Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Nov 1.
Published in final edited form as: Neurochem Res. 2012 Apr 10;37(11):2339–2350. doi: 10.1007/s11064-012-0766-5

Roles of Glutamine Synthetase Inhibition in Epilepsy

Tore Eid 1,, Kevin Behar 2, Ronnie Dhaher 3, Argyle V Bumanglag 4, Tih-Shih W Lee 5
PMCID: PMC3731630  NIHMSID: NIHMS470253  PMID: 22488332

Abstract

Glutamine synthetase (GS, E.C. 6.3.1.2) is a ubiquitous and highly compartmentalized enzyme that is critically involved in several metabolic pathways in the brain, including the glutamine-glutamate-GABA cycle and detoxification of ammonia. GS is normally localized to the cytoplasm of most astrocytes, with elevated concentrations of the enzyme being present in perivascular endfeet and in processes close to excitatory synapses. Interestingly, an increasing number of studies have indicated that the expression, distribution, or activity of brain GS is altered in several brain disorders, including Alzheimer’s disease, schizophrenia, depression, suicidality, and mesial temporal lobe epilepsy (MTLE). Although the metabolic and functional sequelae of brain GS perturbations are not fully understood, it is likely that a deficiency in brain GS will have a significant biological impact due to the critical metabolic role of the enzyme. Furthermore, it is possible that restoration of GS in astrocytes lacking the enzyme could constitute a novel and highly specific therapy for these disorders. The goals of this review are to summarize key features of mammalian GS under normal conditions, and discuss the consequences of GS deficiency in brain disorders, specifically MTLE.

Keywords: Astrocytes, Glutamate, Hippocampus, Seizures, Temporal lobe epilepsy

Introduction

Glutamine synthetase (GS; a.k.a. glutamate ammonia ligase, GLUL, EC 6.3.1.2) is a key enzyme involved in nitrogen metabolism, acid–base homeostasis, and cell signaling across multiple species of prokaryotes and eukaryotes [1-3]. One of the main roles of GS in vertebrates is to metabolize glutamate and ammonia to glutamine according to the following reaction:

Glutamate+NH3.+ATPGlutamine+ADP+Pi.

This reaction is biologically significant because high concentrations of glutamate and ammonia are toxic to the central nervous system (CNS) [4-7]. Moreover, a continuous supply of glutamine is required for several physiological processes, including synthesis of glutamate and GABA, synthesis of proteins, and osmoregulation [8]. Because GS is the only known enzyme in humans capable of synthesizing glutamine, alterations in the expression and activity of the enzyme are likely to have significant biological effects.

Indeed, recent studies have shown that systemic mutations of the GS gene in humans result in brain malformations, seizures, multiorgan failure, and early death [9, 10]. There is also emerging evidence to suggest that brain GS may be implicated in the pathogenesis of several neurological disorders and psychiatric conditions including Alzheimer’s disease [11, 12], hepatic encephalopathy [13], suicide/depression [14, 15], schizophrenia [16], and epilepsy [17-19].

The goal of this review is two-fold; first, to summarize key features of GS under normal conditions, and second, to discuss the potential involvement of GS in brain disorders, particularly epilepsy.

GS is a Ubiquitous, Yet Highly Compartmentalized Enzyme

The importance of GS for life is underscored by its ubiquitous presence in a wide range of organisms, including microbes [20, 21], plants [22], animals (Elliott [51]), and humans [23]. Among the two main forms of GS, GSI and GSII, the latter is present in eukaryotes and prokaryotes. In contrast, GSI has only been detected in prokaryotes [24]. The human GS gene (GLUL) is localized to chromosome 1q31. A GS pseudogene (GLULP) is present on chromosome 9p13, and three GS-like genes are present on 5q33 (GLULL1), on 11p15 (GLULL2), and on 11q24 (GLULL3) [25]. The biological significance of the pseudogene and GS-like genes is not fully understood. Although there are several GLUL gene variants among eukaryotes, the ammonia binding site and the amino acid residues that interact with glutamate, magnesium/manganese ions, and ATP, are highly conserved [26]. Thus, herbicides, drugs, and other toxins that interfere with these sites are likely to do so across multiple species [26].

The GS enzyme in humans is structurally comprised of two ring-like pentamers forming an active decamer [26]. In humans and many vertebrates, the enzyme is abundantly present in a limited set of hepatocytes surrounding the terminal hepatic veins [27], in skeletal muscle cells, and in many (but not all) GFAP-positive astrocytes in the brain [28-30]. An even finer compartmentation of GS protein is evident in immunostained sections of the brain. GS is strongly expressed in astrocytic processes surrounding glutamatergic nerve terminals in the hippocampal formation [31]. GS is also particularly abundant in a laminar pattern in the molecular layer of the dentate gyrus, corresponding to glutamate binding sites [32].

GS is Essential for the Glutamine-Glutamate-GABA Cycle Metabolism

Glutamate and GABA are the primary excitatory and inhibitory neurotransmitters in the brain. To ensure the necessary fidelity of excitatory and inhibitory synaptic transmission, a carefully regulated synthesis, cellular release, and extracellular uptake of glutamate and GABA is required. Thus, an intimate metabolic relationship exists between glutamate and GABA. This relationship, which is known as the glutamine-glutamate-GABA cycle, includes GS and glutamine as well as a number of other metabolic pathways and transporter proteins. The glutamine-glutamate-GABA cycle involves the following steps (Fig. 1): (1) Glutamate in astrocytes is metabolized with ammonia and ATP to form glutamine via the GS reaction described earlier [33]. (2) Glutamine is transported from astrocytes into the extracellular space via specialized transporter proteins, particularly the system N transporters (SN1/SNAT3 and SN2/SNAT5) [34]. (3) Extracellular glutamine is imported to glutamatergic neurons via the system A transporter subtype 2 (SAT2/SNAT2) [35], and to GABA-ergic neurons via system A transporter subtype 1 (SAT1/SNAT1) [36]. (4a) Glutamine in glutamatergic neurons is converted to glutamate via the mitochondrial enzyme phosphate activated glutaminase (PAG) [37, 38], and glutamate is imported into synaptic vesicles via vesicular glutamate transporters (VGLUT) [39-42]. (4b) Glutamate is released from synaptic vesicles followed by binding to extracellular glutamate receptors present on neurons and glial cells. The extracellular glutamate is then taken up primarily by astrocytes via astrocytic glutamate transporters (EAAT1/GLAST and EAAT2/GLT1) [43]. (4c) Finally, glutamate is converted to glutamine by GS in the cytoplasm of astrocytes, thus completing the glutamine-glutamate (or glutamate-glutamine) cycle. (5a) Glutamine taken up by GABAergic neurons is converted to glutamate via PAG and then to GABA via glutamic acid decarboxylase isoforms 65 or 67 (GAD65/67) [44]. (5b) GABA is imported into synaptic vesicles via vesicular GABA transporters (VGAT) [45]. (5c) GABA is released from either the synaptic vesicles or from the cytoplasm through reversal of GABA transporters, then binds to extracellular GABA receptors, and is subsequently taken up by neurons and glial cells via GABA transporters [46, 47]. The GABA transporter subtype GAT3 (slc6a11) is preferentially present on astrocytes, whereas GAT1 and GAT2 are mainly expressed on neurons. (5d) GABA may then enter the TCA cycle as succinate, which eventually gives rise to alpha-ketoglutarate. Glutamate dehydrogenase (GDH) may further convert alpha-ketoglutarate to glutamate, which in astrocytes is metabolized to glutamine via GS [48]. This completes the glutamine-glutamate-GABA cycle.

Fig. 1.

Fig. 1

Diagram of GS and its role in the glutamine-glutamate-GABA cycle and ammonia metabolism. Key enzymes, transporter molecules, and metabolic intermediates are shown. Note the metabolic relationship among astrocytes, GABAergic neurons, and glutamatergic neurons as indicated by the glutamine-glutamate-GABA cycle (purple arrows to the left) and the glutamine-glutamate cycle (red arrows to the right). Abbreviations: ADP adenosine diphosphate, ASAT aspartate aminotransferase, ATP adenosine triphosphate, GABA γ-aminobutyric acid, coA coenzyme A, GAT3 GABA transporter subtype 3, GAD65/67 glutamate decarboxylase isoforms 65/67, GDH glutamate dehydrogenase, MCTs monocarboxylate transporters, PAG phosphate activated glutaminase, SAT1/2 system A transporter subtypes 1/2, SN1 system N transporter subtype 1, VGAT vesicular GABA transporter, VGLUT vesicular glutamate transporter

Importantly, the glutamine-glutamate-GABA cycle represents a simplistic view of a complex process involving numerous additional metabolic pathways closely linked to the cycle. Moreover, a highly efficient spatial arrangement of enzymes and transporter molecules in functional microdomains are likely to exist and adds to the complexity of the cycle concept. However, despite these limitations, the glutamine-glutamate-GABA cycle provides an attractive and testable reference framework for studies of brain metabolism and neurotransmission, as demonstrated by numerous seminal discoveries on the topic [49-51].

GS is Critical for Ammonia Detoxification in the CNS

Considerable amounts of ammonia are formed in the human body every day, mainly from the action of bacterial enzymes on colonic content and from the hydrolysis of glutamine in the small and large intestines [52]. Smaller amounts of ammonia are produced from breakdown of body protein and from metabolism of glutamine to glutamate via PAG [37]. Most of the ammonia derived from the intestines is taken up and degraded by the liver via the urea cycle and hepatic GS. However, even with normal liver function, as much as 35 μmol/L of ammonia can remain in the plasma. The concentration of plasma ammonia is even higher in cases of liver failure, and in patients with congenital defects in GS or the urea cycle. Because ammonia is neurotoxic and readily crosses the blood brain barrier, a system for efficient removal of brain ammonia is required [5, 53, 54]. The brain lacks a urea cycle, and therefore depends on the GS reaction in astrocytes for most of its ammonia removal [55]. The importance of astrocytes in ammonia detoxification is further emphasized by their anatomical relationship with the blood–brain barrier. The perivascular astrocyte endfeet, which surround the abluminal domain of microvessel-associated endothelial cells, constitute a metabolic “buffering” compartment between the blood and the rest of the brain. Thus, blood-derived ammonia enters the astrocytic compartment for metabolism by GS, limiting further progression into the neuronal compartment.

A number of CNS Disorders are Associated with Alterations in GS

Given the ubiquitous presence and discrete distribution of GS it is not surprising that alterations in the enzyme result in significant biological effects. However, it is only recently that the potential involvement of GS in human pathological conditions has begun to be appreciated. Haberle and colleagues reported two cases of congenital GS deficiency due to homozygous mutations in the GS gene (R324C and R341C) [9, 10]. The newborns had extensive brain malformations with almost no cerebral activity on EEG other than short bursts of theta waves and generalized seizures. They exhibited severe enteropathy and necrolytic erythema of the skin and died from cardiac failure and multi-organ failure 2 days and 4 weeks after birth [9, 10]. Glutamine was largely absent from their serum, urine, and cerebrospinal fluid. So far, only one patient with genetic GS deficiency has been known to live beyond early infancy despite having encephalopathy and seizures [56]. The profound morbidity associated with genetic deficiencies in GS is further demonstrated by studies of transgenic mice. Animals with prenatal excisions of the GS gene in all cell types die during early embryonic development [57], while mice with gene deletions selectively in GFAP-positive astrocytes live until postnatal day 3 [58].

Several cases of non-genetic (secondary) deficiencies in GS have been reported. Studies of brains from patients with Alzheimer’s disease demonstrate reduced activity of GS [11, 12], increased concentrations of oxidatively modified GS protein [11, 59, 60], and alterations in the cellular and subcellular distribution of GS protein [61, 62]. Brain GS mRNA is also reduced in patients with depression [63], in depressed suicide subjects [14, 15], and in non-depressed individuals who committed suicide [14]. Alterations in brain GS mRNA are also present in patients with schizophrenia [16]. Finally, we and others discovered that GS protein and activity are severely reduced in the hippocampal formation [18, 19] and amygdala [64] in patients with mesial temporal lobe epilepsy (MTLE). The significance of the latter findings will be discussed below.

GS is Deficient in Astrocytes in Patients with MTLE

MTLE is one of the most common forms of drug-resistant, localization-related epilepsies in humans. The seizures in MTLE appear to involve a network of temporal lobe and limbic structures including the hippocampal formation, amygdala, entorhinal cortex, lateral temporal neocortex, medial thalamus, and inferior frontal lobes [65]. Furthermore, one of the hallmarks of MTLE is a well-characterized pathology known as hippocampal sclerosis, which is recognized by a pattern of selective neuronal loss and glial alterations in the hippocampal formation [66-68].

Studies using in vivo brain microdialysis in patients with MTLE have shown that the sclerotic and epileptogenic hippocampal formation contains five-fold higher concentrations of extracellular glutamate interictally, when compared with the non-sclerotic and non-epileptogenic hippocampal formation [69, 70]. Moreover, during a seizure, the extracellular glutamate concentration increases six-fold above the interictal level in the hippocampal formation, and remains markedly elevated for several hours after the cessation of seizure activity [71]. Finally, the interictal extracellular glutamate concentration is considerably higher in patients with hippocampal sclerosis (MTLE) than in patients without this pathology (non-MTLE), despite the 60–80 % neuronal loss and doubling of glial density in the sclerotic hippocampal formation [70, 72-74]. The glutamate excess in MTLE is likely to be of critical importance for the pathophysiology of the disease due to the excitatory and excitotoxic properties of the neurotransmitter [7, 75-77]. Therefore, a key question is: What causes the glutamate excess in MTLE?

GS is severely deficient in the sclerotic hippocampal formation in patients with MTLE (Fig. 2) [18, 19]. Analysis of surgically resected brain tissue from patients with MTLE revealed a nearly 40 % loss of GS protein and enzyme activity in astrocytes in the sclerotic hippocampal formation. A similar loss of GS was not present in surgically resected hippocampal formations from patients with other types of temporal lobe epilepsy (non-MTLE) or in hippocampal formations from diseased subjects with no history of epilepsy [18]. These findings led us to hypothesize that a deficiency in glutamine synthetase in astrocytes slows the glutamate-glutamine cycle metabolism and causes chronic accumulation of glutamate in astrocytes and the extracellular space. Studies of patients with MTLE using magnetic resonance spectroscopy have supported this hypothesis by demonstrating slowed glutamine-glutamate cycling in the sclerotic versus the non-sclerotic hippocampal formation [78]. Furthermore, sustained inhibition of hippocampal glutamine synthetase using continuous infusion of methionine sulfoximine (MSO) in the hippocampal formation of rats results in recurrent seizures, increased concentrations of glutamate in hippocampal astrocytes, and neuropathological changes that resemble hippocampal sclerosis in some animals (Fig. 3) [17, 79, 80].

Fig. 2.

Fig. 2

GS is deficient in astrocytes in the hippocampal formation in patients with MTLE. a–c Immunohistochemistry of GS in the hippocampal formation from a non-epilepsy (autopsy control) subject reveals intense labeling for GS in the cell body and processes of astrocytes throughout the hippocampal formation (arrows in b and c). Note that the labeling extends into the finest and most distal astrocyte processes. d–f: In contrast, GS is virtually absent from astrocytes in several areas of the hippocampal formation in patients with MTLE, particularly in CA1 (f). In areas where GS is present (such as in the subiculum), the protein frequently does not extend into the most distal astrocyte processes (arrow in e). The loss of GS was confirmed by Western blots and enzyme activity studies. Scale bar in A = 0.5 mm (same magnification as d), B = 100 μm (same magnification as c, e and f). Abbreviations: alv alveus, CA1 hippocampal subfield Cornu Ammonis 1, dg dentate gyrus, sub subiculum. (Adapted from [18] and reproduced with permission from The Lancet/Elsevier B.V.)

Fig. 3.

Fig. 3

Chronic infusion of MSO into the hippocampal formation of rats causes inhibition of GS, and in some cases neuropathological changes that resemble hippocampal sclerosis as reported earlier [17, 80]. a–d: a silver staining technique for labeling degenerating neurons and fiber tracts reveals numerous stained (injured) neurons and fibers preferentially in area CA1 in a rat infused with MSO into the hippocampal formation (b, d) whereas a rat infused with 0.9 % NaCl does not reveal stained (injured) tissue elements (a, c). Magnification: A = B; C = D. Abbreviations: CA1-3 hippocampal subfields Cornu Ammonis 1-3, MSO methionine sulfoximine. (Adapted from [17] and reproduced with permission from Brain/Oxford University Press)

What are the Metabolic and Functional Consequences of Brain GS Deficiency?

Because GS is an integral part of a complex and highly compartmentalized metabolic system, it is likely that alterations in the expression and activity of GS will result in multifaceted metabolic and functional effects. Furthermore, due to the tight homeostatic control of most metabolic pathways, it is also plausible that alterations in GS are accompanied by one or more counter-regulatory (“compensatory”) mechanisms. Thus, to theoretically predict the downstream effect of alterations in GS is difficult, and only carefully designed studies are expected to accurately address this issue. We will therefore limit our discussion of possible consequences of GS deficiency to a few plausible scenarios that either have been or can be tested experimentally.

Effects on Glutamine, Glutamate, and GABA

Numerous studies of patients and animals with genetic GS deficiencies, and experiments involving chemical inhibition of GS through enzyme blockers such as methionine sulfoximine (MSO), have demonstrated that a loss of GS protein or enzyme activity leads to reduced synthesis of glutamine [10, 56, 57, 81, 82]. Because glutamine is critically involved in the glutamine-glutamate-GABA cycle, a highly relevant question is whether a lack of glutamine will affect the concentrations of glutamate and GABA, the main excitatory and inhibitory neurotransmitters in the brain.

It can be argued that loss of GS in the brain leads to decreased metabolism of glutamate to glutamine with accumulation of glutamate in the cytoplasm of astrocytes. Indeed, glutamate increases and glutamine decreases in astrocytes in organotypic slice cultures of the hippocampus after treatment with MSO [82]. Glutamate also increases in hippocampal astrocytes after chronic infusion of MSO into the hippocampus in rats [79]. Possible consequences of the increased astrocytic glutamate are: (1) impaired uptake of extracellular glutamate due to a reduced concentration gradient of the amino acid across the plasma membrane [18]; (2) release of glutamate into the extracellular space by astrocytes [83-85]; and (3) metabolism by oxidation of excess astrocytic glutamate via other pathways such as the TCA cycle [50, 86]. The contribution of each of these mechanisms to the increased astrocytic and extracellular fluid glutamate observed in vivo in MTLE remains to be established.

Glutamine is used for the synthesis of glutamate in neurons via the PAG reaction [37, 87]; hence, inhibition of GS in normal animals leads to acute decreases in neuronal and extracellular brain glutamate [81, 88, 89], similar to findings seen with the PAG inhibitor 6-diazo-5-oxo-L-norleucine [90]. These studies may appear to contradict our hypothesis that a deficiency in GS leads to increased extracellular glutamate in MTLE [18]. However, it should be noted that the cited studies were performed in normal animals using acute and widespread inhibition of GS by MSO. This situation is quite different from patients with drug-resistant MTLE which is characterized by several years (sometimes decades) of seizure activity, profound neuropathological changes in the hippocampal formation, and spatially restricted deficiencies in GS in the hippocampal formation (mainly in CA1, CA3, and the dentate hilus) [18, 19, 66, 67]. In human MTLE, it is possible that compensatory events specific to the epileptogenic hippocampal formation provide sufficient glutamine for neuronal synthesis of glutamate, perhaps by increased uptake of glutamine from the blood or via other mechanisms. Furthermore, the loss of GS in human MTLE is spatially restricted to specific areas such as CA1, CA3, and the dentate hilus, whereas other regions such as CA2 and subiculum are less affected and therefore may produce glutamine [18]. Recent studies have also suggested that GS is redistributed at the subcellular level in MTLE. In chronically epileptic pilocarpine-treated rats, GS is expressed more in proximal astrocytic branches than in distal branches, and GS expressing astrocytic cell bodies are located in closer proximity to vascular walls [91]. This finding suggests a highly compartmentalized action (and lack of action) of GS in the epileptogenic hippocampal formation with potentially different effects on the glutamine-glutamate metabolism in areas near microvessels compared to areas near synapses.

GABAergic neurotransmission is also influenced by inhibition of GS, partly because glutamine is a precursor for the synthesis of GABA. Blockade of GS by MSO results in decreased synthesis of neuronal GABA [92] and impaired release of synaptic GABA under physiological conditions [93]. Furthermore, inhibition of GS by MSO directly influences GABAergic neurotransmission by altering GABA(A) subunit composition; however, the mechanism and functional significance of this effect is not known [94].

Effects on Brain Excitability

Several studies indicate that inhibition of, or deficiency in GS can increase brain excitability and lead to seizures. More than 60 years ago it was observed that domestic animals developed seizures after being fed bleached (agenized) flour [95]. The bleaching process oxidized methionine in the flour to MSO [96], and later investigations demonstrated that systemic administration of MSO to rats and mice inhibits GS and results in severe convulsive seizures [97-100]. Moreover, chronic infusion of MSO locally into the hippocampus of rats results in inhibition of GS, and subsequent recurrent seizures [17, 80]. Finally, patients with mutations of the GS gene typically develop severe, intractable seizures [9, 10].

Although the studies described above strongly implicate a deficiency in GS in the pathophysiology of epilepsy, the definite role of GS in MTLE remains to be fully established. Importantly, most studies investigating the role of GS in epilepsy involve widespread (systemic) deficiencies in GS, whereas the deficiency observed in MTLE is highly localized to specific anatomical and subcellular compartments in the brain [18, 19, 64, 91]. The observation that intrahippocampal infusion of MSO in rats leads to a clinical and pathological state similar to MTLE suggests that a focal deficiency in brain GS is important in the disease process (Fig. 3) [17, 80]. However, the effects of MSO are not limited to GS inhibition, and it is possible that the convulsant properties of the chemical are caused by other mechanisms. For example, treatment of animals with MSO increases the content of glycogen in astrocytes [97] and depletes the tissue stores of glutathione [101]. Alterations in brain glutathione [102] and glycogen (discussed later) [98, 103] have been associated with seizures; however, the exact role of these compounds in epilepsy are not completely understood. MSO can also induce glutamate release independent of GS inhibition in cortical slices [104, 105]. Further studies, using more specific inhibitors or targeted gene knockouts of GS are therefore needed to fully address the role of GS inhibition in epilepsy.

Effects on Ammonia and Cell Swelling

Because GS is one of the key enzymes responsible for metabolism of brain ammonia, it is expected that ammonia is increased in areas of the brain where GS is deficient, such as the amygdala, and the hippocampal areas CA1, CA3, and dentate hilus in MTLE [18, 19, 64]. However, the metabolic and functional consequences of such highly localized ammonia elevations are not known, because most studies of brain ammonia have involved conditions of severe hyperammonemia, which is typically observed in conditions of liver failure (hepatic encephalopathy) and urea cycle defects. These studies have suggested that GS serves as an important and beneficial detoxifying enzyme for physiological amounts of brain ammonia. However, when plasma ammonia is present at high concentrations, metabolism via the GS pathway may be detrimental to the brain. This notion is based on the idea that the excessive amounts of glutamine produced by GS during hyperammonemia serves as a “Trojan horse” for the following reasons [106]. First, accumulation of glutamine in astrocytes leads to swelling of the cells with cytotoxic brain edema and possible release of glutamate as clinically important consequences [8, 107]. Second, the increased brain glutamine may be taken up by neurons and converted back to glutamate and ammonia via PAG, which is preferentially localized to neuronal mitochondria [37, 87, 108]. Accumulation of ammonia in mitochondria has been proposed to induce oxidative/nitrosative stress and result in opening of the mitochondrial transition pore. These events can lead to cell dysfunction and cell death [106]. Notably, administration of MSO to animal models of hyperammonemia ameliorates many of the toxic effects of this condition, suggesting that GS inhibition may in fact be neuroprotective during conditions of hyperammonemia [13]. The effects of GS inhibition are clearly multifaceted and further studies are required to assess the functional consequences of localized increases of brain ammonia thought to be present in MTLE.

Effects on Energy Metabolism

Interestingly, seizures induced acutely by systemic convulsant doses of MSO in ventilated rodents differ from those produced by GABA receptor antagonists (e.g., bicuculline, pentylenetetrazol, or flurothyl) in their effects on brain glucose and energy metabolism. Whereas GABA antagonists induce rapid loss of phosphocreatine and glycogen, and large increases in lactate (~6–8-fold)—all signs of intense glycolytic activation [109-114], MSO-induced seizures do not produce such changes. Instead, levels of phosphocreatine and ATP are maintained at or near normal during MSO-induced seizures, while levels of glucose and glycogen are elevated, with only a modest rise in lactate (<twofold) [98]. Because GS is a major pathway for free ammonia disposal, inhibition of GS by systemic injection of MSO leads to hyperammonemia, which in itself can induce convulsions [115]; MSO however, counteracts many of the effects of hyperammonemia such as astrocyte swelling and a rise in extracellular potassium ions and glutamine [13].

While the difference in metabolic response to MSO has led to questioning whether GS inhibition is related to seizure generation, we note that unlike GABA-antagonist induced seizures, which play out with an intact astroglial glutamate uptake and clearance system, i.e., an operational glutamine-glutamate-GABA cycle [116], MSO blocks the major pathway of glial glutamate clearance and neuronal glutamate/GABA replenishment. Thus, the increases in glucose and glycogen, and diminished lactate observed during MSO-induced seizures could be explained if the metabolic pathways are linked directly to the energetics of astroglial glutamate clearance [117-120].

An intriguing link between brain glycogen metabolism and glutamine synthesis is highlighted by the dramatic effect of MSO on glycogen levels. Glycogen is the major storage form of glucose, comprising a heterogeneously sized population of high molecular weight molecules, but is limited in supply to approximately 3–10 μmol/g. Glycogen is normally synthesized only in astrocytes [121, 122], but formation is seen in some neurons under pathophysiological conditions since the necessary enzymes are present. Glycogen levels reflect the concerted actions of glycogen synthase (E.C.2.4.1.11) and glycogen phosphorylase (E.C.2.4.1.1), and both enzymes are highly regulated via cyclic-AMP dependent protein phosphorylation involving protein kinases, phosphatases, and allosteric modulators, including IMP, AMP, and glucose. In MSO-treated mice the rise in glycogen at seizure-onset has been attributed to reduction in the activated form of phosphorylase, suggesting that decreased degradation is involved [103]. Glycogen accumulation is greatest in the perivascular endfeet [123], which is also a site of high expression of glutamate transporters and GS. Increased gluconeogenesis from glutamate has been proposed to explain MSO-induced glycogen accumulation [124], although a study using primary cultures of astrocytes found that direct synthesis of glycogen from glucose in the medium could account for this [120]. These investigators first noted the possible link between GS inhibition (and the interruption of the flow of glutamate-to-glutamine) and glycogen accumulation. Of note, MSO-induced glycogen accumulation was not seen in white matter [98], which has been shown to have much lower rates of glutamine-glutamate-GABA cycling compared to grey-matter regions that are enriched in nerve terminals [125]. MSO-induced glycogen accumulation is also much greater in vivo, where glutamine synthesis and cycling rates are high [126], in contrast to brain slices where only small increases were seen (14–28 %) [98, 127], an observation consistent with the slow rate of glutamine synthesis from neuron-derived glutamate reported in the unstimulated slice [128]. Noteworthy is the recent report of high glycogen levels in epileptic hippocampus of quickly frozen tissue removed during neurosurgery for intractable TLE [129], although its relation to neuropathology is unclear because only ‘normal’ appearing cortical grey and white matter (and not hippocampal tissue) was available for comparison. Thus, it remains to be shown whether glycogen levels are elevated in the human hippocampus in TLE.

Brain glycogen concentration shows an inverse relationship with activity. Higher levels are seen under deep anesthesia [130] and during sleep, with lower levels observed under waking states and sleep deprivation [131]. Glycogen is rapidly degraded during physiologic brain activation and its turnover is increased with activity [132-135]. In studies of the rat optic nerve in vitro, glucose deprivation leads to glycogen breakdown in astrocytes and formation of lactate, which can be utilized by axons [136]. The shunting of glucose through glycogen in astrocytes has been proposed as a possible explanation of the value of the oxygen-to-glucose index (OGI) and its change during increased activity [118], effectively linking the flow of glucose through astroglial glycogen as a source of rapid glycolytic ATP formation needed for the extrusion of sodium ions accumulated during glutamate uptake for rapid ECF glutamate clearance [117, 118, 135]. Astrocytes may also provide glycogen-derived glucose or lactate to neurons as oxidative fuels [137-139], as well as support de novo synthesis of glutamate and glutamine in astrocytes to replenish neuronal neurotransmitter pools [140-142].

Glycogen accumulation has been shown to delay the rate of propagation of spreading depression induced by insults such as high potassium or oxygen-glucose deprivation [127], which could play an important role in delaying the onset and spread of seizures [143, 144]. Thus, understanding the MSO-induced increase in glycogen levels could have potential therapeutic benefits. Because the chronic MSO-infused hippocampal formation in rats replicate some of the finding in human MTLE, future studies are needed to assess the levels of glycogen in this epilepsy model to more firmly link glial deficits in glutamine-glutamate cycling to glycogen and epilepsy.

Conclusions

GS is a ubiquitous and highly compartmentalized enzyme that is critically involved in several metabolic pathways, including the glutamine-glutamate-GABA cycle and metabolism of ammonia in the brain. Not surprisingly, an increasing number of studies suggest that alterations in the expression or activity of GS may underlie a variety of brain disorders, including Alzheimer’s disease, depression, schizophrenia, and epilepsy. However, because GS is an integral part of a complex metabolic system, the effects of GS perturbations are multifaceted and difficult to predict. Furthermore, the type of perturbation varies among each specific disorder, and includes alterations in GS mRNA, posttranslational modifications of the protein, decreased enzyme activity, redistribution of the protein within astrocytes, aberrant expression in neurons, and changes limited to specific areas of the brain. Controlled studies using targeted mutations of the GS gene, or highly specific, focally delivered enzyme inhibitors, are expected to better address the role of GS during physiological and pathological conditions. If a deficiency in GS is indeed critically involved in the development of brain disorders such as MTLE, then restoration of GS in astrocytes deficient in the enzyme, could represent a novel and potentially efficacious therapeutic approach.

Acknowledgments

The authors are supported by grants from the National Institutes of Health (NIH): NINDS K08 NS058674 and R01 NS070824 to TE and NIMH R01 MH095104 to KB. This work was also made possible by CTSA Grant Number UL1 RR024139 from the National Center for Advancing Translational Sciences (NCATS), a component of the National Institutes of Health (NIH), and NIH roadmap for Medical Research. The contents of the publication are solely the responsibility of the authors and do not necessarily represent the official view of NCATS or NIH.

Contributor Information

Tore Eid, Email: Tore.eid@yale.edu, Department of Laboratory Medicine, Yale University School of Medicine, 330 Cedar Street, P.O. Box 208035, New Haven, CT 06520-8035, USA.

Kevin Behar, Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06520-8035, USA.

Ronnie Dhaher, Department of Laboratory Medicine, Yale University School of Medicine, 330 Cedar Street, P.O. Box 208035, New Haven, CT 06520-8035, USA.

Argyle V. Bumanglag, Department of Neurosurgery, Yale University School of Medicine, New Haven, CT 06520-8035, USA

Tih-Shih W. Lee, Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06520-8035, USA

References

  • 1.Elliott WH. Adenosinetriphosphate in glutamine synthesis. Nature. 1948;161(4082):128. [PubMed] [Google Scholar]
  • 2.Speck JF. The enzymatic synthesis of glutamine, a reaction utilizing adenosine triphosphate. J Biol Chem. 1949;179(3):1405–1426. [PubMed] [Google Scholar]
  • 3.Wu C. Glutamine synthetase. I. A comparative study of its distribution in animals and its inhibition by Dl-Allo-Delta-Hydroxylysine. Comp Biochem Physiol. 1963;34:335–351. doi: 10.1016/0010-406x(63)90169-5. [DOI] [PubMed] [Google Scholar]
  • 4.Albrecht J, Jones EA. Hepatic encephalopathy: molecular mechanisms underlying the clinical syndrome. J Neurol Sci. 1999;170(2):138–146. doi: 10.1016/s0022-510x(99)00169-0. [DOI] [PubMed] [Google Scholar]
  • 5.Butterworth RF. Pathophysiology of hepatic encephalopathy: a new look at ammonia. Metab Brain Dis. 2002;17(4):221–227. doi: 10.1023/a:1021989230535. [DOI] [PubMed] [Google Scholar]
  • 6.Choi DW, Hartley DM. Calcium and glutamate-induced cortical neuronal death. In: Waxman SG, editor. Molecular and Cellular Approaches to the Treatment of Neurologic Disease. Raven Press; New York: 1993. [PubMed] [Google Scholar]
  • 7.Olney JW, Sharpe LG, Feigin RD. Glutamate-induced brain damage in infant primates. J Neuropathol Exp Neurol. 1972;31(3):464–488. doi: 10.1097/00005072-197207000-00006. [DOI] [PubMed] [Google Scholar]
  • 8.Norenberg MD, Jayakumar AR, Rama Rao KV, Panickar KS. New concepts in the mechanism of ammonia-induced astrocyte swelling. Metab Brain Dis. 2007;22(3–4):219–234. doi: 10.1007/s11011-007-9062-5. [DOI] [PubMed] [Google Scholar]
  • 9.Haberle J, Gorg B, Rutsch F, Schmidt E, Toutain A, Benoist JF, Gelot A, Suc AL, Hohne W, Schliess F, Haussinger D, Koch HG. Congenital glutamine deficiency with glutamine synthetase mutations. N Engl J Med. 2005;353(18):1926–1933. doi: 10.1056/NEJMoa050456. 10.1056/NEJMoa050456. [DOI] [PubMed] [Google Scholar]
  • 10.Haberle J, Gorg B, Toutain A, Rutsch F, Benoist JF, Gelot A, Suc AL, Koch HG, Schliess F, Haussinger D. Inborn error of amino acid synthesis: human glutamine synthetase deficiency. J Inherit Metab Dis. 2006;29(2–3):352–358. doi: 10.1007/s10545-006-0256-5. 10.1007/s10545-006-0256-5. [DOI] [PubMed] [Google Scholar]
  • 11.Hensley K, Hall N, Subramaniam R, Cole P, Harris M, Aksenov M, Aksenova M, Gabbita SP, Wu JF, Carney JM, et al. Brain regional correspondence between Alzheimer’s disease histopathology and biomarkers of protein oxidation. J Neurochem. 1995;65(5):2146–2156. doi: 10.1046/j.1471-4159.1995.65052146.x. [DOI] [PubMed] [Google Scholar]
  • 12.Smith CD, Carney JM, Starke-Reed PE, Oliver CN, Stadtman ER, Floyd RA, Markesbery WR. Excess brain protein oxidation and enzyme dysfunction in normal aging and in Alzheimer disease. Proc Natl Acad Sci USA. 1991;88(23):10540–10543. doi: 10.1073/pnas.88.23.10540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Brusilow SW, Koehler RC, Traystman RJ, Cooper AJ. Astrocyte glutamine synthetase: importance in hyperammonemic syndromes and potential target for therapy. Neurotherapeutics. 2010;7(4):452–470. doi: 10.1016/j.nurt.2010.05.015. 10.1016/j.nurt.2010.05.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Klempan TA, Sequeira A, Canetti L, Lalovic A, Ernst C, ffrench-Mullen J, Turecki G. Altered expression of genes involved in ATP biosynthesis and GABAergic neurotransmission in the ventral prefrontal cortex of suicides with and without major depression. Mol psychiatry. 2009;14(2):175–189. doi: 10.1038/sj.mp.4002110. 10.1038/sj.mp.4002110. [DOI] [PubMed] [Google Scholar]
  • 15.Sequeira A, Mamdani F, Ernst C, Vawter MP, Bunney WE, Lebel V, Rehal S, Klempan T, Gratton A, Benkelfat C, Rouleau GA, Mechawar N, Turecki G. Global brain gene expression analysis links glutamatergic and GABAergic alterations to suicide and major depression. PLoS ONE. 2009;4(8):e6585. doi: 10.1371/journal.pone.0006585. 10.1371/journal.pone.0006585. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Bruneau EG, McCullumsmith RE, Haroutunian V, Davis KL, Meador-Woodruff JH. Increased expression of glutaminase and glutamine synthetase mRNA in the thalamus in schizophrenia. Schizophr Res. 2005;75(1):27–34. doi: 10.1016/j.schres.2004.12.012. [DOI] [PubMed] [Google Scholar]
  • 17.Eid T, Ghosh A, Wang Y, Beckstrom H, Zaveri HP, Lee TS, Lai JC, Malthankar-Phatak GH, de Lanerolle NC. Recurrent seizures and brain pathology after inhibition of glutamine synthetase in the hippocampus in rats. Brain. 2008;131(Pt 8):2061–2070. doi: 10.1093/brain/awn133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Eid T, Thomas MJ, Spencer DD, Runden-Pran E, Lai JC, Malthankar GV, Kim JH, Danbolt NC, Ottersen OP, de Lanerolle NC. Loss of glutamine synthetase in the human epileptogenic hippocampus: possible mechanism for raised extracellular glutamate in mesial temporal lobe epilepsy. Lancet. 2004;363(9402):28–37. doi: 10.1016/s0140-6736(03)15166-5. [DOI] [PubMed] [Google Scholar]
  • 19.van der Hel WS, Notenboom RG, Bos IW, van Rijen PC, van Veelen CW, de Graan PN. Reduced glutamine synthetase in hippocampal areas with neuron loss in temporal lobe epilepsy. Neurology. 2005;64(2):326–333. doi: 10.1212/01.WNL.0000149636.44660.99. [DOI] [PubMed] [Google Scholar]
  • 20.Elliott WH. Studies on the enzymic synthesis of glutamine. Biochem J. 1951;49(1):106–112. doi: 10.1042/bj0490106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Fry BA. Glutamine synthesis by Micrococcus pyogenes var. aureus. Biochem J. 1955;59(4):579–589. doi: 10.1042/bj0590579. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Elliott WH. Isolation of glutamine synthetase and glutamotransferase from green peas. J Biol Chem. 1953;201(2):661–672. [PubMed] [Google Scholar]
  • 23.Tumani H, Shen GQ, Peter JB. Purification and immunocharacterization of human brain glutamine synthetase and its detection in cerebrospinal fluid and serum by a sandwich enzyme immunoassay. J Immunol Methods. 1995;188(1):155–163. doi: 10.1016/0022-1759(95)00215-4. [DOI] [PubMed] [Google Scholar]
  • 24.Kumada Y, Benson DR, Hillemann D, Hosted TJ, Rochefort DA, Thompson CJ, Wohlleben W, Tateno Y. Evolution of the glutamine synthetase gene, one of the oldest existing and functioning genes. Proc Natl Acad Sci USA. 1993;90(7):3009–3013. doi: 10.1073/pnas.90.7.3009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Wang Y, Kudoh J, Kubota R, Asakawa S, Minoshima S, Shimizu N. Chromosomal mapping of a family of human glutamine synthetase genes: functional gene (GLUL) on 1q25, pseudogene (GLULP) on 9p13, and three related genes (GLULL1, GLULL2, GLULL3) on 5q33, 11p15, and 11q24. G Genomics. 1996;37(2):195–199. doi: 10.1006/geno.1996.0542. [DOI] [PubMed] [Google Scholar]
  • 26.Krajewski WW, Collins R, Holmberg-Schiavone L, Jones TA, Karlberg T, Mowbray SL. Crystal structures of mammalian glutamine synthetases illustrate substrate-induced conformational changes and provide opportunities for drug and herbicide design. J Mol Biol. 2008;375(1):217–228. doi: 10.1016/j.jmb.2007.10.029. [DOI] [PubMed] [Google Scholar]
  • 27.Gebhardt R, Mecke D. Heterogeneous distribution of glutamine synthetase among rat liver parenchymal cells in situ and in primary culture. EMBO J. 1983;2(4):567–570. doi: 10.1002/j.1460-2075.1983.tb01464.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Blondet B, Hantaz-Ambroise D, Ait-Ikhlef A, Cambier D, Murawsky M, Rieger F. Astrocytosis in wobbler mouse spinal cord involves a population of astrocytes which is glutamine synthetase-negative. Neurosci Lett. 1995;183(3):179–182. doi: 10.1016/0304-3940(94)11145-9. [DOI] [PubMed] [Google Scholar]
  • 29.Norenberg MD. Distribution of glutamine synthetase in the rat central nervous system. J Histochem Cytochem. 1979;27:756–762. doi: 10.1177/27.3.39099. [DOI] [PubMed] [Google Scholar]
  • 30.Riepe RE, Norenberg MD. Glutamine synthetase in the developing rat retina: an immunohistochemical study. Exp Eye Res. 1978;27(4):435–444. doi: 10.1016/0014-4835(78)90022-2. [DOI] [PubMed] [Google Scholar]
  • 31.Derouiche A, Frotscher M. Astroglial processes around identified glutamatergic synapses contain glutamine synthetase: evidence for transmitter degradation. Brain Res. 1991;552(2):346–350. doi: 10.1016/0006-8993(91)90103-3. [DOI] [PubMed] [Google Scholar]
  • 32.Derouiche A, Ohm TG. Glutamine synthetase immunoreactivity in the human hippocampus is lamina-specific. Neurosci Lett. 1994;165(1–2):179–182. doi: 10.1016/0304-3940(94)90739-0. [DOI] [PubMed] [Google Scholar]
  • 33.Martinez-Hernandez A, Bell KP, Norenberg MD. Glutamine synthetase: glial localization in brain. Science. 1977;195(4284):1356–1358. doi: 10.1126/science.14400. [DOI] [PubMed] [Google Scholar]
  • 34.Chaudhry FA, Reimer RJ, Edwards RH. The glutamine commute: take the N line and transfer to the A. Journal cell Biol. 2002;157(3):349–355. doi: 10.1083/jcb.200201070. 10.1083/jcb.200201070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Jenstad M, Quazi AZ, Zilberter M, Haglerod C, Berghuis P, Saddique N, Goiny M, Buntup D, Davanger S, FM SH, Barnes CA, McNaughton BL, Ottersen OP, Storm-Mathisen J, Harkany T, Chaudhry FA. System A transporter SAT2 mediates replenishment of dendritic glutamate pools controlling retrograde signaling by glutamate. Cereb Cortex. 2009;19(5):1092–1106. doi: 10.1093/cercor/bhn151. 10.1093/cercor/bhn151. [DOI] [PubMed] [Google Scholar]
  • 36.Solbu TT, Bjorkmo M, Berghuis P, Harkany T, Chaudhry FA. SAT1, a glutamine transporter, is preferentially expressed in GABAergic neurons. Front Neuroanat. 2010;4:1. doi: 10.3389/neuro.05.001.2010. 10.3389/neuro.05.001.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Kvamme E, Roberg B, Torgner IA. Kinetics and localization of phosphate activated glutaminase. J Neurosci Res. 2001;66:951–958. doi: 10.1002/jnr.10041. [DOI] [PubMed] [Google Scholar]
  • 38.Svenneby G. Pig brain glutaminase: purification and identification of different enzyme forms. J Neurochem. 1970;17(11):1591–1599. doi: 10.1111/j.1471-4159.1970.tb03729.x. [DOI] [PubMed] [Google Scholar]
  • 39.Bellocchio EE, Hu H, Pohorille A, Chan J, Pickel VM, Edwards RH. The localization of the brain-specific inorganic phosphate transporter suggests a specific presynaptic role in glutamatergic transmission. J Neurosci. 1998;18(21):8648–8659. doi: 10.1523/JNEUROSCI.18-21-08648.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Fremeau RT, Jr, Burman J, Qureshi T, Tran CH, Proctor J, Johnson J, Zhang H, Sulzer D, Copenhagen DR, Storm-Mathisen J, Reimer RJ, Chaudhry FA, Edwards RH. The identification of vesicular glutamate transporter 3 suggests novel modes of signaling by glutamate. Proc Natl Acad Sci USA. 2002;99(22):14488–14493. doi: 10.1073/pnas.222546799. 10.1073/pnas.222546799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Fremeau RT, Jr, Troyer MD, Pahner I, Nygaard GO, Tran CH, Reimer RJ, Bellocchio EE, Fortin D, Storm-Mathisen J, Edwards RH. The expression of vesicular glutamate transporters defines two classes of excitatory synapse. Neuron. 2001;31(2):247–260. doi: 10.1016/s0896-6273(01)00344-0. [DOI] [PubMed] [Google Scholar]
  • 42.Takamori S, Malherbe P, Broger C, Jahn R. Molecular cloning and functional characterization of human vesicular glutamate transporter 3. EMBO Rep. 2002;3(8):798–803. doi: 10.1093/embo-reports/kvf159. 10.1093/embo-reports/kvf159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Danbolt NC. Glutamate uptake. Prog Neurobiol. 2001;65:1–105. doi: 10.1016/s0301-0082(00)00067-8. [DOI] [PubMed] [Google Scholar]
  • 44.Bu DF, Erlander MG, Hitz BC, Tillakaratne NJ, Kaufman DL, Wagner-McPherson CB, Evans GA, Tobin AJ. Two human glutamate decarboxylases, 65-kDa GAD and 67-kDa GAD, are each encoded by a single gene. Proc Natl Acad Sci USA. 1992;89(6):2115–2119. doi: 10.1073/pnas.89.6.2115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Jin H, Wu H, Osterhaus G, Wei J, Davis K, Sha D, Floor E, Hsu CC, Kopke RD, Wu JY. Demonstration of functional coupling between γ-aminobutyric acid (GABA) synthesis and vesicular GABA transport into synaptic vesicles. Proc Natl Acad Sci USA. 2003;100(7):4293–4298. doi: 10.1073/pnas.0730698100. 10.1073/pnas.0730698100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Eulenburg V, Gomeza J. Neurotransmitter transporters expressed in glial cells as regulators of synapse function. Brain Res Rev. 2010;63(1–2):103–112. doi: 10.1016/j.brainresrev.2010.01.003. 10.1016/j.brainresrev.2010.01.003. [DOI] [PubMed] [Google Scholar]
  • 47.Richerson GB, Wu Y. Dynamic equilibrium of neuro-transmitter transporters: not just for reuptake anymore. J Neurophysiol. 2003;90(3):1363–1374. doi: 10.1152/jn.00317.2003. 10.1152/jn.00317.2003. [DOI] [PubMed] [Google Scholar]
  • 48.Dennis SC, Lai JC, Clark JB. Comparative studies on glutamate metabolism in synpatic and non-synaptic rat brain mitochondria. Biochem J. 1977;164(3):727–736. doi: 10.1042/bj1640727. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Albrecht J, Sidoryk-Wegrzynowicz M, Zielinska M, Aschner M. Roles of glutamine in neurotransmission. Neuron Glia Biol. 2011;6(4):263–276. doi: 10.1017/S1740925X11000093. [DOI] [PubMed] [Google Scholar]
  • 50.Hertz L. Astrocytic energy metabolism and glutamate formation–relevance for 13C-NMR spectroscopy and importance of cytosolic/mitochondrial trafficking. Magn Reson Imaging. 2011;29(10):1319–1329. doi: 10.1016/j.mri.2011.04.013. 10.1016/j.mri.2011.04.013. [DOI] [PubMed] [Google Scholar]
  • 51.Rothman DL, De Feyter HM, de Graaf RA, Mason GF, Behar KL. 13C MRS studies of neuroenergetics and neurotransmitter cycling in humans. NMR Biomed. 2011;24(8):943–957. doi: 10.1002/nbm.1772. 10.1002/nbm.1772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Kirk E. Amino acid metabolism in liver disease. Acta Med Scand Suppl. 1936;77:1–147. [Google Scholar]
  • 53.Gabuzda GJ, Jr, Phillips GB, Davidson CS. Reversible toxic manifestations in patients with cirrhosis of the liver given cation-exchange resins. N Engl J Med. 1952;246(4):124–130. doi: 10.1056/NEJM195201242460402. 10.1056/NEJM195201242460402. [DOI] [PubMed] [Google Scholar]
  • 54.Phillips GB, Schwartz R, Gabuzda GJ, Jr, Davidson CS. The syndrome of impending hepatic coma in patients with cirrhosis of the liver given certain nitrogenous substances. N Engl J Med. 1952;247(7):239–246. doi: 10.1056/NEJM195208142470703. 10.1056/NEJM195208142470703. [DOI] [PubMed] [Google Scholar]
  • 55.Cooper AJ, Plum F. Biochemistry and physiology of brain ammonia. Physiol Rev. 1987;67(2):440–519. doi: 10.1152/physrev.1987.67.2.440. [DOI] [PubMed] [Google Scholar]
  • 56.Haberle J, Shahbeck N, Ibrahim K, Hoffmann GF, Ben-Omran T. Natural course of glutamine synthetase deficiency in a 3 years old patient. Mol Genet Metab. 2011;103(1):89–91. doi: 10.1016/j.ymgme.2011.02.001. 10.1016/j.ymgme.2011.02.001. [DOI] [PubMed] [Google Scholar]
  • 57.He Y, Hakvoort TB, Vermeulen JL, Lamers WH, Van Roon MA. Glutamine synthetase is essential in early mouse embryogenesis. Dev Dyn. 2007;236(7):1865–1875. doi: 10.1002/dvdy.21185. 10.1002/dvdy.21185. [DOI] [PubMed] [Google Scholar]
  • 58.He Y, Hakvoort TB, Vermeulen JL, Labruyere WT, De Waart DR, Van Der Hel WS, Ruijter JM, Uylings HB, Lamers WH. Glutamine synthetase deficiency in murine astrocytes results in neonatal death. Glia. 2010;58(6):741–754. doi: 10.1002/glia.20960. 10.1002/glia.20960. [DOI] [PubMed] [Google Scholar]
  • 59.Butterfield DA, Hensley K, Cole P, Subramaniam R, Aksenov M, Aksenova M, Bummer PM, Haley BE, Carney JM. Oxidatively induced structural alteration of glutamine synthetase assessed by analysis of spin label incorporation kinetics: relevance to Alzheimer’s disease. J Neurochem. 1997;68(6):2451–2457. doi: 10.1046/j.1471-4159.1997.68062451.x. [DOI] [PubMed] [Google Scholar]
  • 60.Butterfield DA, Poon HF, St Clair D, Keller JN, Pierce WM, Klein JB, Markesbery WR. Redox proteomics identification of oxidatively modified hippocampal proteins in mild cognitive impairment: insights into the development of Alzheimer’s disease. Neurobiol Dis. 2006;22(2):223–232. doi: 10.1016/j.nbd.2005.11.002. [DOI] [PubMed] [Google Scholar]
  • 61.Robinson SR. Neuronal expression of glutamine synthetase in Alzheimer’s disease indicates a profound impairment of metabolic interactions with astrocytes. Neurochem Int. 2000;36(4–5):471–482. doi: 10.1016/s0197-0186(99)00150-3. [DOI] [PubMed] [Google Scholar]
  • 62.Robinson SR. Changes in the cellular distribution of glutamine synthetase in Alzheimer’s disease. J Neurosci Res. 2001;66(5):972–980. doi: 10.1002/jnr.10057. [DOI] [PubMed] [Google Scholar]
  • 63.Choudary PV, Molnar M, Evans SJ, Tomita H, Li JZ, Vawter MP, Myers RM, Bunney WE, Jr, Akil H, Watson SJ, Jones EG. Altered cortical glutamatergic and GABAergic signal transmission with glial involvement in depression. Proc Natl Acad Sci USA. 2005;102(43):15653–15658. doi: 10.1073/pnas.0507901102. 10.1073/pnas.0507901102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Steffens M, Huppertz HJ, Zentner J, Chauzit E, Feuerstein TJ. Unchanged glutamine synthetase activity and increased NMDA receptor density in epileptic human neocortex: implications for the pathophysiology of epilepsy. Neurochem Int. 2005;47(6):379–384. doi: 10.1016/j.neuint.2005.06.001. [DOI] [PubMed] [Google Scholar]
  • 65.Spencer SS. Neural networks in human epilepsy: evidence of and implications for treatment. Epilepsia. 2002;43(3):219–227. doi: 10.1046/j.1528-1157.2002.26901.x. [DOI] [PubMed] [Google Scholar]
  • 66.Gloor P. Mesial temporal sclerosis: Historical background and an overview from a modern perspective. In: Luders H, editor. Epilepsy Surgery. Raven Press; New York: 1991. pp. 689–703. [Google Scholar]
  • 67.Sommer W. Erkrankung des Ammonshorns als aetiologisches Moment der Epilepsie. Arch Psychiatr Nervenkr. 1880;10:631–675. [Google Scholar]
  • 68.Margerison JH, Corsellis JAN. Epilepsy and the temporal lobes. Brain. 1966;89:499–530. doi: 10.1093/brain/89.3.499. [DOI] [PubMed] [Google Scholar]
  • 69.Cavus I, Kasoff WS, Cassaday MP, Jacob R, Gueorguieva R, Sherwin RS, Krystal JH, Spencer DD, Abi-Saab WM. Extracellular metabolites in the cortex and hippocampus of epileptic patients. Ann Neurol. 2005;57(2):226–235. doi: 10.1002/ana.20380. [DOI] [PubMed] [Google Scholar]
  • 70.Cavus I, Pan JW, Hetherington HP, Abi-Saab W, Zaveri HP, Vives KP, Krystal JH, Spencer SS, Spencer DD. Decreased hippocampal volume on MRI is associated with increased extracellular glutamate in epilepsy patients. Epilepsia. 2008;49(8):1358–1366. doi: 10.1111/j.1528-1167.2008.01603.x. [DOI] [PubMed] [Google Scholar]
  • 71.During MJ, Spencer DD. Extracellular hippocampal glutamate and spontaneous seizure in the conscious human brain. Lancet. 1993;341(8861):1607–1610. doi: 10.1016/0140-6736(93)90754-5. [DOI] [PubMed] [Google Scholar]
  • 72.Kim JH, Je S, Petroff OA, Spencer SS, Hwang JY, Spencer DD. Hippocampal glial density in temporal lobe epilepsy. Epilepsia. 2004;45(S7):S33–S34. [Google Scholar]
  • 73.Petroff OA, Cavus I, Kim JH, Spencer DD. Interictal extracellular glutamate concentrations are increased in hippocampal sclerosis. Ann Neurol. 2004;56(S8):S43. [Google Scholar]
  • 74.Petroff OA, Errante LD, Kim JH, Spencer DD. N-acetyl-aspartate, total creatine, and myo-inositol in the epileptogenic human hippocampus. Neurology. 2003;60(10):1646–1651. doi: 10.1212/01.wnl.0000068020.85450.8b. [DOI] [PubMed] [Google Scholar]
  • 75.Ben-Ari Y. Limbic seizure and brain damage produced by kainic acid: mechanisms and relevance to human temporal lobe epilepsy. Neuroscience. 1985;14(2):375–403. doi: 10.1016/0306-4522(85)90299-4. [DOI] [PubMed] [Google Scholar]
  • 76.Nadler J, Cuthbertson G. Kainic acid neurotoxicity toward the hippocampal formation: dependence on specific excitatory pathways. Brain Res. 1980;195:47–56. doi: 10.1016/0006-8993(80)90865-3. [DOI] [PubMed] [Google Scholar]
  • 77.Olney JW. Neurotoxicity of excitatory amino acids. In: McGeer EG, Olney JW, McGeer PL, editors. Kainic Acid as a Tool in Neurobiology. Raven Press; New York: 1978. pp. 37–70. [Google Scholar]
  • 78.Petroff OA, Errante LD, Rothman DL, Kim JH, Spencer DD. Glutamate-glutamine cycling in the epileptic human hippocampus. Epilepsia. 2002;43(7):703–710. doi: 10.1046/j.1528-1157.2002.38901.x. [DOI] [PubMed] [Google Scholar]
  • 79.Perez E, Lauritzen F, Wang Y, Kang D, Ottersen OP, Eid T. Ultrastructural distribution of glutamate in the glutamine-synthetase-deficient epileptogenic rat hippocampus. Neurosci Meet Plan. 2010;254:220. [Google Scholar]
  • 80.Wang Y, Zaveri HP, Lee TS, Eid T. The development of recurrent seizures after continuous intrahippocampal infusion of methionine sulfoximine in rats: a video-intracranial electroencephalographic study. Exp Neurol. 2009;220(2):293–302. doi: 10.1016/j.expneurol.2009.08.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Bottcher T, Goiny M, Bering J, Domhof S, Nau R, Ungerstedt U. Regional differences in glutamine synthetase inhibition by l-methionine sulfoximine: a microdialysis study in the rabbit brain. Exp Brain Res. 2003;150(2):194–200. doi: 10.1007/s00221-003-1401-0. [DOI] [PubMed] [Google Scholar]
  • 82.Laake JH, Slyngstad TA, Haug FM, Ottersen OP. Glutamine from glial cells is essential for the maintenance of the nerve terminal pool of glutamate: immunogold evidence from hippocampal slice cultures. J Neurochem. 1995;65(2):871–881. doi: 10.1046/j.1471-4159.1995.65020871.x. [DOI] [PubMed] [Google Scholar]
  • 83.Fellin T, Pascual O, Gobbo S, Pozzan T, Haydon PG, Carmignoto G. Neuronal synchrony mediated by astrocytic glutamate through activation of extrasynaptic NMDA receptors. Neuron. 2004;43(5):729–743. doi: 10.1016/j.neuron.2004.08.011. [DOI] [PubMed] [Google Scholar]
  • 84.Hamilton NB, Attwell D. Do astrocytes really exocytose neurotransmitters? Nat Rev Neurosci. 2010;11(4):227–238. doi: 10.1038/nrn2803. 10.1038/nrn2803. [DOI] [PubMed] [Google Scholar]
  • 85.Tian GF, Azmi H, Takano T, Xu Q, Peng W, Lin J, Oberheim N, Lou N, Wang X, Zielke HR, Kang J, Nedergaard M. An astrocytic basis of epilepsy. Nat Med. 2005;11(9):973–981. doi: 10.1038/nm1277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.McKenna MC, Sonnewald U, Huang X, Stevenson J, Zielke HR. Exogenous glutamate concentration regulates the metabolic fate of glutamate in astrocytes. J Neurochem. 1996;66(1):386–393. doi: 10.1046/j.1471-4159.1996.66010386.x. [DOI] [PubMed] [Google Scholar]
  • 87.Kvamme E, Roberg B, Torgner IA. Glutamine transport in brain mitochondria. Neurochem Int. 2000;37(2–3):131–138. doi: 10.1016/s0197-0186(00)00016-4. [DOI] [PubMed] [Google Scholar]
  • 88.Paulsen RE, Fonnum F. Role of glial cells for the basal and Ca2+-dependent K+-evoked release of transmitter amino acids investigated by microdialysis. J Neurochem. 1989;52(6):1823–1829. doi: 10.1111/j.1471-4159.1989.tb07263.x. [DOI] [PubMed] [Google Scholar]
  • 89.Rothstein JD, Tabakoff B. Alteration of striatal glutamate release after glutamine synthetase inhibition. J Neurochem. 1984;43(5):1438–1446. doi: 10.1111/j.1471-4159.1984.tb05406.x. [DOI] [PubMed] [Google Scholar]
  • 90.Conti F, Minelli A. Glutamate immunoreactivity in rat cerebral cortex is reversibly abolished by 6-diazo-5-oxo-L-norleucine (DON), an inhibitor of phosphate-activated glutaminase. J Histochem Cytochem. 1994;42(6):717–726. doi: 10.1177/42.6.7910617. [DOI] [PubMed] [Google Scholar]
  • 91.Papageorgiou IE, Gabriel S, Fetani AF, Kann O, Heinemann U. Redistribution of astrocytic glutamine synthetase in the hippocampus of chronic epileptic rats. Glia. 2011;59(11):1706–1718. doi: 10.1002/glia.21217. 10.1002/glia.21217. [DOI] [PubMed] [Google Scholar]
  • 92.Sonnewald U, Westergaard N, Schousboe A, Svendsen JS, Unsgard G, Petersen SB. Direct demonstration by [13C]NMR spectroscopy that glutamine from astrocytes is a precursor for GABA synthesis in neurons. Neurochem Int. 1993;22(1):19–29. doi: 10.1016/0197-0186(93)90064-c. [DOI] [PubMed] [Google Scholar]
  • 93.Liang SL, Carlson GC, Coulter DA. Dynamic regulation of synaptic GABA release by the glutamate-glutamine cycle in hippocampal area CA1. J Neurosci. 2006;26(33):8537–8548. doi: 10.1523/JNEUROSCI.0329-06.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Cremer CM, Bidmon HJ, Gorg B, Palomero-Gallagher N, Escobar JL, Speckmann EJ, Zilles K. Inhibition of glutamate/glutamine cycle in vivo results in decreased benzodiazepine binding and differentially regulated GABAergic subunit expression in the rat brain. Epilepsia. 2010 doi: 10.1111/j.1528-1167.2010.02562.x. 10.1111/j.1528-1167.2010.02562.x. [DOI] [PubMed] [Google Scholar]
  • 95.Silver ML, Johnson RE, et al. White bread and epilepsy in animals. J Am Med Assoc. 1947;135(12):757–760. doi: 10.1001/jama.1947.02890120011005. [DOI] [PubMed] [Google Scholar]
  • 96.Campbell PN, Work TS, Mellanby E. The isolation of a toxic substance from agenized wheat flour. Biochem J. 1951;48(1):106–113. doi: 10.1042/bj0480106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Bernard-Helary K, Ardourel MY, Hevor T, Cloix JF. In vivo and in vitro glycogenic effects of methionine sulfoximine are different in two inbred strains of mice. Brain Res. 2002;929(2):147–155. doi: 10.1016/s0006-8993(01)03380-7. [DOI] [PubMed] [Google Scholar]
  • 98.Folbergrova J, Passonneau JV, Lowry OH, Schulz DW. Glycogen, ammonia and related metabolities in the brain during seizures evoked by methionine sulphoximine. J Neurochem. 1969;16(2):191–203. doi: 10.1111/j.1471-4159.1969.tb05937.x. [DOI] [PubMed] [Google Scholar]
  • 99.Rowe WB, Meister A. Identification of l-methionine-S-sulfoximine as the convulsant isomer of methionine sulfoximine. Proc Natl Acad Sci USA. 1970;66(2):500–506. doi: 10.1073/pnas.66.2.500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Sellinger OZ, Azcurra JM, Ohlsson WG. Methionine sulfoximine seizures. 8. The dissociation of the convulsant and glutamine synthetase inhibitory effects. J Pharmacol Exp Ther. 1968;164(1):212–222. [PubMed] [Google Scholar]
  • 101.Shaw CA, Bains JS. Synergistic versus antagonistic actions of glutamate and glutathione: the role of excitotoxicity and oxidative stress in neuronal disease. Cell Mol Biol (Noisyle-grand) 2002;48(2):127–136. [PubMed] [Google Scholar]
  • 102.Prigol M, Bruning CA, Nogueira CW, Zeni G. The role of the glutathione system in seizures induced by diphenyl diselenide in rat pups. Chem Biol Interact. 2011;193(1):65–70. doi: 10.1016/j.cbi.2011.05.004. 10.1016/j.cbi.2011.05.004. [DOI] [PubMed] [Google Scholar]
  • 103.Folbergrova J. Glycogen and glycogen phosphorylase in the cerebral cortex of mice under the influence of methionine sulphoximine. J Neurochem. 1973;20(2):547–557. doi: 10.1111/j.1471-4159.1973.tb12154.x. [DOI] [PubMed] [Google Scholar]
  • 104.Kam K, Nicoll R. Excitatory synaptic transmission persists independently of the glutamate-glutamine cycle. J Neurosci. 2007;27(34):9192–9200. doi: 10.1523/JNEUROSCI.1198-07.2007. 10.1523/JNEUROSCI.1198-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Shaw CA, Bains JS, Pasqualotto BA, Curry K. Methionine sulfoximine shows excitotoxic actions in rat cortical slices. Can J Physiol Pharmacol. 1999;77(11):871–877. [PubMed] [Google Scholar]
  • 106.Albrecht J, Norenberg MD. Glutamine: a Trojan horse in ammonia neurotoxicity. Hepatology (Baltimore, Md) 2006;44(4):788–794. doi: 10.1002/hep.21357. [DOI] [PubMed] [Google Scholar]
  • 107.Kimelberg HK, Goderie SK, Higman S, Pang S, Waniewski RA. Swelling-induced release of glutamate, aspartate, and taurine from astrocyte cultures. J Neurosci. 1990;10:1583–1591. doi: 10.1523/JNEUROSCI.10-05-01583.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Laake JH, Takumi Y, Eidet J, Torgner IA, Roberg B, Kvamme E, Ottersen OP. Postembedding immunogold labelling reveals subcellular localization and pathway-specific enrichment of phosphate activated glutaminase in rat cerebellum. Neurosci. 1999;88:1137–1151. doi: 10.1016/s0306-4522(98)00298-x. [DOI] [PubMed] [Google Scholar]
  • 109.Chapman AG, Evans MC. Cortical GABA turnover during bicuculline seizures in rats. J Neurochem. 1983;41(3):886–889. doi: 10.1111/j.1471-4159.1983.tb04823.x. [DOI] [PubMed] [Google Scholar]
  • 110.Duffy TE, Howse DC, Plum F. Cerebral energy metabolism during experimental status epilepticus. J Neurochem. 1975;24(5):925–934. doi: 10.1111/j.1471-4159.1975.tb03657.x. [DOI] [PubMed] [Google Scholar]
  • 111.Folbergrova J, Ingvar M, Nevander G, Siesjo BK. Cerebral metabolic changes during and following fluorothyl-induced seizures in ventilated rats. J Neurochem. 1985;44(5):1419–1426. doi: 10.1111/j.1471-4159.1985.tb08778.x. [DOI] [PubMed] [Google Scholar]
  • 112.Ingvar M, Soderfeldt B, Folbergrova J, Kalimo H, Olsson Y, Siesjo BK. Metabolic, circulatory, and structural alterations in the rat brain induced by sustained pentylenetetrazole seizures. Epilepsia. 1984;25(2):191–204. doi: 10.1111/j.1528-1157.1984.tb04176.x. [DOI] [PubMed] [Google Scholar]
  • 113.Petroff OA, Prichard JW, Behar KL, Alger JR, Shulman RG. In vivo phosphorus nuclear magnetic resonance spectroscopy in status epilepticus. Ann Neurol. 1984;16(2):169–177. doi: 10.1002/ana.410160203. 10.1002/ana.410160203. [DOI] [PubMed] [Google Scholar]
  • 114.Young RS, Chen B, Petroff OA, Gore JC, Cowan BE, Novotny EJ, Jr, Wong M, Zuckerman K. The effect of diazepam on neonatal seizure: in vivo 31P and 1H NMR study. Pediatr Res. 1989;25(1):27–31. doi: 10.1203/00006450-198901000-00006. [DOI] [PubMed] [Google Scholar]
  • 115.Voorhies TM, Ehrlich ME, Duffy TE, Petito CK, Plum F. Acute hyperammonemia in the young primate: physiologic and neuropathologic correlates. Pediatr Res. 1983;17(12):970–975. doi: 10.1203/00006450-198312000-00009. [DOI] [PubMed] [Google Scholar]
  • 116.Patel AB, de Graaf RA, Mason GF, Kanamatsu T, Rothman DL, Shulman GI, Behar KL. Glutamatergic neurotransmission and neuronal glucose oxidation are coupled during intense neuronal activation. J Cereb Blood Flow Metabol. 2004;24(9):972–985. doi: 10.1097/01.WCB.0000126234.16188.71. 00004647-200409000-00003. [DOI] [PubMed] [Google Scholar]
  • 117.Pellerin L, Magistretti PJ. Glutamate uptake into astrocytes stimulates aerobic glycolysis: a mechanism coupling neuronal activity to glucose utilization. Proc Natl Acad Sci USA. 1994;91(22):10625–10629. doi: 10.1073/pnas.91.22.10625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Shulman RG, Hyder F, Rothman DL. Cerebral energetics and the glycogen shunt: neurochemical basis of functional imaging. Proc Natl Acad Sci USA. 2001;98(11):6417–6422. doi: 10.1073/pnas.101129298. 10.1073/pnas.101129298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Sibson NR, Mason GF, Shen J, Cline GW, Herskovits AZ, Wall JE, Behar KL, Rothman DL, Shulman RG. In vivo 13C NMR measurement of neurotransmitter glutamate cycling, anaplerosis and TCA cycle flux in rat brain during [2-13C] glucose infusion. J Neurochem. 2001;76(4):975–989. doi: 10.1046/j.1471-4159.2001.00074.x. [DOI] [PubMed] [Google Scholar]
  • 120.Swanson RA, Yu ACH, Chan PH, Sharp FR. Glutamate increases glycogen content and reduces glucose utilization in primary astrocyte culture. J Neurochem. 1990;54:490–496. doi: 10.1111/j.1471-4159.1990.tb01898.x. [DOI] [PubMed] [Google Scholar]
  • 121.Brown AM, Ransom BR. Astrocyte glycogen and brain energy metabolism. Glia. 2007;55(12):1263–1271. doi: 10.1002/glia.20557. [DOI] [PubMed] [Google Scholar]
  • 122.Gruetter R. Glycogen: the forgotten cerebral energy store. J Neurosci Res. 2003;74(2):179–183. doi: 10.1002/jnr.10785. 10.1002/jnr.10785. [DOI] [PubMed] [Google Scholar]
  • 123.Willard-Mack CL, Koehler RC, Hirata T, Cork LC, Takahashi H, Traystman RJ, Brusilow SW. Inhibition of glutamine synthetase reduces ammonia-induced astrocyte swelling in rat. Neuroscience. 1996;71(2):589–599. doi: 10.1016/0306-4522(95)00462-9. [DOI] [PubMed] [Google Scholar]
  • 124.Hevor TK, Delorme P, Beauvillain JC. Glycogen synthesis and immunocytochemical study of fructose-1,6-biphosphatase in methionine sulfoximine epileptogenic rodent brain. J Cereb Blood Flow Metab. 1986;6(3):292–297. doi: 10.1038/jcbfm.1986.51. 10.1038/jcbfm.1986.51. [DOI] [PubMed] [Google Scholar]
  • 125.de Graaf RA, Mason GF, Patel AB, Rothman DL, Behar KL. Regional glucose metabolism and glutamatergic neuro-transmission in rat brain in vivo. Proc Natl Acad Sci USA. 2004;101(34):12700–12705. doi: 10.1073/pnas.0405065101. 10.1073/pnas.0405065101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Sibson NR, Dhankhar A, Mason GF, Rothman DL, Behar KL, Shulman RG. Stoichiometric coupling of brain glucose metabolism and glutamatergic neuronal activity. Proc Natl Acad Sci USA. 1998;95(1):316–321. doi: 10.1073/pnas.95.1.316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Seidel JL, Shuttleworth CW. Contribution of astrocyte glycogen stores to progression of spreading depression and related events in hippocampal slices. Neuroscience. 2011;192:295–303. doi: 10.1016/j.neuroscience.2011.05.006. 10.1016/j.neuroscience.2011.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Badar-Goffer RS, Ben-Yoseph O, Bachelard HS, Morris PG. Neuronal-glial metabolism under depolarizing conditions. A 13C-n.m.r. study. Biochem J. 1992;282(1):225–230. doi: 10.1042/bj2820225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Dalsgaard MK, Madsen FF, Secher NH, Laursen H, Quistorff B. High glycogen levels in the hippocampus of patients with epilepsy. J Cereb Blood Flow Metab. 2007;27(6):1137–1141. doi: 10.1038/sj.jcbfm.9600426. [DOI] [PubMed] [Google Scholar]
  • 130.Phelps CH. Barbiturate-induced glycogen accumulation in brain. An electron microscopic study Brain Res. 1972;39(1):225–234. doi: 10.1016/0006-8993(72)90797-4. [DOI] [PubMed] [Google Scholar]
  • 131.Kong J, Shepel PN, Holden CP, Mackiewicz M, Pack AI, Geiger JD. Brain glycogen decreases with increased periods of wakefulness: implications for homeostatic drive to sleep. J Neurosci. 2002;22(13):5581–5587. doi: 10.1523/JNEUROSCI.22-13-05581.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Dienel GA, Ball KK, Cruz NF. A glycogen phosphorylase inhibitor selectively enhances local rates of glucose utilization in brain during sensory stimulation of conscious rats: implications for glycogen turnover. J Neurochem. 2007;102(2):466–478. doi: 10.1111/j.1471-4159.2007.04595.x. 10.1111/j.1471-4159.2007.04595.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Orkand PM, Bracho H, Orkand RK. Glial metabolism: alteration by potassium levels comparable to those during neural activity. Brain Res. 1973;55(2):467–471. doi: 10.1016/0006-8993(73)90315-6. [DOI] [PubMed] [Google Scholar]
  • 134.Swanson RA. Physiologic coupling of glial glycogen metabolism to neuronal activity in brain. Can J Physiol Pharmacol. 1992;70(Suppl):S138–S144. doi: 10.1139/y92-255. [DOI] [PubMed] [Google Scholar]
  • 135.Swanson RA, Morton MM, Sagar SM, Sharp FR. Sensory stimulation induces local cerebral glycogenolysis: demonstration by autoradiography. Neuroscience. 1992;51(2):451–461. doi: 10.1016/0306-4522(92)90329-z. [DOI] [PubMed] [Google Scholar]
  • 136.Wender R, Brown AM, Fern R, Swanson RA, Farrell K, Ransom BR. Astrocytic glycogen influences axon function and survival during glucose deprivation in central white matter. J Neurosci. 2000;20(18):6804–6810. doi: 10.1523/JNEUROSCI.20-18-06804.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.DiNuzzo M, Maraviglia B, Giove F. Why does the brain (not) have glycogen? BioEssays. 2011;33(5):319–326. doi: 10.1002/bies.201000151. 10.1002/bies.201000151. [DOI] [PubMed] [Google Scholar]
  • 138.Rossi DJ, Brady JD, Mohr C. Astrocyte metabolism and signaling during brain ischemia. Nat Neurosci. 2007;10(11):1377–1386. doi: 10.1038/nn2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Tekkok SB, Brown AM, Westenbroek R, Pellerin L, Ransom BR. Transfer of glycogen-derived lactate from astrocytes to axons via specific monocarboxylate transporters supports mouse optic nerve activity. J Neurosci Res. 2005;81(5):644–652. doi: 10.1002/jnr.20573. 10.1002/jnr.20573. [DOI] [PubMed] [Google Scholar]
  • 140.Gibbs ME, Anderson DG, Hertz L. Inhibition of glycogenolysis in astrocytes interrupts memory consolidation in young chickens. Glia. 2006;54(3):214–222. doi: 10.1002/glia.20377. 10.1002/glia.20377. [DOI] [PubMed] [Google Scholar]
  • 141.Gibbs ME, Lloyd HG, Santa T, Hertz L. Glycogen is a preferred glutamate precursor during learning in 1-day-old chick: biochemical and behavioral evidence. J Neurosci Res. 2007;85(15):3326–3333. doi: 10.1002/jnr.21307. 10.1002/jnr.21307. [DOI] [PubMed] [Google Scholar]
  • 142.Hertz L, O’Dowd BS, Ng KT, Gibbs ME. Reciprocal changes in forebrain contents of glycogen and of glutamate/glutamine during early memory consolidation in the day-old chick. Brain Res. 2003;994(2):226–233. doi: 10.1016/j.brainres.2003.09.044. [DOI] [PubMed] [Google Scholar]
  • 143.Bernard-Helary K, Lapouble E, Ardourel M, Hevor T, Cloix JF. Correlation between brain glycogen and convulsive state in mice submitted to methionine sulfoximine. Life Sci. 2000;67(14):1773–1781. doi: 10.1016/s0024-3205(00)00756-6. [DOI] [PubMed] [Google Scholar]
  • 144.Cloix JF, Hevor T. Epilepsy, regulation of brain energy metabolism and neurotransmission. Curr Med Chem. 2009;16(7):841–853. doi: 10.2174/092986709787549316. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES