisThe Laboratory Diagnosis Of Tuberous Sclerosis
The Laboratory Diagnosis Of Tuberous Sclerosis
 
Ferretti et al., BBI 2016
Ferretti et al., BBI 2016Ferretti et al., BBI 2016
Ferretti et al., BBI 2016
 
Bio341.Final.new
Bio341.Final.newBio341.Final.new
Bio341.Final.new
 
Molecular biology seminar - mlmh-3
Molecular biology   seminar - mlmh-3Molecular biology   seminar - mlmh-3
Molecular biology seminar - mlmh-3
 
Human Genetics Paper
Human Genetics PaperHuman Genetics Paper
Human Genetics Paper
 
BDSRA 2015 CLN10 Burrow, Spaeth, Sisk, Hallinan
BDSRA 2015 CLN10 Burrow, Spaeth, Sisk, HallinanBDSRA 2015 CLN10 Burrow, Spaeth, Sisk, Hallinan
BDSRA 2015 CLN10 Burrow, Spaeth, Sisk, Hallinan
 
Dr. Francesc Palau - 'Neuropatías periféricas hereditarias'
Dr. Francesc Palau - 'Neuropatías periféricas hereditarias'Dr. Francesc Palau - 'Neuropatías periféricas hereditarias'
Dr. Francesc Palau - 'Neuropatías periféricas hereditarias'
 
Diapos seminario microglía
Diapos seminario microglía Diapos seminario microglía
Diapos seminario microglía
 
Marc Dhenain Alzforum Webinar - Dec 7, 2016
Marc Dhenain Alzforum Webinar - Dec 7, 2016Marc Dhenain Alzforum Webinar - Dec 7, 2016
Marc Dhenain Alzforum Webinar - Dec 7, 2016
 
Hypothermia
HypothermiaHypothermia
Hypothermia
 
stem cell and Traumatic brain injury
stem cell and Traumatic brain injurystem cell and Traumatic brain injury
stem cell and Traumatic brain injury
 
BDSRA 2015 CLN1 Hofmann
BDSRA 2015 CLN1 HofmannBDSRA 2015 CLN1 Hofmann
BDSRA 2015 CLN1 Hofmann
 
Scientifi c Journal of Multiple Sclerosis
Scientifi c Journal of Multiple SclerosisScientifi c Journal of Multiple Sclerosis
Scientifi c Journal of Multiple Sclerosis
 

Similar to Localized hippocampal glutamine synthetase knockout a novel model of mesial temporal lobe epilepsy

Epileptogenesis
EpileptogenesisEpileptogenesis
Epileptogenesis
Ahmed Mamdouh
 
Study of anticonvulsant activity of quinidine in albino rats using pentylenet...
Study of anticonvulsant activity of quinidine in albino rats using pentylenet...Study of anticonvulsant activity of quinidine in albino rats using pentylenet...
Study of anticonvulsant activity of quinidine in albino rats using pentylenet...
iosrjce
 
Epileptogenesis
EpileptogenesisEpileptogenesis
Epileptogenesis
Vamsi Krishna Koneru
 
Ncomms5563
Ncomms5563Ncomms5563
Ncomms5563
You-Yin Chen
 
Axonal Polyneuropathy with Anti-Caspr1 Igg1 Paranodal Antibodies
Axonal Polyneuropathy with Anti-Caspr1 Igg1 Paranodal AntibodiesAxonal Polyneuropathy with Anti-Caspr1 Igg1 Paranodal Antibodies
Axonal Polyneuropathy with Anti-Caspr1 Igg1 Paranodal Antibodies
semualkaira
 
958461
958461958461
Myasthenia gravis
Myasthenia gravisMyasthenia gravis
Myasthenia gravis
Tikal Kansara
 
Myasthenia Gravis e-Medicine Article
Myasthenia Gravis e-Medicine Article Myasthenia Gravis e-Medicine Article
Myasthenia Gravis e-Medicine Article
Imhotep Virtual Medical School
 
Articulo TRPC6
Articulo TRPC6Articulo TRPC6
Articulo TRPC6
Jorge Parodi
 
INVESTIGATION THE EFFECT OF PENTHYLENTETRAZOLE INDUCING EPILEPSY MODEL USING ...
INVESTIGATION THE EFFECT OF PENTHYLENTETRAZOLE INDUCING EPILEPSY MODEL USING ...INVESTIGATION THE EFFECT OF PENTHYLENTETRAZOLE INDUCING EPILEPSY MODEL USING ...
INVESTIGATION THE EFFECT OF PENTHYLENTETRAZOLE INDUCING EPILEPSY MODEL USING ...
Self-employed researcher
 
Lab diagnosis of Autoimmune Encephalitis
Lab diagnosis of Autoimmune EncephalitisLab diagnosis of Autoimmune Encephalitis
Lab diagnosis of Autoimmune Encephalitis
Santosh Dash
 
A case of Neuromyelitis optica as a presenting manifestation of Systemic Lupu...
A case of Neuromyelitis optica as a presenting manifestation of Systemic Lupu...A case of Neuromyelitis optica as a presenting manifestation of Systemic Lupu...
A case of Neuromyelitis optica as a presenting manifestation of Systemic Lupu...
Apollo Hospitals
 
Lnc rna meg3 promotes glaucomatous retinal ganglion cell apoptosis via upregu...
Lnc rna meg3 promotes glaucomatous retinal ganglion cell apoptosis via upregu...Lnc rna meg3 promotes glaucomatous retinal ganglion cell apoptosis via upregu...
Lnc rna meg3 promotes glaucomatous retinal ganglion cell apoptosis via upregu...
Clinical Surgery Research Communications
 
vaibhav presentation on the epilepsy 29-12-2001.pptx
vaibhav presentation on the epilepsy 29-12-2001.pptxvaibhav presentation on the epilepsy 29-12-2001.pptx
vaibhav presentation on the epilepsy 29-12-2001.pptx
ssuserb83b8a
 
Review Nature 399 A15
Review Nature 399 A15Review Nature 399 A15
Review Nature 399 A15
MADHU.MATHEWS
 
A Case of CIDP
A Case of CIDPA Case of CIDP
Changes in the Bladder After Spinal Cord Injury and Expression of VEGF and AP...
Changes in the Bladder After Spinal Cord Injury and Expression of VEGF and AP...Changes in the Bladder After Spinal Cord Injury and Expression of VEGF and AP...
Changes in the Bladder After Spinal Cord Injury and Expression of VEGF and AP...
ANALYTICAL AND QUANTITATIVE CYTOPATHOLOGY AND HISTOPATHOLOGY
 
UTF-8''Final Assessing post-synaptic partners of Dentate Granule Cells in a M...
UTF-8''Final Assessing post-synaptic partners of Dentate Granule Cells in a M...UTF-8''Final Assessing post-synaptic partners of Dentate Granule Cells in a M...
UTF-8''Final Assessing post-synaptic partners of Dentate Granule Cells in a M...
Grant Pizzo
 
Crimson Publishers-More than Meets the Eye: Biological Clocks
 Crimson Publishers-More than Meets the Eye: Biological Clocks Crimson Publishers-More than Meets the Eye: Biological Clocks
Crimson Publishers-More than Meets the Eye: Biological Clocks
CrimsonpublishersMSOR
 
140514 - HD synaptic characterization
140514 - HD synaptic characterization140514 - HD synaptic characterization
140514 - HD synaptic characterization
AJ Keefe
 

Similar to Localized hippocampal glutamine synthetase knockout a novel model of mesial temporal lobe epilepsy (20)

Epileptogenesis
EpileptogenesisEpileptogenesis
Epileptogenesis
 
Study of anticonvulsant activity of quinidine in albino rats using pentylenet...
Study of anticonvulsant activity of quinidine in albino rats using pentylenet...Study of anticonvulsant activity of quinidine in albino rats using pentylenet...
Study of anticonvulsant activity of quinidine in albino rats using pentylenet...
 
Epileptogenesis
EpileptogenesisEpileptogenesis
Epileptogenesis
 
Ncomms5563
Ncomms5563Ncomms5563
Ncomms5563
 
Axonal Polyneuropathy with Anti-Caspr1 Igg1 Paranodal Antibodies
Axonal Polyneuropathy with Anti-Caspr1 Igg1 Paranodal AntibodiesAxonal Polyneuropathy with Anti-Caspr1 Igg1 Paranodal Antibodies
Axonal Polyneuropathy with Anti-Caspr1 Igg1 Paranodal Antibodies
 
958461
958461958461
958461
 
Myasthenia gravis
Myasthenia gravisMyasthenia gravis
Myasthenia gravis
 
Myasthenia Gravis e-Medicine Article
Myasthenia Gravis e-Medicine Article Myasthenia Gravis e-Medicine Article
Myasthenia Gravis e-Medicine Article
 
Articulo TRPC6
Articulo TRPC6Articulo TRPC6
Articulo TRPC6
 
INVESTIGATION THE EFFECT OF PENTHYLENTETRAZOLE INDUCING EPILEPSY MODEL USING ...
INVESTIGATION THE EFFECT OF PENTHYLENTETRAZOLE INDUCING EPILEPSY MODEL USING ...INVESTIGATION THE EFFECT OF PENTHYLENTETRAZOLE INDUCING EPILEPSY MODEL USING ...
INVESTIGATION THE EFFECT OF PENTHYLENTETRAZOLE INDUCING EPILEPSY MODEL USING ...
 
Lab diagnosis of Autoimmune Encephalitis
Lab diagnosis of Autoimmune EncephalitisLab diagnosis of Autoimmune Encephalitis
Lab diagnosis of Autoimmune Encephalitis
 
A case of Neuromyelitis optica as a presenting manifestation of Systemic Lupu...
A case of Neuromyelitis optica as a presenting manifestation of Systemic Lupu...A case of Neuromyelitis optica as a presenting manifestation of Systemic Lupu...
A case of Neuromyelitis optica as a presenting manifestation of Systemic Lupu...
 
Lnc rna meg3 promotes glaucomatous retinal ganglion cell apoptosis via upregu...
Lnc rna meg3 promotes glaucomatous retinal ganglion cell apoptosis via upregu...Lnc rna meg3 promotes glaucomatous retinal ganglion cell apoptosis via upregu...
Lnc rna meg3 promotes glaucomatous retinal ganglion cell apoptosis via upregu...
 
vaibhav presentation on the epilepsy 29-12-2001.pptx
vaibhav presentation on the epilepsy 29-12-2001.pptxvaibhav presentation on the epilepsy 29-12-2001.pptx
vaibhav presentation on the epilepsy 29-12-2001.pptx
 
Review Nature 399 A15
Review Nature 399 A15Review Nature 399 A15
Review Nature 399 A15
 
A Case of CIDP
A Case of CIDPA Case of CIDP
A Case of CIDP
 
Changes in the Bladder After Spinal Cord Injury and Expression of VEGF and AP...
Changes in the Bladder After Spinal Cord Injury and Expression of VEGF and AP...Changes in the Bladder After Spinal Cord Injury and Expression of VEGF and AP...
Changes in the Bladder After Spinal Cord Injury and Expression of VEGF and AP...
 
UTF-8''Final Assessing post-synaptic partners of Dentate Granule Cells in a M...
UTF-8''Final Assessing post-synaptic partners of Dentate Granule Cells in a M...UTF-8''Final Assessing post-synaptic partners of Dentate Granule Cells in a M...
UTF-8''Final Assessing post-synaptic partners of Dentate Granule Cells in a M...
 
Crimson Publishers-More than Meets the Eye: Biological Clocks
 Crimson Publishers-More than Meets the Eye: Biological Clocks Crimson Publishers-More than Meets the Eye: Biological Clocks
Crimson Publishers-More than Meets the Eye: Biological Clocks
 
140514 - HD synaptic characterization
140514 - HD synaptic characterization140514 - HD synaptic characterization
140514 - HD synaptic characterization
 

More from https://www.facebook.com/garmentspace

Một số biện pháp phát triển ngôn ngữ cho trẻ 5 -6 tuổi trường Mẫu Giáo Minh T...
Một số biện pháp phát triển ngôn ngữ cho trẻ 5 -6 tuổi trường Mẫu Giáo Minh T...Một số biện pháp phát triển ngôn ngữ cho trẻ 5 -6 tuổi trường Mẫu Giáo Minh T...
Một số biện pháp phát triển ngôn ngữ cho trẻ 5 -6 tuổi trường Mẫu Giáo Minh T...
https://www.facebook.com/garmentspace
 
Một số kiến nghị và giải pháp nhằm nâng cao hiệu quả khai thác của đại lý bảo...
Một số kiến nghị và giải pháp nhằm nâng cao hiệu quả khai thác của đại lý bảo...Một số kiến nghị và giải pháp nhằm nâng cao hiệu quả khai thác của đại lý bảo...
Một số kiến nghị và giải pháp nhằm nâng cao hiệu quả khai thác của đại lý bảo...
https://www.facebook.com/garmentspace
 
Một số giải pháp Marketing thúc đẩy tình hình tiêu thụ sản phẩm tại Công ty T...
Một số giải pháp Marketing thúc đẩy tình hình tiêu thụ sản phẩm tại Công ty T...Một số giải pháp Marketing thúc đẩy tình hình tiêu thụ sản phẩm tại Công ty T...
Một số giải pháp Marketing thúc đẩy tình hình tiêu thụ sản phẩm tại Công ty T...
https://www.facebook.com/garmentspace
 
Nghiên cứu tính toán cung cấp điện cho hệ thống xử lý nước thải nhà máy thép ...
Nghiên cứu tính toán cung cấp điện cho hệ thống xử lý nước thải nhà máy thép ...Nghiên cứu tính toán cung cấp điện cho hệ thống xử lý nước thải nhà máy thép ...
Nghiên cứu tính toán cung cấp điện cho hệ thống xử lý nước thải nhà máy thép ...
https://www.facebook.com/garmentspace
 
Hoàn thiện hoạt đông phân phối xe máy Honda Việt Nam tại Công ty TNHH Thương ...
Hoàn thiện hoạt đông phân phối xe máy Honda Việt Nam tại Công ty TNHH Thương ...Hoàn thiện hoạt đông phân phối xe máy Honda Việt Nam tại Công ty TNHH Thương ...
Hoàn thiện hoạt đông phân phối xe máy Honda Việt Nam tại Công ty TNHH Thương ...
https://www.facebook.com/garmentspace
 
Hạch toán tiền lương và các khoản trích theo lương tại công ty viễn thông Hà ...
Hạch toán tiền lương và các khoản trích theo lương tại công ty viễn thông Hà ...Hạch toán tiền lương và các khoản trích theo lương tại công ty viễn thông Hà ...
Hạch toán tiền lương và các khoản trích theo lương tại công ty viễn thông Hà ...
https://www.facebook.com/garmentspace
 
Website tin tức – Quảng cáo tiếp thị.pdf
Website tin tức – Quảng cáo tiếp thị.pdfWebsite tin tức – Quảng cáo tiếp thị.pdf
Website tin tức – Quảng cáo tiếp thị.pdf
https://www.facebook.com/garmentspace
 
Xác định bảng giá đất trên một số tuyến đường quận 5 Thành phố Hồ Chí Minh.pdf
Xác định bảng giá đất trên một số tuyến đường quận 5 Thành phố Hồ Chí Minh.pdfXác định bảng giá đất trên một số tuyến đường quận 5 Thành phố Hồ Chí Minh.pdf
Xác định bảng giá đất trên một số tuyến đường quận 5 Thành phố Hồ Chí Minh.pdf
https://www.facebook.com/garmentspace
 
Thực hiện hệ thống MIMO STBC trên Board FPGA Arria V.pdf
Thực hiện hệ thống MIMO STBC trên Board FPGA Arria V.pdfThực hiện hệ thống MIMO STBC trên Board FPGA Arria V.pdf
Thực hiện hệ thống MIMO STBC trên Board FPGA Arria V.pdf
https://www.facebook.com/garmentspace
 
Vai trò và tác dụng của các nguyên tố vi lượng.doc
Vai trò và tác dụng của các nguyên tố vi lượng.docVai trò và tác dụng của các nguyên tố vi lượng.doc
Vai trò và tác dụng của các nguyên tố vi lượng.doc
https://www.facebook.com/garmentspace
 
Phân tích hệ thống HACCP áp dụng tại công ty Acecook.doc
Phân tích hệ thống HACCP áp dụng tại công ty Acecook.docPhân tích hệ thống HACCP áp dụng tại công ty Acecook.doc
Phân tích hệ thống HACCP áp dụng tại công ty Acecook.doc
https://www.facebook.com/garmentspace
 
Nghiên cứu hiện trạng và đề xuất giải pháp quản lý chất thải sinh hoạt trên đ...
Nghiên cứu hiện trạng và đề xuất giải pháp quản lý chất thải sinh hoạt trên đ...Nghiên cứu hiện trạng và đề xuất giải pháp quản lý chất thải sinh hoạt trên đ...
Nghiên cứu hiện trạng và đề xuất giải pháp quản lý chất thải sinh hoạt trên đ...
https://www.facebook.com/garmentspace
 
Nghiên cứu đề xuất các giải pháp cải tiến hệ thống quản lý môi trường theo IS...
Nghiên cứu đề xuất các giải pháp cải tiến hệ thống quản lý môi trường theo IS...Nghiên cứu đề xuất các giải pháp cải tiến hệ thống quản lý môi trường theo IS...
Nghiên cứu đề xuất các giải pháp cải tiến hệ thống quản lý môi trường theo IS...
https://www.facebook.com/garmentspace
 
Nâng cao công tác quản trị rủi ro cho vay tại Ngân hàng Nông nghiệp và Phát t...
Nâng cao công tác quản trị rủi ro cho vay tại Ngân hàng Nông nghiệp và Phát t...Nâng cao công tác quản trị rủi ro cho vay tại Ngân hàng Nông nghiệp và Phát t...
Nâng cao công tác quản trị rủi ro cho vay tại Ngân hàng Nông nghiệp và Phát t...
https://www.facebook.com/garmentspace
 
Nâng cao công tác quản trị vốn lưu động tại Công ty TNHH Thương mại Dịch vụ K...
Nâng cao công tác quản trị vốn lưu động tại Công ty TNHH Thương mại Dịch vụ K...Nâng cao công tác quản trị vốn lưu động tại Công ty TNHH Thương mại Dịch vụ K...
Nâng cao công tác quản trị vốn lưu động tại Công ty TNHH Thương mại Dịch vụ K...
https://www.facebook.com/garmentspace
 
Một số giải pháp tạo động lực làm việc cho công nhân thông qua việc thực hiện...
Một số giải pháp tạo động lực làm việc cho công nhân thông qua việc thực hiện...Một số giải pháp tạo động lực làm việc cho công nhân thông qua việc thực hiện...
Một số giải pháp tạo động lực làm việc cho công nhân thông qua việc thực hiện...
https://www.facebook.com/garmentspace
 
Một số giải pháp nhằm nâng hiệu quả sử dụng vốn lưu động của Công ty Cổ phần ...
Một số giải pháp nhằm nâng hiệu quả sử dụng vốn lưu động của Công ty Cổ phần ...Một số giải pháp nhằm nâng hiệu quả sử dụng vốn lưu động của Công ty Cổ phần ...
Một số giải pháp nhằm nâng hiệu quả sử dụng vốn lưu động của Công ty Cổ phần ...
https://www.facebook.com/garmentspace
 
Một số giải pháp nhằm nâng cao hiệu quả các biện pháp marketing mix tại Công ...
Một số giải pháp nhằm nâng cao hiệu quả các biện pháp marketing mix tại Công ...Một số giải pháp nhằm nâng cao hiệu quả các biện pháp marketing mix tại Công ...
Một số giải pháp nhằm nâng cao hiệu quả các biện pháp marketing mix tại Công ...
https://www.facebook.com/garmentspace
 
Một số giải pháp nhằm nâng cao hiệu quả hoạt động truyền thông Marketing tại ...
Một số giải pháp nhằm nâng cao hiệu quả hoạt động truyền thông Marketing tại ...Một số giải pháp nhằm nâng cao hiệu quả hoạt động truyền thông Marketing tại ...
Một số giải pháp nhằm nâng cao hiệu quả hoạt động truyền thông Marketing tại ...
https://www.facebook.com/garmentspace
 
Một số giải pháp nâng cao hiệu quả sử dụng vốn tại Công ty Cổ phần Thiết bị B...
Một số giải pháp nâng cao hiệu quả sử dụng vốn tại Công ty Cổ phần Thiết bị B...Một số giải pháp nâng cao hiệu quả sử dụng vốn tại Công ty Cổ phần Thiết bị B...
Một số giải pháp nâng cao hiệu quả sử dụng vốn tại Công ty Cổ phần Thiết bị B...
https://www.facebook.com/garmentspace
 

More from https://www.facebook.com/garmentspace (20)

Một số biện pháp phát triển ngôn ngữ cho trẻ 5 -6 tuổi trường Mẫu Giáo Minh T...
Một số biện pháp phát triển ngôn ngữ cho trẻ 5 -6 tuổi trường Mẫu Giáo Minh T...Một số biện pháp phát triển ngôn ngữ cho trẻ 5 -6 tuổi trường Mẫu Giáo Minh T...
Một số biện pháp phát triển ngôn ngữ cho trẻ 5 -6 tuổi trường Mẫu Giáo Minh T...
 
Một số kiến nghị và giải pháp nhằm nâng cao hiệu quả khai thác của đại lý bảo...
Một số kiến nghị và giải pháp nhằm nâng cao hiệu quả khai thác của đại lý bảo...Một số kiến nghị và giải pháp nhằm nâng cao hiệu quả khai thác của đại lý bảo...
Một số kiến nghị và giải pháp nhằm nâng cao hiệu quả khai thác của đại lý bảo...
 
Một số giải pháp Marketing thúc đẩy tình hình tiêu thụ sản phẩm tại Công ty T...
Một số giải pháp Marketing thúc đẩy tình hình tiêu thụ sản phẩm tại Công ty T...Một số giải pháp Marketing thúc đẩy tình hình tiêu thụ sản phẩm tại Công ty T...
Một số giải pháp Marketing thúc đẩy tình hình tiêu thụ sản phẩm tại Công ty T...
 
Nghiên cứu tính toán cung cấp điện cho hệ thống xử lý nước thải nhà máy thép ...
Nghiên cứu tính toán cung cấp điện cho hệ thống xử lý nước thải nhà máy thép ...Nghiên cứu tính toán cung cấp điện cho hệ thống xử lý nước thải nhà máy thép ...
Nghiên cứu tính toán cung cấp điện cho hệ thống xử lý nước thải nhà máy thép ...
 
Hoàn thiện hoạt đông phân phối xe máy Honda Việt Nam tại Công ty TNHH Thương ...
Hoàn thiện hoạt đông phân phối xe máy Honda Việt Nam tại Công ty TNHH Thương ...Hoàn thiện hoạt đông phân phối xe máy Honda Việt Nam tại Công ty TNHH Thương ...
Hoàn thiện hoạt đông phân phối xe máy Honda Việt Nam tại Công ty TNHH Thương ...
 
Hạch toán tiền lương và các khoản trích theo lương tại công ty viễn thông Hà ...
Hạch toán tiền lương và các khoản trích theo lương tại công ty viễn thông Hà ...Hạch toán tiền lương và các khoản trích theo lương tại công ty viễn thông Hà ...
Hạch toán tiền lương và các khoản trích theo lương tại công ty viễn thông Hà ...
 
Website tin tức – Quảng cáo tiếp thị.pdf
Website tin tức – Quảng cáo tiếp thị.pdfWebsite tin tức – Quảng cáo tiếp thị.pdf
Website tin tức – Quảng cáo tiếp thị.pdf
 
Xác định bảng giá đất trên một số tuyến đường quận 5 Thành phố Hồ Chí Minh.pdf
Xác định bảng giá đất trên một số tuyến đường quận 5 Thành phố Hồ Chí Minh.pdfXác định bảng giá đất trên một số tuyến đường quận 5 Thành phố Hồ Chí Minh.pdf
Xác định bảng giá đất trên một số tuyến đường quận 5 Thành phố Hồ Chí Minh.pdf
 
Thực hiện hệ thống MIMO STBC trên Board FPGA Arria V.pdf
Thực hiện hệ thống MIMO STBC trên Board FPGA Arria V.pdfThực hiện hệ thống MIMO STBC trên Board FPGA Arria V.pdf
Thực hiện hệ thống MIMO STBC trên Board FPGA Arria V.pdf
 
Vai trò và tác dụng của các nguyên tố vi lượng.doc
Vai trò và tác dụng của các nguyên tố vi lượng.docVai trò và tác dụng của các nguyên tố vi lượng.doc
Vai trò và tác dụng của các nguyên tố vi lượng.doc
 
Phân tích hệ thống HACCP áp dụng tại công ty Acecook.doc
Phân tích hệ thống HACCP áp dụng tại công ty Acecook.docPhân tích hệ thống HACCP áp dụng tại công ty Acecook.doc
Phân tích hệ thống HACCP áp dụng tại công ty Acecook.doc
 
Nghiên cứu hiện trạng và đề xuất giải pháp quản lý chất thải sinh hoạt trên đ...
Nghiên cứu hiện trạng và đề xuất giải pháp quản lý chất thải sinh hoạt trên đ...Nghiên cứu hiện trạng và đề xuất giải pháp quản lý chất thải sinh hoạt trên đ...
Nghiên cứu hiện trạng và đề xuất giải pháp quản lý chất thải sinh hoạt trên đ...
 
Nghiên cứu đề xuất các giải pháp cải tiến hệ thống quản lý môi trường theo IS...
Nghiên cứu đề xuất các giải pháp cải tiến hệ thống quản lý môi trường theo IS...Nghiên cứu đề xuất các giải pháp cải tiến hệ thống quản lý môi trường theo IS...
Nghiên cứu đề xuất các giải pháp cải tiến hệ thống quản lý môi trường theo IS...
 
Nâng cao công tác quản trị rủi ro cho vay tại Ngân hàng Nông nghiệp và Phát t...
Nâng cao công tác quản trị rủi ro cho vay tại Ngân hàng Nông nghiệp và Phát t...Nâng cao công tác quản trị rủi ro cho vay tại Ngân hàng Nông nghiệp và Phát t...
Nâng cao công tác quản trị rủi ro cho vay tại Ngân hàng Nông nghiệp và Phát t...
 
Nâng cao công tác quản trị vốn lưu động tại Công ty TNHH Thương mại Dịch vụ K...
Nâng cao công tác quản trị vốn lưu động tại Công ty TNHH Thương mại Dịch vụ K...Nâng cao công tác quản trị vốn lưu động tại Công ty TNHH Thương mại Dịch vụ K...
Nâng cao công tác quản trị vốn lưu động tại Công ty TNHH Thương mại Dịch vụ K...
 
Một số giải pháp tạo động lực làm việc cho công nhân thông qua việc thực hiện...
Một số giải pháp tạo động lực làm việc cho công nhân thông qua việc thực hiện...Một số giải pháp tạo động lực làm việc cho công nhân thông qua việc thực hiện...
Một số giải pháp tạo động lực làm việc cho công nhân thông qua việc thực hiện...
 
Một số giải pháp nhằm nâng hiệu quả sử dụng vốn lưu động của Công ty Cổ phần ...
Một số giải pháp nhằm nâng hiệu quả sử dụng vốn lưu động của Công ty Cổ phần ...Một số giải pháp nhằm nâng hiệu quả sử dụng vốn lưu động của Công ty Cổ phần ...
Một số giải pháp nhằm nâng hiệu quả sử dụng vốn lưu động của Công ty Cổ phần ...
 
Một số giải pháp nhằm nâng cao hiệu quả các biện pháp marketing mix tại Công ...
Một số giải pháp nhằm nâng cao hiệu quả các biện pháp marketing mix tại Công ...Một số giải pháp nhằm nâng cao hiệu quả các biện pháp marketing mix tại Công ...
Một số giải pháp nhằm nâng cao hiệu quả các biện pháp marketing mix tại Công ...
 
Một số giải pháp nhằm nâng cao hiệu quả hoạt động truyền thông Marketing tại ...
Một số giải pháp nhằm nâng cao hiệu quả hoạt động truyền thông Marketing tại ...Một số giải pháp nhằm nâng cao hiệu quả hoạt động truyền thông Marketing tại ...
Một số giải pháp nhằm nâng cao hiệu quả hoạt động truyền thông Marketing tại ...
 
Một số giải pháp nâng cao hiệu quả sử dụng vốn tại Công ty Cổ phần Thiết bị B...
Một số giải pháp nâng cao hiệu quả sử dụng vốn tại Công ty Cổ phần Thiết bị B...Một số giải pháp nâng cao hiệu quả sử dụng vốn tại Công ty Cổ phần Thiết bị B...
Một số giải pháp nâng cao hiệu quả sử dụng vốn tại Công ty Cổ phần Thiết bị B...
 

Recently uploaded

How to Fix the Import Error in the Odoo 17
How to Fix the Import Error in the Odoo 17How to Fix the Import Error in the Odoo 17
How to Fix the Import Error in the Odoo 17
Celine George
 
DRUGS AND ITS classification slide share
DRUGS AND ITS classification slide shareDRUGS AND ITS classification slide share
DRUGS AND ITS classification slide share
taiba qazi
 
Walmart Business+ and Spark Good for Nonprofits.pdf
Walmart Business+ and Spark Good for Nonprofits.pdfWalmart Business+ and Spark Good for Nonprofits.pdf
Walmart Business+ and Spark Good for Nonprofits.pdf
TechSoup
 
World environment day ppt For 5 June 2024
World environment day ppt For 5 June 2024World environment day ppt For 5 June 2024
World environment day ppt For 5 June 2024
ak6969907
 
Pride Month Slides 2024 David Douglas School District
Pride Month Slides 2024 David Douglas School DistrictPride Month Slides 2024 David Douglas School District
Pride Month Slides 2024 David Douglas School District
David Douglas School District
 
How to Build a Module in Odoo 17 Using the Scaffold Method
How to Build a Module in Odoo 17 Using the Scaffold MethodHow to Build a Module in Odoo 17 Using the Scaffold Method
How to Build a Module in Odoo 17 Using the Scaffold Method
Celine George
 
PCOS corelations and management through Ayurveda.
PCOS corelations and management through Ayurveda.PCOS corelations and management through Ayurveda.
PCOS corelations and management through Ayurveda.
Dr. Shivangi Singh Parihar
 
How to Manage Your Lost Opportunities in Odoo 17 CRM
How to Manage Your Lost Opportunities in Odoo 17 CRMHow to Manage Your Lost Opportunities in Odoo 17 CRM
How to Manage Your Lost Opportunities in Odoo 17 CRM
Celine George
 
MARY JANE WILSON, A “BOA MÃE” .
MARY JANE WILSON, A “BOA MÃE”           .MARY JANE WILSON, A “BOA MÃE”           .
MARY JANE WILSON, A “BOA MÃE” .
Colégio Santa Teresinha
 
S1-Introduction-Biopesticides in ICM.pptx
S1-Introduction-Biopesticides in ICM.pptxS1-Introduction-Biopesticides in ICM.pptx
S1-Introduction-Biopesticides in ICM.pptx
tarandeep35
 
LAND USE LAND COVER AND NDVI OF MIRZAPUR DISTRICT, UP
LAND USE LAND COVER AND NDVI OF MIRZAPUR DISTRICT, UPLAND USE LAND COVER AND NDVI OF MIRZAPUR DISTRICT, UP
LAND USE LAND COVER AND NDVI OF MIRZAPUR DISTRICT, UP
RAHUL
 
Azure Interview Questions and Answers PDF By ScholarHat
Azure Interview Questions and Answers PDF By ScholarHatAzure Interview Questions and Answers PDF By ScholarHat
Azure Interview Questions and Answers PDF By ScholarHat
Scholarhat
 
Film vocab for eal 3 students: Australia the movie
Film vocab for eal 3 students: Australia the movieFilm vocab for eal 3 students: Australia the movie
Film vocab for eal 3 students: Australia the movie
Nicholas Montgomery
 
How to Add Chatter in the odoo 17 ERP Module
How to Add Chatter in the odoo 17 ERP ModuleHow to Add Chatter in the odoo 17 ERP Module
How to Add Chatter in the odoo 17 ERP Module
Celine George
 
Advanced Java[Extra Concepts, Not Difficult].docx
Advanced Java[Extra Concepts, Not Difficult].docxAdvanced Java[Extra Concepts, Not Difficult].docx
Advanced Java[Extra Concepts, Not Difficult].docx
adhitya5119
 
The History of Stoke Newington Street Names
The History of Stoke Newington Street NamesThe History of Stoke Newington Street Names
The History of Stoke Newington Street Names
History of Stoke Newington
 
Chapter 4 - Islamic Financial Institutions in Malaysia.pptx
Chapter 4 - Islamic Financial Institutions in Malaysia.pptxChapter 4 - Islamic Financial Institutions in Malaysia.pptx
Chapter 4 - Islamic Financial Institutions in Malaysia.pptx
Mohd Adib Abd Muin, Senior Lecturer at Universiti Utara Malaysia
 
Hindi varnamala | hindi alphabet PPT.pdf
Hindi varnamala | hindi alphabet PPT.pdfHindi varnamala | hindi alphabet PPT.pdf
Hindi varnamala | hindi alphabet PPT.pdf
Dr. Mulla Adam Ali
 
How to Setup Warehouse & Location in Odoo 17 Inventory
How to Setup Warehouse & Location in Odoo 17 InventoryHow to Setup Warehouse & Location in Odoo 17 Inventory
How to Setup Warehouse & Location in Odoo 17 Inventory
Celine George
 
Exploiting Artificial Intelligence for Empowering Researchers and Faculty, In...
Exploiting Artificial Intelligence for Empowering Researchers and Faculty, In...Exploiting Artificial Intelligence for Empowering Researchers and Faculty, In...
Exploiting Artificial Intelligence for Empowering Researchers and Faculty, In...
Dr. Vinod Kumar Kanvaria
 

Recently uploaded (20)

How to Fix the Import Error in the Odoo 17
How to Fix the Import Error in the Odoo 17How to Fix the Import Error in the Odoo 17
How to Fix the Import Error in the Odoo 17
 
DRUGS AND ITS classification slide share
DRUGS AND ITS classification slide shareDRUGS AND ITS classification slide share
DRUGS AND ITS classification slide share
 
Walmart Business+ and Spark Good for Nonprofits.pdf
Walmart Business+ and Spark Good for Nonprofits.pdfWalmart Business+ and Spark Good for Nonprofits.pdf
Walmart Business+ and Spark Good for Nonprofits.pdf
 
World environment day ppt For 5 June 2024
World environment day ppt For 5 June 2024World environment day ppt For 5 June 2024
World environment day ppt For 5 June 2024
 
Pride Month Slides 2024 David Douglas School District
Pride Month Slides 2024 David Douglas School DistrictPride Month Slides 2024 David Douglas School District
Pride Month Slides 2024 David Douglas School District
 
How to Build a Module in Odoo 17 Using the Scaffold Method
How to Build a Module in Odoo 17 Using the Scaffold MethodHow to Build a Module in Odoo 17 Using the Scaffold Method
How to Build a Module in Odoo 17 Using the Scaffold Method
 
PCOS corelations and management through Ayurveda.
PCOS corelations and management through Ayurveda.PCOS corelations and management through Ayurveda.
PCOS corelations and management through Ayurveda.
 
How to Manage Your Lost Opportunities in Odoo 17 CRM
How to Manage Your Lost Opportunities in Odoo 17 CRMHow to Manage Your Lost Opportunities in Odoo 17 CRM
How to Manage Your Lost Opportunities in Odoo 17 CRM
 
MARY JANE WILSON, A “BOA MÃE” .
MARY JANE WILSON, A “BOA MÃE”           .MARY JANE WILSON, A “BOA MÃE”           .
MARY JANE WILSON, A “BOA MÃE” .
 
S1-Introduction-Biopesticides in ICM.pptx
S1-Introduction-Biopesticides in ICM.pptxS1-Introduction-Biopesticides in ICM.pptx
S1-Introduction-Biopesticides in ICM.pptx
 
LAND USE LAND COVER AND NDVI OF MIRZAPUR DISTRICT, UP
LAND USE LAND COVER AND NDVI OF MIRZAPUR DISTRICT, UPLAND USE LAND COVER AND NDVI OF MIRZAPUR DISTRICT, UP
LAND USE LAND COVER AND NDVI OF MIRZAPUR DISTRICT, UP
 
Azure Interview Questions and Answers PDF By ScholarHat
Azure Interview Questions and Answers PDF By ScholarHatAzure Interview Questions and Answers PDF By ScholarHat
Azure Interview Questions and Answers PDF By ScholarHat
 
Film vocab for eal 3 students: Australia the movie
Film vocab for eal 3 students: Australia the movieFilm vocab for eal 3 students: Australia the movie
Film vocab for eal 3 students: Australia the movie
 
How to Add Chatter in the odoo 17 ERP Module
How to Add Chatter in the odoo 17 ERP ModuleHow to Add Chatter in the odoo 17 ERP Module
How to Add Chatter in the odoo 17 ERP Module
 
Advanced Java[Extra Concepts, Not Difficult].docx
Advanced Java[Extra Concepts, Not Difficult].docxAdvanced Java[Extra Concepts, Not Difficult].docx
Advanced Java[Extra Concepts, Not Difficult].docx
 
The History of Stoke Newington Street Names
The History of Stoke Newington Street NamesThe History of Stoke Newington Street Names
The History of Stoke Newington Street Names
 
Chapter 4 - Islamic Financial Institutions in Malaysia.pptx
Chapter 4 - Islamic Financial Institutions in Malaysia.pptxChapter 4 - Islamic Financial Institutions in Malaysia.pptx
Chapter 4 - Islamic Financial Institutions in Malaysia.pptx
 
Hindi varnamala | hindi alphabet PPT.pdf
Hindi varnamala | hindi alphabet PPT.pdfHindi varnamala | hindi alphabet PPT.pdf
Hindi varnamala | hindi alphabet PPT.pdf
 
How to Setup Warehouse & Location in Odoo 17 Inventory
How to Setup Warehouse & Location in Odoo 17 InventoryHow to Setup Warehouse & Location in Odoo 17 Inventory
How to Setup Warehouse & Location in Odoo 17 Inventory
 
Exploiting Artificial Intelligence for Empowering Researchers and Faculty, In...
Exploiting Artificial Intelligence for Empowering Researchers and Faculty, In...Exploiting Artificial Intelligence for Empowering Researchers and Faculty, In...
Exploiting Artificial Intelligence for Empowering Researchers and Faculty, In...
 

Localized hippocampal glutamine synthetase knockout a novel model of mesial temporal lobe epilepsy

  • 1. Yale University EliScholar – A Digital Platform for Scholarly Publishing at Yale Yale Medicine Thesis Digital Library School of Medicine January 2019 Localized Hippocampal Glutamine Synthetase Knockout: A Novel Model Of Mesial Temporal Lobe Epilepsy Maxwell Gerard Farina Follow this and additional works at: https://elischolar.library.yale.edu/ymtdl This Open Access Thesis is brought to you for free and open access by the School of Medicine at EliScholar – A Digital Platform for Scholarly Publishing at Yale. It has been accepted for inclusion in Yale Medicine Thesis Digital Library by an authorized administrator of EliScholar – A Digital Platform for Scholarly Publishing at Yale. For more information, please contact elischolar@yale.edu. Recommended Citation Farina, Maxwell Gerard, "Localized Hippocampal Glutamine Synthetase Knockout: A Novel Model Of Mesial Temporal Lobe Epilepsy" (2019). Yale Medicine Thesis Digital Library. 3491. https://elischolar.library.yale.edu/ymtdl/3491
  • 2. Localized Hippocampal Glutamine Synthetase Knockout: a Novel Model of Mesial Temporal Lobe Epilepsy a thesis submitted to the Yale University School of Medicine in partial fulfillment for the degree of Doctor of Medicine Maxwell G. Farina Advisor: Tore Eid, MD, PhD Thesis Committee Members: Peter Tattersall, PhD, Nihal DeLanerolle, DPhil, DSc & Ellen Foxman, MD, PhD May 2019
  • 3. Abstract LOCALIZED HIPPOCAMPAL GLUTAMINE SYNTHETASE KNOCKOUT: A NOVEL MODEL OF MESIAL TEMPORAL LOBE EPILEPSY. Maxwell Farina and Tore Eid. Departments of Laboratory Medicine and Neurosurgery, Yale University, School of Medicine, New Haven, CT. The purpose of this study was to create and optimize a model of mesial temporal lobe epilepsy through selective depletion of glutamine synthetase (GS) in the mouse hippocampus. Following validation of the model, preliminary studies attempted to characterize morphological astrocytic and synaptic changes that re- sult from GS deficiency. Aim 1 established a novel mouse model of GS knockout in hippocampal astrocytes. Aim 2 tested whether localized hippocampal knockout of GS causes mice to exhibit an epilepsy-like phe- notype. Aim 3 characterized the cellular effects of localized GS loss. To generate the knockout, Glul-floxed C57BL/6J mice were injected with four different adeno-associated viral vectors containing Cre-recombinase expression cassettes. Mice were also implanted with intracranial depth or screw electrodes and monitored for spontaneous seizures using 24-hour video-EEG recording for two weeks. To assess for provoked seizure sensitivity, seizures were induced with pentylenetetrazol (PTZ) prior to perfusion fixation. Brains were per- fused, sectioned, and immunostained for analysis using standard and STED fluorescence microscopy. Knock- out of GS, as evidenced by loss of GS immunoreactivity, was found over a greater area in brain regions in- jected with the AAV5 CMV and AAV8 GFAP serotypes. In addition, within each GS knockout region, AAV8 GFAP exhibited a significantly greater efficiency of knockdown compared to AAV5 CMV Legacy and AAV8 CMV (83.1% decreased fluorescence intensity, p=0.0003) and compared to AAV5 CMV (20.2% decreased fluorescence intensity, p=0.018). AAV8 GFAP exhibited near perfect target specificity (98.7% of GFP+ cells were astrocytes), while AAV5 CMV Legacy, AAV5 CMV, and AAV8 CMV targeted mostly neu- rons with varied degrees astrocyte labeling detected (10.0%, 21.3%, and 12.7% astrocytes, respectively. Sixty percent (3/5) of mice injected with AAV8 GFAP exhibited an epilepsy-like phenotype including sponta- neous recurrent seizures that were clustered in the morning hours. Twenty-five percent (1/4) of control mice seized spontaneously over the same period. Additionally, focal GS knockout mice demonstrated significantly lower time to initial clonic twitch following PTZ administration compared to control mice (mean ± SEM: 41.2 ± 3.2 seconds vs. 65.83 ± 12.9 seconds, respectively; p=0.044). The effect on time to convulsive seizure was not statistically significant, though there was a trend of knockout animals proceeding to convulsions in less time (74.2 ± 9.4 seconds vs. 100.0 ± 18.0 seconds, p=0.20). Finally, examination of synaptic mark- ers revealed decreased expression of PSD-95 surrounding GS- astrocytes compared to GS+ astrocytes, with sampled relative intensity of 0.57 ± 0.04 (p=0.002). Relative intensity (RI) of synaptophysin and gephyrin appeared to be unchanged in the sampled areas (synaptophysin RI 0.94 ± 0.15, p=0.87; gephyrin RI 0.94 ± 0.04, p=0.23). In this study, we created a novel model of mesial temporal lobe epilepsy by selectively knock- ing out GS in the hippocampal astrocytes of mice. Development of this monogenetic knockout model with effects restricted to the hippocampus and adjacent structures has the potential to more fully elucidate the impact of GS loss in this treatment-resistant disease. Initial examination of synaptic markers in GS depleted areas highlights the importance of glutamatergic synaptic transmission in epilepsy pathology. ii
  • 4. Acknowledgments This work was made possible by the knowledge, skills, and support conferred by countless friends and mentors through the years. In particular, I am thankful to Tore Eid, who exemplifies selfless mentorship; my father, who taught me the importance of ask- ing questions; and my mother, who volunteered to help my fourth-grade science class dissect a shark, and in doing so, instilled in me an unending love for science, its system- atic search for truth, and the uniquely fundamental way in which it connects us to one another. Financial support for the work presented in this thesis was provided in part by the Na- tional Heart, Lung, and Blood Institute of the National Institutes of Health (Maxwell Farina, award number T35HL007649) and NIH grant S10-OD020142 (Yale Confocal Mi- croscopy Core). iii
  • 5. Abbreviations AAV Adeno-associated virus AMPA α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid ATP Adenosine triphosphate CA 1-3 Cornu ammonis subfields 1-3 of the hippocampus CMV Cytomegalovirus CNS Central nervous system DH Dentate hilus of the hippocampus EAAT Excitatory amino acid transporters EC Entorhinal cortex GABA γ-aminobutyric acid GFAP Glial fibrillary acid protein GFP Green fluorescent protein GLUL Glutamine ammonia ligase Gr Granule cell layer of the dentate gyrus GS Glutamine synthetase LV Lateral ventricle Mol Molecular layer of the dentate gyrus MSO Methionine sulfoximine MTLE Mesial temporal lobe epilepsy NMDA n-methyl-D-aspartate PTZ Pentylenetetrazol SUB Subiculum TCA Tricarboxylic acid iv
  • 6. Contents Introduction 2 Mesial Temporal Lobe Epilepsy . . . . . . . . . . . . . . . . . . . . . . . . . . 2 Glutamine Synthetase . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5 Chemical Models of Mesial Temporal Lobe Epilepsy . . . . . . . . . . . . . . . 9 Genetic Models of Glutamine Synthetase Depletion . . . . . . . . . . . . . . . 12 Specific Aims 17 Methods 18 Animals and Reagents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18 Surgery: Viral Injection and Electrode Implantation . . . . . . . . . . . . . . . . 20 Video-EEG Monitoring and Seizure Precipitation Studies . . . . . . . . . . . . 23 Fixation, Immunofluorescence, and Microscopy . . . . . . . . . . . . . . . . . . 23 Statistical Analysis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24 Results 25 Knockout of Glutamine Synthetase . . . . . . . . . . . . . . . . . . . . . . . . 25 Seizure Findings . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 29 Synaptic Changes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32 Discussion 34 References 38 1
  • 7. Introduction Epilepsy is a chronic neurological disorder characterized by recurrent, unprovoked seizures; i.e. sudden and transient episodes of abnormal electrical brain activity that result in a change in clinical state. The clinical presentation of epilepsy is widely varied, as seizure activity can take many forms including staring, unresponsiveness, stereotyped movements, loss of muscle tone, stiffness, and limb-jerking. Likely due in part to its potentially dra- matic appearance, epilepsy is one of the oldest recognized health conditions, with extensive descriptions originating on Babylonian cuneiform tablets dating to 4000 BC [1]. Today, it is estimated that epilepsy affects approximately 2% of the worldwide population [2]. Of these cases, about one-third are refractory (i.e., inadequately controlled by ≥2 appropriately selected antiepileptic drugs) [3]; some forms of epilepsy are refractory at much higher rates. Indeed, there are dozens of “epilepsy syndromes,” each of which is characterized by consis- tently occurring seizure type, age of onset, electroencephalographic findings, precipitating factors, genetic markers, clinical course, prognosis, and response to pharmacotherapy. Mesial Temporal Lobe Epilepsy Of the epilepsy syndromes, mesial temporal lobe epilepsy (MLTE) is the most common treatment resistant variant in adults [4]. It is estimated that 70% of MTLE patients are in- adequately controlled with medication alone [2]. Detailed epidemiological data on MTLE is scarce, as an incomplete understanding of disease pathophysiology has led to imprecise
  • 8. INTRODUCTION 3 nomenclature and a lack of consensus on diagnostic criteria [5]. However, a recent meta- analysis inferred an incidence of 8.9 cases per 100,000 people per year, a prevalence of 1.9 cases per 1000 people, and an estimated patient population of 615,600 individuals in the United States alone [6]. MTLE most commonly starts before the age of 18 and gradually increases in intensity and pharmacoresistance over time [7]. Patients often have a past med- ical history that includes childhood febrile convulsions. While the causes of MTLE remain to be elucidated, the most widely accepted theory posits that these childhood febrile con- vulsions or other predisposing injuries act as an early insult that culminates in hippocampal damage and eventual development of MTLE [5]. Though this theory, initially posited by Meyer in 1954, serves as a useful framework for understanding MTLE, it is important to highlight that a large number of MTLE patients have no history of predisposing injury that precedes onset of the epileptic syndrome. The clinical presentation of MTLE is varied, as the entity referred to as MTLE is widely believed to be a heterogeneous collection of different pathophysiologies; however, cer- tain patterns appear to be more characteristic of MTLE patients. MTLE seizure episodes often begin with a vegetative aura, often described as “an epigastric or substernal rising sensation,” [5]. Other common aura symptoms include a sudden sense of fear, delusions, hallucinations, and olfactory or gustatory sensations [7]. As the complex partial seizure be- gins, behavioral arrest and staring occur. Next follow automatisms including lip smacking and chewing, and while motor symptoms are less common in MTLE, dystonic postur- ing of the contralateral arm does occur and is useful as a lateralizing feature [5]. The com- plex partial seizure typically continues for 1-2 minutes and can include head deviation, and clonic-tonic activity, uncommonly culminating in convulsion. While the postictal period is variable, it is not uncommon for patients to exhibit significant confusion and (in the case of dominant temporal lobe onset) several minutes of postictal aphasia. Postictal memory impairment can also occur in MTLE, sometimes rendering the patient amnestic for several hours despite apparently normal behavior.
  • 9. INTRODUCTION 4 Epilepsy syndromes are largely classified by EEG patterns. Though neither EEG find- ings nor clinical presentation are pathognomonic for MTLE, interictal scalp electroen- cephalogram often demonstrates temporal-lobe spike and sharp waves and focal slowing [5]. Hippocampal sclerosis – the atrophy and scarring of the hippocampus – often cited as the hallmark of MTLE, is also difficult to recognize using magnetic resonance imaging, as these changes are often subtle and bilateral, interfering with the qualitative asymmetry re- quired to identify the pathology. All of these factors make definitive diagnosis of MTLE challenging. Untreated MTLE is often described as a chronic, progressive disorder in which seizures increase in duration and intensity over time [5]. While MTLE is typically very respon- sive to pharmacotherapy at the onset of disease, seizures frequently become pharmacoresis- tant by early adulthood. In addition to often debilitating seizures, patients are commonly plagued by additional long-term sequelae of MTLE including global cognitive dysfunction, impaired episodic memory recall, poor working memory performance, executive dysfunc- tion, impaired task-switching, decreased alertness, and difficulties with language and word retrieval [8]. MTLE patients also exhibit features of depression, anxiety, and obsessive- compulsive disorder with greater frequency than the general population [9]. In particular, depression is a widely known comorbidity of temporal lobe epilepsy, with an incidence of 30% and prevalence of 50% among MTLE patients [10]. Indeed, suicide is the leading cause of death in patients with refractory MTLE; one study found that of the deaths of en- rolled MTLE patients during a 9-year follow up, 50% were suicides [11]. Suicidal ideation is a major driver of the finding that MTLE patients had a standardized mortality ratio of 4.86. The pathogenesis of MTLE is poorly understood. Broadly, based on our understand- ing of neuronal function, it can be inferred that epilepsy is caused by a departure from the homeostatic balance of excitatory and inhibitory forces in human neuronal networks that predisposes neurons to inappropriate synchronous excitation [5]. This imbalance can re-
  • 10. INTRODUCTION 5 sult from a preponderance of excitatory signaling or a paucity of inhibitory signaling, and in reality, MTLE is likely caused by a complex combination of both of these (and other) factors. Investigation of excitatory pathways reveals several etiological hypotheses includ- ing mossy fiber sprouting which forms new recurrent excitatory connections in the den- tate gyrus [12], increased expression of certain voltage-gated sodium channels [13], and in- creased expression of the AMPA and NMDA glutamate receptors [14]. Hypotheses regard- ing compromise of balancing inhibitory signaling include loss of hippocampal interneu- rons [15], shortened duration of inhibitory GABAA synaptic potentials [16], and lack of GABAB-mediated use-dependent synaptic depression [17]. Potential extra-neuronal eti- ologies include impairment of the blood-brain-barrier [18], shifts in hormonal neuromod- ulators [19], and a host of astrocyte-related changes in neurotransmitter metabolism, ion redistribution, and direct synaptic interaction [2]. In short, the pathophysiology of MTLE is staggeringly complex and likely represents a multifactorial system which spans dozens of cell types, receptors, and molecules. While recent years have brought a vast amount of progress in the understanding of these various pathways, much work remains. A large number of MTLE cases are poorly controlled, and there have been no new epilepsy drug therapies developed in the last decade. Further work on animal models of MTLE and cor- relation to human disease will be crucial to categorizing the various subtypes of temporal lobe epilepsy, identifying their most relevant and high-impact target pathways, and design- ing molecules and interventions that improve the symptomology and long-term survival of patients who suffer from MTLE. Glutamine Synthetase In 1993, During and Spencer reported that extracellular hippocampal glutamate levels were elevated in MTLE patients not only following complex partial seizures but also preceding any electroencephalographic or clinical evidence of seizure [20]. Such human evidence,
  • 11. INTRODUCTION 6 taken with the findings that administration of glutamate analogues in animal models is suf- ficient to trigger seizures [21], implicates high levels of extracellular glutamate as a pos- sible causative factor in epilepsy. A subsequent study utilizing magnetic resonance spec- troscopy found that epileptic hippocampus contains both increased glutamate levels and decreased glutamine levels [22]. These findings pointed Eid and colleagues towards poten- tial involvement of glutamine synthetase, an enzyme found in astrocytes that functions to convert excitatory neurotransmitter glutamate into glutamine. In an effort to understand the origin of the pathological metabolic disturbances, Eid and colleagues examined tis- sue levels of glutamine synthetase in the human hippocampus, and in 2004, they reported that compared to control tissues, glutamine synthetase expression and enzymatic activity were reduced by 40% and 38%, respectively, in MTLE [23]. The prospect of identifying a well-characterized enzymatic defect as a contributor of seizure generation has significant implications for therapeutic target identification, thus glutamine synthetase has become an important subject of investigation in recent years. Glutamine synthetase (GS) is an enzyme encoded by the strikingly ubiquitous gluta- mate ammonia ligase (Glul) gene, which is found in all known living species. The eukary- otic variant of GS is structured as two adjacent pentameric rings consisting of ten identical subunits [24], and the enzyme serves as a catalyst of the ATP-dependent synthesis of glu- tamine from substrates glutamate and ammonia. In mammals, GS expression is limited to specific tissues: liver, kidney, pancreas, adipose tissue, skin, and the central nervous system (CNS) [25]. Within the CNS, GS is exclusively expressed in astrocytes [26], emphasizing the indispensable role that astrocytes play in the maintenance of the homeostatic cellular and metabolic environment of the CNS. This astrocytic regulation of extracellular metabolite levels—particularly in proximity to the synapse—is most relevant to epileptogenesis through the glutamate-glutamine cycle. In a recent textbook chapter, Eid et. al., succinctly describe the glutamate-glutamine cycle in the following steps:
  • 12. INTRODUCTION 7 1. Exocytosis of glutamate from axon terminals into the synaptic cleft. 2. Binding of glutamate to postsynaptic glutamate receptors. 3. Diffusion of glutamate away from the synapse with possible binding to extrasynaptic receptors and uptake into astrocytes or neurons via high-affinity excitatory amino acid transporters (EAATs). 4. Conversion of glutamate and ammonia to glutamine in the astroglial cytoplasm via the enzyme glutamine synthetase. 5. Shuttling of glutamine to neurons via glutamine transporters. 6. Conversion of glutamine to glutamate and ammonia via the enzyme glutaminase. 7. Concentration of glutamate in synaptic vesicles via vesicular glutamate transporters. [25] As is evident in Figure 1, one astrocytic function is to remove glutamate – the most abundant neurotransmitter in the vertebrate nervous system [27] – from the synaptic cleft. This astrocytic intake of synaptic glutamate serves two purposes: 1) ensuring a precise post- synaptic glutamate response (and modulation of time course, pattern, and extent of said response) [28], and 2) prevention of the neuronal death that occurs in the presence of el- evated glutamate levels for extended periods (a phenomenon referred to as excitotoxicity) [29]. Once inside the astrocyte, glutamate is degraded by a variety of pathways including conversion to glutathione and metabolism by the TCA cycle (into lactate, aspartate, or recycled glutamate), though it appears that the largest fraction ( 50%) is converted to glu- tamine by GS [30]. Finally, the astrocyte exports glutamine for use by neurons, which can easily convert glutamine to functional glutamate via glutaminase. Glutamine is a preferable metabolite for extracellular transport, as it is not associated with the same excitotoxicity phenomenon as glutamate.
  • 13. INTRODUCTION 8 Figure 1. The Glutamate- Glutamine cycle. Image from Eid, T., et al. (2016). ”The Glutamate-Glutamine Cycle in Epilepsy.” Adv Neu- robiol 13: 351-400. Genetic cases of GS loss in humans are extremely rare, though findings from these pa- tients support the importance of GS in healthy CNS function. Of the three noted cases of homozygous Glul mutation, all patients developed severe epileptic encephalopathy, and two of the thee experienced fatal multi-organ failure and resulting neonatal death [31, 32]. Individuals with Alzheimer’s dementia have also been found to have reduced expression of GS [33], and abnormalities of tissue GS levels have been associated with many other condi- tions including amyotrophic lateral sclerosis [34], Huntington’s disease [35], depression [36], schizophrenia [37], and suicidal ideation [38]. Given the multitude of studies linking GS to human disease as well as the enzyme’s compelling biochemical relationship to CNS health and functional neurotransmission, much attention has been placed on further elucidation of the specific role GS has in epileptogenesis, seizure initiation, and seizure propagation. However, human studies are often limited in sample size, access to relevant tissues, and, of course, interventional scope. Thus, animal models of epilepsy have been and remain crucial to the experimental process of determining the precise relationship between glutamine synthetase and human disease.
  • 14. INTRODUCTION 9 Chemical Models of Mesial Temporal Lobe Epilepsy For over 50 years, animal models have provided insight into the mechanisms of epilepsy and formed the foundation for discovering novel anti-epileptic drugs found in the clinic today [39]. However, this suite of pharmacotherapies continues to be largely ineffective in the treatment of refractory epilepsies—particularly MTLE. Some hypothesize that the lack of advancement in MTLE treatment stems from fundamental differences between fea- tures of epilepsy animal models in widespread use and the features specific to MTLE [40]. Staying cognizant of this important observation, the following discussion will focus on the most common models with high relevance to MTLE specifically. In addition, it is impor- tant to note that there are a vast number of other, non-chemical animal models of epilepsy which will not be discussed here. While models of induction such as traumatic brain in- jury, ischemic brain damage, and hyperthermia are undoubtedly useful, their widespread and often non-specific effects complicate the study of complex, interconnected biochemi- cal pathways [41]. One of the earliest examples of an MTLE model was first described by Ben-Ari and Lagowska, who injected kainic acid into the unilateral rat amygdala [42]. Kainic acid is a cyclic analog of glutamate that produces excitatory post-synaptic potentials upon binding as an agonist to the ionotropic kainate receptor and as a partial agonist to AMPA receptors [43]. Kainic acid can be administered locally as an injection to the desired brain region or via systemic routes including subcutaneous, intraperitoneal, and intravenous [44]. While systemic administration certainly simplifies the experimental procedure, it also carries sev- eral disadvantages compared to direct CNS delivery: 1) extrahippocampal neuronal damage is far more extensive than found in human MTLE, 2) unlike the typically unilateral hip- pocampal damage seen in MTLE, systemic kainic acid damages both hippocampi, and 3) animal mortality is high due to the systemic effects of kainic acid [45]. In contrast, intracra- nial administration of kainic acid largely avoids these shortcomings, and in particular, in- trahippocampal administration in mice closely resembles human MTLE both in terms of
  • 15. INTRODUCTION 10 histological findings and seizure patterns. Following unilateral injection of 0.2 µg of kainic acid into the dorsal hippocampus, mice enter status epilepticus which is typically noncon- vulsive [46]. Following a two week latency period, mice enter a chronic epileptic phase in which EEG demonstrates isolated high-voltage sharp waves and hippocampal paroxysmal discharges which are restricted to the ipsilateral hippocampus [47]. In the study by Riban et. al., all mice developed spontaneous chronic seizures, and approximately half of the co- hort experienced secondary generalization. Histopathological findings in this model are highly consistent with findings in human MTLE including neuronal loss in CA1 and CA3 subfields and dentate hilus as well as reactive gliosis predominantly restricted to the ipsilat- eral hippocampus [46, 47]. Pilocarpine, a cholinergic muscarinic agonist, was first used as a rodent epileptogenic shortly after kainic acid [48]. As seen with kainic acid, pilocarpine can be administered through a variety of systemic and intracerebral routes, however administration via intrahip- pocampal injection results in less extra-hippocampal damage, lower subject mortality, more consistent induction of initial status epilepticus, and more prompt recovery from status epilepticus [49]. In a large study, following unilateral injection of 2.4 µg of pilocarpine into the hippocampus, 76% of animals experienced status epilepticus, which took a form similar to the initial status epilepticus in the kainic acid model [50]. After a period of la- tency averaging two weeks (range 2-30 days), 71% of animals demonstrated spontaneous recurring seizures. In the pilocarpine model, abnormal EEG discharges typically begin in the hippocampus and are associated with orofacial automatisms prior to development of head and forelimb clonus, rearing, and falling [51]. As in human MTLE progression, seizure frequency increases over time in the pilocarpine model [52]. Upon neuropatho- logical examination, findings include neuronal loss in CA1, CA3, and dentate hilus, as well as mossy fiber sprouting in the dentate gyrus [51]. Though extra-hippocampal damage is more limited in local injection compared to systemic administration, the hippocampal injection technique does yield some neurodegeneration in the cortex, thalamus, and amyg-
  • 16. INTRODUCTION 11 dala [49]. The kainic acid and pilocarpine models induce neuronal damage in similar re- gions, however the timing of such damage is quite different, as pilocarpine induced neural loss is fast-appearing, whereas neurodegeneration is delayed following administration of kainic acid [53]. The kainic acid and pilocarpine models of epilepsy have maintained their relevance for decades, and they remain standards for the investigation of MTLE. However, understand- ing the exact relevance of GS in epileptogenesis requires more specific methods of epilepsy induction, particularly when attempting to understand whether GS plays a causative role in the disease. In an effort to determine the direct effects of GS loss in vivo, in 2008, Eid and colleagues infused methionine sulfoximine (MSO) into the hippocampi of otherwise normal rats [54]. MSO, an irreversible glutamine synthetase inhibitor, is first phosphory- lated by and then irreversibly binds to the enzyme [55]. Ten days after initiation of contin- uous MSO infusion into the unilateral hippocampus, GS activity was found to be signifi- cantly reduced compared to saline infused controls (82% reduction in low-dose MSO [.625 µg/hr] and 97% reduction in high-dose MSO [2.5 µg/hr]) [54]. Interestingly, GS activity in the contralateral hippocampus was significantly reduced in the high-dose animals but was unchanged in low-dose animals; drug diffusion effects may explain this finding. Remarkably, 98% of MSO-infused animals were noted to have recurrent behavioral seizures, whereas none of the saline infused animals seized [54]. This finding is the first compelling evidence suggesting that GS depletion is sufficient to cause epileptogenesis. Further, the seizure patterns seen in the MSO-infused animals mimicked patterns seen in MTLE patients: 1) seizures were found to be temporally clustered, 2) EEG suggested a mild degree of secondary generalization (i.e., mean 5.3 depth-electrode detected seizures per day vs. mean 1.0 epidural electrode detected seizures per day), and 3) seizure behaviors included immobilization, chewing and whisker twitching, forelimb clonus, and rearing and falling. Upon histological examination of brain sections, MSO-infused animals demon- strated dose-dependent neuronal loss in the hippocampus and adjacent structures. A subset
  • 17. INTRODUCTION 12 of animals showed MTLE-like pathology including hippocampal shrinkage, CA1-CA3 neuronal loss, and relative sparing of neurons in the subiculum and granule cell layer of the dentate gyrus. Interestingly, many animals that received the low dose of MSO exhibited minimal neuronal loss despite extensive seizure activity, and the authors suggest that such a result may indicate that GS depletion is more relevant to MTLE development than the often-discussed mesial temporal sclerosis pattern. The aforementioned MSO study has highlighted the relationship between GS and MTLE as a crucial realm of investigation. Indeed, subsequent experiments have remon- strated the model’s validity [56], addressed new areas of MTLE pathology [57], and as- sessed the relevance of therapeutic interventions [58]. However, as with almost all chemical agents, selectivity and off-target effects must be discussed. MSO is known to deplete tissue glutathione [59], increase astrocyte glycogen [60], and activate neurons independent of its action on GS [61]. While the 2008 MSO study accounted for glutathione depletion, and found that there was no discernible effect on tissue glutathione in the low-dose MSO an- imals [54], the control of other off-target effects of MSO poses a significant challenge. In order to truly verify and further study the effects of specific absence of GS alone, genetic approaches to GS depletion must be undertaken. Genetic Models of Glutamine Synthetase Depletion Since the first use of the Cre-lox recombination system in mammalian cells in 1988 [62], this remarkable technology has gained widespread acceptance and validation as a tool for the selective deletion of genes in certain model organisms. Following the establishment of the NIH’s Neuroscience Research Cre-driver projects [63], the first model of Cre-lox mediated GS knockout was reported in 2007 [64]. The researchers found that in a whole- body (i.e., non-conditional) knockout of GS, the mice were not viable; embryo death oc- curred when the embryo migrates from the oviduct into the uterus (embryonic day 3.5).
  • 18. INTRODUCTION 13 This finding was largely unsurprising, as GS plays many crucial functions—both synthetic and detoxifying—in several tissues throughout the body. In order to improve viability of the mice and further study the effects of GS knockout, a follow-up experiment from the same group restricted Cre expression to GFAP+ cells, thus limiting the GS knockout to CNS astrocytes [65]. These mice with CNS knockout of GS were reported to be the first viable animal model of genetic deletion of GS, however, even this more limited knockout model resulted in early death (approximately post-natal day 3) that precluded the study of potential epileptogenesis. Very recently, Zhou et. al., reported creation of a novel GS knockout that was further restricted, yielding viable mice that seize spontaneously [66]. By taking advantage of the Emx1-Cre mouse line described previously [67], GS knockout was restricted to astrocytes in the hippocampus and neocortex. These Emx1-Cre GS knockout mice were found to exhibit recurrent spontaneous seizures, thus establishing the method as a tool for studying epileptogenesis; indeed, the preliminary study itself shed light on several aspects of MTLE pathology. Perhaps most notably, the researchers found that following deletion of GS, changes in brain biochemistry as well as glial and vascular morphology were present for several weeks prior to the development of recurrent seizures. This temporal dissociation in a GS-specific epilepsy model provides strong evidence that GS-related epileptogenesis is unlikely to be mediated by an excitotoxic mechanism, and instead, is related to “a patho- logical process involving reactive astrocytes and impaired neurovascular coupling,” [66]. The Emx1-Cre GS knockout is certainly the most precise model of GS-mediated MTLE to date. In addition, the high level of similarity between Emx1-Cre GS knock- out and human MTLE tissue pathology and phenotype provides strong validation to the gliopathy hypothesis of epileptogenesis [68]. However, the most notable limitation of the Emx1-Cre knockout model is the overly-widespread distribution of GS knockout. Tis- sue specific Cre-recombinase expression is restricted by the viability and availability of transgenic mice with Cre expression driven by a particular gene of interest (e.g., GFAP or
  • 19. INTRODUCTION 14 Emx1, as described above). Further spatial specificity of Cre expression is only feasible by alternate techniques. In the Emx1-Cre GS knockout model, the observed strong deletion of GS in the neocortex, while expected, is not representative of human MTLE, in which GS depletion is typically present in the hippocampus and amygdala [23]. In fact, assessment of human MTLE neocortex has revealed that GS activity appears to be unchanged in this region [69]. In addition, tissue-specific Cre expression systems do not allow for lateraliza- tion of the desired knockout, and the bilateral GS loss found in the Emx1-Cre model can- not be refined to more closely approximate the unilateral pathology often found in MTLE. In order to overcome these limitations of tissue-specific Cre-driven knockout, our goal was to generate GS-knockout mice using local delivery of Cre via a direct injection of Cre-expressing viral vector. The recombination of loxP sites driven by Cre derived from a vector rather than from crossing with a Cre-expressing mouse line was first described in 1995, when an adenovirus was used to transfect cells in vitro in an effort to prove that such extrinsic methods of delivery were capable of inducing recombination of a floxed gene [70]. Early studies using the technique faced substantial complications due to the immunogenicity of adenovirus and other vectors, however in 2002, a newly engineered adeno-associated virus (AAV) was used successfully in vivo to express the reporter gene green fluorescent protein (GFP) in the mouse brain [71]. AAVs and lentivirus vectors are unique in that they both have been engineered to remove all viral genes, thus rendering them minimally immunogenic and virtually nontoxic [72, 73]. While many viral vectors rely on active cell division in order to incorporate vector DNA into the host genome and sustain protein production, AAVs and lentiviruses have the ability to transduce both divid- ing and non-dividing cells, making them ideal candidates for transduction of the CNS, in which many cell populations are non-dividing. AAVs retain this ability by maintaining the viral genome episomally within the host cell [74], whereas lentiviruses have the unique ca- pability of entering the nucleus in order to incorporate the plasmid in the absence of cell division [75]. One of the most significant points of differentiation between lentiviruses and
  • 20. INTRODUCTION 15 AAVs is their capsid size, as lentivirus has a typical diameter of 100nm and AAVs have a diameter of approximately 20nm. Indeed, this substantial size difference has implications for vector diffusibility through tissue following injection; the smaller AAV demonstrates 3- to 5-fold greater viral spread than comparable lentivirus injections [76, 77]. The increased diffusibility of AAVs come at the expense of payload, as more complex or multi-genetic expression cassettes are not capable of fitting within the smaller AAV genome of approxi- mately 4.7 kbp [78]. Given the relatively small size of the Cre and reporter GFP expression cassettes, the im- portance of maximum viral spread from injection site, and the extensive customizability of AAV packages, we elected to utilize AAVs in this study. However, within the AAV family, There are numerous catalogued AAV serotypes, each of which has a unique capsid struc- ture and surface proteins that influence cellular tropism and specificity [79]. In a highly exhaustive review of transduction characteristics of the six most common AAV serotypes currently used, Aschauer, Kreuz, and Rumpel present useful head-to-head comparisons in efficiency of transgene expression, cell-type specificity, impact on microglia, and capabil- ity for retrograde axonal transport [80]. Among the findings presented was evidence that serotypes AAV5 and AAV8 were both highly effective in transduction of astrocytes. When examined specifically, background-scaled reporter fluorescence of hippocampal astrocytes was approximately 10-fold greater in AAV8 transduced cells compared to AAV5 transduced cells, though both serotypes performed well compared to most other AAV types. As this extensive dataset of serotype comparisons was conducted entirely with expression cassettes driven by CMV promoters, we selected three of our four investigational viruses to utilize CMV as well. In addition, given research suggesting that AAVs utilizing the intermedi- ate filament glial fibrillary acidic protein (GFAP) improve selectivity of astrocytes and may also improve transgene expression [81], we also elected to include an AAV8 serotype with GFAP promoters. By comparing the knockdown efficiency of what appear to be four of the most effective astrocytic transducers, we aim to identify the optimal vehicle for knock-
  • 21. INTRODUCTION 16 down of GS in the mouse hippocampus via a methodology that allows for the spatial and temporal control required for precise emulation of human MTLE.
  • 22. Specific Aims The purpose of this study was to create and optimize a model of mesial temporal lobe epilepsy through selective depletion of glutamine synthetase (GS) in the mouse hippocam- pus. In addition, preliminary studies characterized morphological astrocytic and synaptic changes that result from the GS deficiency. The goal of Aim 1 was to establish a novel mouse model of glutamine synthetase (GS) knockout in hippocampal astrocytes. Cohorts of homozygous GS-floxed C57BL/6 mice were injected with Cre-expressing virus into the bilateral dentate gyri, subiculi, and en- torhinal cortices. Each cohort was injected with a virus of different serotype and promoter sequence in order to determine which virus maximally depletes GS at the injection sites. The goal of Aim 2 was to test whether localized hippocampal knockout of GS causes mice to exhibit an epilepsy-like phenotype. Knockout and control mice were monitored for severity and frequency of seizures using 24-hour video EEG recordings, and prior to sacrifice, seizure proclivity was measured during a pentylenetetrazol injection study. Such studies evaluated hippocampal GS knockout as a useful model of human epilepsy. The goal of Aim 3 was to characterize the cellular effects of local GS loss. Given GS’s role in converting glutamate to glutamine, proteins such as gephyrin (GABA synapse marker), PSD-95 (glutamate synapse marker), and synaptophysin (global synapse marker) are hypothesized to display altered tissue distribution. We will examine this using stimu- lated emission depletion confocal microscopy. Mice created in Aim 1 allow us to identify downstream effects of GS knockout and better characterize the process of epileptogenesis.
  • 23. Methods MGF performed all procedures described below with the exception of the preparation of GS recombineering vector. Roni Dhaher assisted with seizure precipitation studies. Some of the surgeries were performed by Mani Sandhu. Animals and Reagents All animal care and use procedures were approved by the Institutional Animal Care and Use Committee of Yale University, and experiments were performed in accordance with current guidelines. Care was taken to avoid suffering and minimize the number of exper- imental animals required for the study. Mice were housed on a 12-hour light/dark cycle in individually ventilated cages at constant temperature (22 ± 0.7 °C) and humidity (56 ± 6%) and were fed with Harlan Teklad 2018 (Harlan Laboratories Inc., Indianapolis, IN, USA) with access to food and water ad libitum. Biopsies from ear or tail were collected for determining genotype. In order to create a conditional knockout of GS, male and female C57BL/6J Glulf/f mice were generated (see Figure 2). GS targeting vector was prepared by recombineer- ing as described by Lee et. al., [82]. Briefly, approximately 12 kb of Glul genomic frag- ment containing the entire Glul sequence was retrieved from the bacterial artificial chro- mosome (BAC) clone, RP24-326N10 (obtained from the BACPAC Resources Center at the Children’s Hospital Oakland Research Institute, Oakland, CA) by gap repair. The 5’LoxP site was inserted in intron 1 approximately 628 nucleotides 5’ of exon 2, and the
  • 24. METHODS 19 Figure 2. Conditional knock- out generation. Selective deletion of the Glul gene in the mesial temporal lobe was achieved by injecting Cre- expressing virus directly into the hippocampi of Glul-floxed mice. second 3’LoxP sequence together with the Frt-PGKneo-Frt selectable marker was in- serted in intron 6 approximately 140 nucleotides 5’ of exon 7. This vector containing ap- proximately 4kb and 2.9kb of 5’-long and 3’-short arms, respectively, was then linearized by NotI digestion, purified, and then electroporated into ES cells, which were derived from F1(129sv/C57BL/6J) blastocyst. ES cells were cultured in the presence of G418 and Gancyclovir after electroporation according to Wurst and Joyner [83], and drug resis- tant colonies were picked and cultured in 96-well plates. Drug resistant ES clones were screened by nested long-range PCR using primers specific to genomic sequences outside the homology arms and LoxP sites to identify targeted ES clones. Targeted clones were expanded and screened again to confirm their identity prior to the generation of chimeric animals by aggregation with CD1 morula. Chimeric males were then bred with ROSA26- Flpe female [84] to remove the PGKneo cassette to generate the final Glul floxed allele. The resulting floxed F1 mice were then backcrossed with C57BL/6 mice for over 20 gen- erations. Site specific recombination of the loxP sites was achieved using a variety of viruses ex- pressing Cre recombinase and GFP under various promoters in two separate expression
  • 25. METHODS 20 cassettes (see example in Figure 3). The AAV5 CMV Legacy virus (SignaGen Laboratories, Rockville, MD), which expressed Cre and GFP under CMV promoters within a capsid from AAV serotype 5, was readily available from prior experiments conducted in our lab. To compare viral transfection efficiency, three other virus types were acquired through custom order from the Gene Therapy Center Vector Core at the University of North Car- olina (Chapel Hill, NC). These viruses include AAV5 CMV (an additional AAV serotype 5 expressing Cre and GFP under separate CMV promoters), AAV8 CMV (an AAV serotype 8 expressing Cre and GFP under separate CMV promoters), and AAV8 GFAP (an AAV serotype 8 expressing Cre and GFP under separate GFAP promoters). In addition, a corre- sponding control virus was ordered for each of the four experimental viruses. Each control virus was procured from the same source and was of identical construction to its corre- sponding experimental virus other than lacking the Cre expression cassette. Prior to injec- tion, all viruses were diluted to the same concentration to account for titer mismatch. Primary antibodies used in this study included chicken anti-GFP (Aves Labs, Tigard, OR), rabbit anti-GS (Sigma, St. Louis, MO), mouse anti-synaptophysin (Sigma), mouse anti-gephyrin (Synaptic Systems, Göttingen, Germany), and mouse anti-PSD-95 (EMD Millipore, Billerica, MA). Secondary antibodies Alexa Fluor 488 goat anti-chicken IgG, Alexa Fluor 555 goat anti-rabbit IgG, Alexa Fluor 647 goat anti-mouse IgG, and Pro- Long Diamond Antifade Mountant were from Life Technologies (Eugene, OR). All other reagents, unless otherwise noted, were obtained from Sigma Chemical Co. (St. Louis, MO). Surgery: Viral Injection and Electrode Implantation Mice were inducted into anesthesia with 5% Isoflurane (Baxter, Deerfield, IL) in O2 and placed in a stereotaxic frame (David Kopf Instruments, Tujunga, CA). For the duration of surgery, Isoflurane was maintained at 1.5-3% and titrated based on respiratory status and re-
  • 26. METHODS 21 Figure 3. Vector map of the viral plasmid construct, showing AAV5 CMV as an example. Upon entry into CMV-expressing cells, transcription and translation yields functional Cre and eGFP pro- tein. sponse to intermittent leg pinch. Burr holes were made into the skull bilaterally, and virus was injected into the brain parenchyma using the following validated coordinates using bregma as reference: Dentate Gyrus- AP -3.2mm, ML ±3.1mm, DV -3.5mm; Subiculum- AP -3.7mm, ML ±3.5mm, DV -3.44mm; Entorhinal Cortex- AP -4.1mm, ML ±3.8mm, DV -3.5mm. Virus was delivered stereotaxically using a 5µL 26s gauge syringe (Hamil- ton, Reno, NV). At each of the six injection sites per animal, 0.5µL of virus was injected at a rate of 0.05µL per 30 seconds. In order to minimize viral reflux upon needle removal, following delivery at each injection site, the needle was left to rest for 2 minutes and then raised out of the parenchyma at a rate not exceeding 0.5mm per minute.
  • 27. METHODS 22 In the viral comparison study, 16 animals were injected as above, split equally (n=2 each) between Cre-expressing and control viruses of the serotypes AAV5 CMV (Legacy), AAV5 CMV, AAV8 CMV, and AAV8 GFAP. Concurrent with viral injection, stainless steel screw electrodes (Plastics One, Roanoke, VA) were positioned in the epidural space over the parietal cortex bilaterally. A third screw electrode was placed in the epidural space over the cerebellum as a reference, and a fourth screw electrode was implanted in the occipital bone as a ground. The female ends of the electrodes were inserted into a pedestal (Plastics One) which was cemented onto the skull with UV light cured acrylated urethane adhesive (Loctite 3106 Light Cure Adhesive, Henkel Corp., Rocky Hill, CT) to form a headcap. Post-operative analgesia was achieved using a single injection of Meloxicam SR at a dose of 4 mg/kg administered prior to removal of Isoflurane. In the video-EEG study, 16 animals were injected with virus as described above, split equally between Cre-expressing and control viruses only of serotype AAV8 GFAP (based on findings of the viral comparison study; see Results section). Four weeks after viral in- jection surgery, recording electrodes were implanted in various arrangements: one subset was implanted with bilateral depth electrodes located in each hippocampus (n=6), another subset was implanted with unilateral screw electrodes over the parietal cortex (n=7), and a final subset was implanted with a single recording depth electrode in the hippocampus and a reference depth electrode in the white matter of the cerebellum (n=3). Headcaps were created as described above. Analgesia was achieved using one 0.05mg/kg pre-op dose of buprenorphine (Reckitt Benckiser Healthcare, Hull, England), one 0.05mg/kg post- op dose of buprenorphine, and approximately 1mg/kg/day of Meloxicam in the drinking water for 48 hours after surgery.
  • 28. METHODS 23 Video-EEG Monitoring and Seizure Precipitation Studies Four to six weeks after viral injection surgery, mice were placed individually in custom- made Plexiglas cages. A 6-channel cable was connected to the animal’s electrode pedestal on one end and to a commutator (Plastics One) on the other. A second cable connected the commutator to the digital EEG recording unit (CEEGraph Vision LTM, Natus/Bio- Logic Systems Corp., San Carlos, CA). Digital cameras with infrared light detection ca- pability were used to record animal behavior. The digital video signal was encoded and synchronized to the digital EEG signals. Seizures were identified by visual inspection of the EEG record. The Racine criteria [85] were modified and used to classify the seizures from the video records, as follows: Stage 1, immobilization, eye blinking, twitching of the vib- rissae, and mouth movements; Stage 2, head nodding, often accompanied by severe facial clonus; Stage 3, forelimb clonus; Stage 4, rearing without falling; Stage 5, rearing, falling, and generalized convulsions; Stage 6, violent generalized convulsions with repeated jump- ing. Ten weeks after viral injection, seizure precipitation studies were conducted by measur- ing the time to first clonic twitch and time to generalized convulsions following intraperi- toneal injection of the convulsant agent pentylenetetrazol (Sigma Aldrich, St. Louis, MO) at a dose of 90mg/kg (diluted to 9mg/mL in normal saline). Following induction of con- vulsive seizure, animals were fully anesthetized using Isoflurane and sacrificed as described below. Fixation, Immunofluorescence, and Microscopy For staining, mice were fully anesthetized with Isoflurane and perfused transcardially with saline (to remove intravascular blood) followed by 4% formaldehyde in 0.1M PB, pH 7.4 for 5 minutes. The brains were rapidly removed from the skull and post-fixed in the same fixative overnight at 4°C and then sectioned on a Vibratome at 50µm. Sections
  • 29. METHODS 24 were rinsed (3 x 5 minutes) in TBST (TBS with 0.5% Triton X-100), treated with 1M ethanolamine in 0.1M NaPi pH 7.4 for 30 minutes, washed in TBST (3 x 5 minutes), in- cubated for 1 hour in TBST containing 10% newborn calf serum and 3% bovine serum albumin followed by overnight incubation with primary antibodies. Next, sections were washed with TBST (3 x 10 minutes), incubated with secondary antibodies (Alexa 488 di- luted to 1:1000, Alexa 555 and Alexa 647 diluted to 1:500), washed with TBST (3 x 10 minutes), and mounted onto gel-coated slides with ProLong Diamond Antifade Mountant. Samples were examined using an Olympus BX61 wide-angle fluorescence microscope with Olympus DP71 camera attachment for image recording. Super-resolution images were acquired using a Leica TCS SP8 Gated STED 3X at maximal total magnification of approximately 5000x. Triple-labeling studies were conducted using a custom proto- col consisting of sequential scans in order of descending STED depletion wavelength, as follows: Alexa 647 (pulsed white-light laser tuned to 647nm, HyD photodetector tuned to 680-730nm, 775nm STED depletion beam), Alexa 555 (pulsed white-light laser tuned to 555nm, HyD photodetector tuned to 550-620nm, 660nm STED depletion beam), and Alexa 488 (pulsed white-light laser tuned to 488nm, HyD photodetector tuned to 500- 570nm, 592nm STED depletion beam). Statistical Analysis Statistical analyses and graph plotting were conducted in Igor Pro 8.02 (WaveMetrics). Statistical significance was assessed by Student’s t-test. Differences between groups were judged to be significant when p-values were smaller than 0.05. Error bars indicate the mean ± standard error of the mean (SEM) except when stated. No samples or animals were excluded from the analysis. For PTZ seizure precipitation studies, experimenters were blinded to the viral vector used in each animal when recording time to seizure events.
  • 30. Results Knockout of Glutamine Synthetase To evaluate whether injection of each virus into the mesial temporal lobe resulted in vi- able transfection of CNS cells, we first stained formaldehyde-fixed brain sections for GFP, the reporter protein carried by each viral plasmid. GFP fluorescence was visualized in all animals, confirming that the selected injection coordinates resulted in viral transfection of the dentate gyrus, subiculum, and entorhinal cortex (see Figure 4a). Despite maintaining consistent injection sites and quantity of virus injected for all viral serotypes, the degree of observed transfection area varied substantially between virus types (see Figure 4b), sug- gesting that different virus types may have differing abilities to diffuse through tissues or to transfect cells in regions of lower viral concentration (i.e., at the periphery of the injection field). Transfection area was determined by measuring the number of pixels that emitted FITC filtered fluorescence (excitation 470-490nm, emission 510nm long-pass) at an in- tensity above that of background emission and below that of obvious artifact (e.g., edge effect). Measurements were taken at the section level which appeared to have maximal affected area based on qualitative comparison and spanned an entire 40x field in order to accommodate assessment of all three injection sites in one hemisphere. The most striking finding was that the AAV8 GFAP virus appeared to transfect a substantially larger area than both AAV5 CMV viruses and the AAV8 CMV virus (approximately 2.5 x 105 pixels vs. 1.2 x 105 , 1.4 x 105 , and 7.4 x 104 pixels, respectively).
  • 31. RESULTS 26 Figure 4. Injection locations and viral spread. (A) GFP is detected in transfected cells of the hip- pocampus and adjacent structures in a representative 40x field. EC- entorhinal cortex, DH- dentate hilus, Gr- granule cell layer of the dentate gyrus, Mol- molecular layer of the dentate gyrus, SUB- subiculum, LV- lateral ventricle. Scale bar = 0.5mm (B) Viral spread is measured as GFP+ pixels present in the mesial temporal lobe. We first confirmed that GS knockout was restricted to areas containing virus-transfected cells by assessing co-localization of GFP and GS (see Figure 5). Next, area of GS knockout was quantified for each virus at three magnifications in order to assess the proportion of knockout in the entire mesial temporal lobe (40x), in the hippocampal formation (100x), and in the dentate gyrus (200x). Area was determined by measuring the number of pixels emitting TRITC filtered fluorescence (excitation 520-550nm, emission 580nm long-pass) at an intensity above that of background emission and below that of distant areas of tis- sue exhibiting normal expression of GS. As seen in Figure 6, the AAV5 CMV and AAV8 GFAP viruses decreased expression of GS in a substantially greater area than the other two viruses. Next, the efficiency of GS knockout was assessed for each virus type. In order to calcu- late the extent of knockdown, five regions of interest (circular, radius 8 pixels) were placed in the areas of apparently maximal knockdown visualized in the most qualitatively affected 40x field, and mean TRITC wavelength fluorescence intensity was measured in these re-
  • 32. RESULTS 27 Figure 5. Representative section of brain transfected with AAV8 GFAP virus. Green represents virus-transfected cells, while red represents presence of GS. GS knockout is seen only in areas with transfected cells. Scale bar = 0.2mm gions (see Figure 7a). The intensity was scaled by a control figure measured as the average fluorescence intensity of three regions of interest which appeared to exhibit normal GS ex- pression in the same section. The resulting relative intensity figure is the complement of knockdown efficiency, expressed as a percentage. As seen in Figure 7b, knockdown ef- ficiency was not statistically significant for AAV5 CMV Legacy (95% CI -14.5% to 2.4%, p=0.12) or AAV8 CMV (95% CI -10.7% to 2.9%, p=0.19). Both the AAV5 CMV virus and the AAV8 GFAP virus knocked down GS significantly (respectively, 95% CIs 47.8% to 78.1% and 78.8% to 87.5%, p=0.0003 and 0.000007). In addition, the increased knock- down of AAV8 GFAP over AAV5 CMV was statistically significant (p=0.018). Finally, each virus’ selectivity for astrocytes was assessed, as cell-type specificity is an im- portant factor that mitigates confounding cellular effects of off-target Cre and GFP ex- pression and improves visualization of cells of interest in microscopy studies. Selectivity
  • 33. RESULTS 28 800 600 400 200 Pixels (x10 3 ) AAV5 CMV (Legacy) AAV5 CMV AAV8 CMV AAV8 GFAP at 40x at 100x at 200x Figure 6. Area of GS knockout for each virus at various magnifications. AAV5 CMV and AAV8 GFAP outperformed the AAV5 CMV Legacy and AAV8 CMV viruses. Figure 7. Quantification of GS knockout intensity. (A) Example of ROI selection in a section transfected with AAV5 CMV. ROIs 1-5 are placed in areas of knockout whereas ROIs 6-8 are used as control regions to scale intensity measures. Red stained areas are not infected with virus and ex- press GS normally. Scale bar = 0.5mm (B) Knockdown efficiency following infection with each virus type (*P<0.05 and **P<0.001).
  • 34. RESULTS 29 Figure 8. Assessment of astrocyte specificity in transfected cell population. (A) Representative field from AAV8 GFAP demonstrating astrocyte morphology in nearly all GFP+ cells. Scale bar = 0.1mm (B) Proportion of GFP+ cells demonstrating astrocyte morphology after transfection with each virus type. was assessed by manual count of GFP+ cells within a representative 200x field within the dentate gyrus and classification of cells as “astrocyte” or “other” based on morphology. As expected, AAV8 GFAP exhibited near perfect target specificity (98.7% of GFP+ cells were astrocytes). As seen in Figure 8, AAV5 CMV Legacy, AAV5 CMV, and AAV8 CMV tar- geted mostly neurons with varied degrees astrocyte labeling detected (10.0%, 21.3%, and 12.7% astrocytes, respectively.) Seizure Findings To determine if intrahippocampal knockout of GS is sufficient to cause recurrent sponta- neous seizures, mice injected with Cre-expressing AAV8 GFAP (n=5) and control virus (n=4) were monitored for two weeks with continuous video and intracranial EEG record- ing. Epileptiform activity was assessed visually (example in Figure 9). Of the 6 animals im- planted with bilateral hippocampal depth electrodes, 5 died due to post-operative compli- cations prior to commencement of EEG recording. Thus, the remaining 10 animals were implanted either with screw electrodes only (n=7) or a single recording hippocampal depth
  • 35. RESULTS 30 Figure 9. Example EEG tracing during Racine stage 3 seizure. (A) Seizure progression and termi- nation is demonstrated in top tracing with normal control trace below for comparison. Scale bar represents 5 seconds. (B) Highlighted area in (A) is expanded to detail epileptic waveform. electrode (n=3). Seizures were noted to occur in 60% (3/5) of the animals injected with Cre-expressing virus and in 25% (1/4) of the animals injected with control virus (see Table 1). Seizures tended to occur in clusters (11/19 seizure events occurred within 2-hours of another seizure event in the same animal) and in the early morning hours (14/19 seizure events occurred between midnight at 8:00 AM. Observed behaviors during seizure events were varied between animals, however, individual animals tended to exhibit similar features during each event. Common seizure behaviors included facial automatisms, tail extension, lordosis, rearing, and uncontrolled jumping. In addition to comparing development of spontaneous seizures, sensitivity to provo- cation of seizures was also measured. Fifteen mice transfected with one of the four ex- perimental viruses (n=9) or four control viruses (n=6) were observed to determine time to first myoclonic twitch and time to convulsive seizure following intraperitoneal injec- tion of PTZ. As seen in Figure 10, animals with GS knocked out exhibited the first clonic twitch after an average of 41.2 ± 3.2 seconds (mean ± SEM), which was significantly less time than control animals, averaging a delay of 65.83 ± 12.9 seconds (p=0.044). The effect
  • 36. RESULTS 31 Table 1. Day-by-day report of noted spontaneous behavioral seizures in the 9 continuous video- EEG-recorded mice. Dark grey boxes indicate that recording was stopped due to death of the animal or deterioration of clinical state requiring euthanasia. Electrode arrangements are as follows: Depth, Bil. depth recording electrodes in both hippocampi; Depth, Uni.- depth recording electrode in a single hippocampus and a depth reference electrode in the cerebellum; Screw- screw recording electrode above the parietal cortex. on time to convulsive seizure was not statistically significant, though there was a trend of knockout animals proceeding to convulsions in less time (74.2 ± 9.4 seconds vs. 100.0 ± 18.0 seconds, p=0.20). 120 100 80 60 40 20 0 Delay from PTZ Administration (s) * Clonic Twitch Convulsions GS Knockout Control Figure 10. PTZ-induced precipitation of pro- voked seizures. GS knockout mice demon- strate increased susceptibility to provoked seizures, as illustrated by a shorter time to clonic twitch compared to mice transfected with control virus (p<0.05). There was a sim- ilar trend in time to convulsion, though this effect was not significant (p=0.20).
  • 37. RESULTS 32 Synaptic Changes A preliminary assessment of observed synaptic changes caused by GS loss was also per- formed by performing fluorescence STED microscopy on brain sections from mice in- jected with AAV8 GFAP Cre-expressing virus. Sections were triple-labeled with primary antibodies against GFP, GS, and one of three synaptic markers: synaptophysin, PSD-95, or gephyrin. First, changes in synaptic marker expression between GS+ and GS- areas were assessed visually in a 200x field containing the dentate gyrus (Figure 10a). The expected absence synaptophysin, PSD-95, and gephyrin within the granule cell layer of the dentate gyrus was noted, and no other gross changes were apparent. Next, more nuanced changes in synaptic marker expression were assessed by measuring the synaptic fluorescence in- tensity in selected regions within the visible territories of both GFP-/GS+ (unaffected) and GFP+/GS- (knockout) astrocytes. Expression of PSD-95 appeared to be decreased in synapses proximal to GS- astrocytes, with sampled relative intensity of 0.57 ± 0.04 (mean ± SEM, p=0.002, see Figure 10b). Relative expression of synaptophysin and gephyrin appeared to be unchanged in the sampled areas (synaptophysin relative expression 0.94 ± 0.15, p=0.87; gephyrin relative expression 0.94 ± 0.04, p=0.23).
  • 38. RESULTS 33 Figure 11. Assessment of synaptic marker changes associated with loss of GS. (A) Example 200x field illustrating staining pattern of synaptophysin in the dentate gyrus, which is grossly unchanged in areas with GS knockdown. (B) Quantification of relative intensity of synaptic markers in normal GS+ astrocytes (arrow in panel A) compared to GFP+/GS- astrocytes (arrowhead in panel A). PSD-95 expression appears to be reduced in GS- astrocytes (p=0.002), while synaptophysin and gephyrin expression appear to be unchanged. (C)Comparison of a synaptophysin colocalization with single astrocytic process of a GS+ astrocyte (left panel) and a GS knockout astrocyte (right panel) at 5000x magnification. Scale bars = 5µm
  • 39. Discussion Development of a monogenetic knockout model with effects restricted to the hip- pocampus and adjacent structures has the potential to more fully elucidate the impact of GS loss in this treatment-resistant disease. In this study, we demonstrated the use of a novel model of MTLE by selectively knocking out GS in the hippocampal astrocytes of mice. Studies of MTLE in humans and in other experimental models show that loss of GS is closely associated with MTLE pathology [23, 25, 54], however the exact role of GS in MTLE and the biochemical pathways through which it may exert its effects remain un- clear. In order to first maximize GS knockout, we compared the expression patterns of GS after Glul-floxed C57BL/6J mice were injected with four different adeno-associated viral vectors containing Cre-recombinase expression cassettes. Injection volumes and viral titers were matched between viruses, however knockout of GS was found over a greater area in the AAV5 CMV and AAV8 GFAP serotypes. In addition, within each GS knockout re- gion, AAV8 GFAP exhibited a significantly greater efficiency of knockdown compared to other serotypes and most selectively targeted astrocytes. Thus, AAV8 GFAP was selected for use in video-EEG and synaptic microscopy studies. When hippocampal GS knockout mice were assessed with video-EEG over a 12-day period, 60% (3/5) animals exhibited a phenotype comparable to human MTLE including spontaneous recurrent seizures clustered in the morning hours. 25% (1/4) of control animals also seized recurrently when moni-
  • 40. DISCUSSION 35 tored over the same period. Additionally, GS knockout mice appeared more susceptible to provoked seizures, as delay to clonic twitch following PTZ injection was reduced by 24 seconds (p=0.044). Finally, examination of synaptic markers in the territory of GS+ and GS- astrocytes revealed a significant decrease in PSD-95, while expression of synaptophysin and gephyrin appeared unchanged. Several limitations of the research presented must be discussed in order to most appro- priately interpret the study results. First, the small sample size prevents definitive conclu- sions from being drawn in the seizure proclivity studies, particularly given the development of spontaneous seizures in one of the control animals injected with non-Cre-expressing virus. It should be noted that the epileptic control animal was the sole surviving animal of the 6 mice implanted with bilateral depth electrodes, which may suggest that the seizures resulted from physical trauma to the brain parenchyma during electrode implantation. As- certaining the cause of this (and any subsequent) control animal seizures will be crucial in validation of the model and confirmation that seizures in experimental animals are caused by GS depletion rather than physical trauma associated with viral injection and/or cellu- lar effects of viral transfection or intracellular GFP. We also encountered challenges with a high incidence of premature headcap removal (5/9 mice). This further limited data col- lected for video-EEG, seizure precipitation, and histology studies, and, importantly, po- tentially exposed experimental animals to undue discomfort. Subsequent iterations of EEG implantation should attempt to use alternate materials or methods of headcap mounting in order to reduce the frequency of such events. Small sample size also limits the general- izability of microscopy data, which in some cases, required analysis of brain sections from a single animal transfected with certain viral serotypes. Finally, while the virally mediated localization of GS knockout has the potential to fully mimic the spatial pattern of GS loss seen in human MTLE, this iteration of the model does not. The experimenters elected to knockout GS in the dentate gyrus, subiculum, and entorhinal cortex bilaterally as an ini- tial proof-of-concept, however subsequent iterations will more closely model the typically
  • 41. DISCUSSION 36 unilateral findings of GS depletion. The results presented lay the foundation for informative follow-up investigations. First, bolstering of sample size will clarify the efficacy of virus-mediated GS knockout in elicit- ing spontaneous seizures and will allow more granular study of epileptogenesis at various time points following GS loss. The extent of GS knockdown should also be confirmed using tissue-based methods such as laser capture micro-dissection followed by qPCR. As- sessing local knockout mice for comorbidities found in human MTLE, such as depression and anxiety, will also lend further evaluate how well the model mimics human disease. Additionally, following thorough validation of the local GS knockout model, more ex- pansive microscopy experiments should be undertaken to fully capitalize on the poten- tial advantages conferred by super-resolution microscopy techniques such as STED. Mi- croscopy studies will include the examination of the changes induced by GS-loss on vas- culature, molecular transporters, microglia, and synapses, including further investigation of our finding that PSD-95 expression appeared to be reduced in the vicinity of GS knock- out astrocytes. Given PSD-95’s known binding activity with neural potassium channels and glutamate receptors AMPA and NMDA [86] as well as the well-characterized BDNF me- diated downregulation of NMDA receptor expression in the context of excitotoxicity [87], our findings may suggest that such changes may vary within each cell-to-cell microenvi- ronment. Additional examination of this observed PSD-95 downregulation and apparent sparing of synaptophysin (which is nearly ubiquitous in all synapses) and gephyrin (found at inhibitory synapses) may shed light on the specific importance of excitatory synaptic reg- ulation in epileptogenesis. Further studies will also produce more information about the prevalence of seizures in local GS knockout mice, and should there be GS knockout mice which do not seize (as there were in this study), the unique cellular features present in such mice may provide useful long-term insights for therapeutic targeting and drug develop- ment. The most notable shortcoming of the virus-mediated localized knockout of GS is the
  • 42. DISCUSSION 37 highly technical and time-consuming nature of subject generation. Even after Glul-floxed mice have been created and bred to produce homozygous-floxed animals, injection of virus is a relatively involved process which requires careful diligence and time. The necessity to use stereotaxic technique, deliver virus extremely gradually, and repeat the procedure over multiple injection sites results in a multi-hour surgical procedure for each individ- ual subject which benefits negligibly from batching of procedures. For this reason, local GS knockout may not be an ideal model for all MTLE investigations. However, the novel precision of the technique does address recent critiques about indiscriminate selection of animal models [88]. This approach also improves on the emulative accuracy of models cur- rently in use in epilepsy basic science and translational research, which has been cited as a contributor to the recent paucity of novel therapeutics in the field of treatment-resistant epilepsies such as MTLE [2]. The novel model presented here has the capability to mimic the spatial and temporal disease pattern of human MTLE with greater accuracy and flex- ibility than any previously reported GS-depletion model of epilepsy, and with further validation, virus-mediated knockout of hippocampal GS has the potential to become the gold-standard model in MTLE research.
  • 43. References 1. Wilson, J.K. and E.H. Reynolds, Translation and analysis of a cuneiform text form- ing part of a Babylonian treatise on epilepsy. Medical history, 1990. 34(2): p. 185- 198. 2. Steinhauser, C., M. Grunnet, and G. Carmignoto, Crucial role of astrocytes in tem- poral lobe epilepsy. Neuroscience, 2016. 323: p. 157-69. 3. Stafstrom, C.E. and L. Carmant, Seizures and Epilepsy: An Overview for Neurosci- entists. Cold Spring Harbor Perspectives in Medicine, 2015. 5(6). 4. Bernhardt, B., et al., Imaging structural and functional brain networks in temporal lobe epilepsy. Frontiers in Human Neuroscience, 2013. 7(624). 5. Thom, M. and E.H. Bertram, Chapter 14 - Temporal lobe epilepsy, in Handbook of Clin- ical Neurology, H. Stefan and W.H. Theodore, Editors. 2012, Elsevier. p. 225-240. 6. Asadi-Pooya, A.A., et al., Prevalence and Incidence of Drug-Resistant Mesial Tem- poral Lobe Epilepsy in the United States. World Neurosurgery, 2017. 99: p. 662- 666. 7. Brodie, M.S., SC; Kwan, P, Epilepsy. 5 ed. Fast Facts. 2012: Health Press Limited. 146. 8. Allone, C., et al., Neuroimaging and cognitive functions in temporal lobe epilepsy: A review of the literature. Journal of the Neurological Sciences, 2017. 381: p. 7-15. 9. Hermann, B.P., et al., Comorbid Psychiatric Symptoms in Temporal Lobe Epilepsy: Association with Chronicity of Epilepsy and Impact on Quality of Life. Epilepsy & Behavior, 2000. 1(3): p. 184-190. 10. Garcia, C.S., Depression in temporal lobe epilepsy: a review of prevalence, clinical features, and management considerations. Epilepsy research and treatment, 2012. 2012: p. 1-12. 11. Andrade-Machado, R., et al., Mortality in patients with refractory temporal lobe epilepsy at a tertiary center in Cuba. Epilepsy & Behavior, 2015. 53: p. 154-160. 12. Sutula, T., Mossy fiber synaptic reorganization in the epileptic human temporal lobe. Annals of neurology. 26(3): p. 321-330.
  • 44. REFERENCES 39 13. Aronica, E., et al., Induction of neonatal-sodium channel II and III (α-isoform mR- NAs in neurons and microglia after status epilepticus in the rat hippocampus. Euro- pean Journal of Neuroscience, 2001. 13(6): p. 1261-1266. 14. Mathern, G.W., Hippocampal AMPA and NMDA mRNA levels and subunit im- munoreactivity in human temporal lobe epilepsy patients and a rodent model of chronic mesial limbic epilepsy. Epilepsy research. 32(1-2): p. 154-171. 15. de Lanerolle, N.C., Hippocampal interneuron loss and plasticity in human temporal lobe epilepsy. Brain research. 495(2): p. 387-395. 16. Mangan, P.S. and E.H. Bertram Iii, Shortened-duration GABA(A) receptor- mediated synaptic potentials underlie enhanced CA1 excitability in a chronic model of temporal lobe epilepsy. Neuroscience, 1997. 80(4): p. 1101-1111. 17. Mangan, P.S. and E.W. Lothman, Profound disturbances of pre- and postsynaptic GABA(B)-receptor-mediated processes in region CA1 in a chronic model of tempo- ral lobe epilepsy. Journal of Neurophysiology, 1996. 76(2): p. 1282-1296. 18. Rigau, V., et al., Angiogenesis is associated with blood-brain barrier permeability in temporal lobe epilepsy. Brain, 2007. 130(7): p. 1942-1956. 19. Mtchedlishvili, Z., E.H. Bertram, and J. Kapur, Diminished allopregnanolone en- hancement of GABAA receptor currents in a rat model of chronic temporal lobe epilepsy. Journal of Physiology, 2001. 537(2): p. 453-465. 20. During, M.J., Extracellular hippocampal glutamate and spontaneous seizure in the conscious human brain. Lancet. 341(8861): p. 1607-1610. 21. Olney, J.W., Excitotoxic mechanisms of epileptic brain damage. Advances in neurol- ogy, 1986. 44: p. 857-877. 22. Petroff, O.A.C., Glutamate-glutamine cycling in the epileptic human hippocampus. Epilepsia (Copenhagen). 43(7): p. 703-710. 23. Eid, T., et al., Loss of glutamine synthetase in the human epileptogenic hippocam- pus: possible mechanism for raised extracellular glutamate in mesial temporal lobe epilepsy. The Lancet, 2004. 363(9402): p. 28-37. 24. Krajewski, W.W., et al., Crystal structures of mammalian glutamine synthetases illus- trate substrate-induced conformational changes and provide opportunities for drug and herbicide design. (1089-8638 (Electronic)). 25. Eid, T., et al., The Glutamate-Glutamine Cycle in Epilepsy. Adv Neurobiol, 2016. 13: p. 351-400. 26. Norenberg Md Fau - Martinez-Hernandez, A. and A. Martinez-Hernandez, Fine structural localization of glutamine synthetase in astrocytes of rat brain. (0006-8993 (Print)).
  • 45. REFERENCES 40 27. Meldrum, B.S., Glutamate as a neurotransmitter in the brain: review of physiology and pathology. (0022-3166 (Print)). 28. Danbolt, N.C., Glutamate uptake. (0301-0082 (Print)). 29. Olney, J.W., Brain lesions, obesity, and other disturbances in mice treated with monosodium glutamate. (0036-8075 (Print)). 30. McKenna, M.C., The glutamate-glutamine cycle is not stoichiometric: fates of glu- tamate in brain. Journal of neuroscience research. 85(15): p. 3347-3358. 31. Haberle, J., et al., Congenital glutamine deficiency with glutamine synthetase muta- tions. (1533-4406 (Electronic)). 32. Haberle, J., et al., Natural course of glutamine synthetase deficiency in a 3 year old patient. (1096-7206 (Electronic)). 33. Le Prince, G., et al., Glutamine synthetase (GS) expression is reduced in senile de- mentia of the Alzheimer type. 1995(0364-3190 (Print)). 34. Tumani, H., et al., Glutamine synthetase in cerebrospinal fluid, serum, and brain: a diagnostic marker for Alzheimer disease? 1999(0003-9942 (Print)). 35. Carter, C.J., Loss of glutamine synthetase activity in the brain in Huntington’s dis- ease. 1981(0140-6736 (Print)). 36. Rajkowska, G., Astrocyte pathology in major depressive disorder: insights from hu- man postmortem brain tissue. Current drug targets, 2013. 14(11): p. 1225-1236. 37. Bruneau, E.G., Increased expression of glutaminase and glutamine synthetase mRNA in the thalamus in schizophrenia. Schizophrenia research, 2005. 75(1): p. 27-34. 38. Kalkman, H.O., Circumstantial evidence for a role of glutamine-synthetase in sui- cide. Medical hypotheses, 2011. 76(6): p. 905-907. 39. Löscher, W., Critical review of current animal models of seizures and epilepsy used in the discovery and development of new antiepileptic drugs. Seizure, 2011. 20(5): p. 359-368. 40. Depaulis, A. and S. Hamelin, Animal models for mesiotemporal lobe epilepsy: The end of a misunderstanding? Revue Neurologique, 2015. 171(3): p. 217-226. 41. Löscher, W., Animal models of epilepsy and epileptic seizures. Antiepileptic Drugs. Handbook of Experimental Pharmacology, 1999: p. 19-62. 42. Ben-Ari, Y. and J. Lagowska, Epileptogenic action of intra-amygdaloid injection of kainic acid. Comptes rendus hebdomadaires des seances de l’Academie des sciences. Serie D: Sciences naturelles, 1978. 287(8): p. 813-816.
  • 46. REFERENCES 41 43. Fritsch, B., et al., Role of GluK1 Kainate Receptors in Seizures, Epileptic Dis- charges, and Epileptogenesis. The Journal of Neuroscience, 2014. 34(17): p. 5765. 44. Lévesque, M. and M. Avoli, The kainic acid model of temporal lobe epilepsy. Neu- roscience & Biobehavioral Reviews, 2013. 37(10, Part 2): p. 2887-2899. 45. Jefferys, J., C. Steinhäuser, and P. Bedner, Chemically-induced TLE models: Topical application. Journal of Neuroscience Methods, 2016. 260: p. 53-61. 46. Bouilleret, V., et al., Recurrent seizures and hippocampal sclerosis following intrahippocampal kainate injection in adult mice: Electroencephalography, histopathology and synaptic reorganization similar to mesial temporal lobe epilepsy. Neuroscience, 1999. 89(3): p. 717-729. 47. Riban, V., et al., Evolution of hippocampal epileptic activity during the develop- ment of hippocampal sclerosis in a mouse model of temporal lobe epilepsy. Neuro- science, 2002. 112(1): p. 101-111. 48. Turski, W.A., et al., Limbic seizures produced by pilocarpine in rats: Behavioural, electroencephalographic and neuropathological study. Behavioural Brain Research, 1983. 9(3): p. 315-335. 49. Castro, O.W., et al., Comparative neuroanatomical and temporal characterization of FluoroJade-positive neurodegeneration after status epilepticus induced by systemic and intrahippocampal pilocarpine in Wistar rats. Brain Research, 2011. 1374: p. 43- 55. 50. Furtado, M.A., et al., Study of spontaneous recurrent seizures and morphological alterations after status epilepticus induced by intrahippocampal injection of pilo- carpine. Epilepsy Behavior, 2011. 20(2): p. 257-266. 51. De A. Furtado, M., et al., Behavioral, Morphologic, and Electroencephalo- graphic Evaluation of Seizures Induced by Intrahippocampal Microinjection of Pi- locarpine. Epilepsia, 2002. 43(s5): p. 37-39. 52. Arida, R.M., et al., The course of untreated seizures in the pilocarpine model of epilepsy. Epilepsy Research, 1999. 34(2): p. 99-107. 53. Covolan, L. and L.E.A.M. Mello, Temporal profile of neuronal injury following pi- locarpine or kainic acid-induced status epilepticus. Epilepsy Research, 2000. 39(2): p. 133-152. 54. Eid, T., et al., Recurrent seizures and brain pathology after inhibition of glutamine synthetase in the hippocampus in rats. Brain, 2008. 131(Pt 8): p. 2061-70. 55. Krajewski, W.W., S.L. Jones Ta Fau - Mowbray, and S.L. Mowbray, Structure of Mycobacterium tuberculosis glutamine synthetase in complex with a transition-state mimic provides functional insights. Proc Natl Acad Sci, U. S. A., 2005(0027-8424 (Print)).
  • 47. REFERENCES 42 56. Dhaher, R., et al., Effects of site-specific infusions of methionine sulfoximine on the temporal progression of seizures in a rat model of mesial temporal lobe epilepsy. Epilepsy, Res, 2015(1872-6844 (Electronic)). 57. Wang, H., et al., Network evolution in mesial temporal lobe epilepsy revealed by diffusion tensor imaging. Epilepsia, 2017(1528-1167 (Electronic)). 58. Dhaher, R., et al., 5-aminovaleric acid suppresses the development of severe seizures in the methionine sulfoximine model of mesial temporal lobe epilepsy. Neurobiol, Dis, 2014(1095-953X (Electronic)). 59. Shaw, C.A. and J.S. Bains, Synergistic versus antagonistic actions of glutamate and glutathione: the role of excitotoxicity and oxidative stress in neuronal disease. Cellu- lar and Molecular Biology, 2002. 48(2): p. 127-136. 60. Bernard-Hélary, K., et al., In vivo and in vitro glycogenic effects of methionine sul- foximine are different in two inbred strains of mice. Brain Research, 2002. 929(2): p. 147-155. 61. Kam, K. and R. Nicoll, Excitatory synaptic transmission persists independently of the glutamate-glutamine cycle. Journal of Neuroscience, 2007. 27(34): p. 9192- 9200. 62. Sauer, B. and N. Henderson, Site-specific DNA recombination in mammalian cells by the Cre recombinase of bacteriophage P1. Proceedings of the National Academy of Sciences, 1988. 85(14): p. 5166. 63. NIH blueprint for neuroscience: Cre driver network. Available from: http://www.neuroscienceblueprint.nih.gov/factSheet/CreDriver.htm. 64. He, Y., et al., Glutamine synthetase is essential in early mouse embryogenesis. De- velopmental Dynamics, 2007. 236(7): p. 1865-1875. 65. He, Y., et al., Glutamine synthetase deficiency in murine astrocytes results in neona- tal death. GLIA, 2010. 58(6): p. 741-754. 66. Zhou, Y., et al., Selective deletion of glutamine synthetase in the mouse cerebral cortex induces glial dysfunction and vascular impairment that precede epilepsy and neurodegeneration. Neurochem Int, 2018. 67. Gorski, J.A., et al., Cortical Excitatory Neurons and Glia, But Not GABAergic Neurons, Are Produced in the Emx1-Expressing Lineage. The Journal of Neuro- science, 2002. 22(15): p. 6309. 68. Coulter, D.A. and T. Eid, Astrocytic regulation of glutamate homeostasis in epilepsy. Glia, 2012. 60(8): p. 1215-1226. 69. Steffens, M., et al., Unchanged glutamine synthetase activity and increased NMDA receptor density in epileptic human neocortex: Implications for the pathophysiology of epilepsy. Neurochemistry International, 2005. 47(6): p. 379-384.
  • 48. REFERENCES 43 70. Anton, M. and F.L. Graham, Site-specific recombination mediated by an adenovirus vector expressing the Cre recombinase protein: a molecular switch for control of gene expression. Journal of Virology, 1995. 69(8): p. 4600. 71. Kaspar, B.K., et al., Adeno-associated virus effectively mediates conditional gene modification in the brain. Proceedings of the National Academy of Sciences, 2002. 99(4): p. 2320. 72. Blömer, U., et al., Highly efficient and sustained gene transfer in adult neurons with a lentivirus vector. Journal of Virology, 1997. 71(9): p. 6641. 73. Kaspar, B.K., et al., Targeted Retrograde Gene Delivery for Neuronal Protection. Molecular Therapy, 2002. 5(1): p. 50-56. 74. Salganik, M., M.L. Hirsch, and R.J. Samulski, Adeno-associated virus as a mammalian DNA vector. Microbiology spectrum, 2015. 3(4). 75. Sakuma, T., M.A. Barry, and Y. Ikeda, Lentiviral vectors: Basic to translational. Biochemical Journal, 2012. 443(3): p. 603-618. 76. Cearley, C.N. and J.H. Wolfe, Transduction characteristics of adeno-associated virus vectors expressing cap serotypes 7, 8, 9, and Rh10 in the mouse brain. Molecular Therapy, 2006. 13(3): p. 528-537. 77. Linterman, K.S., et al., Lentiviral-mediated gene transfer to the sheep brain: Impli- cations for gene therapy in batten disease. Human Gene Therapy, 2011. 22(8): p. 1011-1020. 78. Blessing, D. and N. Déglon, Adeno-associated virus and lentivirus vectors: a refined toolkit for the central nervous system. Current Opinion in Virology, 2016. 21: p. 61-66. 79. Agbandje-McKenna, M. and J. Kleinschmidt, AAV capsid structure and cell interactions, in Methods in Molecular Biology. 2011. p. 47-92. 80. Aschauer, D.F., S. Kreuz, and S. Rumpel, Analysis of Transduction Efficiency, Tropism and Axonal Transport of AAV Serotypes 1, 2, 5, 6, 8 and 9 in the Mouse Brain. PLOS ONE, 2013. 8(9): p. e76310. 81. von Jonquieres, G., et al., Glial Promoter Selectivity following AAV-Delivery to the Immature Brain. PLOS ONE, 2013. 8(6): p. e65646. 82. Lee, E.C., et al., A highly efficient Escherichia coli-based chromosome engineering system adapted for recombinogenic targeting and subcloning of BAC DNA. Ge- nomics, 2001. 73(0888-7543 (Print)). 83. Wurst, W.J., A.L., Production of Targeted Embryonic Stem Cell Clones, in Targeting, Gene, A Practical Approach, A.L. Joyner, Editor. 1999, Oxford University Press: New York.
  • 49. REFERENCES 44 84. Soriano, P., The PDGF� receptor is required for neural crest cell development and for normal patterning of the somites. Development, 1997. 124(14): p. 2691-2700. 85. Racine, R.J., et al., Rates of motor seizure development in rats subjected to electri- cal brain stimulation: strain and inter-stimulation interval effects. Electroencephalog- raphy and Clinical Neurophysiology, 1973. 35(5): p. 553-556. 86. Sheng, M. and C. Sala, PDZ Domains and the Organization of Supramolecular Complexes. Annual Review of Neuroscience, 2001. 24(1): p. 1-29. 87. Brandoli, C., et al., Brain-Derived Neurotrophic Factor and Basic Fibroblast Growth Factor Downregulate NMDA Receptor Function in Cerebellar Granule Cells. The Journal of Neuroscience, 1998. 18(19): p. 7953. 88. Denayer, T., T. Stöhr, and M. Van Roy, Animal models in translational medicine: Validation and prediction. New Horizons in Translational Medicine, 2014. 2(1): p. 5-11.